ArticlePDF AvailableLiterature Review

Cytolethal Distending Toxin Sequence and Activity in the Enterohepatic Pathogen Helicobacter hepaticus

American Society for Microbiology
Infection and Immunity
Authors:

Abstract and Figures

Little is known about the molecular pathogenesis of hepatitis and enterocolitis caused by enterohepatic Helicobacter species. Sonicates of the murine pathogen Helicobacter hepaticus were found to cause progressive cell distension, accumulation of filamentous actin, and G(2)/M cell cycle arrest in HeLa cell monolayers. The genes encoding this cytotoxic activity were cloned from H. hepaticus. Three open reading frames with closest homology to cdtA, cdtB, and cdtC from Campylobacter jejuni were identified. Sonicates of a laboratory strain of Escherichia coli carrying the cloned cdtABC gene cluster from H. hepaticus reproduced the cytotoxic activities seen with sonicates of H. hepaticus. Cytolethal distending toxin activity is a potential virulence determinant of H. hepaticus that may play a role in the pathogenesis of Helicobacter-associated hepatitis and enterocolitis.
This content is subject to copyright. Terms and conditions apply.
INFECTION AND IMMUNITY,
0019-9567/00/$04.000Jan. 2000, p. 184–191 Vol. 68, No. 1
Copyright © 2000, American Society for Microbiology. All Rights Reserved.
Cytolethal Distending Toxin Sequence and Activity in the
Enterohepatic Pathogen Helicobacter hepaticus
VINCENT B. YOUNG,
1,2
KIMBERLY A. KNOX,
3
AND DAVID B. SCHAUER
1,3
*
Division of Bioengineering and Environmental Health
1
and Division of Comparative Medicine,
3
Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, and Infectious Diseases Unit, Department of Medicine,
Massachusetts General Hospital, Boston, Massachusetts 02114
2
Received 27 July 1999/Returned for modification 10 September 1999/Accepted 14 October 1999
Little is known about the molecular pathogenesis of hepatitis and enterocolitis caused by enterohepatic
Helicobacter species. Sonicates of the murine pathogen Helicobacter hepaticus were found to cause progressive
cell distension, accumulation of filamentous actin, and G
2
/M cell cycle arrest in HeLa cell monolayers. The
genes encoding this cytotoxic activity were cloned from H. hepaticus. Three open reading frames with closest
homology to cdtA,cdtB, and cdtC from Campylobacter jejuni were identified. Sonicates of a laboratory strain of
Escherichia coli carrying the cloned cdtABC gene cluster from H. hepaticus reproduced the cytotoxic activities
seen with sonicates of H. hepaticus. Cytolethal distending toxin activity is a potential virulence determinant of
H. hepaticus that may play a role in the pathogenesis of Helicobacter-associated hepatitis and enterocolitis.
Enterohepatic Helicobacter species (EHS) are emerging as
important pathogens in the genus Helicobacter (15). In contrast
to Helicobacter pylori and other gastric Helicobacter species, the
EHS colonize the lower gastrointestinal tract, including the
ileum, cecum, colon, and biliary tree. However, in a manner
similar to the gastric Helicobacter species, the EHS cause per-
sistent infections associated with chronic inflammation and
epithelial cell hyperproliferation that can lead to neoplastic
disease (15, 19, 36).
H. hepaticus is an EHS that causes chronic active hepatitis
and typhlocolitis in immunocompetent mice (18, 37). In male
mice of susceptible strains, H. hepaticus infection leads to
chronic active hepatitis and liver cancer (16, 18, 20, 36). In
addition, H. hepaticus infection is sufficient to induce inflam-
matory bowel disease in certain immunodeficient mice (9, 23,
34, 35).
Little is known about the molecular pathogenesis of the
EHS. Urease, which has been demonstrated to be a virulence
factor in the gastric Helicobacter species H. pylori and H. mus-
telae, is not present in all of the EHS. H. hepaticus strains do
possess urease activity (16), but the role of urease in coloni-
zation or disease is not clear. Genes homologous to vacA,
encoding the vacuolating cytotoxin (Vac) (13), and cag, the
cytotoxin-associated genes that are part of a pathogenicity is-
land in H. pylori (1, 10), have not been definitively identified in
any EHS.
A cytolethal distending toxin (CDT) has been described in a
number of mucosal pathogens, including Campylobacter jejuni
(21) and other Campylobacter species (27), certain Escherichia
coli strains (7, 22), Shigella dysenteriae (26), Haemophilus du-
creyi (12), and Actinobacillus actinomycetemcomitans (31).
CDT causes progressive cell enlargement and eventual death.
The mechanism of CDT activity is reported to involve G
2
/M
cell cycle arrest in target cells, possibly by preventing activation
of cdc2 (11, 38). Additionally, the appearance of abnormal
accumulations of polymerized actin has been reported in Chi-
nese hamster ovary cells treated with the CDT from E. coli
9142-88 (5).
Campylobacter species are closely related to members of the
genus Helicobacter. Members of both genera are microaerobic,
motile, spiral- to curved-shaped, gram-negative bacteria that
colonize the mucus of the gastrointestinal tract. C. jejuni and
other Campylobacter species are an important cause of acute
gastroenteritis (3). EHS have also been recognized to cause
gastroenteritis, and similarities between these two groups of
organisms have resulted in misidentification of some EHS as
Campylobacter species in clinical and epidemiologic studies (6,
8, 28, 29). It has been suggested that CDT plays a role in the
pathogenesis of C. jejuni-induced gastroenteritis (38). Given
the similarities between campylobacters and helicobacters,
particularly the EHS, we examined H. hepaticus for nucleotide
sequence homology to the cdtABC gene cluster and for CDT
activity.
MATERIALS AND METHODS
Bacteria and cell lines. H. hepaticus ATCC 51449 was obtained from the
American Type Culture Collection (ATCC), Manassas, Va., and was cultured on
tryptic soy agar plates supplemented with 5% sheep blood. A microaerobic
environment was maintained in vented GasPak jars which were evacuated to 20
mm Hg and then equilibrated with a gas mixture consisting of 80% N
2
, 10% H
2
,
and 10% CO
2
. An incubation temperature of 37°C was used for growth. Long-
term storage of bacteria was at 70°C in tryptic soy broth with 30% glycerol.
E. coli XL-1 Blue and SOLR were obtained from Stratagene (La Jolla, Calif.)
and maintained on Luria-Bertani broth agar plates with the appropriate antibi-
otic selection.
HeLa cells (CCL-2) were obtained from the ATCC and cultured in Dulbecco’s
modified Eagle’s medium supplemented with glutamine and 10% fetal calf se-
rum.
PCR and DNA sequence determination. Genomic DNA from plate-grown
bacteria was isolated by using a Qiagen QIAamp kit for small-scale preparations
or a Qiagen genomic G-100 kit for large-scale purification (Qiagen Inc., Santa
Clarita, Calif.). Kits were used in accordance with the recommendations of the
manufacturer.
The degenerate primers VAT2 and WMI1, originally used to identify the cdtB
gene in C. jejuni (27), were synthesized (IDT, Coraville, Iowa) and used for PCR.
PCR was performed by using Pharmacia Ready-To-Go PCR beads (Amersham
Pharmacia Biotech Inc., Piscataway, N.J.). Reactions were set up with 1 l
(approximately 100 ng) of template DNA, 20 pmol of each primer, and enough
water for a total volume of 25 l. This yielded a reaction containing 1.5 U of Taq
polymerase, 10 mM Tris-HCl (pH 9.0 at room temperature), 50 mM KCl, 1.5
mM MgCl
2
, 200 M each nucleotide, and stabilizers, including bovine serum
albumin (BSA). The reaction mixtures were overlaid with 50 l of mineral oil
and subjected to amplification in a DNA thermal cycler (Perkin-Elmer model
* Corresponding author. Mailing address: MIT, Rm. 56-787, Cam-
bridge, MA 02139. Phone: (617) 253-8113. Fax: (617) 258-0225. E-
mail: schauer@mit.edu.
184
FIG. 1. Nucleotide sequence of the cdtABC gene cluster from H. hepaticus. The three presumed ribosomal binding sites (underlined) for each open reading frame
are shown along with the deduced amino acid sequences. Also indicated (bold) are the sites at which the degenerate PCR primers VAT2 and WMI1 bind. Forward
primer VAT2 and the predicted site of reverse primer WMI1 are expected to yield a 505-bp amplicon. The actual binding site of the reverse primer WMI1* along with
forward primer VAT2 produced an observed product of 768 bp.
185
480; PE Biosystems, Foster City, Calif.). The cycling conditions were as follows:
initial denaturation at 94°C for 4 min, followed by 30 cycles of denaturation at
94°C for 1 min, annealing at 42°C for 1.5 min, and extension at 72°C for 1.5 min.
A final extension at 72°C for 4 min was performed. Ten microliters of each
reaction mixture was analyzed by electrophoresis in a 1.0% agarose gel and
visualized after staining with ethidium bromide.
Bands of interest were excised from the gel and purified by using a gel band
purification kit (Amersham Pharmacia Biotech) in accordance with the recom-
mendations of the manufacturer. Purified fragments were ligated into the Pro-
mega pGEM T-easy vector (Promega, Madison, Wis.).
DNA sequencing on an ABI model 377 PRISM automated DNA sequencer
was performed by the core laboratory at the Massachusetts General Hospital
(Boston). DNA sequence analysis was performed on a Macintosh G3 computer
using the MacVector 6.5 software package (Oxford Molecular, Campbell, Calif.).
Preparation of bacterial sonicates. H. hepaticus cultures grown for 48 h on
three 100-mm-diameter plates were harvested into 1 ml of phosphate-buffered
saline (PBS). The bacteria were disrupted by six 30-s pulses on ice with a
VirSonic 50 sonicator (Virtis, Gardiner, N.Y.). Debris was removed by centrif-
ugation at 16,000 gin an Eppendorf model 5415 centrifuge (Eppendorf
Scientific, Westbury, N.Y.), followed by filtration through a 0.2-m-pore-size
filter. Aliquots of the preparations were stored at 70°C.
Cultures of E. coli harboring the cloned cdtABC gene cluster from H. hepaticus
were grown for 18 h in 5 ml of Luria-Bertani broth supplemented with ampicillin
at 100 g/ml. Bacteria were harvested by centrifugation and then suspended in
1 ml of PBS. Preparation of sonicates was then performed as described above.
Tissue culture assay for CDT activity. HeLa cells were seeded onto 13-mm-
diameter circular glass coverslips in 24-well tissue culture plates at a density of
210
3
per well. Twenty microliters of bacterial sonicate was added to each well,
and the plates were incubated in 5% CO
2
at 37°C. At appropriate time points,
coverslips were washed with PBS and then stained with Diff-Quik modified
Wright stain (Baxter Healthcare, Miami, Fla.) and mounted for visualization by
light microscopy.
Immunofluorescence microscopy. Coverslips were washed with PBS and then
fixed with a solution of 3.7% formaldehyde in PBS for 10 min at room temper-
ature. After washing with PBS, cells were permeabilized with a solution of 0.1%
Triton X-100 in PBS for 10 min at room temperature. The coverslips were
washed again with PBS and then stored at 4°C in PBS with 0.5% BSA until they
were stained.
Polymerized actin was stained with phalloidin labeled with Texas red (Molec-
ular Probes, Eugene, Oreg.), and the nuclei were stained with Hoechst 33342 as
described previously (39). Photographs were taken on a Nikon Labophot micro-
scope (Nikon, USA, Melville, N.Y.) with T-Max 100 film (Kodak, Rochester,
N.Y.).
Flow cytometry. Twenty-five-square-centimeter tissue culture flasks were
seeded with 3 10
5
HeLa cells. One hundred microliters of bacterial sonicate
was added to each flask, and then the flasks were incubated in 5% CO
2
at 37°C.
After 24, 48, and 72 h, cells were removed by trypsinization and transferred to a
1.5-ml microcentrifuge tube. Cells were pelleted by centrifugation at 735 gfor
3 min and resuspended in 3% polyethylene glycol 8000–2.5 g of propidium
iodide per ml–9 U of RNase per ml–0.1% Triton X-100–0.001% BSA in 4 mM
sodium citrate. The cells were incubated for 20 min at 37°C and then mixed with
an equal volume of 3% polyethylene glycol 8000–2.5 g of propidium iodide per
ml–0.1% Triton X-100–0.001% BSA in 0.4 M NaCl. Cells were incubated at 4°C
for at least 1 h before performance of DNA content analysis on a FACScan flow
cytometer (Becton Dickinson, Franklin Lakes, N.J.) using the Cell Quant soft-
ware for data acquisition. Data analysis was performed by using the ModFit
program on 10
4
cells for each experiment.
Genomic library construction and screening. Two genomic H. hepaticus li-
braries were constructed by using the insertion vector ZAPII (Stratagene).
Briefly, genomic DNA from H. hepaticus ATCC 51449 was partially digested with
FIG. 2. Comparison of the predicted amino acid sequences of CdtA, CdtB, and CdtC from H. hepaticus ATCC 51449 (Hh) and C. jejuni 81-176 (Cj). Colons indicate
identical amino acids, and conserved amino acids are indicated by periods. Dashes indicate gaps introduced by the MacVector ClustalW alignment program.
186 YOUNG ET AL. INFECT.IMMUN.
the restriction enzyme Tsp509I. DNA with a length of 5 kb was ligated into the
EcoRI site of the vector. The libraries were screened by DNA hybridization using
the PCR amplicon generated by amplification of H. hepaticus genomic DNA with
the primers VAT2 and WMI1 (27). An [-
32
P]dCTP-labeled probe was gener-
ated by using a random primer DNA labeling kit (Ready-To-Go DNA labeling
beads; Amersham Pharmacia Biotech). The radioactive probe was used to screen
10
5
recombinant bacteriophage. Probe-positive plaques were identified and sub-
cloned into the plasmid vector pBluescript SK() using the in vivo excision and
recircularization features of the ZAPII vector. E. coli bacteria carrying these
recombinant plasmids were further characterized by restriction mapping and by
screening for CDT activity.
Nucleotide sequence accession number. The nucleotide sequence of the H.
hepaticus cdtABC gene cluster has been entered in the GenBank database under
accession no. AF163667.
RESULTS
H. hepaticus possesses cdtABC nucleotide sequence homol-
ogy. The degenerate primers VAT2 and WMI1 (27) amplify a
494-bp fragment of the cdtB gene from C. jejuni. Amplification
of H. hepaticus genomic DNA with these primers produced a
larger than expected amplicon of approximately 750 bp (data
not shown). The complete nucleotide sequence of this ampli-
con was determined, and the deduced amino acid sequence
exhibited significant homology to the published CdtB sequence
of C. jejuni. The predicted H. hepaticus peptide fragment ex-
hibited 57% identity and 72% similarity to the C. jejuni gene
product. The larger than expected size of the amplicon was a
consequence of the WMI1 primer annealing to a site 264 bp
downstream of the anticipated target site (Fig. 1).
By using this PCR product, lambda clones containing ho-
mology to cdtB were isolated and corresponding plasmid
clones were generated. DNA sequence analysis of these clones
revealed that H. hepaticus has three closely linked open read-
ing frames corresponding to the entire cdtABC gene cluster
(Fig. 1). The deduced amino acid sequences had the closest
homology to the proteins encoded by the cdtABC gene cluster
from C. jejuni (Fig. 2). The amino acid homologies were as
follows: CdtA, 30% identity and 44% similarity; CdtB, 56%
identity and 72% similarity; CdtC, 40% identity and 55% sim-
ilarity.
Sonicates of H. hepaticus cause morphologic changes in
HeLa cell monolayers. Because the presence of cdtABC nucle-
otide sequence homology was demonstrated in DNA from H.
hepaticus, we sought to determine whether sonicates of this
organism exhibited CDT activity. HeLa cells treated with soni-
cates of H. hepaticus showed marked cellular distension (Fig.
3B). The distended cells also exhibited nuclear enlargement,
and approximately 15% of the affected cells were found to be
multinucleated. Occasionally, nuclear irregularities and frag-
mentation were also seen in HeLa cells treated with sonicates
of H. hepaticus.
The cytoplasmic distension induced by sonicates of H. he-
paticus was readily apparent, but the extent of cellular margins
was underestimated by using light microscopy following the
application of this modified Wright stain. To further examine
the cellular structure of treated cells and to detect associated
cytoskeletal changes, monolayers were stained with fluores-
cently labeled phalloidin to visualize filamentous actin (F-ac-
tin). Phalloidin staining revealed the full extent of the enlarge-
ment of cells in monolayers treated with sonicates of H.
hepaticus (Fig. 4). There also appeared to be an increase in the
amount of F-actin present in cells in affected monolayers. Nu-
clear staining with the fluorescent DNA stain Hoechst 33342
confirmed the nuclear enlargement, multinucleation, and nu-
clear fragmentation visualized by light microscopy.
Sonicates of H. hepaticus cause G
2
/M cell cycle arrest in
cultured cells. To examine whether the morphologic changes
observed in cells treated with sonicates of H. hepaticus were
also associated with cell cycle arrest, the DNA content of
treated cells was determined by flow cytometry. In untreated
cell monolayers, the fraction of cells with a DNA content of 4N
was consistently 8 to 10% (Fig. 5A). In monolayers treated
with sonicates of H. hepaticus, an increase in the fraction of
cells with a DNA content of 4N was seen 24 h after sonicate
addition (Fig. 5D). The fraction of cells with a DNA content of
4N increased at 48 h and reached a maximum by 72 after
sonicate addition (Fig. 5G and J). In addition, by 72 h, there
was a significant fraction of cells with a DNA content of 8N
among cells treated with H. hepaticus sonicates. Examination
of the size of these cells, as judged by fluorescent width, indi-
cates that these are probably multinucleated cells, as opposed
to cellular aggregates. This is consistent with the observation
that multinucleated cells were present in treated monolayers
(Fig. 3).
Cytopathic activity of E. coli carrying the cdtABC gene clus-
ter from H. hepaticus. Sonicates of the 15 E. coli clones har-
boring the recombinant plasmids generated from the genomic
library screen were examined for a cytopathic effect on HeLa
cell monolayers. Sonicates of 3 of the 15 clones produced a
cytopathic effect on cultured HeLa cell monolayers which was
indistinguishable from that produced by sonicates of H. hepati-
cus. Representative clones were selected for further character-
ization. HeLa cell monolayers treated with sonicates of E. coli
carrying the H. hepaticus cdt locus were examined for cytoskel-
etal and nuclear rearrangements over time by fluorescence
FIG. 3. Cytopathic effect of H. hepaticus CDT on HeLa cells. Compared to
untreated cells (A), cells treated with sonicates from H. hepaticus (B) exhibited
marked cytoplasmic distension along with nuclear enlargement, multinucleation,
and nuclear fragmentation. Magnification, 250.
VOL. 68, 2000 HELICOBACTER HEPATICUS CDT 187
microscopy. At 24 h after sonicate addition, nuclear fragmen-
tation was observed (Fig. 5F) but the majority of cells still had
a normal size and actin ultrastructure. By 48 h, more nuclear
abnormalities were observed (Fig. 5I) and cell distension be-
came apparent, along with an increase in the amount of F-actin
(Fig. 5H). At 72 h, the cells in monolayers treated with soni-
cates of an E. coli CDT clone (Fig. 5K and L) were indistin-
guishable from those treated with H. hepaticus sonicates (Fig.
3).Cell cycle analysis demonstrated that the cytopathic effect
produced by the E. coli clones was also accompanied by G
2
/M
cell cycle arrest (Fig. 6). Mapping of the inserts from a number
of clones demonstrated that induction of a cytopathic effect
and cell cycle arrest required the presence of the entire
cdtABC gene cluster (Fig. 6).
DISCUSSION
In this study, we identified and characterized CDT activity in
the EHS H. hepaticus. The term CDT was coined to describe
an activity in culture supernatants of certain E. coli strains, as
well as C. jejuni, that causes progressive distension and even-
tually death of cultured mammalian cells (21, 22). We demon-
strate here that sonicates of H. hepaticus induce G
2
/M cell
cycle arrest in cultured HeLa cells and cause progressive cel-
lular enlargement of HeLa cells, accompanied by the appear-
ance of abnormal accumulations of F-actin. Coupled with the
presence of DNA sequences homologous to the cdtABC gene
cluster from C. jejuni and the cytopathic activity of E. coli
strains carrying the cloned H. hepaticus genes, these results
indicate that H. hepaticus possesses a toxin that is a novel
member of the CDT family.
The predicted CDT gene products from H. hepaticus had the
closest homology to the CDT from C. jejuni. As reported
previously (27, 31), the greatest homology is seen in the CdtB
amino acid sequence. This may indicate a conserved function
for the CdtB subunit; however, our results are in agreement
with others indicating that all three gene products are neces-
sary for cytotoxic activity in laboratory strains of E. coli.
The exact role of CDT in pathogenesis has not been clearly
determined. It has been proposed that CDT plays a role in the
pathogenesis of diarrheal illness. CDT activity in E. coli was
originally described in clinical isolates associated with gastro-
enteritis (22). A study of children with acute diarrhea showed
a trend toward an increased rate of isolation of CDT-produc-
ing E. coli among children with diarrhea compared to controls,
but this did not reach statistical significance (2). It should be
noted that several different virotypes of E. coli are associated
FIG. 4. Cytoplasmic and nuclear distension of HeLa cells treated with sonicates of H. hepaticus. Photomicrographs of untreated cells (A and B) and cells treated
with sonicates of H. hepaticus (C and D) for 72 h are shown. Fixed and permeabilized cells were double labeled for epifluorescence microscopy with Texas red-labeled
phalloidin (A and C) and Hoechst 33342 (B and D). Treated cells exhibited increased cell size and prominent stress fiber-like structures (C) as well, as increased nuclear
size (D). Magnification, 500.
188 YOUNG ET AL. INFECT.IMMUN.
with diarrheal illness, each with distinct virulence determinants
(24). The presence of CDT in a particular E. coli strain may
represent only one of several virulence factors required for
gastrointestinal pathogenesis. Conversely, CDT has been dem-
onstrated in all C. jejuni isolates, as well as in other members
of the genus Campylobacter (14, 27). Demonstration of a role
for CDT in gastrointestinal pathogenesis mediated by C. jejuni
has not been reported. The well-characterized colonization
models for C. jejuni may not be optimal for establishing the
contribution of CDT to disease outcome, since they do not
reproduce the clinical syndrome of gastroenteritis associated
with C. jejuni infection. Partially purified preparations of the
FIG. 5. Time dependence of DNA content and cytopathic effect of HeLa cells treated with bacterial sonicates with CDT activity. Compared to control cells (A),
cells treated with sonicates from H. hepaticus (D, G, and J) showed a progressive increase in the fraction of cells with a DNA content of 4 N, with a maximal effect
being reached 72 h after sonicate addition (J). Compared to control cells (B and C), cells treated with sonicates from an E. coli strain harboring the cloned H. hepaticus
cdt locus showed a progressive increase in size with accumulations of polymerized actin (E, H, and K) and progressive nuclear abnormalities (F, I, and L). These changes
in morphology mirrored the progressive cell cycle block observed by flow cytometry. Magnification, 400. PI, propidium iodide.
VOL. 68, 2000 HELICOBACTER HEPATICUS CDT 189
CDT from S. dysenteriae expressed in a laboratory strain of E.
coli have been shown to induce watery diarrhea in suckling
mice (25). However, the role of CDT in intact S. dysenteriae or
in diarrheagenic E. coli has not been demonstrated. An iso-
genic H. ducreyi cdtC mutant has been shown to maintain
virulence in the temperature-dependent rabbit model of ex-
perimental chancroid (30).
CDT is a candidate virulence factor in the EHS H. hepaticus
that may play a role in the pathogenesis of gastrointestinal
disease caused by these organisms. In mice, infection with H.
hepaticus is associated with a proliferative typhlitis and prolif-
erative hepatitis. In vitro, CDT appears to induce cell cycle
arrest, which suggests that the true targets of CDT activity are
not enterocytes or hepatocytes. It is possible that CDT causes
arrest of a cell type that inhibits epithelial cell proliferation.
Alternatively, CDT in H. hepaticus may have a role in modu-
lation of the immune response that allows persistence of the
organism. However, it should be noted that CDT activity and
nucleotide sequence homology do not appear to be present in
all EHS that cause gastroenteritis. We have identified CDT
activity and a homologue of the cdtB locus in the EHS H.
pullorum but have failed to demonstrate either CDT activity or
DNA homology to cdtB in the EHS H. cinaedi or H. fennelliae
(data not shown). It also remains to be determined if CDT is
a candidate virulence determinant for any of the gastric Heli-
cobacter species, but CDT homology is not present in the
genomic sequence of H. pylori (4, 33).
Another cytotoxic activity called granulating cytotoxin has
been described previously in H. hepaticus (32). This activity is
distinct from the Vac characterized in H. pylori. This cytotoxic
activity can be demonstrated on the CCL9.1 mouse liver cell
line and is characterized by the appearance of cytoplasmic
granules in intoxicated cells. The role of this toxin in patho-
genesis is also unknown. Other cell lines, including HeLa cells,
do not display cytopathic effects when treated with granulating
cytotoxin. The lack of effect on HeLa cells, which were used to
demonstrate CDT activity, indicates that the previously de-
scribed granulating cytotoxin is distinct from the CDT activity
described here.
We are currently characterizing CDT genes and activities in
other Helicobacter species. We have also developed methods
for targeted gene disruption in EHS (unpublished results). By
using these techniques and our well-characterized small-ani-
mal models of EHS infection and disease (9, 17, 37), we expect
to define the role of CDT in the pathogenesis of Helicobacter-
associated experimental inflammatory bowel disease and he-
patic disease.
ACKNOWLEDGMENTS
This work was supported by Public Health Service grants AI01398 to
V.B.Y. and DK52413 to D.B.S. from the National Institutes of Health.
We thank James G. Fox and Stephen B. Calderwood for critical
review of the manuscript and Glenn Paradis for help with flow cytom-
etry.
REFERENCES
1. Akopyants, N. S., S. W. Clifton, D. Kersulyte, J. E. Crabtree, B. E. Youree,
C. A. Reece, N. O. Bukanov, E. S. Drazek, B. A. Roe, and D. E. Berg. 1998.
Analyses of the cag pathogenicity island of Helicobacter pylori. Mol. Micro-
biol. 28:37–53.
2. Albert, M. J., S. M. Faruque, A. S. Faruque, K. A. Bettelheim, P. K. Neogi,
N. A. Bhuiyan, and J. B. Kaper. 1996. Controlled study of cytolethal dis-
tending toxin-producing Escherichia coli infections in Bangladeshi children.
J. Clin. Microbiol. 34:717–719.
3. Allos, B. M., and M. J. Blaser. 1995. Campylobacter jejuni and the expanding
spectrum of related infections. Clin. Infect. Dis. 20:1092–1099.
4. Alm, R. A., L. S. Ling, D. T. Moir, B. L. King, E. D. Brown, P. C. Doig, D. R.
Smith, B. Noonan, B. C. Guild, B. L. deJonge, G. Carmel, P. J. Tummino, A.
Caruso, M. Uria-Nickelsen, D. M. Mills, C. Ives, R. Gibson, D. Merberg,
S. D. Mills, Q. Jiang, D. E. Taylor, G. F. Vovis, and T. J. Trust. 1999.
Genomic-sequence comparison of two unrelated isolates of the human gas-
tric pathogen Helicobacter pylori. Nature 397:176–180.
5. Aragon, V., K. Chao, and L. A. Dreyfus. 1997. Effect of cytolethal distending
toxin on F-actin assembly and cell division in Chinese hamster ovary cells.
Infect. Immun. 65:3774–3780.
6. Atabay, H. I., J. E. Corry, and S. L. On. 1998. Identification of unusual
Campylobacter-like isolates from poultry products as Helicobacter pullorum.
J. Appl. Microbiol. 84:1017–1024.
7. Bouzari, S., and A. Varghese. 1990. Cytolethal distending toxin (CLDT)
production by enteropathogenic Escherichia coli (EPEC). FEMS Microbiol.
Lett. 59:193–198.
8. Burnens, A. P., J. Stanley, R. Morgenstern, and J. Nicolet. 1994. Gastroen-
teritis associated with Helicobacter pullorum. Lancet 344:1569–1570.
9. Cahill, R. J., C. J. Foltz, J. G. Fox, C. A. Dangler, F. Powrie, and D. B.
Schauer. 1997. Inflammatory bowel disease: an immunity-mediated condi-
tion triggered by bacterial infection with Helicobacter hepaticus. Infect. Im-
mun. 65:3126–3131.
10. Censini, S., C. Lange, Z. Xiang, J. E. Crabtree, P. Ghiara, M. Borodovsky,
R. Rappuoli, and A. Covacci. 1996. cag, a pathogenicity island of Helicobacter
pylori, encodes type I-specific and disease-associated virulence factors. Proc.
Natl. Acad. Sci. USA 93:14648–14653.
11. Comayras, C., C. Tasca, S. Y. Peres, B. Ducommun, E. Oswald, and J. De
Rycke. 1997. Escherichia coli cytolethal distending toxin blocks the HeLa cell
cycle at the G
2
/M transition by preventing cdc2 protein kinase dephosphor-
ylation and activation. Infect. Immun. 65:5088–5095.
12. Cope, L. D., S. Lumbley, J. L. Latimer, J. Klesney-Tait, M. K. Stevens, L. S.
Johnson, M. Purven, R. S. Munson, Jr., T. Lagergard, J. D. Radolf, and E. J.
Hansen. 1997. A diffusible cytotoxin of Haemophilus ducreyi. Proc. Natl.
Acad. Sci. USA 94:4056–4061.
FIG. 6. Cytolethal distending toxin activity exhibited by sonicates of E. coli clones carrying plasmids containing various sections of the H. hepaticus cdtABC gene
cluster. The ability of sonicates of each clone to cause a typical CDT cytopathic effect and cell cycle arrest in cultured HeLa cell monolayers is indicated. Cell cycle
arrest is indicated by the percentage of cells in G
2
/M relative to the percentage seen in monolayers treated with H. hepaticus sonicates. Only sonicates of a clone
(pVBY9) that carries a plasmid with the entire cdtABC gene cluster were able to cause a cytopathic effect or cell cycle arrest. ND, not done.
190 YOUNG ET AL. INFECT.IMMUN.
13. Cover, T. L. 1996. The vacuolating cytotoxin of Helicobacter pylori. Mol.
Microbiol. 20:241–246.
14. Eyigor, A., K. A. Dawson, B. E. Langlois, and C. L. Pickett. 1999. Cytolethal
distending toxin genes in Campylobacter jejuni and Campylobacter coli iso-
lates: detection and analysis by PCR. J. Clin. Microbiol. 37:1646–1650.
15. Fox, J. G. 1997. The expanding genus of Helicobacter: pathogenic and zoo-
notic potential. Semin. Gastrointest. Dis. 8:124–141.
16. Fox, J. G., F. E. Dewhirst, J. G. Tully, B. J. Paster, L. Yan, N. S. Taylor, M. J.
Collins, Jr., P. L. Gorelick, and J. M. Ward. 1994. Helicobacter hepaticus sp.
nov., a microaerophilic bacterium isolated from livers and intestinal mucosal
scrapings from mice. J. Clin. Microbiol. 32:1238–1245.
17. Fox, J. G., X. Li, L. Yan, R. J. Cahill, R. Hurley, R. Lewis, and J. C. Murphy.
1996. Chronic proliferative hepatitis in A/JCr mice associated with persistent
Helicobacter hepaticus infection: a model of helicobacter-induced carcino-
genesis. Infect. Immun. 64:1548–1558.
18. Fox, J. G., L. Yan, B. Shames, J. Campbell, J. C. Murphy, and X. Li. 1996.
Persistent hepatitis and enterocolitis in germfree mice infected with Helico-
bacter hepaticus. Infect. Immun. 64:3673–3681.
19. Fox, J. G., L. L. Yan, F. E. Dewhirst, B. J. Paster, B. Shames, J. C. Murphy,
A. Hayward, J. C. Belcher, and E. N. Mendes. 1995. Helicobacter bilis sp.
nov., a novel Helicobacter species isolated from bile, livers, and intestines of
aged, inbred mice. J. Clin. Microbiol. 33:445–454.
20. Hailey, J. R., J. K. Haseman, J. R. Bucher, A. E. Radovsky, D. E. Malarkey,
R. T. Miller, A. Nyska, and R. R. Maronpot. 1998. Impact of Helicobacter
hepaticus infection in B6C3F1 mice from twelve National Toxicology Pro-
gram two-year carcinogenesis studies. Toxicol. Pathol. 26:602–611.
21. Johnson, W. M., and H. Lior. 1988. A new heat-labile cytolethal distending
toxin (CLDT) produced by Campylobacter spp. Microb. Pathog. 4:115–126.
22. Johnson, W. M., and H. Lior. 1988. A new heat-labile cytolethal distending
toxin (CLDT) produced by Escherichia coli isolates from clinical material.
Microb. Pathog. 4:103–113.
23. Kullberg, M. C., J. M. Ward, P. L. Gorelick, P. Caspar, S. Hieny, A. Cheever,
D. Jankovic, and A. Sher. 1998. Helicobacter hepaticus triggers colitis in
specific-pathogen-free interleukin-10 (IL-10)-deficient mice through an IL-
12- and gamma interferon-dependent mechanism. Infect. Immun. 66:5157–
5166.
24. Nataro, J. P., and J. B. Kaper. 1998. Diarrheagenic Escherichia coli. Clin.
Microbiol. Rev. 11:142–201.
25. Okuda, J., M. Fukumoto, Y. Takeda, and M. Nishibuchi. 1997. Examination
of diarrheagenicity of cytolethal distending toxin: suckling mouse response to
the products of the cdtABC genes of Shigella dysenteriae. Infect. Immun.
65:428–433.
26. Okuda, J., H. Kurazono, and Y. Takeda. 1995. Distribution of the cytolethal
distending toxin A gene (cdtA) among species of Shigella and Vibrio, and
cloning and sequencing of the cdt gene from Shigella dysenteriae. Microb.
Pathog. 18:167–172.
27. Pickett, C. L., E. C. Pesci, D. L. Cottle, G. Russell, A. N. Erdem, and H.
Zeytin. 1996. Prevalence of cytolethal distending toxin production in Campy-
lobacter jejuni and relatedness of Campylobacter sp. cdtB gene. Infect. Im-
mun. 64:2070–2078.
28. Stanley, J., D. Linton, A. P. Burnens, F. E. Dewhirst, S. L. On, A. Porter,
R. J. Owen, and M. Costas. 1994. Helicobacter pullorum sp. nov.-genotype
and phenotype of a new species isolated from poultry and from human
patients with gastroenteritis. Microbiology 140:3441–3449.
29. Steinbrueckner, B., G. Haerter, K. Pelz, S. Weiner, J. A. Rump, W. Deissler,
S. Bereswill, and M. Kist. 1997. Isolation of Helicobacter pullorum from
patients with enteritis. Scand. J. Infect. Dis. 29:315–318.
30. Stevens, M. K., J. L. Latimer, S. R. Lumbley, C. K. Ward, L. D. Cope, T.
Lagergard, and E. J. Hansen. 1999. Characterization of a Haemophilus
ducreyi mutant deficient in expression of cytolethal distending toxin. Infect.
Immun. 67:3900–3908.
31. Sugai, M., T. Kawamoto, S. Y. Peres, Y. Ueno, H. Komatsuzawa, T. Fujiwara,
H. Kurihara, H. Suginaka, and E. Oswald. 1998. The cell cycle-specific
growth-inhibitory factor produced by Actinobacillus actinomycetemcomitans
is a cytolethal distending toxin. Infect. Immun. 66:5008–5019.
32. Taylor, N. S., J. G. Fox, and L. Yan. 1995. In-vitro hepatotoxic factor in
Helicobacter hepaticus,H. pylori and other Helicobacter species. J. Med.
Microbiol. 42:48–52.
33. Tomb, J. F., O. White, A. R. Kerlavage, R. A. Clayton, G. G. Sutton, R. D.
Fleischmann, K. A. Ketchum, H. P. Klenk, S. Gill, B. A. Dougherty, K.
Nelson, J. Quackenbush, L. Zhou, E. F. Kirkness, S. Peterson, B. Loftus, D.
Richardson, R. Dodson, H. G. Khalak, A. Glodek, K. McKenney, L. M.
Fitzegerald, N. Lee, M. D. Adams, and J. C. Venter. 1997. The complete
genome sequence of the gastric pathogen Helicobacter pylori. Nature 388:
539–547.
34. von Freeden-Jeffry, U., N. Davidson, R. Wiler, M. Fort, S. Burdach, and R.
Murray. 1998. IL-7 deficiency prevents development of a non-T cell non-B
cell-mediated colitis. J. Immunol. 161:5673–5680.
35. Ward, J. M., M. R. Anver, D. C. Haines, J. M. Melhorn, P. Gorelick, L. Yan,
and J. G. Fox. 1996. Inflammatory large bowel disease in immunodeficient
mice naturally infected with Helicobacter hepaticus. Lab. Anim. Sci. 46:15–20.
36. Ward, J. M., J. G. Fox, M. R. Anver, D. C. Haines, C. V. George, M. J.
Collins, Jr., P. L. Gorelick, K. Nagashima, M. A. Gonda, and R. V. Gilden.
1994. Chronic active hepatitis and associated liver tumors in mice caused by
a persistent bacterial infection with a novel Helicobacter species. J. Natl.
Cancer Inst. 86:1222–1227.
37. Whary, M. T., T. J. Morgan, C. A. Dangler, K. J. Gaudes, N. S. Taylor, and
J. G. Fox. 1998. Chronic active hepatitis induced by Helicobacter hepaticus in
the A/JCr mouse is associated with a Th1 cell-mediated immune response.
Infect. Immun. 66:3142–3148.
38. Whitehouse, C. A., P. B. Balbo, E. C. Pesci, D. L. Cottle, P. M. Mirabito, and
C. L. Pickett. 1998. Campylobacter jejuni cytolethal distending toxin causes a
G
2
-phase cell cycle block. Infect. Immun. 66:1934–1940.
39. Young, V. B., S. Falkow, and G. K. Schoolnik. 1992. The invasin protein of
Yersinia enterocolitica: internalization of invasin-bearing bacteria by eukary-
otic cells is associated with reorganization of the cytoskeleton. J. Cell Biol.
116:197–207.
Editor: P. E. Orndorff
VOL. 68, 2000 HELICOBACTER HEPATICUS CDT 191
... CDT is a toxin that is sensitive to trypsin and is not thermostable (inactivated by heating at 70°C for 15 minutes). The genetic analysis demonstrated that all three CDT genes are required to be expressed in the producing bacterium for the generation of active (cytotoxic) CDT (Pickett et al., 1994) (Pickett et al., 1996) (Cope et al., 1997) (Sugai et al., 1998 (Young et al., 2000). ...
... The induction of the cytopathogenic effects of the CdtB was verified by immunofluorescence and cell cycle analyses. In both cell lines, the H. hepaticus CdtB induced the previously reported effects of the CDT in all of the cells (Figures 1A,B), i.e., a cellular and nuclear enlargement, with the formation of cortical actin-rich large lamellipodia and actin stress fibers (Young et al., 2000), as well as the nuclear translocation of NF-κB . A high increase in the percentage of cells in G2/M phase was observed in response to the CdtB in both cell lines (Figures 1C,D), reflecting the effectiveness of the puromycin selection. ...
... In the present study, the direct effects of the active CdtB subunit of the CDT were evaluated using colon and liver epithelial cell lines allowing the inducible expression of the H. hepaticus CdtB. In vitro, this lentiviral model reproduced the well-known effects of the H. hepaticus CdtB, i.e., a major remodeling of the cytoskeleton as well as the nuclear translocation of NF-κB (Young et al., 2000;. The transduced cells were then engrafted into immunodeficient mice and the CdtB subunit was conditionally expressed. ...
Thesis
L’homme est fréquemment exposé à des génotoxines bactériennes impliquées dans les cancers digestifs, comme la colibactine et la toxine CDT (Cytolethal Distending Toxin). Ces toxines endommagent l'ADN des cellules hôtes et constituent un facteur prédisposant au développement de cancers. Les Helicobacter entérohépatiques, tels que Helicobacter hepaticus et Helicobacter pullorum, sont associés à plusieurs maladies intestinales et hépatiques. Leur principal facteur de virulence est la toxine CDT. Nous avons étudié les mécanismes d'action de la génotoxine CDT dans l'activation des processus procancéreux et la survie cellulaire, en utilisant l'infection à Helicobacter hepaticus comme modèle. Nous avons démontré que le remodelage nucléaire induit par la sous-unité active CdtB de la CDT de Helicobacter implique la surexpression de l'oncoprotéine MAFB qui est ensuite associée à une forte localisation nucléaire et périnucléaire dans les noyaux distendus en réponse à la CDT/CdtB, ainsi qu’à la formation de gros lamellipodes, des extensions cellulaires connues pour être impliquées dans la migration cellulaire et la production de métastases. Nous avons également montré que le remodelage nucléaire induit par CDT/CdtB peut être associé à la formation de réticulum nucléoplasmique (NR) enrichi en particules ribonucléoprotéiques. Ces structures dynamiques et transitoires induites par la génotoxine pourraient correspondre à une passerelle privilégiée pour la synthèse d'ARNm sélectionnés, qui seraient préférentiellement transportés du noyau à travers les pores nucléaires et traduits à l'intérieur du NR. Le NR induit par la CDT (et aussi par la colibactine) pourrait permettre à la cellule de s'arrêter et de réparer les dommages à l'ADN causés par les génotoxines bactériennes afin de maintenir la survie cellulaire. L'identification à base de MicroArrays des gènes régulés par la CdtB a aussi montré une régulation de l'autophagie par cette toxine. Nous avons montré que la CdtB stimulait le flux autophagique et induisait une autophagie de survie suite aux dommages à l'ADN induits par la CDT/CdtB. Nous avons montré que la CDT/CdtB favorise la formation d'agrégats cytoplasmiques SQSTM1/P62 parfois profondément invaginés dans les noyaux distendus ainsi que des vésicules extra-cellulaires SQSTM1/P62-positives. La formation de NR suite aux dommages à l'ADN induits par la CDT est associée à l'induction de l'autophagie, qui joue un rôle de survie dans ce contexte. Les particules SQSTM1/P62 ont un rôle clé dans ces effets. Ces résultats offrent de nouvelles perspectives dans le contexte de la formation du NR et de la survie cellulaire en réponse aux dommages à l'ADN, une caractéristique commune à de nombreux cancers, qui apparaissent non seulement en réponse aux dommages à l'ADN induits par les thérapies mais aussi plus tôt en réponse aux bactéries génotoxiques. De nouvelles études sont nécessaires pour identifier les structures cytosoliques entourées et englouties par P62/SQSTM1 ainsi que pour déchiffrer le rôle de cette protéine cargo dans la survie des cellules dont l'ADN est endommagé et, plus particulièrement, son rôle dans la formation de NR à la lumière de sa fonction de protéine de liaison à l’ARN récemment démontrée. Les résultats issus de cette thèse confortent l’idée que les génotoxines bactériennes pourraient être à l’origine de cancers.
... -Helicobacter hepaticus (HhepCDT) (Young et al. 2000), une bactérie de l'intestin et du foie retrouvée principalement chez la souris et causant elle aussi des entérocolites. La prévalence des gènes de CDT varie en fonction de l'espèce et de la souche de bactérie, mais également de la pathologie associée. ...
... Ces effets cellulaires ont donné son nom à la Cytolethal Distending Toxin. Des changements morphologiques analogues ont été observés pour diverses lignées cellulaires adhérentes et avec des CDT de différentes origines (Cope et al. 1997 ;Cortes-Bratti et al. 1999 ;Okuda et al. 1995 ;Young et al. 2000). Les cellules distendues peuvent être entre 4 et 7 fois plus grosses que des cellules non-traitées, avec un noyau distendu jusqu'à 2,5 fois (Aragon et al. 1997 ;De Rycke et al. 2000 ;Peres et al. 1997 ;Whitehouse et al. 1998). ...
Thesis
La Cytolethal Distending Toxin (CDT) est un facteur de virulence produit par de nombreuses bactéries pathogènes Gram-négatives. La CDT est impliquée dans la pathogénicité des bactéries productrices et promeut la persistance à long terme de l’infection et de l’inflammation. De plus, la CDT est capable d’endommager l’ADN des cellules infectées, ce qui la caractérise comme étant une génotoxine. La CDT induit donc de l’instabilité génétique qui, en synergie avec la promotion de l’inflammation, est probablement à l’origine de l’association constatée entre CDT et cancérogenèse. L’action de la toxine repose sur la sous-unité catalytique CdtB, mais des doutes persistent sur son mode d’action exact. En effet, sur la base d’alignements de séquence et structure avec la DNase I, il a été suggéré que la sous-unité CdtB est une nucléase et que son activité est évaluable par un test in vitro de dégradation d’un plasmide bactérien. Ce test est largement utilisé dans la littérature, mais des incohérences dans l’activité nucléase mesurée ont été rapportées. D’autre part, des alignements de séquence avec certaines phosphatases ciblant des phosphoinositides ont permis d’envisager une activité similaire pour la CdtB. Cependant, l’importance respective de chacune de ces deux activités biochimiques dans la toxicité de CDT n’est pas bien caractérisée et, de manière plus générale, leur impact sur les pathologies associées à CDT est également peu compris à ce jour. Afin de mieux caractériser les activités biochimiques de CdtB, nous avons généré et utilisé diverses toxines mutantes. Dans une première étude, une comparaison des effets cellulaires et du test de digestion de plasmide in vitro a permis de déterminer que ce dernier est inadapté à l’étude de l’activité biochimique de CdtB. Suite à ce travail, nous suggérons l’utilisation de tests cellulaires, reposant particulier sur l’évaluation de la réponse aux dommages à l’ADN induite par CDT, pour définir l’activité de la toxine. Dans un second temps, ces tests cellulaires ont été utilisés pour évaluer l’impact de différentes mutations sur l’activité de CdtB. Une partie de ces mutations a eu pour but de découpler les deux activités, nucléases et phosphatases, en ciblant notamment des résidus potentiellement impliqués dans la liaison aux substrats. Grâce à ces mutants, nous avons déterminé que la toxicité est principalement liée à l’activité nucléase tandis que l’activité phosphatase est probablement impliquée dans le transport intracellulaire de la toxine. Les autres mutants générés ciblaient une région strictement conservée avec une endonucléase humaine présentant des caractéristiques similaires à CdtB et les résultats préliminaires sont présentés. Enfin, une dernière partie de ma thèse portait sur l’étude de l’impact de CDT sur le déroulement du cycle cellulaire et sur l’instabilité génomique. Ce projet, basé sur l’utilisation d’une technique d’imagerie en temps réel de cellules vivantes, a mis en évidence l’induction par CDT de défauts en mitose et plus particulièrement un allongement de la transition métaphase / anaphase. En conclusion, ce travail de thèse a permis d’une part de mieux caractériser les liens entre activités biochimiques de CdtB et effets toxiques de CDT et d’autre part d’étudier son implication dans l’induction d’instabilité génomique. De manière générale, cette caractérisation du mode d’action de CDT représente une première étape vers une meilleure compréhension de la pathogénèse qu’elle induit.
... Senescent cells might finally die or escape senescence [207], leading to de-polyploidisation and cell survival [182] (dotted arrow). ⑨ ⑨ CDT-induced cell cycle arrest is associated with a profound nuclear and actin cytoskeleton remodeling with stress fiber formation [184,208,209]. Nuclear NET1 is dephosphorylated leading to RhOA activation which subsequently phosphorylates p38 Mitogen-Activated Protein Kinase (MAPK) and its downstream effector protein MAPK-activated protein kinase 2 which promotes the formation of actin stress fibers and delays cell death [210,211]. ...
Article
The Helicobacter genus actually comprises 46 validly published species divided into two main clades: gastric and enterohepatic Helicobacters. These bacteria colonize alternative sites of the digestive system in animals and humans, and contribute to inflammation and cancers. In humans, Helicobacter infection is mainly related to H. pylori, a gastric pathogen infecting more than half of the world's population, leading to chronic inflammation of the gastric mucosa that can evolve into two types of gastric cancers: gastric adenocarcinomas and gastric MALT lymphoma. In addition, H. pylori but also non-H. pylori Helicobacter infection has been associated to many extra-gastric malignancies. This review focuses on H. pylori and its role in gastric cancers and extra-gastric diseases as well as malignancies induced by non-H. pylori Helicobacters. Their different virulence factors and their involvement in carcinogenesis is discussed. This review highlights the importance of both gastric and enterohepatic Helicobacters in gastrointestinal and liver cancers.
... Javiana data set) bootstrap repetitions. iTOL v4 was used for editing of phylogenetic trees (67,68). ...
Article
Full-text available
While previous reports had suggested that the typhoid toxin (TT) could potentially use ArtB as an alternate binding subunit, this was thought to play a minor role in the evolution and biology of the toxin. In this study, we establish that both TT genes and artBSalmonella entericaentericaSalmonella
Thesis
The mammalian gastrointestinal tract is a complex ecosystem shaped by interactions between the host, indigenous gut microbiota, and external world. When colonizing the gut, bacteria must overcome barriers imposed by the intestinal environment, such as host immune responses and microbiota-mediated nutrient limitation. Thus, understanding bacterial colonization requires determining how the gut landscape interacts with microbes attempting to associate with the established community. However, the myriad interactions between elements of the gastrointestinal ecosystem make it difficult to detect emergent properties of the system when studied solely via reductive methods. Therefore, a systems biology approach, which unites biological and computational modeling to investigate complex systems by analyzing the behavior and relationships of all elements of the system, may be the best way to obtain an integrative perspective of the colonization process. This dissertation encompasses multiple projects that collectively follow a systems biology framework to interrogate how the intestinal environment regulates bacterial colonization of the gut. I first explore the challenges faced by a bacterium as it initially colonizes the gut using human intestinal organoids (HIOs) and germ-free mice. I show that HIOs pose a more restrictive environment to bacteria than the murine gut. These results demonstrate that different experimental systems model unique aspects of host-microbe interactions in the gut, thus underscoring the need to benchmark such systems to inform their incorporation into integrative experimental frameworks. I conclude that the murine gut is best suited for increasingly holistic investigations of the gut ecosystem moving forward. To that end, I next explore bacterial colonization in a complex intestinal landscape encompassing both host and microbiota elements. I focus on colonization by the enteric pathogen Clostridioides difficile in the setting of inflammatory bowel disease (IBD). While susceptibility to C. difficile infection (CDI) typically follows the administration of antibiotics, patients with IBD exhibit increased incidence of CDI, even in the absence of antibiotic treatment. However, the mechanisms underlying this susceptibility are unclear. To explore these mechanisms, I leverage murine and computational models to demonstrate that IBD-associated intestinal inflammation alters microbiota composition to permit C. difficile colonization. These results advance our understanding of the host-microbiota interface as it relates to C. difficile colonization in the setting of inflammation. More broadly, this dissertation highlights the value of a systems biology paradigm for gaining insights into ecosystem elements regulating the colonization process. Ultimately, such insights could inform the development of strategies to promote or prevent intestinal colonization by symbiotic and pathogenic bacteria, respectively.
Article
The gut microbiota has recently emerged as a unique mechanism of immunotherapeutic resistance or response within certain cancer patients. Certain adherent bacterial species that reside along the epithelial barrier within the gastrointestinal tract have been shown to be the most immunogenic and include several species within the Helicobacteraceae family. The role of these microbes in cancer remains controversial and varies according to species, immune status, and cancer type. Here, we hypothesize that the functional characteristics rather than the bacterial species of Helicobacteraceae dictate the type of immune response with either a benefit or a detriment to overall cancer progression.
Article
Hepatocytes injury caused by cytolethal distending toxin (CDT) are major events during helicobacter hepaticus (H.hepaticus) infection. Recent study showed that pre-survival autophagy was promoted against CdtB subunit induced DNA damage. In the present study, we demonstrated that inflammatory cytokines IL-6, IL-1β, TNF-α, IFN-α, IFN-γ expression and STAT phosphorylation were promoted by CdtB. Besides, CdtB decreased cell viability while promote apoptosis in mouse liver (AML12) cells. Especially, apoptotic protein caspase-9, caspase-3 and PARP were activated while the ratio of Bcl-2/Bax was decreased after CdtB treatment. Moreover, apoptosis induced by CdtB was inhibited due to Erk/p38 MAPK signaling pathway suppression performed with SB203580 or U0126. Meanwhile, we found that CdtB increased autophagic marker levels accompanied by Akt/mTOR/P70S6K signaling pathway in a dose dependent manner. To assess the correlation between autophagy and apoptosis induced by H.hepaticus, chloroquine (CQ, 50 μM) was employed to inhibit autophagy. The result showed that inhibition of autophagy with CQ treatment promoted apoptosis induced by CdtB. Altogether, all these results suggest that CdtB triggers apoptosis via MAPK/Erk/p38 signaling pathway in caspase dependent manner, which was prevented by autophagy in AML12 cells. Collectively, our findings provide new insights into the virulence potential of CdtB on the molecular pathogenesis throughout H.hepaticus infection.
Article
Full-text available
Cytolethal Distending Toxin (CDT) belongs to the AB toxin family and is produced by a plethora of Gram-negative bacteria. Eight human-affecting enteropathogens harbor CDT that causes irritable bowel syndrome (IBS), dysentery, chancroid, and periodontitis worldwide. They have a novel molecular mode of action as they interfere in the eukaryotic cell-cycle progression leading to G2/M arrest and apoptosis. CDT, the first bacterial genotoxin described, is encoded in a single operon possessing three proteins, CdtA, CdtB, and CdtC. CdtA and CdtC are needed for the binding of the CDT toxin complex to the cholesterol-rich lipid domains of the host cell while the CdtB is the active moiety. Sequence and 3D structural-based analysis of CdtB showed similarities with nucleases and phosphatases, it was hypothesized that CdtB exercises a biochemical function identical to both these enzymes. CDT is secreted through the outer membrane vesicles from the producing bacteria. It is internalized in the target cells via clathrin-dependent endocytosis and translocated to the host cell nucleus through the Golgi complex and ER. This study discusses the virulence role of CDT, causing pathogenicity by acting as a tri-perditious complex in the CDT-producing species with an emphasis on its potential role as a biomarker and an anti-tumor agent. Graphic abstract
Article
The genus Helicobacter defined just over 30 years ago, is a highly diverse and fast-growing group of bacteria that are able to persistently colonize a wide range of animals. The members of this genus are subdivided into two groups with different ecological niches, associated pathologies, and phylogenetic relationships: the gastric Helicobacter (GH) and the enterohepatic Helicobacter (EHH) species. Although GH have been mostly studied, EHH species have become increasingly important as emerging human pathogens and potential zoonotic agents in the last years. This group of bacteria has been associated with the development of several diseases in humans from acute pathologies like gastroenteritis to chronic pathologies that include inflammatory bowel disease, and liver and gallbladder diseases. However, their reservoirs, as well as their routes of transmission, have not been well established yet. Therefore, this review summarizes the current knowledge of taxonomy, epidemiology, and clinical role of the EHH group.
Article
Full-text available
The incidence of C. difficile infection (CDI) has increased significantly among patients with IBD, independently of antibiotic use, yet the relationship between IBD and increased risk for CDI remains to be understood. Our study sought to describe and utilize an antibiotic-independent mouse model to specifically explore the relationship between the IBD-associated gut and susceptibility to C. difficile colonization and CDI development.
Article
Full-text available
Little is known about the virulence mechanisms employed by Haemophilus ducreyi in the production of genital ulcers. This Gram-negative bacterium previously has been shown to produce a soluble cytotoxic activity that kills HeLa and HEp-2 cells. We have now identified a cluster of three H. ducreyi genes that encode this cytotoxic activity. The predicted proteins encoded by these genes are most similar to the products of the Escherichia coli cdtABC genes that comprise the cytolethal distending toxin (CDT) of this enteric pathogen. Eleven of 12 H. ducreyi strains were shown to possess this gene cluster and culture supernatants from these strains readily killed HeLa cells. The culture supernatant from a single strain of H. ducreyi that lacked these genes was unable to kill HeLa cells. When the H. ducreyi cdtABC gene cluster was cloned into E. coli, culture supernatant from the recombinant E. coli clone killed HeLa cells. A monoclonal antibody that neutralized this soluble cytotoxic activity of H. ducreyi was shown to bind to the H. ducreyi cdtC gene product. This soluble H. ducreyi cytotoxin may play a role in the development or persistence of the ulcerative lesions characteristic of chancroid.
Article
Full-text available
Yersinia enterocolitica, a facultative intracellular pathogen of mammals, readily enters (i.e., invades) cultured eukaryotic cells, a process that can be conferred by the cloned inv locus of the species. We have studied the mechanism by which the product of inv, a microbial outer membrane protein termed "invasin," mediates the internalization of bacteria by HEp-2 cells and chicken embryo fibroblasts. Invasin-bearing bacteria initially bound the filopodia and the leading edges of cultured cells. Multiple points of contact between the bacterial surface and the surface of the cell ensued and led to the internalization of the bacterium within an endocytic vacuole; the same multi-step process could be induced by an inert particle coated with invasin-containing membranes. Both adherence and internalization were blocked by an antisera directed against the beta 1 integrin cell-adherence molecule. Ultrastructural studies of detergent-insoluble cytoskeletons from infected cells and immunofluorescence microscopy of phalloidin-labeled cells showed alterations in the structure of the cytoskeleton during the internalization process including the accumulation of polymerized actin around entering bacteria. Bacterial entry was prevented by cytochalasin D indicating that the internalization process requires actin microfilament function. Possible linkages between beta 1 containing integrins and the cytoskeleton were examined during the internalization process through the use of protein-specific antibodies and immunofluorescence microscopy. Like actin, the actin-associated proteins filamin, talin and the beta 1 integrin subunit were also found to accumulate around entering bacteria. These findings suggest that the invasin-mediated internalization process is associated with cytoskeletal reorganization.
Article
Full-text available
cagA, a gene that codes for an immunodominant antigen, is present only in Helicobacter pylori strains that are associated with severe forms of gastroduodenal disease (type I strains). We found that the genetic locus that contains cagA (cag) is part of a 40-kb DNA insertion that likely was acquired horizontally and integrated into the chromosomal glutamate racemase gene. This pathogenicity island is flanked by direct repeats of 31 bp. In some strains, cag is split into a right segment (cagI) and a left segment (cagII) by a novel insertion sequence (IS605). In a minority of H. pylori strains, cagI and cagII are separated by an intervening chromosomal sequence. Nucleotide sequencing of the 23,508 base pairs that form the cagI region and the extreme 3' end of the cagII region reveals the presence of 19 ORFs that code for proteins predicted to be mostly membrane associated with one gene (cagE), which is similar to the toxin-secretion gene of Bordetella pertussis, ptlC, and the transport systems required for plasmid transfer, including the virB4 gene of Agrobacterium tumefaciens. Transposon inactivation of several of the cagI genes abolishes induction of IL-8 expression in gastric epithelial cell lines. Thus, we believe the cag region may encode a novel H. pylori secretion system for the export of virulence determinants.
Article
Escherichia coli is the predominant nonpathogenic facultative flora of the human intestine. Some E. coli strains, however, have developed the ability to cause disease of the gastrointestinal, urinary, or central nervous system in even the most robust human hosts. Diarrheagenic strains of E. coli can be divided into at least six different categories with corresponding distinct pathogenic schemes. Taken together, these organisms probably represent the most common cause of pediatric diarrhea worldwide. Several distinct clinical syndromes accompany infection with diarrheagenic E. coli categories, including traveler’s diarrhea (enterotoxigenic E. coli), hemorrhagic colitis and hemolytic-uremic syndrome (enterohemorrhagic E. coli), persistent diarrhea (enteroaggregative E. coli), and watery diarrhea of infants (enteropathogenic E. coli). This review discusses the current level of understanding of the pathogenesis of the diarrheagenic E. coli strains and describes how their pathogenic schemes underlie the clinical manifestations, diagnostic approach, and epidemiologic investigation of these important pathogens.
Article
Escherichia coli is the predominant nonpathogenic facultative flora of the human intestine. Some E. coli strains, however, have developed the ability to cause disease of the gastrointestinal, urinary, or central nervous system in even the most robust human hosts. Diarrheagenic strains of E. coli can be divided into at least six different categories with corresponding distinct pathogenic schemes. Taken together, these organisms probably represent the most common cause of pediatric diarrhea worldwide. Several distinct clinical syndromes accompany infection with diarrheagenic E. coli categories, including traveler's diarrhea (enterotoxigenic E. coli), hemorrhagic colitis and hemolytic-uremic syndrome (enterohemorrhagic E. coli), persistent diarrhea (enteroaggregative E. coli), and watery diarrhea of infants (entero-pathogenic E. coli). This review discusses the current level of understanding of the pathogenesis of the diarrheagenic E. coli strains and describes how their pathogenic schemes underlie the clinical manifestations, diagnostic approach, and epidemiologic investigation of these important pathogens.
Article
Helicobacter pylori, the causative agent of chronic superficial gastritis and duodenal ulcer disease in humans, produces a unique cytotoxin (VacA) that induces cytoplasmic vacuolation in eukaryotic cells. The structural organization and processing of the vacuolating cytotoxin are characteristic of a family of proteins exemplified by Neisseria gonorrhoeae IgA protease. Although only 50% of H. pylori isolates produce detectable cytotoxin activity in vitro, vacA homologues are present in virtually all isolates. Several families of vacA alleles have been identified, and there is a strong correlation between presence of specific vacA genotypes, cytotoxin activity, and peptic ulceration. Experiments in a mouse model of H. pylori-induced gastric damage indicate that the cytotoxin plays an important role in inducing gastric epithelial necrosis.
Article
Helicobacter pylori, the causative agent of chronic superficial gastritis and duodenal ulcer disease in humans, produces a unique cytotoxin (VacA) that induces cytoplasmic vacuolation in eukaryotic cells. The structural organization and processing of the vacuolating cytotoxin are characteristic of a family of proteins exemplified by Neisseria gonorrhoeae IgA protease. Although only 50% of H. pylori isolates produce detectable cytotoxin activity in vitro, vacA homologues are present in virtually all isolates. Several families of vacA alleles have been identified, and there is a strong correlation between presence of specific vacA genotypes, cytotoxin activity, and peptic ulceration. Experiments in a mouse model of H. pylori-induced gastric damage indicate that the cytotoxin plays an important role in inducing gastric epithelial necrosis.
cag, a pathogenicity island of Helicobacter pylori, encodes type I-specific and disease-associated virulence factors
  • S Censini
  • C Lange
  • Z Xiang
  • J E Crabtree
  • P Ghiara
  • M Borodovsky
  • R Rappuoli
  • Censini S.
Inflammatory large bowel disease in immunodeficient mice naturally infected with Helicobacter hepaticus
  • J M Ward
  • M R Anver
  • D C Haines
  • J M Melhorn
  • P Gorelick
  • Yan L Fox
  • Ward J. M.