Sie sind auf Seite 1von 9

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 3, Issue of January 16, pp.

2324 –2331, 2004


Printed in U.S.A.

Interactions between p-33 and p-55 Domains of the Helicobacter


pylori Vacuolating Cytotoxin (VacA)*
Received for publication, September 12, 2003
Published, JBC Papers in Press, October 30, 2003, DOI 10.1074/jbc.M310159200

Victor J. Torres‡§, Mark S. McClain¶, and Timothy L. Cover‡¶储**


From the ‡Department of Microbiology and Immunology and the ¶Department of Medicine, Vanderbilt University
School of Medicine, Nashville, Tennessee 37232-2605 and the 储Department of Veterans Affairs Medical Center,
Nashville, Tennessee 37212

The VacA toxin secreted by Helicobacter pylori is con- Most virulent H. pylori strains secrete a cytotoxin known as
sidered to be an important virulence factor in the patho- VacA into the extracellular space (5– 8). Several lines of evi-
genesis of peptic ulcer disease and gastric cancer. VacA dence indicate that VacA contributes to the capacity of H. py-
monomers self-assemble into water-soluble oligomeric lori to colonize the stomach and that this toxin is an important
structures and can form anion-selective membrane virulence factor in the pathogenesis of peptic ulceration and
channels. The goal of this study was to characterize gastric cancer (6 –12). The most extensively characterized ac-
VacA-VacA interactions that may mediate assembly of tivity of VacA is its capacity to induce vacuolation in mamma-
VacA monomers into higher order structures. We inves- lian cells (5, 13). The membranes of these VacA-induced vacu-

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


tigated potential interactions between two domains of oles contain markers for late endosomal and lysosomal
VacA (termed p-33 and p-55) by using a yeast two-hybrid
compartments (14). The precise mechanism of VacA-induced
system. p-33/p-55 interactions were detected in this sys-
vacuole formation is not completely understood but is thought
tem, whereas p-33/p-33 and p-55/p-55 interactions were
to involve binding of the toxin to the plasma membrane, inter-
not detected. Several p-33 proteins containing internal
deletion mutations were unable to interact with wild- nalization of the toxin, and action of the toxin in an intracel-
type p-55 in the yeast two-hybrid system. Introduction of lular site (6 – 8). One model proposes that VacA forms anion-
these same deletion mutations into the H. pylori vacA selective channels in the membrane of late endocytic
gene resulted in secretion of mutant VacA proteins that compartments (6 – 8, 15, 16). In addition to inducing the forma-
failed to assemble into large oligomeric structures and tion of intracellular vacuoles, VacA causes multiple other ef-
that lacked vacuolating toxic activity for HeLa cells. Ad- fects on target cells, including depolarization of the membrane
ditional mapping studies in the yeast two-hybrid system potential (16, 17), permeabilization of epithelial monolayers
indicated that only the N-terminal portion of the p-55 (18), apoptosis (19 –22), detachment of epithelial cells from the
domain is required for p-33/p-55 interactions. To charac- basement membrane (12), interference with the process of an-
terize further p-33/p-55 interactions, we engineered an tigen presentation (23), and inhibition of T lymphocyte activa-
H. pylori strain that produced a VacA toxin containing an tion (24).
enterokinase cleavage site located between the p-33 and The vacA gene is translated as a 139 –140-kDa protoxin and
p-55 domains. Enterokinase treatment resulted in com- shows no striking similarity to any other known bacterial toxin
plete proteolysis of VacA into p-33 and p-55 domains, (5, 11, 25, 26). Upon expression, this protoxin undergoes N- and
which remained physically associated within oligomeric C-terminal processing to yield a mature 88-kDa secreted VacA
structures and retained vacuolating cytotoxin activity. toxin (27). The mature secreted 88-kDa VacA toxin can undergo
These results provide evidence that interactions be-
spontaneous proteolytic degradation into fragments that are
tween p-33 and p-55 domains play an important role in
about 33 and 55 kDa in mass (11, 27–29). This degradation has
VacA assembly into oligomeric structures.
been observed especially when preparations of the purified
toxin are stored for prolonged periods (11, 28). The site of
proteolytic cleavage is predicted to be located within a hydro-
Helicobacter pylori is a Gram-negative, spiral-shaped mi-
philic surface-exposed loop (11, 27). It has been presumed that
croaerophilic bacterium that colonizes the gastric mucosa of
the two fragments (termed p-33 and p-55) represent two do-
more than 50% of the human population (1). Colonization of the
mains or subunits of VacA (11, 30, 31). It should be noted that
gastric mucosa by H. pylori results in mucosal inflammation
the nomenclature for describing these fragments has not been
and is recognized as a major risk factor for the development of
uniform; several previous studies have described 37- and 58-
peptic ulcer disease, distal gastric adenocarcinoma, and gastric
kDa fragments (11, 30, 32–36). In the current study, we desig-
lymphoma (2– 4).
nate the two fragments as p-33 and p-55, based on the results
of mass spectrometry analysis (27).
* This work was supported in part by National Institutes of Health Previous studies have identified specific functions that are
Grants AI39657 and DK53623 and by the Medical Research Depart- attributable to either the p-33 or the p-55 domain. Several lines
ment of the Department of Veterans Affairs (to T. L. C.). The costs of of evidence indicate that amino acid sequences within the p-55
publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked “advertisement” domain (located at the C terminus of the mature secreted toxin)
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. mediate binding of VacA to host cells (28, 31, 37, 38). VacA
§ Supported by a structural biology predoctoral fellowship from the binding to cells is inhibited by antiserum reactive with the p-55
Vanderbilt University Center for Structural Biology. fragment but not inhibited by antiserum to the p-33 fragment
** To whom correspondence should be addressed: Division of Infec-
tious Diseases, A3310 MCN, Vanderbilt University School of Medicine,
(28). Also, a truncated form of VacA containing mainly the p-55
Nashville, TN 37232. Tel.: 615-322-2035; Fax: 615-343-6160; E-mail: VacA fragment was reported to bind to target cells in a manner
timothy.L.cover@vanderbilt.edu. similar to the full-length VacA (31). Amino acid sequences

2324 This paper is available on line at http://www.jbc.org


H. pylori VacA 2325
located within a hydrophobic region near the N terminus TABLE I
(within the p-33 domain) are required for membrane channel Oligonucleotides used in this study
formation by VacA (39, 40). When expressed in transiently Primer Nucleotide sequence
name
transfected cells, neither the p-33 nor the p-55 domain alone is
sufficient for vacuolating toxin activity (32, 34, 41). In tran- AND510a 5⬘-TATAGCCCGGGCCGCCTTTTTTACAACCGTGAT
siently transfected cells, expression of the entire p-33 domain AND511 5⬘-CCCCTCGACTTATTTAGCACCACTTTGAGAAG
AND512 5⬘-CCCCGGATCCGCCTTTTTTACAACCGTGAT
as well as about 111 amino acids from the N terminus of the AND515a 5⬘-CCCCGTCGACTTAAGCGTAGCTAGCGAAACGCG
p-55 domain is required for vacuolating toxin activity (32). AND516 5⬘-CCCCGGATCCAACGACAAACAAGAGAGCAG
Secreted VacA proteins assemble into large water-soluble AND6098 5⬘-CCCCGAATTCAACGACAAACAAGAGAGCAG
and flower-shaped structures composed predominantly of OP4175 5⬘-CCCCGGATCCGCCTTTTTTACAACCCTTGG
OP4176 5⬘-CCCCGTCGACTTACGTATCAATACCTTTAAAAT
12–14 VacA monomers (29, 30, 42). Assembly of VacA into
BAR1556 5⬘-CCCCGAATTCGGTAATGGTGGTTTCAACAC
oligomeric structures is presumably required for membrane BAR1557 5⬘-CCCCGAATTCACTAGGTCAATCTTTTCTGG
channel formation. This hypothesis is supported by electron BAR1558 5⬘-CCCCGTCGACTTAGCCAGTTTCCAAACGCACG
microscopy studies in which VacA associated with membranes BAR1559 5⬘-CCCCGTCGACTTAGATTTTCGCTTTCAATAAAACA
appears as oligomeric structures (15, 42), as well as electro- OP2891 5⬘-CCGACTACAAGGATGACGACGACAAAG
OP2892 5⬘-CTTTGTCGTCGTCATCCTTGTAGTCGG
physiologic studies in which the kinetics of membrane channel
formation by VacA have been investigated (43). Thus far, the
process by which VacA assembles into higher order structures on SD medium supplemented with the required amino acids and glu-
has not been studied in any detail. Therefore, the goal of this cose at 30 °C as described in the GAL4 Two-hybrid Phagemid manual
study was to characterize the interactions that may mediate (Stratagene). For a positive protein-protein interaction control, we co-
transformed the yeast with pBD-WT and pAD-WT plasmids, which
assembly of VacA into oligomeric structures. We describe here
encode fusion proteins consisting of amino acids 132–236 of wild-type
several lines of evidence indicating that interactions occur be- ␭cI, fragment C, together with either the GAL4-BD or -AD, respectively.

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


tween the p-33 and p-55 domains of VacA, and we identify For a negative control (i.e. two proteins that do not interact), we used
candidate regions within these two domains that are required the pAD-WT plasmid co-transformed with a pBD-pLamin C plasmid,
for these interactions. In addition, we propose that interactions which expresses the BD of GAL4 fused to amino acids 60 –230 of human
between p-33 and p-55 domains are essential for the assembly lamin C. We also used plasmids pAD-Mut and pBD-Mut, which encode
a mutated (E233K) ␭cI, fragment C, fused to either the GAL4-AD or
of VacA into oligomeric structures and for VacA vacuolating
-BD. The cI-E233K mutation interferes with the interaction between
cytotoxin activity. the cI monomers, resulting in a protein-protein interaction that is
EXPERIMENTAL PROCEDURES weaker than that of wild-type proteins. Plasmids expressing positive
and negative interaction control proteins were obtained from
Bacterial and Yeast Strains, Media, and Growth Condition—H. py- Stratagene.
lori strain 60190 (ATCC 49503) was grown on trypticase soy agar plates One colony of each co-transformant was grown in 2 ml of SD medium
containing 5% sheep blood at 37 °C in ambient air containing 5% CO2. containing 2% (w/v) glucose and lacking Trp and Leu (SD⫺Leu,⫺Trp)
Liquid cultures were grown in sulfite-free Brucella broth supplemented at 30 °C with aeration for ⬃24 h. Cultures were then normalized to an
with either 5% fetal bovine serum or 0.5% activated charcoal. Esche- A600 of about 0.26 in a final volume of 100 ␮l of SD⫺Leu,⫺Trp,⫺His
richia coli XL1-Blue (Stratagene) was used for plasmid propagation and broth, and 10-fold serially diluted into SD⫺Leu,⫺Trp,⫺His broth in a
was grown on Luria-Bertani (LB) broth or LB agar at 37 °C. Yeast microtiter plate. Diluted cultures (5 ␮l) were seeded on duplicate
two-hybrid experiments were performed with Saccharomyces cerevisiae SD⫺Leu,⫺Trp plates (selection for plasmids) and SD⫺Leu,⫺Trp,⫺His
strain YRG-2 (MAT␣ ura3-52 his3-200 ade2-101 lys2-801 trp1-901 plates (selection for protein-protein interactions) and incubated at
leu2-3,112 gal4-542 gal80-538 LYS2::UASGAL1-TATAGAL1-HIS3 30 °C for 3–10 days.
URA3::UASGAL4 17mers(x3)-TATACYC1-lacZ) (Stratagene). Yeast strains To confirm the occurrence of protein-protein interactions, a ␤-galac-
were grown in rich medium (yeast extract, peptone, dextrose (YPAD)) or tosidase liquid assay was performed using the Yeast ␤-Galactosidase
in synthetic defined (SD) minimal medium (supplemented with re- Assay Kit as described by the manufacturer (Pierce). Briefly, individual
quired amino acids and glucose) at 30 °C as described in the GAL4 co-transformants were grown in 2 ml of SD⫺Leu,⫺Trp medium con-
Two-hybrid Phagemid manual (Stratagene). taining 2% (w/v) glucose at 30 °C with aeration for ⬃16 –24 h until
Construction of VacA Yeast Two-hybrid Vectors Containing vacA cultures reached mid-log phase (A600 0.4 – 0.5). 150-␮l aliquots of the
Fragments—vacA sequences encoding the wild-type p-33 and p-55 VacA cultures were then mixed with 150 ␮l of the working solution (equal
domains were PCR-amplified from genomic DNA of H. pylori strain volume of 2⫻ ␤-Galactosidase Assay Buffer and Y-PERTM Reagent) and
60190 and cloned into plasmids encoding the transcription activation then incubated at 37 °C until solutions turned yellow (⬃1– 4 h), at
domain (pAD)1 and/or the DNA binding domain (pBD) of the GAL4 which time reactions were stopped by the addition of 175 ␮l of ␤-Ga-
Two-hybrid Phagemid system (Stratagene). The sequences of the prim- lactosidase Assay Stop Solution. The time required for yellow color
ers used for cloning the different vacA fragments into the yeast two- development was recorded for each tube. The A410 values of clarified
hybrid vectors are described in Table I. Primers combinations, restric- reaction supernatants (200 ␮l) were measured in a 96-well plate, and
tion site used for cloning the PCR products into the pAD and pBD the ␤-galactosidase activity was calculated by using the Miller equation
vectors, and the amino acid numbers of the VacA fragments are de- as described in the Yeast ␤-Galactosidase Assay Protocol (Pierce). The
scribed in Table II. values presented are the average of at least three independent co-
To facilitate the introduction of in-frame deletion mutations into the transformants (means ⫾ S.D.). Statistical significance was analyzed
pAD33 plasmid, vacA sequences encoding p-33 mutant domains were using Student’s t test.
PCR-amplified from genomic DNA of previously described H. pylori Introduction of DNA Encoding a FLAG Tag Epitope and Enteroki-
strains harboring the specific in-frame vacA deletions (39) and cloned nase Site into the vacA Gene of H. pylori 60190 —To modify the vacA
into the pAD vector (Table II). vacA sequences encoding p-55 fragments gene so that it encoded a VacA protein containing a FLAG tag (DYK-
were PCR-amplified from genomic DNA of H. pylori strain 60190 (Table DADDDK) and an enterokinase cleavage site (after the underlined K),
II) and were cloned into the pBD vector. The entire vacA fragment in complementary primers OP2891 and OP2892 encoding the FLAG
each p-33 and p-55 encoding plasmid was analyzed by nucleotide se- epitope (Table I) were annealed and ligated into the unique StuI site of
quence analysis in order to verify that no unintended mutations had plasmid pA178 (45). The resulting plasmid, pA178-FLAG, contained a
been introduced. sequence encoding the FLAG epitope in the proper orientation inserted
Yeast Methods—Yeast strains were co-transformed with 400 ng of between amino acids 317 and 318 of VacA. Plasmid pA178-FLAG was
individual plasmids using the lithium acetate method (44) and cultured then used to transform H. pylori strain VM022 (39), and transformants
were selected by growth on 5.5% sucrose plates, as described previously
1
The abbreviations used are: pAD, plasmids encoding the GAL4 (39, 46). Analysis of a single transformant (H. pylori VM088) by PCR
transcription-activation domain; pBD, plasmids encoding the GAL4 and by DNA sequence analysis confirmed that the sequence encoding
DNA binding domain; CHAPS, 3-[(3-cholamidopropyl)-dimethyl- the FLAG epitope and the enterokinase cleavage site had been intro-
ammonio]-1-propanesulfonate. duced into the desired location.
2326 H. pylori VacA
er; and (ii) the DNA binding domain (BD) present on the
plasmid pBD-GAL4-Cam (pBD), which encodes for the TRP1
gene as a selectable marker. If two fusion proteins interact in
this system, they will bring in close proximity the GAL4 tran-
scription activation domain and the GAL4 DNA binding do-
main to the GAL4/GAL1 promoters, which in turn will initiate
the transcription of the HIS3 and lacZ reporter genes. Protein-
FIG. 1. Proteolytic degradation of VacA into 33- and 55-kDa protein interactions are then detected by the ability of co-
fragments. Proteins precipitated from broth culture supernatant of transformed yeast cells to grow in selective medium lacking
H. pylori 60190 by a 50% saturated solution of ammonium sulfate (lanes Leu, Trp, and His (SD⫺Leu,⫺Trp,⫺His) and by production of
A and B) or purified VacA (lanes C and D) from H. pylori 60190 were ␤-galactosidase activity.
electrophoresed on a 10% SDS-polyacrylamide gel, transferred to a
nitrocellulose membrane, and immunoblotted with polyclonal anti- vacA sequences encoding p-33 and p-55 fragments were
VacA serum. These preparations exhibit varying degrees of spontane- cloned into plasmids encoding the transcription activation do-
ous VacA proteolysis into p-33 and p-55 fragments. main (pAD) and the DNA binding domain (pBD), generating
plasmids pAD33, pBD33, pAD55, and pBD55 (Table II). In
Purification of Oligomeric Forms of VacA—VacA was purified from order to test whether interactions among VacA fragments could
broth culture supernatants of H. pylori as described previously (29). be detected in this system, we co-transformed all four possible
Briefly, broth culture supernatant proteins were concentrated by pre- combinations of the p-33 and p-55 yeast two-hybrid plasmids
cipitation with a 50% saturated solution of ammonium sulfate and
into the yeast reporter strain (Fig. 2A). Serial dilutions of
resuspended in phosphate-buffered saline. Oligomeric VacA (⬃1,000
kDa) was purified by gel filtration chromatography using a Superose co-transformed yeast were then tested for their ability to grow
6HR 16/50 column. To analyze the oligomeric state of the enterokinase- on SD medium lacking Leu and Trp, as well as SD medium
treated FLAG-VacA, 40 ␮g of cleaved toxin was applied to a Superose

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


lacking Leu, Trp, and His. All of the co-transformed yeast grew
6HR 16/50 column, and fractions (2 ml each) were collected. Fractions on the former media, which confirmed that the transformation
were analyzed by immunoblot analysis with an anti-VacA serum. had been successful and provided an indication of the relative
Proteolysis of FLAG-VacA with Enterokinase—Purified FLAG-VacA
was proteolytically cleaved with enterokinase as described by the Re-
number of co-transformed yeast cells present in each dilution.
combinant Enterokinase Kit manual (Novagen). Briefly, 10 ␮g of puri- Yeast co-transformed with plasmids encoding p-33 and p-55
fied FLAG-VacA was incubated with 1 unit of enterokinase and 5 ␮l of fusion proteins grew in SD medium lacking Leu, Trp, and His,
10⫻ enterokinase cleavage buffer in a final volume of 50 ␮l for ⱖ16 h at indicating that there was an interaction between these fusion
room temperature. For mock treatment, FLAG-VacA was treated in the proteins (Fig. 2B). In contrast, yeast co-transformed with plas-
same manner but without enterokinase. Proteolysis of the full-length
mids encoding p-33/p-33 or p-55/p-55 fusion proteins did not
FLAG-VacA was assessed by immunoblot analysis using an anti-VacA
serum or the M2 monoclonal anti-FLAG antibody (Sigma). grow on SD medium lacking Leu, Trp, and His (Fig. 2B). The
Cell Culture and Vacuolating Assays—HeLa cells were grown in p-33/p-55 interaction observed within co-transformed yeast
minimal essential medium (modified Eagle’s medium containing was further confirmed by the analysis of ␤-galactosidase activ-
Earle’s salts) supplemented with 10% fetal bovine serum. For vacuolat- ity, which reflects the activation of the lacZ reporter gene (Fig.
ing assays, HeLa cells were seeded (1.5 ⫻ 104) into 96-well plates 24 h 2C). The p-33/p-55 interaction was independent of the protein
prior to each experiment. Protein concentrations of the purified VacA
toxins were determined by using a Micro-BCA assay (Pierce). Serial fused (AD or BD) to the p-33 or p-55 fragment, because both
dilutions of purified VacA toxins (standardized by protein concentra- combinations of tested plasmids (pAD33/pBD55 and pAD55/
tion) were incubated with cells in serum-free medium containing 10 mM pBD33) were able to activate the HIS3 and lacZ reporters (Fig.
ammonium chloride as described previously (5). Purified VacA prepa- 2, B and C). Furthermore, none of the fusion constructs were
rations (VacA, FLAG-VacA, and cleaved FLAG-VacA) were acid-acti- able to activate the reporter genes when transformed alone into
vated by adjusting them to pH 3 by the addition of 250 mM hydrochloric
the yeast reporter strain (data not shown), indicating that the
acid before they were added to cell culture wells (47, 48). After 24 h of
incubation of VacA toxins with the cell monolayers, cells were examined interaction detected in the yeast two-hybrid system is due to a
by inverted light microscopy. Toxins that induced vacuoles in more than specific interaction between p-33 and p-55 VacA fragments.
50% of the cells were scored positive for vacuolating cytotoxic activity. p-33/p-55 Interactions of VacA Mutant Fragments—Previ-
Vacuolating activity was also quantified by a neutral red uptake assay ously, we have reported the construction of H. pylori strains
as described previously (49). Neutral red uptake data are presented as containing in-frame deletion mutations in the vacA gene (39).
A540 values (means ⫾ S.D.). Statistical significance was analyzed by
using Student’s t test. Results from this previous study indicated that a mutant VacA
protein with a deletion of amino acids 6 –27, VacA-(⌬6 –27),
RESULTS could assemble into water-soluble oligomeric structures and
Detection of p-33/p-55 Interactions in a Yeast Two-hybrid suggested that VacA proteins containing ⌬28 –108, ⌬56 – 83,
System—Previous studies have reported that 88-kDa VacA ⌬85–127, or ⌬112–196 deletion mutations were defective in the
monomers commonly undergo spontaneous degradation, yield- capacity for oligomer assembly (39) (Table III). We hypothe-
ing 33- and 55-kDa fragments (Fig. 1) (11, 27, 28). These two sized that the latter mutations might disrupt interactions be-
fragments have been considered to represent two domains or tween p-33 and p-55 domains, and that such a defect might
subunits of VacA (11, 20, 30, 31). To investigate potential account for failure of these VacA mutant proteins to form
interactions among these two VacA fragments, we investigated oligomeric structures. In order to test directly this hypothesis,
whether they were able to interact in a GAL4 transcription we introduced these same deletion mutations (i.e. ⌬6 –27, ⌬28 –
activator yeast two-hybrid system (GAL4 Two-hybrid Phage- 108, ⌬56 – 83, ⌬85–127, and ⌬112–196) into the pAD33 plas-
mid; Stratagene) (50). This particular yeast two-hybrid system mid, as described under “Experimental Procedures” (Table II).
was selected based on the low expression of the bait and prey The pAD33 plasmid was chosen for mutagenesis based on the
fusion proteins which, in turn, is thought to reduce the number observation that higher reporter activity was detected when
of false positive interactions (51). In this system, the yeast wild-type p-33 was fused to the transcription activation domain
GAL4 transcription activator has been divided into the follow- and wild-type p-55 fused to the DNA binding domain, com-
ing two separate functional domains (52): (i) the transcription pared with the opposite orientation (Fig. 2C). Mutant pAD33
activation domain (AD) present on plasmid pAD-GAL4 –2.1 plasmids were co-transformed into yeast along with the wild-
(pAD), which encodes for the LEU2 gene as a selectable mark- type pBD55 plasmid, and the activation of the reporter genes
H. pylori VacA 2327
TABLE II
Yeast two-hybrid vectors containing vacA fragments
Y2H plasmidsa Forward primersb Reverse primersb Restriction enzymesc VacA amino acidsd

pBD33 AND510a AND511 SrfI/SalI 1–312


pAD33 AND512 AND511 BamHI/SalI 1–312
pBD55 AND6098 AND515a EcoRI/SalI 313–821
pAD55 AND516 AND515a BamHI/SalI 313–821
pAD⌬6–27 OP4175 AND511 BamHI/SalI 1–312 (⌬6–27)
pAD⌬28–108 AND512 AND511 BamHI/SalI 1–312 (⌬28–108)
pAD⌬56–83 AND512 AND511 BamHI/SalI 1–312 (⌬56–83)
pAD⌬85–127 AND512 AND511 BamHI/SalI 1–312 (⌬85–127)
pAD⌬112–196 AND512 AND511 BamHI/SalI 1–312 (⌬112–196)
pBD313–700 AND6098 BAR1559 EcoRI/SalI 313–700
pBD313–550 AND6098 BAR1558 EcoRI/SalI 313–550
pBD313–478 AND6098 OP4176 EcoRI/SalI 313–478
pBD550–821 BAR1557 AND515a EcoRI/SalI 550–821
pBD479–821 BAR1556 AND515a EcoRI/SalI 479–821
a
Name of the yeast two-hybrid (Y2H) plasmids expressing VacA fragments.
b
Oligonucleotides used to PCR-amplify the different vacA sequences. Oligonucleotide sequences are described in Table I.
c
Restriction sites used to clone the PCR products into the pAD and pBD yeast two-hybrid plasmids.
d
The VacA amino acid numbering system used in this table is based on designating the first amino acid (alanine) of the mature secreted VacA
toxin of strain 60190 (25) as amino acid 1. ⌬ indicates amino acids that are deleted in the mutant VacA fragments.

TABLE III
Characterization of mutant VacA toxins

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


Y2H Oligomer Cytotoxic
p-33a p-55b interactionc formationd activitye

WT WT ⫹ ⫹ ⫹
⌬6–27 WT ⫹ ⫹ ⫺
⌬28–108 WT ⫺ ⫺ ⫺
⌬56–83 WT ⫺ ⫺ ⫺
⌬85–127 WT ⫺ ⫺ ⫺
⌬112–196 WT ⫺ ⫺ ⫺
a
The p-33 VacA domain contains amino acids 1–312.
b
The p-55 VacA domain contains wild type (WT) amino acids 313–
821.
c
Interaction between p-33 and p-55 VacA fragments was determined
using the yeast two-hybrid system (Y2H) as described in the text.
d
Oligomer formation of full-length VacA toxins secreted by H. pylori
was analyzed by determining whether these proteins eluted as large
oligomeric structures (⬇1,000 kDa) from a Superose 6 HR 16/50 FPLC
column (5, 39).
e
Cytotoxic vacuolating activity of VacA toxins for HeLa cells was
analyzed as described under “Experimental Procedures.”

activate the reporter genes when co-transformed with wild-


type pBD55. The p-55/⌬6 –27p-33 interaction was specific, be-
cause yeast cells co-transformed with pBD33 and pAD⌬6 –
27p-33 were not able to activate the reporter genes (data not
shown). Thus, mutations that disrupted oligomerization of
VacA secreted by H. pylori (39) also disrupted p-33/p-55 inter-
actions in the yeast two-hybrid system (Table III). Interest-
ingly, yeast co-transformed with plasmids expressing
FIG. 2. Interaction of VacA fragments in the yeast two-hybrid
⌬6 –27p-33 and p-55 consistently produced higher levels of
system. A, representation of the different p-33 and p-55 fusion proteins
and plasmid combinations used in the co-transformation experiments. ␤-galactosidase activity than did yeast expressing wild-type
B, yeast cells were co-transformed with the plasmids depicted in A or p-33 and p-55 (Fig. 3B).
with controls plasmids described under “Experimental Procedures.” To further map putative amino acid sequences involved in
Co-transformed yeast cells (normalized based on absorbance) were 10- the p-33/p-55 interaction, we generated different p-55 frag-
fold serially diluted, and identical inocula were plated on two types of
SD agar plates. All of the co-transformed yeast grew on SD⫺Leu,⫺Trp ments (i.e. encoding amino acids 313–700, 313–550, 313– 478,
plates. Protein-protein interactions were assessed by growth of the 550 – 821, and 479 – 821) and cloned them into the pBD vector
co-transformed yeast cells on SD⫺Leu,⫺Trp,⫺His plates. C, ␤-galacto- as described under “Experimental Procedures” (Table II).
sidase liquid culture assay of the co-transformed yeast cells. Activation These new fusion proteins were tested for their ability to in-
of the protein-protein interaction reporters was only detected when
yeast were co-transformed with plasmids encoding wild-type p-33 to- teract with wild-type p-33 as described above (Fig. 4A). Yeast
gether with plasmids encoding wild-type p-55. Results represent the co-transformed with the pAD33 plasmid and plasmids express-
mean ⫾ S.D. from triplicate determinations, each representing a sepa- ing p-55 fragments containing amino acids 313–700, 313–550,
rate colony. *, p ⬍0.05 when compared with the negative control; ⫹ or 313– 478 were able to grow on SD⫺Leu,⫺Trp,⫺His, whereas
cntrl, positive control; ⫺ cntrl, negative control.
yeast co-transformed with the pAD33 plasmid and plasmids
expressing p-55 fragments containing amino acids 479 – 821 or
was analyzed as described above. Wild-type pAD33 and 550 – 821 were not able to grow (Fig. 4B). The former group of
pAD⌬6 –27p-33 were each able to activate both the HIS3 and co-transformed yeast grew slowly on SD⫺Leu,⫺Trp,⫺His
lacZ reporter genes when co-transformed with wild-type plates and expressed a relatively low level of ␤-galactosidase
pBD55 (Fig. 3), whereas the other pAD33 mutants failed to activity (Fig. 4, B and C). This suggests that the interactions
2328 H. pylori VacA

FIG. 3. Interaction between wild-type p-55 and p-33 mutant

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


VacA fragments. A, co-transformed yeast cells (normalized based
on absorbance) were 10-fold serially diluted, and identical inocula
were plated on two types of SD plates. Protein-protein interactions
were assessed by growth of the co-transformed yeast cells on
SD⫺Leu,⫺Trp,⫺His plates. B, ␤-galactosidase liquid culture assay of
the co-transformed yeast cells. Activation of the protein-protein inter-
action reporters was only detected when yeast were co-transformed with
plasmids encoding wild-type p-33 or ⌬6 –27p-33 together with a plasmid
encoding wild-type p-55. Results represent the mean ⫾ S.D. from tripli-
cate colonies. *, p ⬍0.05 when compared with the negative control.

between the 313–700, 313–550, and 313– 478 p-55 fragments FIG. 4. Interaction between wild-type p-33 and fragments of
and p-33 are weaker than the interaction between full-length p-55. A, representation of the different p-55 fragments used in the
wild-type p-55 and p-33. When we co-transformed yeast with a co-transformation experiment. B, yeast cells co-transformed with the
pBD55 fragments depicted in A and wild-type pAD33 (normalized based
mutated version of the positive control plasmids (mutant ⫹ on absorbance) were 10-fold serially diluted, and identical inocula were
control described under “Experimental Procedures”) known to plated on two types of SD agar plates. C, ␤-galactosidase liquid culture
encode proteins that interact with relatively low affinity (53), a assay of the co-transformed yeast cells. Protein-protein interactions
similar low level of ␤-galactosidase activity was detected (Fig. were assessed by growth of the co-transformed yeast cells on
SD⫺Leu,⫺Trp,⫺His plates and by a ␤-galactosidase liquid culture as-
4C). The data obtained with these mutant control plasmids say. Sample numbers in C are identical to those in B. Wild-type p-33
support our conclusion that the p-33 interacts weakly with was able to interact with p-55 fragments containing amino acids 313–
313–700, 313–550, and 313– 478 p-55 fragments. 700, 313–550, and 313– 478 but not with amino acids 550 – 821 or
Proteolytic Cleavage of H. pylori VacA into 33- and 55-kDa 479 – 821. ␤-Galactosidase liquid culture assay results represent the
mean ⫾ S.D. from triplicate colonies. *, p ⬍0.05 when compared with
Fragments—We sought to confirm the finding that p-33 inter- the negative control.
acts with p-55 using VacA purified from H. pylori. Numerous
previous studies have demonstrated that VacA proteins pro-
duced by H. pylori can degrade into 33- and 55-kDa fragments 88-kDa VacA protein could be specifically cleaved into frag-
(11, 27, 28). However, it remains unclear whether proteolytic ments similar to the spontaneously arising 33- and 55-kDa
cleavage of the 88-kDa VacA protein into 33- and 55-kDa frag- VacA fragments. Immunoblotting studies with an anti-VacA
ments alters VacA cytotoxic activity or alters its oligomeric serum indicated that an H. pylori strain (VM088) containing
structure. One reason why this issue has not yet been ad- this engineered vacA allele expressed and secreted VacA at
dressed in any detail is that it has been difficult to experimen- levels similar to wild-type H. pylori strain 60190 (data not
tally induce the desired cleavage. VacA preparations commonly shown). The FLAG-VacA toxin was purified from H. pylori
undergo partial proteolytic degradation during storage (Fig. 1), VM088 culture supernatant as large oligomeric structures that
but complete proteolysis of the 88-kDa VacA protein into eluted from a gel filtration column in the same fractions as
smaller fragments occurs uncommonly. Furthermore, sponta- wild-type VacA. Purified FLAG-VacA underwent spontaneous
neous proteolytic degradation of VacA often involves cleavage partial degradation into two forms of the p-33 and p-55 VacA
at multiple sites (27). fragments (Fig. 6B, 1st lane), which is consistent with previous
In order to test experimentally whether cleavage of VacA observations suggesting that proteolysis can occur at multiple
into 33- and 55-kDa fragments alters its activity, we sought to sites (27). As expected, enterokinase treatment resulted in the
develop a system in which a protease could be used to induce complete cleavage of the full-length FLAG-VacA into p-55 and
proteolytic cleavage at a specific site located between the 33- FLAG-tagged p-33 (FLAGp-33) fragments as seen by immuno-
and 55-kDa domains. To accomplish this, we constructed a blot analysis using an anti-VacA serum or an anti-FLAG mono-
modified H. pylori strain in which an oligonucleotide encoding clonal antibody (Fig. 6). In contrast, enterokinase treatment of
the FLAG tag epitope and an enterokinase cleavage site was the wild-type 60190 VacA toxin (lacking the FLAG tag epitope)
inserted into the vacA allele of H. pylori 60190 as described did not induce any detectable cleavage (data not shown).
under “Experimental Procedures” (Fig. 5A). This enterokinase Vacuolating Toxic Activity of Intact FLAG-VacA and Proteo-
cleavage site (Fig. 5B) provided a mechanism by which the lytically Cleaved FLAG-VacA—We next compared the cytotoxic
H. pylori VacA 2329

FIG. 6. Enterokinase treatment of full-length FLAG-VacA re-


sults in production of 33- and 55-kDa VacA fragments. Purified
VacA from H. pylori VM088 (10 ␮g) was either treated with enteroki-
nase (1 unit) or mock-treated as described under “Experimental Proce-
dures.” Toxins (100 ng) were then electrophoresed on a 10% SDS-
polyacrylamide gel, transferred to a nitrocellulose membrane, and
reacted with anti-FLAG M2 monoclonal antibody (Sigma) (A). The blot
was then stripped and reacted with polyclonal anti-VacA serum (B).
Enterokinase treatment induced complete cleavage of full-length
FLAG-VacA into ⬃33- and ⬃55-kDa fragments. In preparations of
FLAG-VacA not treated with enterokinase, the FLAG tag epitope is
detected predominantly in full-length 88-kDa VacA and in the p-55
fragment (A, 1st lane). Only the ⬃33-kDa fragment retained the FLAG
tag epitope after enterokinase treatment (A, 2nd lane).

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


FIG. 5. Construction of H. pylori VM088 using a sacB-based
counter-selection approach. A, the pMM389 plasmid contains a
vacA fragment from H. pylori strain 60190 and a sacB/kan cassette (39,
46). Transformation of H. pylori 60190 with pMM389 yielded a kanamy-
cin-resistant transformant designated H. pylori VM022 (39). H. pylori
VM022 was transformed with pA178-FLAG plasmid; transformants FIG. 7. Vacuolating cytotoxic activity of intact and proteolyti-
were selected on sucrose-containing medium, and a strain (H. pylori cally cleaved FLAG-VacA. Identical aliquots of purified FLAG-VacA
VM088) with a sequence encoding a FLAG tag with an enterokinase site were either treated with enterokinase as described under “Experimen-
inserted into the vacA gene was thereby obtained. B, VacA spontane- tal Procedures” or left untreated. Toxins were then either acid-activated
ously degrades into two fragments, termed p-33 and p-55. Open arrows (pH 3) or left untreated and added to tissue culture medium overlying
indicate the most common site where wild-type VacA from H. pylori HeLa cells. The final VacA concentration of all samples was 5 ␮g/ml.
60190 undergoes spontaneous cleavage and the resulting p-33 and p-55 Vacuolating activity was quantified using a neutral red uptake assay.
fragments (27). Closed arrows indicate the enterokinase cleavage site Enterokinase-treated FLAG-VacA and untreated FLAG-VacA did not
introduced with the FLAG tag epitope and the FLAGp-33 and p-55 differ significantly in vacuolating cytotoxin activity. Furthermore, both
fragments generated after enterokinase cleavage. The amino acid num- enterokinase-treated and untreated FLAG-VacA required acid activa-
bering system used in this figure is based on designating the first amino tion in order to induce vacuolating activity. Results represent the
acid (alanine) of the mature secreted VacA toxin of strain 60190 as mean ⫾ S.D. from triplicate samples and are expressed as a percent of
amino acid 1. neutral red uptake induced by full-length acid-activated FLAG-VacA.

activities of intact FLAG-VacA and FLAG-VacA that had been Oligomeric State of Proteolytically Cleaved FLAG-VacA—
proteolytically cleaved into p-55 and FLAGp-33 fragments. Pu- The secreted FLAG-VacA assembles into large water-soluble
rified oligomeric FLAG-VacA was either treated with enteroki- oligomeric structures (Fig. 8A), in a manner similar to wild-
nase or mock-treated as described under “Experimental Proce- type VacA (5, 29). To determine whether p-33 and p-55 frag-
dures.” Proteins were then acid-activated (a procedure used to ments of VacA remained physically associated following pro-
enhance vacuolating activity) and added to the neutral-pH teolytic cleavage of FLAG-VacA, enterokinase-treated FLAG-
medium overlying HeLa cells (47, 48). The vacuolating activi- VacA was fractionated by gel filtration chromatography, and
ties of intact FLAG-VacA and proteolytically cleaved FLAG- its elution pattern was monitored by immunoblot analysis with
VacA were indistinguishable as determined by microscopic ex- an anti-VacA serum. Proteolytically cleaved FLAG-VacA and
amination (data not shown) and analysis by a neutral red intact FLAG-VacA were found to elute in the same high mo-
uptake assay (Fig. 7). The vacuolating activities of full-length lecular mass fractions (Fig. 8) but not in any of the lower
FLAG-VacA and proteolytically cleaved FLAG-VacA were not molecular mass fractions (data not shown), indicating that
detectably different when tested in a range of toxin concentra- VacA fragments (i.e. FLAGp-33 and p-55) remain associated
tions from 2.5 to 10 ␮g/ml (Fig. 7 and data not shown). within an oligomeric structure (Fig. 8).
VacA purified from wild-type H. pylori strain 60190 exhibits
very little vacuolating toxic activity unless it is acid- or alka- DISCUSSION
line-activated prior to contact with cells (47, 48, 54). We hy- Over the past decade, multiple lines of investigation have
pothesized that proteolytically cleaved VacA might exhibit vac- provided evidence for the occurrence of homotypic interactions
uolating toxic activity even in the absence of acid activation. among VacA proteins (5, 11, 31, 34, 35, 39, 45). The earliest
However, neither intact nor proteolytically cleaved FLAG- evidence suggesting oligomerization of VacA proteins was the
VacA exhibited detectable vacuolating activity in the absence observation that purified VacA from H. pylori broth culture
of acid activation, as determined by microscopic examination supernatant assembles into structures of ⬃1,000 kDa in mass,
(data not shown) and analysis by a neutral red uptake assay corresponding to a buoyant density of about 22 S (5, 29). Ul-
(Fig. 7). trastructural studies have shown that these VacA complexes
2330 H. pylori VacA
provided evidence that an N-terminal hydrophobic portion of
the p-33 fragment could undergo transmembrane protein ho-
modimerization within E. coli membranes (40, 55). The inabil-
ity to detect p-33/p-33 interactions with the yeast two-hybrid
system is consistent with a model in which p-33/p-33 interac-
tions occur only within membranes (40, 55).
In an effort to map putative interacting regions within p-33
and p-55, we used the yeast two-hybrid system to analyze a
series of mutant proteins containing internal deletions, as well
as truncated VacA proteins. The yeast two-hybrid data suggest
that amino acids 28 –196 within the p-33 domain and 313– 478
within the p-55 domain contribute to the p-33/p-55 interaction.
Only one of the p-33 deletion mutants (⌬6 –27p-33) was able to
interact with wild-type p-55. VacA amino acids 6 –27 comprise
a hydrophobic region of VacA that inserts into lipid membranes
and is required for VacA cytotoxic and membrane anion-chan-
FIG. 8. Oligomeric state of full-length FLAG-VacA and cleaved nel activity (39, 40, 55). Interestingly, when co-expressed with
FLAG-VacA. Concentrated broth culture supernatant from H. pylori p-55, the ⌬6 –27p-33 mutant fragment induced higher reporter
strain VM088 (A) and 40 ␮g of purified FLAG-VacA treated with en- activity in the yeast two-hybrid assay than did wild-type p-33.
terokinase as described under “Experimental Procedures” (B) were
A possible explanation for this result could be that the ⌬6 –
applied to a Superose 6 HR FPLC column. Forty fractions of 2 ml each
were collected, with fraction 1 corresponding to the void volume. Sam- 27p-33 mutant fragment is able to translocate to the yeast
ples (40 ␮l) were then electrophoresed on a 10% SDS-polyacrylamide

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


nuclei more efficiently than can the wild-type p-33, since pre-
gel, transferred to a nitrocellulose membrane, and immunoblotted with viously it has been reported (56) that hydrophobic sequences
anti-VacA serum. Full-length VacA as well as enterokinase-cleaved
decrease nuclear protein translocation. Another possibility
VacA eluted in the same fractions with an elution peak at fraction 13,
corresponding to a molecular mass of about 1,000 kDa (5, 29). VacA was could be that the ⌬6 –27p-33 mutant fragment interacts more
not detected in any of the lower molecular mass fractions (data not strongly with the wild-p-55 fragment than does wild-type p-33.
shown). p-55 fragments are detected more prominently than p-33 frag- By constructing a FLAG-VacA toxin, which can be efficiently
ments in B due to the relatively weaker reactivity of the anti-VacA
cleaved into p-33 and p-55 domains, we were able to study the
serum against the p-33 fragment. When the membrane shown in B was
stripped and immunoblotted with polyclonal anti-FLAG antibody, the p-33/p-55 interactions that occur within the VacA oligomer
p-33 band was detected in fractions 8 –16 (data not shown). produced by H. pylori. Our results (Fig. 8) clearly indicate that
p-33 and p-55 fragments remain physically associated after
proteolytic cleavage of VacA. In agreement with this interpre-
have a flower-like shape and that they contain anywhere from
tation, when we incubated proteolytically cleaved FLAG-VacA
6 to 14 subunits (29, 30, 42). These flower-shaped structures
with an Anti-FLAG M2 Affinity Gel (Sigma), both the p-55 and
disassemble into VacA monomers after exposure to acid or
FLAGp-33 fragments were captured (data not shown). Neither
alkaline pH and then reassemble into oligomeric structures
FLAGp-33 nor p-55 fragments could be eluted from the affinity
after neutralization (29, 54). Further evidence for the assembly
of VacA into oligomeric structures has resulted from experi- gel under pH conditions ranging from 7.5 to 4.5, nor with 1%
ments in which two different VacA toxins have been shown to CHAPS, 10% Triton X-100, or 2 M urea, suggesting that there is
form mixed oligomeric structures (34, 39, 45). VacA-VacA in- a stable physical interaction between the p-33 and p-55 VacA
teractions have also been detected by transiently transfecting domains (data not shown). Furthermore, our results demon-
cells with different fluorescently tagged VacA-expressing plas- strate that the vacuolating activity of intact FLAG-VacA and
mids and using fluorescent resonance energy transfer method- fully proteolytically cleaved FLAG-VacA is indistinguishable
ology (35). (Fig. 7). A likely explanation for intact activity of proteolyti-
Although many lines of evidence indicate that VacA can form cally cleaved VacA is that the fragments remain associated and
oligomeric structures, thus far there has been relatively little do not undergo any extensive adverse conformational changes
investigation of the specific VacA-VacA interactions that are (unfolding) following proteolytic cleavage.
required for assembly into higher order structures. In this Two lines of evidence presented in the current study suggest
study we investigated the role of p-33 and p-55 interactions in that the p-33/p-55 interactions play an important role in VacA
oligomer formation by VacA. These two VacA fragments have oligomer assembly. First, the intact oligomeric structure of
been considered to represent two domains or subunits of VacA. proteolytically cleaved VacA indicates that p-33/p-55 interac-
In the present work, we demonstrated that p-33/p-55 interac- tions occur within VacA oligomers. Second, the properties of
tions can be detected using the yeast two-hybrid system. In mutant p-33 fragments in the yeast two-hybrid system corre-
contrast, p-33/p-33 and p-55/p-55 interactions were not de- late perfectly with the capacity of secreted H. pylori mutant
tected in this system. The lack of detectable p-33/p-33 and proteins to form oligomeric structures. This suggests that mu-
p-55/p-55 interactions was not due to the lack of expression of tations disrupting the p-33/p-55 interaction also disrupt the
these fusion proteins in the yeast, because all four fusion pro- formation of large VacA oligomeric structures. It is possible
teins were able to interact when tested for p-33/p-55 interac- that there may be two different types of interactions: (i) in-
tions. Importantly, the failure to detect p-33/p-33 or p-55/p-55 tramolecular interactions between the p-33 and p-55 domains
interactions in the yeast two-hybrid system does not exclude of an individual VacA monomer that might be important for
the possibility that such interactions might occur in other en- proper folding of the VacA monomers within the oligomeric
vironments. A previous study analyzed the properties of a structures, and (ii) intermolecular interaction between p-33
modified p-55 fragment secreted by H. pylori, and reported that and p-55 domains of different 88-kDa molecules to form the
this protein could form homodimers but not large oligomeric larger oligomeric structures. Currently, we are not able to
structures (31). However, the VacA protein analyzed in that differentiate between these two interactions.
study contained at least 27 residues of the p-33 domain in VacA toxins that fail to oligomerize consistently lack vacuo-
addition to the p-55 fragment (31). Another previous study lating cytotoxic activity, suggesting that oligomerization is im-
H. pylori VacA 2331
portant for VacA activity (39). This hypothesis is supported by 18. Papini, E., Satin, B., Norais, N., de Bernard, M., Telford, J. L., Rappuoli, R.,
and Montecucco, C. (1998) J. Clin. Investig. 102, 813– 820
the observation that neither p-33 nor p-55 is able to exhibit 19. Peek, R. M., Jr., Blaser, M. J., Mays, D. J., Forsyth, M. H., Cover, T. L., Song,
vacuolating cytotoxic activity in a transient transfection assay S. Y., Krishna, U., and Pietenpol, J. A. (1999) Cancer Res. 59, 6124 – 6131
20. Galmiche, A., Rassow, J., Doye, A., Cagnol, S., Chambard, J. C., Contamin, S.,
when expressed individually within cells (32, 34, 41). In con- de Thillot, V., Just, I., Ricci, V., Solcia, E., Van Obberghen, E., and Boquet,
trast, when both domains are co-expressed within cells, they P. (2000) EMBO J. 19, 6361– 6370
interact and exhibit vacuolating cytotoxic activity (32, 34). 21. Kuck, D., Kolmerer, B., Iking-Konert, C., Krammer, P. H., Stremmel, W., and
Rudi, J. (2001) Infect. Immun. 69, 5080 –5087
Thus, the data presented in the current study, as well as data 22. Cover, T. L., Krishna, U. S., Israel, D. A., and Peek, R. M., Jr. (2003) Cancer
obtained using other systems, indicate that p-33/p-55 interac- Res. 63, 951–957
tions are essential for VacA assembly into oligomeric struc- 23. Molinari, M., Salio, M., Galli, C., Norais, N., Rappuoli, R., Lanzavecchia, A.,
and Montecucco, C. (1998) J. Exp. Med. 187, 135–140
tures and also for vacuolating cytotoxic activity. 24. Gebert, B., Fischer, W., Weiss, E., Hoffmann, R., and Haas, R. (2003) Science
Oligomerization is an important feature of many proteins, in 301, 1099 –1102
25. Cover, T. L., Tummuru, M. K. R., Cao, P., Thompson, S. A., and Blaser, M. J.
particular pore-forming proteins (57, 58). Examples of these (1994) J. Biol. Chem. 269, 10566 –10573
proteins include perforin, Bcl-2 family members, as well as 26. Schmitt, W., and Haas, R. (1994) Mol. Microbiol. 12, 307–319
several bacterial toxins. Several bacterial pore-forming toxins, 27. Nguyen, V. Q., Caprioli, R. M., and Cover, T. L. (2001) Infect. Immun. 69,
543–546
including protective antigen of Bacillus anthracis, ␣-hemolysin 28. Garner, J. A., and Cover, T. L. (1996) Infect. Immun. 64, 4197– 4203
of Staphylococcus aureus, streptolysin O of Streptococcus pyo- 29. Cover, T. L., Hanson, P. I., and Heuser, J. E. (1997) J. Cell Biol. 138, 759 –769
30. Lupetti, P., Heuser, J. E., Manetti, R., Massari, P., Lanzavecchia, S., Bellon,
genes, ␣-toxin of Clostridium septicum, El Tor cytolysin of P. L., Dallai, R., Rappuoli, R., and Telford, J. L. (1996) J. Cell Biol. 133,
Vibrio cholera, and aerolysin of Aeromonas hydrophila, assem- 801– 807
ble in target cell membranes forming transmembrane channels 31. Reyrat, J. M., Lanzavecchia, S., Lupetti, P., de Bernard, M., Pagliaccia, C.,
Pelicic, V., Charrel, M., Ulivieri, C., Norais, N., Ji, X., Cabiaux, V., Papini,
(59 – 65). These proteins are interesting because, like VacA, E., Rappuoli, R., and Telford, J. L. (1999) J. Mol. Biol. 290, 459 – 470
they are synthesized as water-soluble molecules but then insert 32. Ye, D., Willhite, D. C., and Blanke, S. R. (1999) J. Biol. Chem. 274, 9277–9282
33. Ye, D., and Blanke, S. R. (2000) Infect. Immun. 68, 4354 – 4357
into membranes. Further studies of the processes by which

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


34. Ye, D., and Blanke, S. R. (2002) Mol. Microbiol. 43, 1243–1253
these toxins assemble into oligomeric structures may ulti- 35. Willhite, D. C., Ye, D., and Blanke, S. R. (2002) Infect. Immun. 70, 3824 –3832
mately lead to the development of therapeutic inhibitors that 36. de Bernard, M., Burroni, D., Papini, E., Rappuoli, R., Telford, J., and
Montecucco, C. (1998) Infect. Immun. 66, 6014 – 6016
block the oligomerization and the activity of these toxins 37. Wang, H. J., Chang, P. C., Kuo, C. H., Tzeng, C. S., and Wang, W. C. (1998)
in vivo. Biochem. Biophys. Res. Commun. 250, 397– 402
38. Wang, H. J., and Wang, W. C. (2000) Biochem. Biophys. Res. Commun. 278,
Acknowledgments—We thank Ping Cao, Beverly Hosse, Arlene D. 449 – 454
Vinion-Dubiel, Leslie Morrison, and Mark Hansberger for technical 39. Vinion-Dubiel, A. D., McClain, M. S., Czajkowsky, D. M., Iwamoto, H., Ye, D.,
Cao, P., Schraw, W., Szabo, G., Blanke, S. R., Shao, Z., and Cover, T. L.
assistance and helpful discussions. DNA oligonucleotides were synthe-
(1999) J. Biol. Chem. 274, 37736 –37742
sized by the Vanderbilt University DNA Chemistry Core Facility, and 40. McClain, M. S., Iwamoto, H., Cao, P., Vinion-Dubiel, A. D., Li, Y., Szabo, G.,
DNA sequence analysis was performed by the Vanderbilt University Shao, Z., and Cover, T. L. (2003) J. Biol. Chem. 278, 12101–12108
DNA Sequencing Laboratory. The Vanderbilt University DNA Sequenc- 41. de Bernard, M., Arico, B., Papini, E., Rizzuto, R., Grandi, G., Rappuoli, R., and
ing Laboratory is supported by the Vanderbilt-Ingram Cancer Center. Montecucco, C. (1997) Mol. Microbiol. 26, 665– 674
42. Adrian, M., Cover, T. L., Dubochet, J., and Heuser, J. E. (2002) J. Mol. Biol.
REFERENCES 318, 121–133
1. Warren, J. R., and Marshall, B. J. (1983) Lancet 1, 1273–1275 43. Iwamoto, H., Czajkowsky, D. M., Cover, T. L., Szabo, G., and Shao, Z. (1999)
2. Cover, T. L., Berg, D. E., Blaser, M. J., and Mobley, H. L. T. (2001) in Principles FEBS Lett. 450, 101–104
of Bacterial Pathogenesis (Groisman, E. A., ed) pp. 510 –558, Academic 44. Woods, R. A., and Gietz, R. D. (2001) Methods Mol. Biol. 177, 85–97
Press, San Diego 45. McClain, M. S., Cao, P., Iwamoto, H., Vinion-Dubiel, A. D., Szabo, G., Shao, Z.,
3. Dunn, B. E., Cohen, H., and Blaser, M. J. (1997) Clin. Microbiol. Rev. 10, and Cover, T. L. (2001) J. Bacteriol. 183, 6499 – 6508
720 –741 46. Copass, M., Grandi, G., and Rappuoli, R. (1997) Infect. Immun. 65, 1949 –1952
4. Suerbaum, S., and Michetti, P. (2002) N. Engl. J. Med. 347, 1175–1186 47. de Bernard, M., Papini, E., de Filippis, V., Gottardi, E., Telford, J., Manetti, R.,
5. Cover, T. L., and Blaser, M. J. (1992) J. Biol. Chem. 267, 10570 –10575 Fontana, A., Rappuoli, R., and Montecucco, C. (1995) J. Biol. Chem. 270,
6. Montecucco, C., Papini, E., de Bernard, M., Telford, J. L., and Rappuoli, R. 23937–23940
(1999) in The Comprehensive Sourcebook of Bacterial Protein Toxins (Alouf, 48. McClain, M. S., Schraw, W., Ricci, V., Boquet, P., and Cover, T. L. (2000) Mol.
J. E., and Freer, J. H., eds) pp. 264 –286, Academic Press, San Diego Microbiol. 37, 433– 442
7. Atherton, J. C., Cover, T. L., Papini, E., and Telford, J. L. (2001) in Helico- 49. Cover, T. L., Puryear, W., Pérez-Pérez, G. I., and Blaser, M. J. (1991) Infect.
bacter pylori: Physiology and Genetics (Mobley, H. L. T., Mendz, G. L., and Immun. 59, 1264 –1270
Hazell, S. L., eds) pp. 97–110, American Society for Microbiology, Washing- 50. Fields, S., and Song, O. (1989) Nature 340, 245–246
ton, D. C. 51. James, P. (2001) Methods Mol. Biol. 177, 41– 84
8. Papini, E., Zoratti, M., and Cover, T. L. (2001) Toxicon 39, 1757–1767 52. Ma, J., and Ptashne, M. (1987) Cell 48, 847– 853
9. Figueiredo, C., Machado, J. C., Pharoah, P., Seruca, R., Sousa, S., Carvalho, R., 53. Gimble, F. S., and Sauer, R. T. (1989) J. Mol. Biol. 206, 29 –39
Capelinha, A. F., Quint, W., Caldas, C., van Doorn, L. J., Carneiro, F., and 54. Yahiro, K., Niidome, T., Kimura, M., Hatakeyama, T., Aoyagi, H., Kurazono,
Sobrinho-Simoes, M. (2002) J. Natl. Cancer Inst. 94, 1680 –1687 H., Imagawa, K., Wada, A., Moss, J., and Hirayama, T. (1999) J. Biol.
10. Atherton, J. C., Cao, P., Peek, R. M., Jr., Tummuru, M. K., Blaser, M. J., and Chem. 274, 36693–36699
Cover, T. L. (1995) J. Biol. Chem. 270, 17771–17777 55. McClain, M. S., Cao, P., and Cover, T. L. (2001) Infect. Immun. 69, 1181–1184
11. Telford, J. L., Ghiara, P., Dell’Orco, M., Comanducci, M., Burroni, D., Bugnoli, 56. Boulikas, T. (1993) Crit. Rev. Eukaryotic Gene Expression 3, 193–227
M., Tecce, M. F., Censini, S., Covacci, A., Xiang, Z., Papini, E., Montecucco, 57. Nooren, I. M., and Thornton, J. M. (2003) EMBO J. 22, 3486 –3492
C., Parente, L., and Rappuoli, R. (1994) J. Exp. Med. 179, 1653–1658 58. Gouaux, E. (1997) Curr. Opin. Struct. Biol. 7, 566 –573
12. Fujikawa, A., Shirasaka, D., Yamamoto, S., Ota, H., Yahiro, K., Fukada, M., 59. Lacy, D. B., and Collier, R. J. (2002) Curr. Top. Microbiol. Immunol. 271,
Shintani, T., Wada, A., Aoyama, N., Hirayama, T., Fukamachi, H., and 61– 85
Noda, M. (2003) Nat. Genet. 33, 375–381 60. Bhakdi, S., Bayley, H., Valeva, A., Walev, I., Walker, B., Kehoe, M., and
13. Leunk, R. D., Johnson, P. T., David, B. C., Kraft, W. G., and Morgan, D. R. Palmer, M. (1996) Arch. Microbiol. 165, 73–79
(1988) J. Med. Microbiol. 26, 93–99 61. Parker, M. W., Buckley, J. T., Postma, J. P., Tucker, A. D., Leonard, K., Pattus,
14. Molinari, M., Galli, C., Norais, N., Telford, J. L., Rappuoli, R., Luzio, J. P., and F., and Tsernoglou, D. (1994) Nature 367, 292–295
Montecucco, C. (1997) J. Biol. Chem. 272, 25339 –25344 62. Ramachandran, R., Heuck, A. P., Tweten, R. K., and Johnson, A. E. (2002) Nat.
15. Czajkowsky, D. M., Iwamoto, H., Cover, T. L., and Shao, Z. (1999) Proc. Natl. Struct. Biol. 9, 823– 827
Acad. Sci. U. S. A. 96, 2001–2006 63. Moschioni, M., Tombola, F., de Bernard, M., Coelho, A., Zitzer, A., Zoratti, M.,
16. Szabo, I., Brutsche, S., Tombola, F., Moschioni, M., Satin, B., Telford, J. L., and Montecucco, C. (2002) Cell. Microbiol. 4, 397– 409
Rappuoli, R., Montecucco, C., Papini, E., and Zoratti, M. (1999) EMBO J. 64. van der Goot, F. G., Pattus, F., Wong, K. R., and Buckley, J. T. (1993)
18, 5517–5527 Biochemistry 32, 2636 –2642
17. Schraw, W., Li, Y., McClain, M. S., van der Goot, F. G., and Cover, T. L. (2002) 65. Fivaz, M., Abrami, L., Tsitrin, Y., and van der Goot, F. G. (2001) Toxicon 39,
J. Biol. Chem. 277, 34642–34650 1637–1645
Interactions between p-33 and p-55 Domains of the Helicobacter pylori Vacuolating
Cytotoxin (VacA)
Victor J. Torres, Mark S. McClain and Timothy L. Cover
J. Biol. Chem. 2004, 279:2324-2331.
doi: 10.1074/jbc.M310159200 originally published online October 30, 2003

Access the most updated version of this article at doi: 10.1074/jbc.M310159200

Alerts:
• When this article is cited
• When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

Downloaded from http://www.jbc.org/ by guest on December 22, 2016


This article cites 62 references, 33 of which can be accessed free at
http://www.jbc.org/content/279/3/2324.full.html#ref-list-1

Das könnte Ihnen auch gefallen