You are on page 1of 6

Structural insights into Helicobacter pylori oncoprotein

CagA interaction with β1 integrin


Burcu Kaplan-Türköza,1, Luisa F. Jiménez-Sotob,1, Cyril Dianc, Claudia Ertlb, Han Remautd,e, Arthur Louchea,
Tommaso Tosic, Rainer Haasb,2, and Laurent Terradota,2
a
Centre National de la Recherche Scientifique-Ligue Contre le Cancer, ATIP Avenir Group, Institut de Biologie et Chimie des Protéines, Unité Mixte de
Recherche 5086, Université Lyon, Lyon F-69367, France; bMax von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Department of
Bacteriology, Ludwig-Maximilians-Universität, D-80336 Munich, Germany; cInstitut de Biologie Structurale J. P. Ebel, F-38027 Grenoble Cedex, France;
d
Structural Biology Brussels, Vrije Universiteit Brussels, 1050 Brussels, Belgium; and eStructural and Molecular Microbiology, Vlaams Instituut voor
Biotechnologie (VIB) Department of Structural Biology, VIB, 1050 Brussels, Belgium

Edited* by Scott J. Hultgren, Washington University School of Medicine, St. Louis, MO, and approved July 31, 2012 (received for review April 16, 2012)

Infection with the gastric pathogen Helicobacter pylori is a risk Pro-Ile-Tyr-Ala (EPIYA) motifs that become tyrosine phos-
factor for the development of gastric cancer. Pathogenic strains phorylated by Src and Abl kinases in eukaryotic cells (Fig. 1A).
of H. pylori carry a type IV secretion system (T4SS) responsible Strains of Western origin harbor three different EPIYA phos-
for the injection of the oncoprotein CagA into host cells. H. pylori phorylation motifs, namely A, B, and C (sometimes multiple C
and its cag-T4SS exploit α5β1 integrin as a receptor for CagA trans- motifs) and East Asian strains display A, B, and D motifs (12).
location. Injected CagA localizes to the inner leaflet of the host cell Phosphorylated CagA then binds and activates Src homology
membrane, where it hijacks host cell signaling and induces cyto- 2 domain phosphatase (SHP2) via its SH2 domains, leading to
skeleton reorganization. Here we describe the crystal structure of dephosphorylation and inactivation of Src family kinases, result-
the N-terminal ∼100-kDa subdomain of CagA at 3.6 Å that unveils
ing in morphological transformation and dramatic cytoskeletal
a unique combination of folds. The core domain of the protein
rearrangements in later stages of infection (13). Inactivation of
consists of an extended single-layer β-sheet stabilized by two in-
dependent helical subdomains. The core is followed by a long helix
Src family kinases results in dephosphorylation of vinculin, dis-
that forms a four-helix helical bundle with the C-terminal domain. rupting vinculin interaction with Arp2/3, which leads to de-
Mapping of conserved regions in a set of CagA sequences identi- creased lamellipodia formation and decreased number of focal
fied four conserved surface-exposed patches (CSP1–4), which rep- adhesions. Other CagA effects are phosphorylation-independent.
resent putative hot-spots for protein–protein interactions. The In particular, specific sequences (named MKI) (Fig. 1A) located
proximal part of the single-layer β-sheet, covering CSP4, is in- in the CagA C terminus inhibit Par1b/MARK2 kinase activity
volved in specific binding of CagA to the β1 integrin, as deter- via mimicry of the enzyme’s natural substrate (14). The in-
mined by yeast two-hybrid and in vivo competition assays in H. hibition of the PAR1b/MARK2 perturbs atypical PKC signaling,
pylori cell-culture infection studies. These data provide a structural which results in disruption of tight junctions and loss of cell
basis for the first step of CagA internalization into host cells and polarity (15).
suggest that CagA uses a previously undescribed mechanism to Recent studies have focused on the N-terminal portion of CagA
bind β1 integrin to mediate its own translocation. (residues 1–884, CagA1–884) (Fig. 1A). This domain interacts with
intracellular partners, such as ASPP2 (16), RUNX3 (17), TAK1,
virulence factor | X-ray crystallography | oncogene | pathogenicity | and TRAF6 (18), and thus plays an important role in the de-
gastric ulcer velopment of gastric cancer. Moreover, CagA1–884 contains resi-
dues that bind to the ectodomain of α5β1 integrin, but the

F ifty percent of the world population is infected by Helicobacter


pylori, which causes severe gastric pathologies, including pep-
tic ulceration and gastric cancer (1). The most virulent strains
relevance of this binding for the process of CagA translocation, if
any, was not known (6). Here we present the crystal structure of
CagA1–884, which reveals a unique combination of domains. The
harbor the cag pathogenicity island (cag-PAI), a 40-kb DNA analysis of the structural similarity indicates that the core domain
fragment that encodes the cytotoxin CagA and a type IV secretion resembles the outer surface protein A from Borrelia burgdorferi
system (cag-T4SS). CagA is a unique protein not found in any but the C-terminal helical domain shows some resemblance to
other bacteria besides H. pylori. Transgenic mice expressing the eukaryotic cytoskeleton proteins. A specific single-layer β-sheet
cytotoxin develop gastric polyps and adenocarcinomas, estab- region (SLB) was identified to act as the functional binding do-
lishing CagA as a unique bacterial oncoprotein (2). Upon contact
main of CagA to β1 integrin. We further show that this specific
with the gastric epithelial cells, H. pylori delivers CagA via the cag-
T4SS, a needle-like molecular appendage spanning both bacterial interaction with β1 integrin is essential for the process of CagA
membranes prolonged by a large extracellular pilus (3, 4). Several translocation.
proteins, such as CagL, CagY, and CagA, decorate the H. pylori
cag-T4SS pilus and use α5β1 integrin at focal adhesions as a re-
Author contributions: B.K.-T., L.F.J.-S., R.H., and L.T. designed research; B.K.-T., L.F.J.-S., C.D.,
ceptor to target CagA delivery (5–7). However, the mechanism of C.E., H.R., A.L., and L.T. performed research; T.T. contributed new reagents/analytic tools;
CagA translocation across the eukaryotic membrane via the cag- B.K.-T., L.F.J.-S., C.E., H.R., and R.H. analyzed data; and B.K.-T., L.F.J.-S., H.R., R.H., and L.T.
T4SS conduit has not been established. wrote the paper.
Once injected, CagA localizes at the inner leaflet of the plasma The authors declare no conflict of interest.
membrane at cell-adhesion junctions, where it activates a multi- *This Direct Submission article had a prearranged editor.
faceted attack on host cell signaling. At adherens junctions, CagA Data deposition: The atomic coordinates and structure factors have been deposited in the
forms a complex with cytoskeleton proteins, in particular with E- Protein Data Bank, www.pdb.org (PDB ID code 4GOH).
cadherin (8, 9). Based on high-throughput screening for soluble 1
B.K.-T. and L.F.J.-S. contributed equally to this work.
fragments (10) and on in vivo proteolysis data, CagA can be de- 2
To whom correspondence may be addressed. E-mail: haas@mvp.uni-muenchen.de or
scribed as a protein consisting of two functional domains (11). laurent.terradot@ibcp.fr.
Much attention has been paid to the role of the C-terminal do- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
main (residues 885–1,186). Indeed, this segment bears three Glu- 1073/pnas.1206098109/-/DCSupplemental.

14640–14645 | PNAS | September 4, 2012 | vol. 109 | no. 36 www.pnas.org/cgi/doi/10.1073/pnas.1206098109


Fig. 1. Overall structure of CagA N-terminal do-
main. (A) Schematic representation of CagA (Western
strain 26695). Boxes indicate the domain definition
derived from the CagA1–884 X-ray structure, with D1
colored in red, the SLB in pink, subdomain D2′ in
cyan, D2′′ in blue, D3 in green, and D4 in yellow.
The positions of the EPIYA motifs are indicated by
A, B, and C and those of PAR1-MARK kinase binding
motifs (14) by MKI1 and MKI2. Bars indicate the
positions of the protein fragments used in the study.
(B) Ribbon representation of the structure of
CagA1–884 colored according to A. (C) Topology di-
agram of CagA1–884 structure.

Results spanning residues 305–642 and containing three subdomains. The


Overall Structure of CagA1–884. To gain structural insight into the major part of D2 consists of 11 antiparallel strands (β1–β5 and β8–
mode of action of CagA, we solved the crystal structure of CagA1–884 β13) forming a SLB that has a left-hand twist and is curved at
to 3.6Å (Rfactor/Rfree of 0.32/0.34). The protein crystallized in the strand β8 (Fig. 1B). Interestingly, this SLB is structurally similar to
space group P41212 with one molecule per asymmetric unit. that of the outer surface protein OspA from B. burgdorferi (19)
CagA1–884 has an elongated overall structure consisting of four (DALI Z-score: 5.3) (Fig. S3). The homology region is limited to
domains arranged in a “crescent moon” shape (Fig. 1 B and C, and strands β2–β5 and β8–β11 of CagA that superimpose well to
Fig. S1). Even though seven α-helices are clearly visible (Fig. 1B), strands β6–β13 of OspA (Fig. S3). The resulting structure-based
the N-terminal domain D1 was not included in the refined model sequence comparison indicates that the first four strands are the
because poor quality of the electron density map in this area of the most similar. As a consequence, the surface properties of the
protein prevented unambiguous model building and assignment proximal parts of the two SLBs have comparable hydrophobic
of the sequence. A linker region follows D1, which we estimate to properties (Fig. S3).
be between residues 270 and 300, based on domain topology and A compact subdomain (D2′), formed by a β-hairpin (β6, β7)
limited proteolysis (SI Materials and Methods and Fig. S2). The and helices α8 to α10, is inserted between strand β5 and β8 and
second domain, D2, forms the central core of CagA1–884 structure, buttresses the lower half of the β-sheet (Fig. 1 B and C, and Fig.

Fig. 2. Structure-based sequence alignment of


CagA1–884. Sequences of CagA from strain 26695
(46), V225 (47), HP-No20, and HP-No31 (48), were
selected from the multiple alignment of 80 sequen-
ces generated by CONSURF (49) to illustrate CagA
diversity. Sequence numbering corresponds to CagA
from strain 26695. Conserved and invariant residues
(among 80 CagA sequences) exposed on the surface
of CagA1–884 crystal structure are shaded in magenta
and red, respectively. The exact sequence of strand
β1 could not be determined; the mostly likely can-
MICROBIOLOGY

didate sequence is indicated in black. Secondary


structure elements are indicated above the sequen-
ces and colored according to the domain: D2′, cyan;
SLB, pink; D2′′, purple; D3, green; and D4, yellow.
Blue dots are positioned below residues R624 and
R626, involved in PS binding (25).

Kaplan-Türköz et al. PNAS | September 4, 2012 | vol. 109 | no. 36 | 14641


S1). Finally, the SLB is followed by a helical domain (D2′′) residues R624 and R626, which were previously found to interact
consisting of the short helix α11 and two helices (α12 and α13) with phosphatidylserine (PS) (25) (Fig. 3). The structure reveals
that adopt a hairpin structure that packs against the convex face that additional basic residues of D2′′, such as K613, K614, K617,
of the SLB and stabilizes the upper half of the β-sheet (Figs. 1B and K621 might also contribute to PS binding (Fig. 3B). The
and 2). D3 is a 90 Å-long kinked amphipathic helix (α14) that fourth conserved area CSP4 is discussed below.
protrudes away from the core. The N-terminal moiety of α14
engages into a hydrophobic pocket formed by helices α11, α12, Interaction Between CagA SLB and β1 Integrin Is Required for
and α13 of domain D2′′ (Figs. 1B and 2, and Fig. S1). The car- Translocation. The N-terminal half of the CagA protein (amino
boxyl-terminal part of α14 completes an antiparallel four-helix acids 1–612, CagA1–612) containing domains D1 and most of D2
bundle with α15, α16, and α17 of D4. Helix α17 projects away has previously been shown to interact with α5β1 integrin (6). The
from the bundle and forms a loose hairpin with α18. binding of α5β1 integrin by CagA is believed to be a prerequisite
for CagA internalization into the host cells. To further narrow
Conserved Surface-Located Residues Within CagA Point Toward Four down the β1 integrin-binding domain within CagA1–612, we per-
Functional Areas. Mapping of the surface-exposed invariant and formed a yeast two-hybrid (YTH) protein-interaction screen.
conserved residues to CagA1–884 structure reveals a clustering into The CagA1–612 insert in the YTH bait vector was systematically
four main conserved surface-exposed patches (CSP1–4) (Figs. 2 shortened from both ends to generate eight subclones (Y31–
and 3A). CSP1 is located in helix α18 and the C-terminal portion Y38), producing recombinant CagA proteins differing in length
of helix α17. These helices self-interact with a symmetry-related by 100 aa each (Fig. 4A). The YTH screen revealed interaction
subunit in a head-to-tail manner (buried surface of 543Å2) (Fig. of β1 integrin with three CagA subclones (Y31, Y35, Y36), all of
S4A). CagA was found to form multimers via the MKI sequences which shared the CagA303–404 amino acid sequence (Fig. 4A). In
(also called CagA multimerization sequence-CM) and this mul- the crystal structure, these residues correspond to the first five
timerization increase CagA activity on the SHP2 phosphatase (20, strands of the SLB and part of the D2′ domain (Fig. 1B). This
21). Because the MKI (CM) sequences are not present in the area also coincides with the fourth conserved surface patch,
CagA1–884 fragment, the oligomeric state of CagA1–884 was inves- CSP4, found in the CagA structure (Figs. 2 and 3A).
tigated. Size-exclusion chromatography coupled to multiangle light To corroborate the specific interaction of this region with
scattering shows that CagA1–884 is a monomer in solution (Fig. β1 integrin, competition experiments were performed using
S4B), suggesting that the observed dimerization is likely a result recombinant CagA303–404 (Fig. 4 A and B). CagA303–404 identi-
of crystal packing. However, given the position and conservation fied in the YTH screen was expected to compete with native
of the residues present at this interface, this region might rep- CagA binding of H. pylori and eventually to interfere with CagA
resent a dimerization zone in the context of the full-length CagA translocation into gastric adenocarcinoma (AGS) cells. The cag-
or form a possible hot spot for protein–protein interactions. PAI+ H. pylori strain P12 translocates CagA into AGS cells, as
A second conserved surface patch, CSP2, is located in the distal verified by its EPIYA motif tyrosine phosphorylation (Fig. 4B,
part of the helical bundle formed by the carboxyl-terminal portion lane 1). Using the read-out of CagA phosphorylation, the
of α14, α15, α16, and α17 (Fig. 3A). This left-handed helical purified CagA303–404 strongly inhibited CagA translocation in a
bundle shows similarities with the F-actin binding domain of the dose-dependent manner (Fig. 4B). However, the CagA303–404
Bcr-Abl tyrosine kinase (rmsd 2.4), but also with cytoskeleton fragment was not very stable in solution, likely because it encom-
proteins such as α-catenin (rmsd 2.7), vinculin (rmsd 2.5), and the passs only part of the D2′ domain and thus lacked structural in-
focal adhesion kinase (FAK, rmsd 2.2) (Fig. S5). A remarkable tegrity. To determine whether the SLB or D2′ were involved in β1
and key property of CagA is that it can modify several types of integrin binding, we generated and purified CagA recombinant
cell-to cell junctions (22). In particular, CagA colocalizes with and fragments based on the crystal structure (Fig. 1A) (CagA303–456,
affects FAK and vinculin phosphorylation at FAs (7, 23, 24). CagA369–448) and competition experiments were performed (Fig.
CagA also disrupts the E-cadherin/β-catenin complex at adherens 4C). Binding to AGS cells was detected for all these proteins, but
junctions (8, 9). The structural similarity and sequence conser- not with the same affinity (Fig. 4C) (α-His). Although the same
vation suggests that the CagA helical bundle might be important amount of proteins were incubated, CagA1–273, CagA1–291 (cov-
for interaction of CagA, with some of these membrane proteins ering D1), CagA303–456 (proximal part of the SLB and D2′), and
possibly by mimicry of the F-actin binding domain motif. CagA392–733 (containing the distal part of the SLB and D2′′)
A third area of interest, CSP3, consists of a patch of basic were found to interact more strongly with AGS cells than
residues belonging to α13 of the D2′′ domain. CSP3 includes CagA369–448 (containing only D2′). The binding of the fragment
CagA392–733 on AGS cells might be attributed to the presence of
the PS-binding patch at its surface, because PS is externalized
during H. pylori infection (25). The CagA translocation assay
shows that the purified CagA303–456 fragment inhibited CagA
translocation by H. pylori (Fig. 4C). In the same assay, neither
CagA1–273, CagA1–291, CagA369–448, nor CagA392–733 showed any
inhibitory effect on CagA translocation (Fig. 4C). These results
indicate that the first five strands of the SLB (residues 303–368)
are sufficient to inhibit CagA translocation in our assay. Recom-
binant Yersinia invasin (GST-Inv397), which binds β1 integrin via
a RGD (arginyl-glycyl-aspartic acid)-like motif, did not interfere
either with CagA translocation (Fig. 4C). Taken together with the
YTH interaction data, these functional assays show that the
proximal part of the SLB of CagA binds to β1 integrin and that
Fig. 3. Potential functional sites in CagA structure. (A) Semitransparent
this interaction is an essential step of CagA translocation.
surface representation of the CagA domain D3-D4 (Left) and D2 (Right)
colored in gray with strictly conserved and conserved residues colored in red
Discussion
and pink, respectively. The location of the conserved surface exposed patch
“CSP1–4” are indicated. (B) Surface representation of α12 and α13 colored by CagA is a remarkably versatile oncoprotein produced by H.
electrostatic potential. Residues forming the basic patch potentially involved pylori, known to interact with a plethora of host signaling fac-
in PS binding are indicated. tors and to promote gastric cancer development. The structure

14642 | www.pnas.org/cgi/doi/10.1073/pnas.1206098109 Kaplan-Türköz et al.


Fig. 4. Mapping the CagA-β1 integrin interaction
domains. (A) Schematic representation of CagA frag-
ments interacting with β1 integrin in a YTH screen.
Construction of systematically shortened CagA1–612
clones (Y31–Y38) in the bait vector (gray represents
deleted sequences). The green square denotes the CagA
amino acid sequence essential for interaction of CagA
with β1 integrin. Clones Y32 and Y34 grew on selective
medium without interaction partner (autoactivation).
(B) Recombinant CagA303–404 interferes with CagA
translocation. AGS cells were incubated with 4, 8, or 12
nMol purified CagA303–404 (lanes 2–4) and infected with
H. pylori P12 or not (lane 5). Lysates were run on SDS/
PAGE, blotted, and reacted with antibodies as in-
dicated. (C) AGS cells were incubated with purified
CagA fragments (His-tagged; see Fig. 1B for their exact
location) or recombinant Yersinia invasin (GST-Inv397),
and the structure-derived fragments CagA303–456 or
CagA369–448 and infected with H. pylori P12 strain.
Lysates were run on SDS/PAGE or single gel system,
blotted, and reacted with antibodies, as indicated. (D)
Ribbon diagram of CagA1–884 domain D2 (gray) showing
the β1 integrin interacting portion (magenta). (E) Sur-
face representation of the structure in the same orien-
tation colored according to the electrostatic potential.

of CagA1–884 presented here provides a clear definition of the the RGD motif of fibronectin to bind to β1 integrin (31). CagA
domain organization of the large, stable N-terminal region of does not contain a RGD motif, meaning that CagA binding to β1
CagA, covering about two-thirds of the whole CagA protein. integrin is via a different, so far unexplored mechanism, probably
Some of these domains share different degrees of similarities by binding to a different domain of β1 compared with invasin.
with prokaryotic or eukaryotic proteins, the aim of which might Another H. pylori protein, CagL, was originally reported to bind
be protein mimicry, which could explain how different domains to β1 integrin via a RGD motif (7), whereas other reports showed
of CagA can affect location of the protein in H. pylori as well as a RGD-independent binding of CagL to β1 integrin (6). However,
the eukaryotic target cell (26–29). Another remarkable char- because invasin did not impair CagA translocation, our work
acteristic of CagA N-terminal region is its flexibility, in par- indicates that the RGD binding motif of β1 integrin is not re-
ticular the domain D1 (residues 1–300). Previously, most of the quired for CagA translocation, suggesting that CagA binds to
C-terminal region of CagA was found disordered, even when a different region within β1 integrin.
bound to the Par1b/MARK2 kinase (14). Here we found that The competitive binding of the SLB fragments to β1 integrin
domain D1, as well a number of loops of D2, D3, and D4, are heterodimers on the surface of gastric AGS cells suggests that it
highly flexible. This flexibility could be attenuated in the functionally interferes with binding of full-length CagA during H.
context of the full-length protein, because the N terminus was pylori infection. Which domain of β1 integrin is bound by CagA is
found to interact with the C-terminal domain (28). Neverthe- presently unknown. The SLB of CagA, in particular the β1-
less, both in vitro and in vivo proteolysis studies showed integrin binding domain, revealed an interesting structural sim-
that the protein is rather unstable and heavily prone to deg- ilarity with the outer surface protein OspA of B. burgdorferi.
radation (10, 11). This intrinsic flexibility might play an im- These bacteria cause Lyme disease and use outer surface pro-
portant role for CagA to accommodate interactions with teins to adhere to human cells once transmitted by tick bites
multiple partners. (reviewed in ref. 32). Although an interaction of OspA with β1-
Mapping of conserved residues within the CagA structure integrin receptors has not been described, a direct interaction of
reveals four conserved surface patches (CSP1–4) that represent OspA and OspB with the complement receptor 3 (CR3) has
candidate hot spots within CagA for the interaction with host-cell been reported (33). CR3 is a αMβ2 integrin heterodimer spe-
molecules. CSP3 contains residues that were shown to be involved cifically expressed by immune cells, such as neutrophils, dendritic
in the binding to PS. The binding of CagA CSP3 might contribute cells, and B or T cells, and is required for phagocytosis of B.
to tether CagA at the inner leaflet membrane, where PS is more burgdorferi into human monocytes (34). Therefore, it might be
abundant (25). Another important area is CSP4, which was shown speculated that OspA could interact with β2 integrins via its SLB,
to constitute the binding interface with α5β1 integrin. We provide similarly to the way CagA interacts with β1 integrin. Whether
clear evidence that this interaction is required for internalization CagA binds β2 integrins through this conserved surfaced as well
of CagA into the host cell. In the CagA structure, these residues has yet to be determined.
are part of the SLB, but also of the β6β7 hairpin (Fig. 4D) and Why does CagA, which is the translocated effector protein and
form a large hydrophobic cleft that represents a large interaction not necessarily a structural component of the cag-T4SS, interact
interface for integrin (Fig. 4E). Previous quantification of the with the integrin receptor? The question whether the CagA/β1
CagA/integrin binding by plasmon resonance studies indicated integrin interaction is an epiphenomenon or is essential for
that α5β1 integrin binds CagA approximately 100-fold stronger CagA translocation cannot be studied by gene deletion, because
than its natural ligand, fibronectin (KD of 0.15 nM vs. 15 nM, CagA is the only effector protein of the cag-T4SS. However, the
respectively) (6). In this respect, we note that recombinant Yer- effective blocking of CagA translocation by CagA SLB fragments
MICROBIOLOGY

sinia invasin (GST-Inv397) did not interfere with CagA trans- provided by our study clearly proves that the specific interaction
location (Fig. 4C). Invasin is a well-known bacterial virulence of CagA with the integrin receptor is an essential step for the
factor that binds with high affinity to α5β1 integrin to mediate translocation process of CagA into the host cell. A complete
efficient and rapid internalization of the bacteria into host cells, picture about the intricate interaction of the cag-T4SS and the
especially M cells in the gut (30). The structure of invasin mimics β1-integrin receptor will probably be obtained when the precise

Kaplan-Türköz et al. PNAS | September 4, 2012 | vol. 109 | no. 36 | 14643


binding domains within the integrin, as well as the binding of the Table 2. Phasing and refinement statistics
other components, CagI, CagL, and CagY to the integrin re- Phasing and refinement statistics
ceptor have been determined. Besides being important for
translocation, the CagA N-terminal domain also interacts with Phasing
intracellular partners such as ASPP2 (16), RUNX3 (17), TAK1, No. of sites 8
and TRAF6 (18). The structure of CagA1–884 we describe here Phasing power 1.2
will enable a more in-depth functional characterization of CagA Figure of merit (FOM) 0.27
interactions with these proteins to understand how it promotes FOMdm (59% solvent) 0.57
carcinogenesis, and marks an important first step toward the Refinement
development of specific inhibitors. Resolution range for refinement (Å) 62.7–3.6
No. of unique reflections in refinement 13,349
Materials and Methods Working set 12,679
Cloning, Mutagenesis, Expression, and Purification of CagA1–884, Se-CagA1–884, Test set 670
and Se-CagA1–884 Mutants. The DNA fragment encoding amino acid residues Completeness (%) 91.8
1–884 of CagA (CagA1–884) from H. pylori strain 26695 was cloned into the Monomers/ASU 1
pET101D topo vector (Invitrogen). Single-point mutations (L699M and No. of protein atoms 3,283
L727M) were introduced using the site-directed mutagenesis kit (Stra- Rwork/Rfree 0.3287/0.3413
tagene). BL21 Star (DE3) cells carrying the resulting plasmids were grown
Rmsd bonds (Å) 0.002
in LB media containing ampicillin at 37 °C until an OD600 of 0.7 and protein
Rmsd angles (°) 0.593
expression was induced by the addition of 1 mM isfopropyl-β-D-thio-
galactopyranoside and cells were grown overnight at 20 °C. Briefly, proteins Mol Probity score 2.2
were purified using immobilized metal affinity chromatography, anion ex- Ramachandran plot
change, and gel-filtration chromatography. Selenomethionine-substituted Most favored (%) 95.8
CagA1–884 was obtained following the protocol previously described (35). Allowed (%) and generously allowed (%) 4.0
Se-CagA1–884, Se-CagA1–884 L699M, and Se-CagA1–884 L727M mutants were Outliers (%) 0.2
produced and purified as native except that 1 mM DTT was added in
all buffers.

Crystallization, Structure Determination, and Refinement. CagA1–884, Se- 59% (37). Phasing statistics are given in Table 2. The experimental map
CagA1–884, Se-CagA1–884 L699M, and Se-CagA1–884 L727M crystals were was of excellent quality (Fig. S6) and the model was built using COOT
obtained by using the vapor-diffusion methods in hanging drops. One mi- (40). The atomic positions and TLS (translation, libration, screw)
croliter of protein was mixed with 1.0 μL of reservoir solution containing parameters refined using BUSTER (41) and PHENIX (42) (Tables 1 and 2). The
ethanol 10–14%, (vol/vol), 100 mM sodium cacodylate pH 6–7, and sodium final model includes residues 319–343, 349–478, 489–510, 536–641, 644–
iodide 15 mM. Crystals of 200 μm × 200 μm × 70 μm appeared in 3 to 4 d at 706, 718–736, 738–758, and 764–822. The structure has been deposited in
20 °C and were grown during 2 to 3 wk. Crystals were flash-frozen in liquid the Protein Data Bank (PDB ID code: 4GOH). Figures in the article were
nitrogen after step-wise addition of cryoprotectant solution consisting of generated using Pymol (43).
20% (vol/vol) ethanol, 100 mM sodium cacodylate, 15 mM sodium iodide,
and 18% ethylene glycol. X-ray diffraction data were collected from native Bacterial Strains, Cell Lines, and Culture Conditions. The H. pylori cag-patho-
and selenomethionine derivative crystals at the ID29 beamline of the Eu- genicity island-positive strain P12 was used for infection experiments (44).
ropean Synchrotron Radiation Facility (Grenoble) or at SOLEIL (CagA1–884 H. pylori was grown on GC agar plates (Difco), as described previously (44).
mutants). Crystals belonged to the tetragonal space group P41212 with unit The human gastric cell line AGS (CRL-1739) was used for infection experi-
cell dimension of a = b = 98 Å and c = 244 Å, and diffracted to a resolution ments. Cells were grown in RPMI containing 10% FCS, at 5% CO2, and 37 °C
of 3.6 Å (Native) and 3.9 Å (SAD) (Tables 1 and 2). The diffraction data were and subcultured every 2–3 d.
indexed and integrated using XDS (36) and scaled with SCALA from the CCP4
program suite (37). Data collection statistics are given in Table 1. The YTH Assay. The Invitrogen system consists of the entry vector pDONR207, and
structure of CagA1–884 was solved by the SAD method using selenomethio- destination vectors pGADT7 (prey vector) and pGBKT7 (bait vector). YTH bait
nine derivative data. The position of Se atoms was determined with SHELXD and prey libraries were generated comprising gene sequences covering the
(38), phases calculated with the program SHARP (39), and density modifi- complete external β1-integrin region. For plasmid cloning, the recombina-
cation was performed using SOLOMON and DM with a solvent content of tory cloning procedure, as described by the manufacturer, was used. Bait
and prey plasmids were transformed into the haploid Saccharomyces cer-
evisiae strains Y187 and AH109. Diploid yeast cells were selected after
Table 1. Data collection statistics mating and selection on SD medium lacking tryptophan (Trp) and leucine
(Leu), thus generating all possible combinations of bait and prey plasmids.
Data collection Se-Met Native
After growth on SD-Leu/Trp medium, yeast colonies were transferred to SD-
Space group P41212 P41212 Leu/Trp/His medium to select for interactions. Growth after 3–6 d indicated
Wavelength (Å) 0.97903 0.97627 bait–prey interactions. Additionally, the stringency of this screen was en-
hanced by selection on SD-Leu/Trp/His medium containing the competitive
Cell parameters, 98.2, 98.2, 243.15 97.8, 97.8, 245.4
inhibitor 3-aminotriazole (5 mM).
a, b, c (Å)
Resolution 98.2–3.90 (4.11–3.90) 90.87–3.60 (3.79–3.60)
Antibodies and Reagents. Antibodies against phosphotyrosine were obtained
range (Å) from Santa Cruz (PY99) or Upstate (4G10), and anti-His antibody from
Rmerge 0.07 (0.70) 0.07 (0.85) Genescript. Polyclonal HRP and alkaline phosphatase-conjugated anti-mouse
Rp.i.m. 0.044 (0.44) 0.059 (0.68) IgG and anti-rabbit IgG antisera, HRP-conjugated streptavidin, protease
No. of observed 72,769 (10,315) 46,117 (7,097) inhibitors PMSF, leupeptin, and pepstatin were obtained from Sigma. AK257
reflections and AK268 are rabbit polyclonal antisera against C-terminal or N-terminal
No. of unique 11,530 (1,638) 13,408 (1,974) CagA, respectively, actin-HRP and mouse anti-HIS (GenScript), Integrin-β1
reflections clone LM534 (Chemicon), anti-GST (Sigma).
Mean [I/s(I)] 10.1 (2.2) 6.2 (1.4)
Completeness (%) 99.9 (99.9) 92.8 (96.4) CagA Translocation Assay (Tyrosine Phosphorylation of CagA). Gastric AGS cells
Multiplicity 6.3 (6.3) 3.4 (3.6) were infected with H. pylori at 70–90% confluency with a multiplicity of
infection of 60. For competitive inhibition experiments, recombinant purified
Values in parentheses refer to the indicated resolution shell. Se-Met, sele- CagA proteins (11–19 nmol/1 × 106 cells), or recombinant Yersinia invasin
nomethionine. known to bind to β1 integrin (GST-Inv397, 50 μg/1 × 106 cells, control), were

14644 | www.pnas.org/cgi/doi/10.1073/pnas.1206098109 Kaplan-Türköz et al.


added to the cells (4 °C, 30 min) before the infection with H. pylori. After 3 h ACKNOWLEDGMENTS. We thank Dr. W. Fischer for comments on the
of infection, cells were harvested in PBS with protease inhibitors (pepstatin manuscript; M. P. Candusso for technical assistance for protein purification;
1 μM, leupeptin 1 μM, PMSF 1 mM) and phosphatase inhibitor sodium van- staff members of the European Synchrotron Radiation Facility and SOLEIL
adate (1 mM). Harvested cells were centrifuged (500 × g, 5 min, 4 °C) and beamlines; and acknowledge the use of the UMS3444 crystallogenesis and
centre commun de microanalyse des protéines. This work was funded by the
pellets lysed in RIPA buffer with protease and phosphatase inhibitors and
ATIP-Avenir Program from Centre National de la Recherche Scientifique and
DNase I for later SDS/PAGE or single-gel system (45) in nonreducing, denaturing Ligue Contre le Cancer and by Deutsche Forschungsgemeinschaft SFB914
conditions, and immunoblot analysis. See SI Materials and Methods for further Project B5 and HA 2697/15-1 (to R.H.); and the Odysseus Program from the
details of methods used, and Fig. S7 for the structures of CagA1–884 mutants Fonds Wetenschappelijk Onderzoek–Flanders and Vrije Universiteit Brussels
contoured with the anomalous difference Fourrier electron density maps. Project Grant PRJ9 (to H.R.).

1. Peek RM, Jr., Blaser MJ (2002) Helicobacter pylori and gastrointestinal tract ad- 25. Murata-Kamiya N, Kikuchi K, Hayashi T, Higashi H, Hatakeyama M (2010) Heli-
enocarcinomas. Nat Rev Cancer 2:28–37. cobacter pylori exploits host membrane phosphatidylserine for delivery, localization,
2. Ohnishi N, et al. (2008) Transgenic expression of Helicobacter pylori CagA induces and pathophysiological action of the CagA oncoprotein. Cell Host Microbe 7:399–411.
gastrointestinal and hematopoietic neoplasms in mouse. Proc Natl Acad Sci USA 105: 26. Pelz C, Steininger S, Weiss C, Coscia F, Vogelmann R (2011) A novel inhibitory domain
1003–1008. of Helicobacter pylori protein CagA reduces CagA effects on host cell biology. J Biol
3. Odenbreit S, et al. (2000) Translocation of Helicobacter pylori CagA into gastric epi- Chem 286:8999–9008.
thelial cells by type IV secretion. Science 287:1497–1500. 27. Higashi H, et al. (2005) EPIYA motif is a membrane-targeting signal of Helicobacter
4. Rohde M, Püls J, Buhrdorf R, Fischer W, Haas R (2003) A novel sheathed surface or- pylori virulence factor CagA in mammalian cells. J Biol Chem 280:23130–23137.
ganelle of the Helicobacter pylori Cag type IV secretion system. Mol Microbiol 49: 28. Bagnoli F, Buti L, Tompkins L, Covacci A, Amieva MR (2005) Helicobacter pylori CagA
219–234. induces a transition from polarized to invasive phenotypes in MDCK cells. Proc Natl
5. Terradot L, Waksman G (2011) Architecture of the Helicobacter pylori Cag-type IV Acad Sci USA 102:16339–16344.
29. Couturier MR, Tasca E, Montecucco C, Stein M (2006) Interaction with CagF is required
secretion system. FEBS J 278:1213–1222.
for translocation of CagA into the host via the Helicobacter pylori type IV secretion
6. Jiménez-Soto LF, et al. (2009) Helicobacter pylori type IV secretion apparatus exploits
system. Infect Immun 74:273–281.
beta1 integrin in a novel RGD-independent manner. PLoS Pathog 5:e1000684.
30. Isberg RR, Leong JM (1990) Multiple beta 1 chain integrins are receptors for invasin,
7. Kwok T, et al. (2007) Helicobacter exploits integrin for type IV secretion and kinase
a protein that promotes bacterial penetration into mammalian cells. Cell 60:861–871.
activation. Nature 449:862–866.
31. Hamburger ZA, Brown MS, Isberg RR, Bjorkman PJ (1999) Crystal structure of invasin:
8. Oliveira MJ, et al. (2009) CagA associates with c-Met, E-cadherin, and p120-catenin in
A bacterial integrin-binding protein. Science 286:291–295.
a multiproteic complex that suppresses Helicobacter pylori-induced cell-invasive
32. Radolf JD, Caimano MJ, Stevenson B, Hu LT (2012) Of ticks, mice and men: Un-
phenotype. J Infect Dis 200:745–755.
derstanding the dual-host lifestyle of Lyme disease spirochaetes. Nat Rev Microbiol
9. Murata-Kamiya N, et al. (2007) Helicobacter pylori CagA interacts with E-cadherin and
10:87–99.
deregulates the beta-catenin signal that promotes intestinal transdifferentiation in 33. Garcia RC, Murgia R, Cinco M (2005) Complement receptor 3 binds the Borrelia
gastric epithelial cells. Oncogene 26:4617–4626. burgdorferi outer surface proteins OspA and OspB in an iC3b-independent manner.
10. Angelini A, et al. (2009) Expression of Helicobacter pylori CagA domains by library- Infect Immun 73:6138–6142.
based construct screening. FEBS J 276:816–824. 34. Hawley KL, et al. (2012) CD14 cooperates with complement receptor 3 to mediate
11. Odenbreit S, Gebert B, Püls J, Fischer W, Haas R (2001) Interaction of Helicobacter MyD88-independent phagocytosis of Borrelia burgdorferi. Proc Natl Acad Sci USA
pylori with professional phagocytes: role of the cag pathogenicity island and trans- 109:1228–1232.
location, phosphorylation and processing of CagA. Cell Microbiol 3:21–31. 35. Van Duyne GD, Standaert RF, Karplus PA, Schreiber SL, Clardy J (1993) Atomic
12. Higashi H, et al. (2002) Biological activity of the Helicobacter pylori virulence factor structures of the human immunophilin FKBP-12 complexes with FK506 and rapamy-
CagA is determined by variation in the tyrosine phosphorylation sites. Proc Natl Acad cin. J Mol Biol 229:105–124.
Sci USA 99:14428–14433. 36. Kabsch W (1993) Automatic processing of rotation diffraction data from crystals of
13. Hatakeyama M (2004) Oncogenic mechanisms of the Helicobacter pylori CagA pro- initially unknown symmetry and cell constants. J Appl Cryst 26:795–800.
tein. Nat Rev Cancer 4:688–694. 37. Collaborative Computational Project, Number 4 (1994) The CCP4 suite: Programs for
14. Nesic D, et al. (2010) Helicobacter pylori CagA inhibits PAR1-MARK family kinases by protein crystallography. Acta Crystallogr D Biol Crystallogr 50:760–763.
mimicking host substrates. Nat Struct Mol Biol 17:130–132. 38. Schneider TR, Sheldrick GM (2002) Substructure solution with SHELXD. Acta Crys-
15. Saadat I, et al. (2007) Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt tallogr D Biol Crystallogr 58(Pt 10 Pt 2):1772–1779.
epithelial cell polarity. Nature 447:330–333. 39. de La Fortelle E, Bricogne G (1997) Maximum-likelihood heavy-atom parameter re-
16. Buti L, et al. (2011) Helicobacter pylori cytotoxin-associated gene A (CagA) subverts finement in the MIR and MAD methods. Methods Enzymol 276:472–494.
the apoptosis-stimulating protein of p53 (ASPP2) tumor suppressor pathway of the 40. Emsley P, Cowtan K (2004) Coot: Model-building tools for molecular graphics. Acta
host. Proc Natl Acad Sci USA 108:9238–9243. Crystallogr D Biol Crystallogr 60(Pt 12 Pt 1):2126–2132.
17. Tsang YH, et al. (2010) Helicobacter pylori CagA targets gastric tumor suppressor 41. Blanc E, et al. (2004) Refinement of severely incomplete structures with maxi-
RUNX3 for proteasome-mediated degradation. Oncogene 29:5643–5650. mum likelihood in BUSTER-TNT. Acta Crystallogr D Biol Crystallogr 60(Pt 12 Pt 1):
18. Lamb A, et al. (2009) Helicobacter pylori CagA activates NF-kappaB by targeting TAK1 2210–2221.
42. Adams PD, et al. (2002) PHENIX: Building new software for automated crystal-
for TRAF6-mediated Lys 63 ubiquitination. EMBO Rep 10:1242–1249.
lographic structure determination. Acta Crystallogr D Biol Crystallogr 58(Pt 11):
19. Li H, Dunn JJ, Luft BJ, Lawson CL (1997) Crystal structure of Lyme disease antigen
1948–1954.
outer surface protein A complexed with an Fab. Proc Natl Acad Sci USA 94:3584–3589.
43. DeLano WL (2002) The PyMOL Molecular Graphics System. Available at http://www.
20. Ren S, Higashi H, Lu H, Azuma T, Hatakeyama M (2006) Structural basis and functional
pymol.org.
consequence of Helicobacter pylori CagA multimerization in cells. J Biol Chem 281:
44. Fischer W, et al. (2010) Strain-specific genes of Helicobacter pylori: Genome evolution
32344–32352.
driven by a novel type IV secretion system and genomic island transfer. Nucleic Acids
21. Nagase L, Murata-Kamiya N, Hatakeyama M (2011) Potentiation of Helicobacter py-
Res 38:6089–6101.
lori CagA protein virulence through homodimerization. J Biol Chem 286:
45. Ahn T, Yim SK, Choi HI, Yun CH (2001) Polyacrylamide gel electrophoresis without
33622–33631. a stacking gel: Use of amino acids as electrolytes. Anal Biochem 291:300–303.
22. Wessler S, Gimona M, Rieder G (2011) Regulation of the actin cytoskeleton in Heli- 46. Tomb JF, et al. (1997) The complete genome sequence of the gastric pathogen Hel-
cobacter pylori-induced migration and invasive growth of gastric epithelial cells. Cell icobacter pylori. Nature 388:539–547.
Commun Signal 9:27. 47. Olbermann P, et al. (2010) A global overview of the genetic and functional diversity in
23. Moese S, et al. (2007) The Helicobacter pylori CagA protein disrupts matrix adhesion the Helicobacter pylori cag pathogenicity island. PLoS Genet 6:e1001069.
of gastric epithelial cells by dephosphorylation of vinculin. Cell Microbiol 9: 48. Truong BX, et al. (2009) Diverse characteristics of the CagA gene of Helicobacter
1148–1161. pylori strains collected from patients from southern vietnam with gastric cancer and
24. Tsutsumi R, Takahashi A, Azuma T, Higashi H, Hatakeyama M (2006) Focal adhesion peptic ulcer. J Clin Microbiol 47:4021–4028.
kinase is a substrate and downstream effector of SHP-2 complexed with Helicobacter 49. Glaser F, et al. (2003) ConSurf: Identification of functional regions in proteins by
pylori CagA. Mol Cell Biol 26:261–276. surface-mapping of phylogenetic information. Bioinformatics 19:163–164.
MICROBIOLOGY

Kaplan-Türköz et al. PNAS | September 4, 2012 | vol. 109 | no. 36 | 14645

You might also like