Structure Article

A Helical RGD Motif Promoting Cell Adhesion: Crystal Structures of the Helicobacter pylori Type IV Secretion System Pilus Protein CagL

Stephan Barden,1 Stefanie Lange,1 Nicole Tegtmeyer,3,4 Jens Conradi,2 Norbert Sewald,2 Steffen Backert,3,4 and Hartmut H. Niemann1,* 1Structural Biochemistry, Department of Chemistry, Bielefeld University, 33501 Bielefeld, Germany 2Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, 33501 Bielefeld, Germany 3Institute of Medical Microbiology, OvG University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany 4Department Biology, FAU University Erlangen-Nuremberg, D-91058 Erlangen, Germany *Correspondence: [email protected] http://dx.doi.org/10.1016/j.str.2013.08.018

SUMMARY ploits receptors via a type IV secretion system (T4SS; Kwok et al., 2007). RGD tripeptide motifs frequently mediate ligand T4SSs are macromolecular assemblies that transport proteins binding to . The type IV secretion system or protein-DNA complexes across the bacterial envelope, result- (T4SS) protein CagL of the gastric pathogen Helico- ing in secretion into the medium or transfer into bacterial or eu- bacter pylori also contains an RGD motif. CagL karyotic target cells (Alvarez-Martinez and Christie, 2009; decorates the T4SS pilus and may function as an Fronzes et al., 2009). T4SSs consist of three interlinked parts: adhesin for host cells. Whether CagL binds integrins a cytoplasmic/inner membrane-associated complex composed of three NTPases providing the energy for transport; a channel via its RGD motif is under debate. Here, we present spanning the bacterial envelope, also known as the core com- crystal structures of CagL revealing an elongated plex; and an external pilus. The Agrobacterium tumefaciens T- four-helix bundle that appears evolutionarily unre- DNA transfer system is the prototypic T4SS. It consists of 11 lated to the proposed VirB5 orthologs. The RGD essential VirB proteins (VirB1-VirB11) and VirD4. The virulence- motif is surface-exposed but located within a long associated T4SS of H. pylori is encoded in an 40 kb genomic a helix. This is unprecedented as previously charac- insertion, the cytotoxin-associated gene pathogenicity island terized integrin-binding RGD motifs are located (cagPAI) (Odenbreit et al., 2000). Homologs for most of the 12 within extended or flexible loops. Yet, adhesion of proteins of the A. tumefaciens VirB/D T4SS have been identified gastric epithelial cells to CagL was strictly RGD- among the 27 proteins encoded in the cagPAI, although some dependent. Comparison of seven crystallographi- are poorly defined (Buhrdorf et al., 2003; Covacci et al., 1997). cally independent molecules reveals substantial Little is known about most remaining accessory proteins en- coded by the cagPAI that lack homology to VirB/D proteins (Cen- structural flexibility. Intramolecular disulfide bonds dron and Zanotti, 2011). engineered to reduce CagL flexibility resulted in CagL is one such cagPAI protein that lacks clear sequence more stable protein, but unable to support cell adhe- similarity to any VirB/D protein. CagL localizes to the surface sion. CagL may thus partly unfold during receptor of the T4SS pilus (Kwok et al., 2007) and a DcagL mutant fails binding. to translocate the oncogenic effector protein CagA and induce interleukin-8 (IL-8) secretion in host cells (Fischer et al., 2001). Despite a mere 15% sequence identity, CagL was suggested INTRODUCTION to be an ortholog of VirB5, the prototypical adhesin of the VirB/D system primarily based on its function, size (220 amino Adhesion of animal cells to the (ECM) is a acids), isoelectric point, genomic organization, and three- fundamental process in development, tissue homeostasis, and dimensional modeling (Backert et al., 2008). CagL was alterna- disease. Specific cell surface receptors including the heterodi- tively suggested to be a H. pylori-specific protein that shares meric integrins mediate interactions with ECM proteins including sequence similarity with CagH (Shaffer et al., 2011). CagL can fibronectin (Fn; Hynes, 2002; Singh et al., 2010). Fn alone is suf- be copurified with CagH and CagI, two cagPAI proteins en- ficient to induce spreading of various mammalian cell types. The coded within the same operon as CagL (Pham et al., 2012; Arg-Gly-Asp (RGD) tripeptide motif of Fn is crucial for integrin Shaffer et al., 2011). All three share a conserved C-terminal binding and RGD motifs are also present in other integrin ligands ‘‘S/T-K-I/V-I-V-K’’-hexapeptide and influence pilus formation (Leiss et al., 2008; Ruoslahti, 1996). Some bacterial and viral (Shaffer et al., 2011). pathogens target integrins, facilitating extracellular persistence CagL is the only cagPAI protein with an RGD motif and or entry into host cells (Hauck et al., 2006; Stewart and Nem- may function as a T4SS adhesin that binds to the host inte- erow, 2007; Ulanova et al., 2009). One example is the gastric grins a5b1, aVb5, and aVb3(Conradi et al., 2012a; Jime´ nez- pathogen and group 1 carcinogen Helicobacter pylori, which ex- Soto et al., 2009; Kwok et al., 2007; Wiedemann et al., 2012),

Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved 1931 Structure Structure of HP CagL Reveals a Helical RGD Motif

Table 1. CagL Proteins Used in This Study RESULTS Protein Mutation(s)/Modification Crystallization and Structure Determination of CagL Wild-Type Variants The structure of CagL (amino acids 21–237) was determined in C-HIS CagL C-term. Leu-Glu-His6 two crystal forms. To reduce surface entropy and aid crystalliza- wt CagL N-term. TEV-cleavable His6 tion (Derewenda, 2004), we replaced potentially surface CagLmeth reductive methylation of CagLwt exposed, high entropy amino acids by smaller, low entropy res- N-Terminal Deletion idues to generate the variant CagLKKQEK (Table 1). Functionality KKQEK CagLDN deletion of aa 21–61 of CagL in the T4SS was assessed by genetic complemen- Surface Entropy Reduction Variants tation of CagL in H. pylori and has been proven to be as active as CagLwt (Figure S1 available online). The structure of CagLKKQEK CagLKK69AT K69A and K70T was solved using seleno-methionine (SeMet) derivatized protein CagLKKQEK K69A, K70T, Q147A, E148T, and K149A by multi-wavelength anomalous dispersion (MAD; Table 2). Cu- RGD Variants bic crystals contained one molecule per asymmetric unit. R76A CagL R76A Following reductive methylation of primary amines of wild-type CagLR76K R76K CagL (CagLmeth), we obtained a second, tetragonal crystal CagLG77A G77A form with six molecules in the asymmetric unit. The structure KKQEK CagLD78A D78A was solved by molecular replacement (MR) using CagL Structure-Function Correlation Variants as search model. Data processing and refinement statistics for both crystal forms are provided in Table 3. While the six crystal- CagLI73N I73N lographically independent molecules of CagLmeth are similar to CagLAL80QA A80Q and L81A each other (Table S1), there are major differences to CagLKKQEK. EA87AE CagL E87A and A88E We will use the best defined molecule A of CagLmeth as a refer- G169K CagL G169K ence, and describe important differences for the higher resolu- CagLAS171SN A171S and S172N tion structure of CagLKKQEK where appropriate. C-Terminal Variants CagLC128S C128S CagL Forms an Elongated Helical Bundle ˚ ˚ CagLY225A Y225A CagL is rod-shaped with dimensions of about 90 A by 30 A and D consists of six a helices. Four long a helices (a1, a2, a5, and a6) CagL C deletion of aa 227–237 run roughly parallel to the long axis with two short perpendicular Cysteine Variants helices (a3 and a4) winding around the N-terminal part of a5(Fig- C1 CagL A71C and S172C ures 1 and S1). We will refer to CagL with the long axis oriented C2 CagL N85C and A162C vertically such that both N and C termini point up, positioning the CagLC1+C2 A71C, N85C, S172C, and A162C short helices a3 and a4 at the top. The connections between a1 Substitution and deletion mutants are derived from CagLwt covering aa and a2 and between a5 and a6 form the bottom of the molecule. 21–237. Protein intended for crystallization was TEV-cleaved. Viewed from the top, a1, a2, a5, and a6 are arranged counter- clockwise. The elongated shape and mainly a helical structure of CagL are consistent with the molecular mass estimate of but the RGD dependence of various CagL functions remains 50 kDa from analytical gel filtration (calculated mass from controversial. One group reported RGD-independent IL-8 sequence 25 kDa) and with circular dichroism (CD) spectra of induction and CagA translocation (Jime´ nez-Soto et al., 2009). CagL, respectively (Figures S2A and S3A; Conradi et al., A second group reported RGD motif-independent induction 2012b; Kwok et al., 2007). of the gastrin promoter by CagL (Wiedemann et al., 2012). Although the fold is clearly defined, CagL is characterized by Others found CagA translocation (Kwok et al., 2007), IL-8 in- substantial structural flexibility. The most invariant part of CagL duction (Gorrell et al., 2012), host cell Src and FAK kinase is a three-helix bundle formed by the upper half of a2, a5, and activation (Kwok et al., 2007; Tegtmeyer et al., 2010), and a6 together with a3(Figures 1A and 1B). This CagL core is spreading of various cells on immobilized CagL (Tegtmeyer held together by a cluster of aromatic residues: Phe86, Phe92, et al., 2010) to be RGD motif-dependent. The importance of Phe93, and Phe100 from a2, Tyr163 from a5, and His200, the RGD motif is further underlined by synthetic RGD peptides Phe204, and Tyr207 from a6. Minor structural variations are that interfere with cell adhesion to wild-type CagL (Conradi found in the C terminus of a5, the N terminus of a6, and the con- et al., 2012a) and complement deficiencies of T4SS pilus as- necting short loop. This slightly more flexible region begins with a sembly and CagA phosphorylation in a DcagL H. pylori strain double (Gly168, Gly169) in a 5 and ends with Leu184 in expressing RGD substitution variants of CagL (Kwok et al., a6. The N-terminal part of a2 extends over the loop connecting 2007). a5 and a6 and does not contribute to the three-helix bundle. Here, we report crystal structures of CagL revealing a helical Comparison of the seven independent molecules reveals a hinge conformation of the RGD motif that is remarkably different region in the central part of a2 around Gly77 of the RGD motif, from other structurally resolved integrin-binding RGD motifs. which allows the N-terminal part of a2 to move toward the C ter- We analyzed this distinct structural context of the RGD motif us- minus of a5(Figure 1B). This structural flexibility occurs between ing biochemical, biophysical, and cell adhesion assays. the two different crystal forms and also between different

1932 Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved Structure Structure of HP CagL Reveals a Helical RGD Motif

Table 2. Data Collection and Phasing Statistics Table 3. Data Collection and Refinement Statistics CagLKKQEK (SeMet) Protein CagLKKQEK (SeMet) CagLmeth (Native) High Energy Data collection Protein Peak Inflection Remote Beamline BL14.2 (BESSY) ID23.2 (ESRF) Data Collection Wavelength (A˚ ) 0.97814/0.978247 0.8726 Beamline BL14.2 (BESSY) Space group F23 P43212 Space group F23 Unit cell axis (A˚ ) a = b = c = 181 a = b = 197.12, Wavelength (A˚ ) 0.97962 0.97982 0.97857 c = 106.81 Unit cell axis (A˚ ) a=b=c= a=b=c= a=b=c= Resolution range (A˚ ) 20–2.2 (2.32–2.2) 58.34–3.25 (3.43–3.25) 181.24 181 181.18 Observed reflections 358,914 (33,007) 260,650 (37,776) ˚ Resolution range (A) 41.6-2.7 (2.85-2.70) Unique reflections 24,942 (3,606) 33,724 (4,829) Observed reflections 155,222 153,353 154,109 Multiplicity 14.4 (9.2) 7.7 (7.8) (22,969) (22,267) (22,687) Completeness (%) 99.9 (100) 100 (100) Unique reflections 13,672 13,602 13,668 R 0.139 (0.908) 0.219 (1.081) (2,007) (1,958) (1,996) merge R 0.144 (0.962) 0.235 (1.159) Anomalous 5.8 (5.8) 5.8 (5.8) 5.8 (5.8) meas multiplicity Rpim 0.037 (0.317) 0.084 (0.412) s Anomalous 100 (100) 100 (100) 100 (100) I/ 12.7 (2.3) 8.1 (2.0) completeness (%) CC1/2 0.998 (0.716) 0.995 (0.565)  Rmerge 0.069 (0.234) 0.069 (0.227) 0.073 (0.265) Mosaicity ( ) 0.73 0.39 ˚ 2 Rmeas 0.076 (0.257) 0.076 (0.249) 0.080 (0.292) Wilson B-factor (A ) 39.4 65.8

Rpim 0.031 (0.106) 0.031 (0.103) 0.033 (0.121) Refinement I/s 22.2 (9.3) 22.6 (9.5) 21.8 (8.5) Resolution range (A˚ ) 19.87–2.2 (2.29–2.2) 55.97–3.25 (3.35–3.25)  Mosaicity ( ) 0.54 0.57 0.58 Rcryst (%) 20.75 (29.16) 20.51 (28.72) delAnom between half sets 0.700 (0.213) 0.430 (0.073) 0.525 (0.121) Rfree (%) 24.46 (27.34) 25.13 (35.25) Phasing (after three cycles bucaneer/refmac) No. reflections Correlation coefficient all/weak (%) 55.04/40.05 Work 23,623 (2,614) 31,957 (2632) Site occupancy #1/#6 (search for 1.00/0.20 Test 1,286 (148) 1,706 (138) five sites) No. molecules/atoms Contrast (after 20 cycles SHELXE) 1.23 Protein 1/1,814 6/9,370 Pseudo-free CC (%) 64.84 Ligand 10/52 11/66 Automated model building Water 113 6 No. chains 5 (longest 85 residues) Rmsd No. residues 190 (157 sequenced) Bond (A˚ ) 0.012 0.006  Rcryst (%) 31.86 Angles ( ) 1.29 0.93 Rfree (%) 34.35 Ramachandran plot Favored (%) 98.6 97.66 molecules of CagLmeth. a3 is held in place by p-p-stacking of the Allowed (%) 1.4 2.25 phenolic rings of Tyr103 (C terminus of a2) and Tyr113 (a3) and a Disallowed (%) 0 0.09 hydrogen bond between the amide nitrogen of Leu119 (C termi- Mean B-factor (A˚ 2) 46.2 91.1 nus of a3) and the side chain of Glu140 (a5; Figure 1C). a4is covalently bound to the three-helix bundle by a disulfide bond between Cys128 from a4 and Cys139 from a5. Additionally, a CagLmeth, a1 interacts with the more rigid upper part of the hydrogen bond between Asp132 from the very top of a5 and three-helix bundle. In CagLKKQEK, by contrast, a1 is shifted one Tyr225 from a6 stabilizes the structural arrangement in this re- helix turn toward the N terminus of a2 and a6(Figure 2). In gion. Nevertheless, a4 remains quite flexible and was visible in both structures, amino acids connecting a1 and a2 were disor- only three of the seven crystallographically independent mole- dered. The distance between the last ordered residue of a1 cules, indicating the possibility of partial reduction of the disul- and the first ordered residue of a2is17 A˚ in CagLmeth (from fide bond. Thr51 to Asn58) and CagLKKQEK (from Ser50 to Glu59). The missing residues are sufficient to span this distance in both Helix a1 Packs Loosely against the Three-Helix Bundle cases. The two crystal structures of CagL differ with respect to binding In vitro, CagL was cleaved over time into a larger C-terminal of the N-terminal helix to the three-helix bundle. a1 packs into and a smaller N-terminal fragment containing a1(Figure S2). In a hydrophobic groove formed by a2 and a6 with minor con- gel filtration, the two fragments eluted together, suggesting tributions from a5 (Leu173 and Ala176) in both structures. In that they remain associated. A variant of CagL lacking the

Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved 1933 Structure Structure of HP CagL Reveals a Helical RGD Motif

A B

C

Figure 1. CagL Forms an Elongated Helical Bundle (A) Left: cartoon representation of chain A from CagLmeth colored in blue to red from N to C termini. Amino acids connecting a1 and a2 (Thr52-Asn57) are not resolved and are denoted as a dashed line. The conformation of the N-terminal end of a2 in CagLKKQEK is superimposed in transparent coloring (alignment of aa 80–220). The helical Arg76-Gly77-Asp78 (RGD) motif is highlighted as magenta spheres. Right: the same molecule rotated by 55 counterclockwise around the long axis colored by B-factor from blue (low) to red (high). Aromatic side chains are depicted as sticks (see also Figures S1–S3). (B) Superposition of the seven crystallographically independent molecules of CagL solved in this study. Least-squares fit superposition was done for main chain atoms of amino acids 80–114, 144–166, and 187–219 (see also Table S1). (C) Structural features that stabilize the upper part of CagL. A disulfide bond links Cys128 to Cys139. Coloring as in (A) left.

N-terminal helix (CagLDN) precipitated at concentrations above (see below). Despite its variable association with the rest of 1 mg/ml whereas CagLwt remained soluble even beyond CagL, a1 may thus be important for stability. 10 mg/ml in PBS at 4C. a1 presumably shields the hydrophobic It is not clear whether one of the two observed positions of a1 core from solvent, preventing aggregation. Consistently, the represents the physiological structure. We propose that the buried surface area between a1 and the rest of CagL measuring binding mode of CagLmeth represents the native state because 1,555 A˚ 2 for CagLmeth chain A and 1,686 A˚ 2 for CagLKKQEK is (1) the same binding mode is seen in six crystallographically in- calculated to be 82% hydrophobic in both cases (Hubbard dependent molecules; (2) in five of the six molecules of CagLmeth and Thornton, 1993). Moreover, the melting temperature of a1 is not involved in crystal contacts at all, whereas the C termi- CagLDN in differential scanning fluorimetry (DSF) experiments nus of a1 interacts with a symmetrical molecule in CagLKKQEK; is lower than for CagLwt, as is its ability to promote cell adhesion and (3) in CagLKKQEK two dioxane molecules added during

1934 Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved Structure Structure of HP CagL Reveals a Helical RGD Motif

facilitates formation of a crystal contact around a 2-fold sym- metry axis between the Cb atoms of Ala69 (Figure 3D). This crystal contact involves the Arg76 side chain, which contacts the C terminus of a1 of the symmetry-related molecule (Figures 3C and 3D). Distortion of the helical conformation around the RGD motif may additionally be linked to the shift of a1in CagKKQEK. This shift comes along with an H-bond between the side chain of Tyr34 (a1) and the carbonyl oxygen of Leu75 (Figure 3C), which does not form the typical a-helical intra-main chain H-bond.

Adhesion of MKN-45 Cells to CagL Is Strictly RGD- Dependent Because CagL proposedly is a ligand for integrins, we analyzed adhesion of human gastric adenocarcinoma (MKN-45) cells to plates coated with wild-type CagL or CagL variants, in which each of the three amino acids from the RGD motif was substituted separately (Kwok et al., 2007; Schneider et al., 2011; Tegtmeyer et al., 2010; Table 1). In wells coated with Fn or CagLwt, cells evenly distributed over the surface. In control wells or wells coated with any of the CagL RGD variants, cells Figure 2. Helix a1 Packs Loosely against the Three-Helix Bundle formed aggregates in solution (Figure 4A) and were lost upon KKQEK meth Overlay of CagL aa 80–220 (blue) with chain A of CagL (brown). washing. Quantification by a hexosaminidase assay revealed Background: Surface presentation of CagLmeth a2-a6 in gray with O- and N- KKQEK a that MKN-45 cells adhered to wells coated with 125–500 nM atoms in orange. Labeling of CagL is underlined. In both structures, 1 wt KKQEK packs into a hydrophobic groove formed by a2 and a6 but shifted by about one CagL as well as the crystallization variants CagL and meth helix turn in CagLKKQEK compared to CagLmeth. In CagLmeth, Tyr34 fills a hy- CagL . Signal intensity similar to that of 25 nM Fn was drophobic pocket that is occupied by dioxane (Dio, yellow) in CagLKKQEK (see observed for 500 nM CagL. No adhesion was observed to any also Figure S2). of the RGD variants (Figure 4B). crystallization lie between a1 and the three-helix bundle, one Mutation of Residues around the RGD Motif Does Not filling a hydrophobic pocket occupied by Tyr34 in CagLmeth, Impair Cell Adhesion the other filling a hydrophobic void created by a kink in a2 just The protein surface around the RGD motif is characterized by before the RGD motif (Figure 2). hydrophobic or small amino acids: Ile73 and Ala74 one turn N-terminal of the RGD motif as well as Ala80 and Leu81, Ala84 The RGD Motif Is Helical and Surface Exposed or Ala88 one, two, or three turns toward the C terminus (Fig- The potential integrin-binding RGD motif of CagL (Arg76-Gly77- ure 4C). This hydrophobic patch flanking the RGD motif could Asp78) is centrally located in a2 connecting the rigid upper and be critical for integrin binding. Residues on a5 adjacent to the the flexible lower part (Figure 1A). The RGD motif is surface RGD motif are small as well. Exposed amino acids include exposed, with the Arg facing a1, Gly most accessible and Ser165, Gly168, Gly169, Ala171, and Ser172. To assess the Asp facing a5. In CagLmeth, the RGD motif adopts an a-helical influence of the exposed amino acids in proximity to the RGD conformation (Figure 3A). The Arg76 side chain forms a salt motif, CagL variants CagLI73N, CagLAL80QA, CagLEA87AE, bridge to Asp46 (a1) in five of six molecules, whereas Asp78 CagLG169K, and CagLAS171SN (Table 1) were tested in cell adhe- is mostly free and contacts Asn166 only in molecule F. In the sion assays (Figure 4D). All variants were active in promoting cell structure of CagLKKQEK, the helical conformation of the RGD adhesion with a similar concentration profile as CagLwt. The C motif is distorted with Gly77 adopting a non-a-helical confor- terminus of CagL was similarly tested for adhesion of MKN-45 mation (Figures 3B, 3C, and S4). The Tyr34 side chain H-bonds cells (Figures 1C and 4D). Both CagLC128S, resembling a reduced the Leu75 carbonyl oxygen. The Arg76 carbonyl oxygen and disulfide bond, and the C-terminal deletion variant CagLDC, the Asp78 amide nitrogen point toward the solvent and their lacking the hexapeptide conserved in CagL, CagI, and CagH, H-bonding potentials are saturated by water molecules. A retained full activity. In contrast, CagLY225A showed reduced kink resulting in a2 pushes its N-terminal half closer to the C adhesion, possibly caused by the reduced thermostability of terminus of a5(Figure 1), exposing the RGD motif. The largest CagLY225A due to the loss of the intramolecular interaction with displacement of 2.5 A˚ involves Arg76 while Gly77 and Asp78 Asp132. Finally, CagLC-HIS, described previously (Kwok et al., move by 1.2 a and 1.4 A˚ , respectively (Ca atoms, compared 2007; Saha et al., 2010; Tegtmeyer et al., 2010; Wiedemann to molecule A of CagLmeth). et al., 2012), supported equal adherence as CagLwt. It is not clear why the helical conformation of the RGD motif is distorted in CagLKKQEK. The K69A, K70T double substi- The N Terminus of a2 Lacks Helical Stability tution introduced to obtain this crystal form is located two helix Susceptibility to proteases can identify flexible regions. Trypsin turns N-terminal of the RGD motif, but does not otherwise digestion of CagLC-HIS rapidly generated a slightly smaller frag- affect its structure. However, truncation of the K69 side chain ment that no longer reacted with an antibody directed against

Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved 1935 Structure Structure of HP CagL Reveals a Helical RGD Motif

A B

D

C

Figure 3. A Crystal Contact in CagLKKQEK Distorts the Helical Conformation of the RGD Motif and Induces a Kink in a2 (A) Representation of the regular hydrogen bond network of helix a2 in CagLmeth. (B) Distorted hydrogen bond network of a2 in CagLKKQEK. Tyr34 forms a hydrogen bond to carbonyl oxygen of Leu75. The H-bonding potentials of carbonyl oxygen of Arg76 and amide nitrogen of Asp78 are saturated by water molecules (see also Figure S4). (C) Representation of 2mFo-DFc electron density contoured at 1 s around the distorted RGD motif in CagLKKQEK. (D) Crystal contact between the N termini of a2 of CagLKKQEK (colors as in Figure 1A, left) and its symmetry related molecule (gray). Hydrogen bonds between Arg76 from the RGD motif and the C-terminal residues of a1 are shown as dashed lines, water as red sphere. the His-tag. Consistent with this C-terminal truncation, amino Dijkstra, 1988) to link flexible a2 and a5. In cysteine variant acids Ser232 to Lys237 were not visible in any of the seven mol- CagLC1, a cysteine located six amino acids N-terminal of Gly77 ecules, suggesting that the C terminus of a6 including the links A71C to S172C (Table 1; Figure 5B). In CagLC2 residue conserved hexapeptide is flexible. Continued proteolysis of N85C, eight amino acids C-terminal of Gly77 is linked to CagLC-HIS or CagLwt as well as long-term storage of CagL in A162C (Table 1; Figure 5B). Tryptic proteolysis of CagLC2 (central the absence of exogenously added proteases resulted in forma- disulfide) was indistinguishable from that of CagLwt whereas tion of a dominant fragment of 18.3 kDa (Figures 5A and S2). CagLC1 (more restrained a2) was more resistant to trypsin result- N-terminal sequencing suggested a cleavage C-terminal of ing in a CagLKKQEK-like pattern. DSF (Figure 5C) and temperature Lys70 but was ambiguous. To distinguish between cleavage interval-dependent CD-spectroscopic measurements (Fig- C-terminal of Lys69-Lys70 or after Arg76, we performed tryptic ure S3B) demonstrate that the disulfide bond stabilizes CagLC1. digests of CagLR76A and CagLKKQEK (containing the K69A and It undergoes a structural transition at 49C whereas CagLwt K70A mutations). CagLR76A yielded similar degradation products melted at 43C. Interestingly, CagLR76A and CagLG77A also as CagLwt, while the 18.3 kDa fragment did not form with exhibited a slightly elevated transition temperature of 45C. CagLKKQEK, indicating a cleavage C-terminal of Lys70 rather The melting curve of CagLC2 was qualitatively different from than Arg76 (Figure 5A). Because Lys70 is part of an a helix in that of CagLwt, showing a biphasic behavior, with the second all our structures, cleavage at this site was unexpected and sug- transition and the maximum shifted toward higher temperature. gests that the N-terminal section of a2 may be flexible and lack This possibly reflects a stabilization of the hydrophobic core in helical stability in solution. proximity of the C2 mutation. A variant harboring both disulfides (CagLC1+C2) combined melting characteristics of variants Restraining Flexibility around the RGD Motif Abrogates CagLC1 and CagLC2, indicating independent effects on CagL Cell Adhesion stability. To investigate whether the flexible, protease sensitive part of a2 To assess an influence of the flexibility of a2 on the accessi- affects CagL stability, we introduced disulfide bonds (Hazes and bility of the RGD motif, we performed cell adhesion assays

1936 Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved Structure Structure of HP CagL Reveals a Helical RGD Motif

AB

CD

Figure 4. Adhesion of MKN-45 Cells to Immobilized CagL Depends on the RGD Motif (A) Phase contrast microscopy image showing adhesion of MKN-45 cells to precoated microtiter plates after 2 hr incubation without washing. Five hundred nanomolar CagLwt (upper right; induced a similar microscopic appearance as 25 nM rh Fn; upper left) with an even distribution of adhered cells on the well surface. In contrast, cells did not adhere to negative control wells (lower left) or CagLR76A coated wells (lower right), but aggregated in the medium. Bars represent 50 mm. (B and D) Quantification of the adherence by a hexosaminidase enzyme assay is shown. Absorbances at 405 nm were normalized to the highest signal intensity at 25 nM Fn. Data presented are mean absorbances for four or more independent experiments with four replicates each and corresponding standard deviations (n R 4). One-way ANOVA statistics (OriginPro Software, OriginLabs) compared to CagLwt: n.s., not significant; *p < 0.05; **p < 0.01; ***p < 0.001. (C) Localization of the structure-derived mutations for adhesion assays. Ca-atoms of selected amino acids surrounding the RGD motif are shown as spheres, side chains as sticks. Mutated amino acids are highlighted in magenta. with the disulfide-linked variants. Interestingly, the C1 variant that CagLKKQEK and CagLmeth are physiologically relevant and was strongly attenuated in its ability to induce adhesion of that their structures provide biologically meaningful insight into MKN-45 cells and the C2 variant was completely inactive (Fig- the function of wild-type CagL. ure 5D), indicating that not only accessibility of the RGD motif In previous studies, CagL was proposed to be a VirB5 ortholog is relevant for receptor binding, but also conformational flexibility and a structural model of CagL was generated based on the of a2 around the RGD motif. crystal structure of TraC, a VirB5 protein (Backert et al., 2008). However, the structural data presented here clearly distinguish DISCUSSION CagL from TraC (Yeo et al., 2003). First, CagL consists of an elongated four-helix bundle, whereas TraC contains a three-helix We determined the structure of CagL variants because our bundle with a loose globular appendage. Moreover, the arrange- efforts to determine the structure of wild-type CagL had failed. ment of helices in CagL and TraC is inverted. Second, structural Surface entropy reduction mutations and lysine methylation features such as aromatic residues of the hydrophobic core and are two standard methods to increase the crystallization propen- the disulfide bond of CagL are not conserved in TraC. A DALI sity of proteins or to improve crystal quality (Derewenda, 2004; search (Holm and Rosenstro¨ m, 2010) revealed TraC as largely Walter et al., 2006). Both CagLKKQEK and CagLmeth supported unrelated with a Z-score of 4.9 (root-mean-square deviation cell adhesion similar to CagLwt. CagLKKQEK also complemented [rmsd] of 10.2 A˚ over 130 aligned residues). More related struc- a cagL deletion in H. pylori. Genetic complementation is not tures include diverse helical bundles e.g., of STATs, spectrin re- possible for CagLmeth, but generally, structures of methylated peats, a-actinin, or the large GTPase Mx1 and also the type III proteins are very similar to their nonmethylated counterparts secretion translocator protein SipB from Shigella flexneri and he- (Sledz et al., 2010). Together, these results strongly suggest molysin BL from Bacillus cereus. No obvious sequence

Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved 1937 Structure Structure of HP CagL Reveals a Helical RGD Motif

A B

C D

Figure 5. The N-Terminal End of a2 Containing the RGD Motif Is Flexible (A) Limited proteolysis of CagL at 1 mg/ml with 1:100 trypsin for 45 min and 2 hr, respectively, in TBS on ice (see also Figure S2). (B) Engineered disulfide bonds connect a2toa5. C2 is located near the hydrophobic core of CagL. K69-K70 converted to Ala69-Thr70 in CagLKKQEK is also shown (colors as in Figure 4C). (C) Representative DSF melting curves for CagL variants. The melting of CagL was followed by detection of the relative fluorescence of SYPRO Orange dye using a Real-Time PCR System. Similar melting curves are obtained by CD as shown in Figure S3B. (D) Quantification of MKN-45 cell adherence to CagL-coated plates. The engineered disulfides C1 and C2 significantly inhibited cell adhesion. For a detailed description, see Figure 4. Error bars show SD. homology was found in any structure. Although CagL function- (Del-1) forms the tip of a long protruding loop important for inter- ally resembles VirB5 proteins in other T4SSs, it is probably a pro- action with integrin (Schu¨ rpf et al., 2012). Functional RGD motifs tein specific for the H. pylori cagPAI, as has been previously sug- also occur in flexible loops, as is the case in latent transforming gested (Kutter et al., 2008; Shaffer et al., 2011). growth factor-b, where part of the RGD motif is disordered (Shi The manner in which H. pylori exploits integrins for cell attach- et al., 2011). RGD motifs exposed by viral or bacterial proteins ment and type IV secretion is still under discussion. An RGD- that mediate integrin binding in the infection cycle are likewise dependent model proposes CagL to be located on the surface located in loop regions and are mostly flexible (Emsley et al., of the T4SS pilus mediating the first contact with host cell integ- 1996; Kagawa et al., 2000; Logan et al., 1993; Zubieta et al., rins (Kwok et al., 2007; Tegtmeyer et al., 2010). A competing 2005). Clearly, the RGD motif of CagL does not mimic the loop RGD-independent model proposes a direct interaction of CagA location of other structurally described RGD motifs. Instead, it is with integrin (Jime´ nez-Soto et al., 2009; Kaplan-Tu¨ rko¨ z et al., embedded in a distinct structural context in the middle of a long 2012). The data presented in this study confirm that CagL alone helix. This raises the question of how integrins recognize CagL. is sufficient to induce adhesion of human gastric epithelial MKN- Conformational flexibility around the RGD motif of CagL 45 cells. Moreover, cell adhesion was strictly dependent on the proved to be important for CagL recognition by cells. Disulfide RGD motif because alanine substitutions of any one of the three bonds introduced N-terminal or C-terminal of the RGD motif amino acids abrogated cell adhesion whereas mutation of sur- serve to stabilize the protein but abolish cell adhesion. This face-exposed amino acids in proximity to the RGD motif did agrees with our finding that the N-terminal section of a2 lacks he- not result in loss of function. lical stability. However, it was surprising that the C2 variant did RGD motifs are mostly located in exposed, flexible loop struc- not support cell adhesion because this disulfide bridge is located tures extending beyond the protein surface. The RGD motif of the near the hydrophobic core. Though it would appear unlikely that tenth type III repeat of Fn e.g., is located in a b-hairpin-like loop ex- the hydrophobic core of CagL could unfold upon receptor bind- tending 10 A˚ beyond the core of the domain (Leahy et al., 1996). ing, the effect of the C2 variant does suggest that this may be the The RGD motif of protein developmental endothelial cell locus-1 case.

1938 Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved Structure Structure of HP CagL Reveals a Helical RGD Motif

EXPERIMENTAL PROCEDURES crystals of CagLmeth grew at 10 mg/ml protein with reservoirs containing un- buffered 20% polyethylene glycol (PEG) 3350 with 200 mM of various salts

Cloning of Expression Constructs (e.g., LiCl, KNO3, ammonium tartrate). Crystals were difficult to reproduce A modified pET-28a vector containing the DNA for expression of CagL aa 21- and crystallization time varied from 3 weeks to 12 months. The final crystal C-HIS 237 (H. pylori strain 26995) with a C-terminal Leu-Glu-His6-tag (CagL ) was harvested from a drop containing 200 mM KCl and 22.5% PEG 3350. served as PCR template (Kwok et al., 2007). A fragment encoding CagL amino acids (aa) 21–237 was amplified by PCR and cloned into a pETM-11 vector (G. Crystal Harvesting, Cryo-Protection, and Data Collection Stier, EMBL Heidelberg), facilitating expression of N-terminally His6-tagged Crystals of CagLKKQEK were sensitive to evaporation of liquid or 1,4-dioxane wt CagL aa 21–237 (CagL ) with a linker sequence between the His6-tag and from the crystallization drop. A cryoprotection solution equivalent to the crys- CagL sensitive to the tobacco etch virus (TEV) protease. The surface entropy tallization drop contents plus 20% (v/v) 2,3-butanediol was added to the crys- KKQEK reduction variant CagL was identified by the SERp server (Goldschmidt tallization drop and noncracked crystals were transferred to higher 2,3-buta- et al., 2007) and generated by a modified QuickChange protocol (Stratagene) nediol concentration, mounted in a cryo loop (Hampton Research), and using Phusion polymerase. Similar approaches were used for generation of the flash-cooled in liquid nitrogen. The crystallization drop of CagLmeth was mixed variants listed in Table 1. Truncations of the N or the C termini were generated with glycerol to a final concentration of 20% (v/v) prior to flash-cooling. by amplification of the corresponding vector fragment, followed by DpnI diges- CagLKKQEK diffraction data were collected on BL14.2 operated by the Joint tion and ligation with T4 DNA ligase. All constructs were verified by Berlin MX-Laboratory at the BESSY II electron storage ring (Berlin-Adlershof; sequencing. Mueller et al., 2012). Data sets from CagLmeth were collected at beamline ID23-2 of the European Synchrotron Radiation Facility (ESRF). Protein Expression and Purification All CagL variants were expressed in E. coli BL21-CodonPlus-RIL for 3.5 hr at Data Reduction, Structure Determination, and Refinement 37C in selective lysogeny broth (LB) supplemented with 2 g/l glucose mono- The data sets were either processed with XDS (Kabsch, 2010) or iMOSFLM hydrate and purified from inclusion bodies. The expression of CagL was 1.06 (Battye et al., 2011) and scaled with Scala (Evans, 2006) from the CCP4 induced with 0.5 mM isopropyl b-D-1-thiogalactopyranoside (IPTG) at an op- suite (CCP4, 1994). SHELXC, -D, and -E (Sheldrick, 2010) via the HKL2MAP tical density 600 (OD ) z0.8. Inclusion bodies were isolated at 5,000 3 g in a 600 interface (Pape and Schneider, 2004) were used for anomalous scatterer local- cooled centrifuge, washed twice with PBS, and dissolved in PBS + 8 M urea. ization and phasing of the SeMet derivative of CagLKKQEK. An initial model was Insoluble residual was pelleted at 20,000 3 g. Unfolded CagL was bound to a built with buccaneer (Cowtan, 2006). The structure of CagLmeth was solved by Ni-NTA affinity column and washed with PBS + 8 M urea supplemented suc- MR using Phaser (McCoy et al., 2007). The models were manually modified in cessively with 1 M NaCl, 12.5% glycerol, and 10 mM dithiothreitol (DTT) or COOT (Emsley et al., 2010) and refined with PHENIX (Adams et al., 2010). For 25 mM imidazole. Refolding of CagL was performed at 4C with a 1.5-fold dilu- CagLmeth, the refinement included secondary structure restraints, NCS re- tion series of PBS + 6 M urea with refolding buffer, containing 50 mM straints, and reference structure restraints. Translation/Libration/Screw (TLS) Tris(hydroxymethyl)-aminomethane (Tris), 20 mM NaCl, 0.8 mM KCl, 2 mM groups were identified with the TLSMD server (Painter and Merritt, 2006) reduced glutathione, and 0.2 mM oxidized glutathione at a final pH 8.2 at and included in refinement. LSQKAB was used for structural alignments 4C. Five milliliters of each dilution was applied for seven steps, followed by (Kabsch, 1976). Figures were prepared with the PyMOL Molecular Graphics 25 ml of refolding buffer. CagL was eluted with PBS + 250 mM imidazole. System, Version 0.99rc6 (Schro¨ dinger). The fusion protein was cleaved with TEV protease during dialysis against PBS overnight at 4C. Cleaved CagL was further isolated by negative affinity chromatography using Ni-NTA. Protein used for cell adhesion assays was Cell Adhesion Assays not cleaved. The flow-through or the dialyzed tagged CagL was concentrated MKN-45 cells (Deutsche Sammlung von Mikroorganismen und Zellkultur,  by ultra-filtration (VIVASPIN 20, 5,000 MWCO, Sartorius) and applied to a DSMZ, Braunschweig) were cultivated at 37 C with 5% (v/v) CO2 in RPMI HiLoad 16/60 Superdex75 pg size exclusion chromatography column 1640 medium (PAA) supplemented with 10% fetal calf serum (Lonza), 4 mM (GE Healthcare) using Tris-buffered saline (TBS) for protein intended for cell L-glutamine, 200 U/ml penicillin, and 200 mg/ml streptomycin. The medium was exchanged every 2–3 days recovering nonadherent cells for further culti- adhesion assays or crystallization buffer (20 mM K2HPO4/KH2PO4, pH 7.4, and 20 mM NaCl) for protein intended for crystallization. Monomeric CagL vation by centrifugation. fractions were pooled, concentrated, snap-frozen in liquid nitrogen, and For cell adhesion assays, each well of a transparent F96 MicroWell plate with  stored at À80C. MaxiSorp surface (Nunc) was coated overnight at 4 C with 50 ml 62.5–500 nM SeMet derivatives of CagL were produced in 1 l selective minimal medium CagL or 2.5–25 nM recombinant human (rh) Fn (R&D Systems) diluted in PBS. The wells were then supplemented with 100 ml of 5% (w/v) nonfat dried milk containing 1 g/l NH4Cl, 3 g/l KH2PO4, and 6 g/l Na2HPO4*7H2O supplemented  with 50 mg SeMet and 100 ml of sterile filtered supplement solution containing powder in PBS at pH 8 and incubated for 2 hr at 4 C and finally blocked with 200 ml of fresh milk powder solution for another 2 hr. Harvested nonat- 220 g/l glucose monohydrate, 6.1 g/l MgSO4*7H2O, 0.112 g/l thiamine mono- tached cells and trypsinized detached cells were diluted to 4–5 3 105 cells/ml hydrate, and 0.104 g/l Fe2(SO4)3*H2O. E. coli cells were grown in selective in fresh medium and 10 mM of each MgCl /CaCl /MnCl was added. The milk lysogeny broth with glucose to an OD600 z0.8, pelleted at 4,500 3 g, and re- 2 2 2 suspended in the prepared minimal medium. After 30 minutes, the expression powder solution was discarded and 100 ml of the cell solution was applied. The  of CagL was induced as described previously. plates were incubated without a lid for 2 hr at 37 C with 5% (v/v) CO2. For Methylation of freshly prepared, monomeric, TEV-cleaved CagLwt was per- quantification of the adhered cells, a hexosaminidase enzyme assay (Landeg- formed on ice according to the instructions of the JBS Methylation Kit (Jena ren, 1984) was performed. Nonattached cells were removed by twice washing Biosciences) using Borane-dimethylamine complex (Alfa Aesar). The reaction each well with 200 ml PBS. Sixty microliters of a solution containing 1.9 mM was followed by 3 hr incubation with excess of Tris, concentration by ultra- p-nitrophenol-N-acetyl-b-D-glucosaminide (Sigma-Aldrich), 0.25% (v/v) filtration, and gel filtration chromatography using the crystallization buffer Triton X-100, and 50 mM citrate buffer at pH 5 was added and incubated at described previously. Complete methylation of all primary amines in CagLmeth room temperature for 75 min. The reaction was stopped by adding 90 mlof was verified by MALDI-TOF mass spectrometry. 200 mM glycine-NaOH buffer pH 10.4. Resulting absorbances were measured in a Tecan Infinite200 microplate reader at 405 nm. Each plate contained Crystallization 25 nM Fn as reference and all absorbance readings were normalized to this Initial crystallization conditions were identified using commercially available value after background subtraction. screens from QIAGEN (Hilden, Germany) by sitting drop vapor diffusion. Diffraction quality crystals were grown by hanging drop vapor diffusion with Genetic Complementation of DcagL H. pylori a protein:reservoir ratio of 2:1 at 4C. CagLKKQEK crystallized at 7 mg/ml pro- Mutagenesis and genetic complementation of CagL in H. pylori strain P12, tein with the reservoir containing 100 mM 2-(N-morpholino)ethanesulfonic acid culturing and infection of AGS (gastric adenocarcinoma) cells, quantification (MES) at pH 6.5, 1.2 M ammonium sulfate, and 2%–4% 1,4-dioxane. Initial of the elongation phenotype, and western blotting using anti-phospho-

Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved 1939 Structure Structure of HP CagL Reveals a Helical RGD Motif

CagA, anti-CagA and anti-HA antibodies was performed as described else- Alvarez-Martinez, C.E., and Christie, P.J. (2009). Biological diversity of pro- where (Kwok et al., 2007; Conradi et al., 2012b). karyotic type IV secretion systems. Microbiol. Mol. Biol. Rev. 73, 775–808. Backert, S., Fronzes, R., and Waksman, G. (2008). VirB2 and VirB5 proteins: Differential Scanning Fluorimetry specialized adhesins in bacterial type-IV secretion systems? Trends DSF was measured using a StepOnePlus Real-Time PCR System (Applied Microbiol. 16, 409–413. Biosystems). Experiments were performed with TBS as dilution buffer in Battye, T.G., Kontogiannis, L., Johnson, O., Powell, H.R., and Leslie, A.G. G060/H/1E 96 well PCR plates (Kisker Biotech) sealed with GK480-OS optical (2011). iMOSFLM: a new graphical interface for diffraction-image processing m film (Kisker Biotech). Protein (42.5 l) at 0.0625, 0.125, and 0.25 mg/ml were with MOSFLM. Acta Crystallogr. D Biol. Crystallogr. 67, 271–281. mixed with 2.5 mlof383 SYPRO Orange (Life Technologies) directly in the Buhrdorf, R., Fo¨ rster, C., Haas, R., and Fischer, W. (2003). Topological analysis PCR plates on ice. The plates were centrifuged at 4C for 5 min at 2,000 3 g of a putative virB8 homologue essential for the cag type IV secretion system in and transferred to the real-time PCR cycler. Fluorescence was measured Helicobacter pylori. Int. J. Med. Microbiol. 293, 213–217. using the ROX dye settings from 4Cto97C with a heating speed of 1C/ min. Curves were analyzed using the transformation and analysis files pro- CCP4 (Collaborative Computational Project, Number 4). (1994). The CCP4 vided by Niesen (Niesen et al., 2007) and GraphPad Prism 4 (GraphPad Prism). suite: programs for protein crystallography. Acta Crystallogr. D Biol. All measurements were repeated at least three times with three wells per data Crystallogr. 50, 760–763. point. Cendron, L., and Zanotti, G. (2011). Structural and functional aspects of unique type IV secretory components in the Helicobacter pylori cag-pathogenicity is- CD Spectroscopy land. FEBS J. 278, 1223–1231. CD spectroscopy (Johnson, 1990) was performed in 1 mm path-length quartz Conradi, J., Huber, S., Gaus, K., Mertink, F., Royo Gracia, S., Strijowski, U., cuvettes from 270 nm to 190 nm wavelength with a bandwidth of 1 nm at a Backert, S., and Sewald, N. (2012a). Cyclic RGD peptides interfere with bind- 50 nm/min scanning speed using a J-810 spectropolarimeter (Jasco Instru- ing of the Helicobacter pylori protein CagL to integrins aVb3 and a5b1. Amino ments). Temperature interval-dependent CD spectra were acquired at Acids 43, 219–232. 0.15 mg/ml protein in 1:4 diluted crystallization buffer from 25Cto65C Conradi, J., Tegtmeyer, N., Wozna, M., Wissbrock, M., Michalek, C., Gagell, (75C) with one data point per each 2C. A corresponding buffer baseline C., Cover, T.L., Frank, R., Sewald, N., and Backert, S. (2012b). An RGD helper was subtracted. sequence in CagL of Helicobacter pylori assists in interactions with integrins and injection of CagA. Front. Cell. Infect. Microbiol. 2, 70. ACCESSION NUMBERS Covacci, A., Falkow, S., Berg, D.E., and Rappuoli, R. (1997). Did the inheri- tance of a pathogenicity island modify the virulence of Helicobacter pylori? The Protein Data Bank accession numbers for the model coordinates Trends Microbiol. 5, 205–208. and structure factors of CagLKKQEK and CagLmeth are 3ZCI and 3ZCJ, Cowtan, K. (2006). The Buccaneer software for automated model building. 1. respectively. Tracing protein chains. Acta Crystallogr. D Biol. Crystallogr. 62, 1002–1011. Derewenda, Z.S. (2004). Rational protein crystallization by mutational surface SUPPLEMENTAL INFORMATION engineering. Structure 12, 529–535.

Supplemental Information includes four figures and one table and Emsley, P., Charles, I.G., Fairweather, N.F., and Isaacs, N.W. (1996). Structure can be found with this article online at http://dx.doi.org/10.1016/j.str.2013. of Bordetella pertussis virulence factor P.69 pertactin. Nature 381, 90–92. 08.018. Emsley, P., Lohkamp, B., Scott, W.G., and Cowtan, K. (2010). Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501. AUTHOR CONTRIBUTIONS Evans, P. (2006). Scaling and assessment of data quality. Acta Crystallogr. D Biol. Crystallogr. 62, 72–82. S. Barden and H.N. designed the research, analyzed the data, and wrote the Fischer, W., Pu¨ ls, J., Buhrdorf, R., Gebert, B., Odenbreit, S., and Haas, R. paper; S. Barden and S.L. performed research; N.T. and S. Backert performed (2001). Systematic mutagenesis of the Helicobacter pylori cag pathogenicity and evaluated complementation experiments with H. pylori; S. Backert, J.C., island: essential genes for CagA translocation in host cells and induction of and N.S. initiated the collaboration and provided reagents. interleukin-8. Mol. Microbiol. 42, 1337–1348. Fronzes, R., Christie, P.J., and Waksman, G. (2009). The structural biology of ACKNOWLEDGMENTS type IV secretion systems. Nat. Rev. Microbiol. 7, 703–714. Goldschmidt, L., Cooper, D.R., Derewenda, Z.S., and Eisenberg, D. (2007). We thank Christina Geerds and Ingelore Brandes for excellent technical assis- Toward rational protein crystallization: A Web server for the design of crystal- tance. We thank the beamline staff at BL14.2 of the BESSY II electron storage lizable protein variants. Protein Sci. 16, 1569–1576. ring (Berlin, Germany) and acknowledge coverage of travel expenses for data collection by the Helmholtz Zentrum Berlin. We gratefully acknowledge access Gorrell, R.J., Guan, J., Xin, Y., Tafreshi, M.A., Hutton, M.L., McGuckin, M.A., to beamline ID23-2 of the ESRF (Grenoble, France) and coverage of travel ex- Ferrero, R.L., and Kwok, T. (2012). A novel NOD1- and CagA-independent penses. The work of S. Backert was supported through two DFG grants pathway of interleukin-8 induction mediated by the Helicobacter pylori type (Ba1671/8-1 and project B10 of CRC-796). IV secretion system. Cell. Microbiol. 15, 554–570. Hauck, C.R., Agerer, F., Muenzner, P., and Schmitter, T. (2006). Cellular adhe- Received: April 15, 2013 sion molecules as targets for bacterial infection. Eur. J. Cell Biol. 85, 235–242. Revised: August 8, 2013 Hazes, B., and Dijkstra, B.W. (1988). Model building of disulfide bonds in pro- Accepted: August 17, 2013 teins with known three-dimensional structure. Protein Eng. 2, 119–125. Published: September 26, 2013 Holm, L., and Rosenstro¨ m, P. (2010). Dali server: conservation mapping in 3D. Nucleic Acids Res. 38(Web Server issue), W545-9. REFERENCES Hubbard, S.J., and Thornton, J.M. (1993). NACCESS Computer Program. Adams, P.D., Afonine, P.V., Bunko´ czi, G., Chen, V.B., Davis, I.W., Echols, N., (Department of Biochemistry and Molecular Biology, University College Headd, J.J., Hung, L.W., Kapral, G.J., Grosse-Kunstleve, R.W., et al. (2010). London). PHENIX: a comprehensive Python-based system for macromolecular struc- Hynes, R.O. (2002). Integrins: bidirectional, allosteric signaling machines. Cell ture solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221. 110, 673–687.

1940 Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved Structure Structure of HP CagL Reveals a Helical RGD Motif

Jime´ nez-Soto, L.F., Kutter, S., Sewald, X., Ertl, C., Weiss, E., Kapp, U., Rohde, Pape, T., and Schneider, T.R. (2004). HKL2MAP: a graphcial user interface for M., Pirch, T., Jung, K., Retta, S.F., et al. (2009). Helicobacter pylori type IV phasing with SHELX programs. J. Appl. Cryst. 37, 843–844. secretion apparatus exploits beta1 integrin in a novel RGD-independent Pham, K.T., Weiss, E., Jime´ nez Soto, L.F., Breithaupt, U., Haas, R., and manner. PLoS Pathog. 5, e1000684. Fischer, W. (2012). CagI is an essential component of the Helicobacter pylori Johnson, W.C., Jr. (1990). Protein secondary structure and circular dichroism: Cag type IV secretion system and forms a complex with CagL. PLoS ONE 7, a practical guide. Proteins 7, 205–214. e35341. Kabsch, W. (1976). Solution for best rotation to relate 2 sets of vectors. Acta Ruoslahti, E. (1996). RGD and other recognition sequences for integrins. Annu. Crystallogr. A 32, 922–923. Rev. Cell Dev. Biol. 12, 697–715. Kabsch, W. (2010). Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132. Saha, A., Backert, S., Hammond, C.E., Gooz, M., and Smolka, A.J. (2010). Helicobacter pylori CagL activates ADAM17 to induce repression of the gastric Kagawa, T.F., Cooney, J.C., Baker, H.M., McSweeney, S., Liu, M., Gubba, S., H, K-ATPase alpha subunit. Gastroenterology 139, 239–248. Musser, J.M., and Baker, E.N. (2000). Crystal structure of the zymogen form of the group A Streptococcus virulence factor SpeB: an integrin-binding cysteine Schneider, S., Carra, G., Sahin, U., Hoy, B., Rieder, G., and Wessler, S. (2011). protease. Proc. Natl. Acad. Sci. USA 97, 2235–2240. Complex cellular responses of Helicobacter pylori-colonized gastric adeno- carcinoma cells. Infect. Immun. 79, 2362–2371. Kaplan-Tu¨ rko¨ z, B., Jime´ nez-Soto, L.F., Dian, C., Ertl, C., Remaut, H., Louche, Schu¨ rpf, T., Chen, Q., Liu, J.H., Wang, R., Springer, T.A., and Wang, J.H. A., Tosi, T., Haas, R., and Terradot, L. (2012). Structural insights into (2012). The RGD finger of Del-1 is a unique structural feature critical for integrin Helicobacter pylori oncoprotein CagA interaction with b1 integrin. Proc. Natl. binding. FASEB J. 26, 3412–3420. Acad. Sci. USA 109, 14640–14645. Shaffer, C.L., Gaddy, J.A., Loh, J.T., Johnson, E.M., Hill, S., Hennig, E.E., Kutter, S., Buhrdorf, R., Haas, J., Schneider-Brachert, W., Haas, R., and McClain, M.S., McDonald, W.H., and Cover, T.L. (2011). Helicobacter pylori Fischer, W. (2008). Protein subassemblies of the Helicobacter pylori Cag exploits a unique repertoire of type IV secretion system components for pilus type IV secretion system revealed by localization and interaction studies. assembly at the bacteria-host cell interface. PLoS Pathog. 7, e1002237. J. Bacteriol. 190, 2161–2171. Sheldrick, G.M. (2010). Experimental phasing with SHELXC/D/E: combining Kwok, T., Zabler, D., Urman, S., Rohde, M., Hartig, R., Wessler, S., Misselwitz, chain tracing with density modification. Acta Crystallogr. D Biol. Crystallogr. R., Berger, J., Sewald, N., Ko¨ nig, W., and Backert, S. (2007). Helicobacter 66, 479–485. exploits integrin for type IV secretion and kinase activation. Nature 449, 862–866. Shi, M., Zhu, J., Wang, R., Chen, X., Mi, L., Walz, T., and Springer, T.A. (2011). Latent TGF-b structure and activation. Nature 474, 343–349. Landegren, U. (1984). Measurement of cell numbers by means of the endog- Singh, P., Carraher, C., and Schwarzbauer, J.E. (2010). Assembly of fibro- enous enzyme hexosaminidase. Applications to detection of lymphokines nectin extracellular matrix. Annu. Rev. Cell Dev. Biol. 26, 397–419. and cell surface antigens. J. Immunol. Methods 67, 379–388. Sledz, P., Zheng, H., Murzyn, K., Chruszcz, M., Zimmerman, M.D., Chordia, Leahy, D.J., Aukhil, I., and Erickson, H.P. (1996). 2.0 A crystal structure of a M.D., Joachimiak, A., and Minor, W. (2010). New surface contacts formed four-domain segment of human fibronectin encompassing the RGD loop upon reductive lysine methylation: improving the probability of protein crystal- and synergy region. Cell 84, 155–164. lization. Protein Sci. 19, 1395–1404. Leiss, M., Beckmann, K., Giro´ s, A., Costell, M., and Fa¨ ssler, R. (2008). The role Stewart, P.L., and Nemerow, G.R. (2007). Cell integrins: commonly used of integrin binding sites in fibronectin matrix assembly in vivo. Curr. Opin. Cell receptors for diverse viral pathogens. Trends Microbiol. 15, 500–507. Biol. 20, 502–507. Tegtmeyer, N., Hartig, R., Delahay, R.M., Rohde, M., Brandt, S., Conradi, J., Logan, D., Abu-Ghazaleh, R., Blakemore, W., Curry, S., Jackson, T., King, A., Takahashi, S., Smolka, A.J., Sewald, N., and Backert, S. (2010). A small fibro- Lea, S., Lewis, R., Newman, J., Parry, N., et al. (1993). Structure of a major nectin-mimicking protein from bacteria induces cell spreading and focal adhe- immunogenic site on foot-and-mouth disease virus. Nature 362, 566–568. sion formation. J. Biol. Chem. 285, 23515–23526. McCoy, A.J., Grosse-Kunstleve, R.W., Adams, P.D., Winn, M.D., Storoni, L.C., Ulanova, M., Gravelle, S., and Barnes, R. (2009). The role of epithelial integrin and Read, R.J. (2007). Phaser crystallographic software. J. Appl. Cryst. 40, receptors in recognition of pulmonary pathogens. J. Innate Immun. 1, 4–17. 658–674. Walter, T.S., Meier, C., Assenberg, R., Au, K.F., Ren, J., Verma, A., Nettleship, Mueller, U., Darowski, N., Fuchs, M.R., Fo¨ rster, R., Hellmig, M., Paithankar, J.E., Owens, R.J., Stuart, D.I., and Grimes, J.M. (2006). Lysine methylation as a K.S., Pu¨ hringer, S., Steffien, M., Zocher, G., and Weiss, M.S. (2012). routine rescue strategy for protein crystallization. Structure 14, 1617–1622. Facilities for macromolecular crystallography at the Helmholtz-Zentrum Wiedemann, T., Hofbaur, S., Tegtmeyer, N., Huber, S., Sewald, N., Wessler, Berlin. J. Synchrotron Radiat. 19, 442–449. S., Backert, S., and Rieder, G. (2012). Helicobacter pylori CagL dependent in- Niesen, F.H., Berglund, H., and Vedadi, M. (2007). The use of differential scan- duction of gastrin expression via a novel avb5-integrin-integrin linked kinase ning fluorimetry to detect ligand interactions that promote protein stability. signalling complex. Gut 61, 986–996. Nat. Protoc. 2, 2212–2221. Yeo, H.J., Yuan, Q., Beck, M.R., Baron, C., and Waksman, G. (2003). Structural Odenbreit, S., Pu¨ ls, J., Sedlmaier, B., Gerland, E., Fischer, W., and Haas, R. and functional characterization of the VirB5 protein from the type IV secretion (2000). Translocation of Helicobacter pylori CagA into gastric epithelial cells system encoded by the conjugative plasmid pKM101. Proc. Natl. Acad. Sci. by type IV secretion. Science 287, 1497–1500. USA 100, 15947–15952. Painter, J., and Merritt, E.A. (2006). TLSMD web server for the generation of Zubieta, C., Schoehn, G., Chroboczek, J., and Cusack, S. (2005). The structure multi-group TLS models. J. Appl. Cryst. 39, 109–111. of the human adenovirus 2 penton. Mol. Cell 17, 121–135.

Structure 21, 1931–1941, November 5, 2013 ª2013 Elsevier Ltd All rights reserved 1941