Published November 16, 2011, doi:10.4049/jimmunol.1102394 The Journal of Immunology

Stable Coordination of the Inhibitory Ca2+ Ion at the Metal Ion-Dependent Adhesion Site in CD11b/CD18 by an Antibody-Derived Ligand Aspartate: Implications for Integrin Regulation and Structure-Based Drug Design

Bhuvaneshwari Mahalingam,*,† Kaouther Ajroud,†,1 Jose´ Luis Alonso,† Saurabh Anand,† Brian D. Adair,*,† Alberto L. Horenstein,‡ Fabio Malavasi,‡ Jian-Ping Xiong,*,† and M. Amin Arnaout*,†

A central feature of integrin interaction with physiologic ligands is the monodentate binding of a ligand carboxylate to a Mg2+ ion hexacoordinated at the metal ion-dependent adhesion site (MIDAS) in the integrin A domain. This interaction stabilizes the A domain in the high-affinity state, which is distinguished from the default low-affinity state by tertiary changes in the domain that culminate in . Small molecule ligand-mimetic integrin antagonists act as partial agonists, eliciting similar activating conformational changes in the A domain, which has contributed to paradoxical adhesion and increased patient mortality in large clinical trials. As with other ligand-mimetic integrin antagonists, the function-blocking mAb 107 binds MIDAS of integrin CD11b/ CD18 A domain (CD11bA), but in contrast, it favors the inhibitory Ca2+ ion over the Mg2+ ion at MIDAS. We determined the crystal structures of the Fab fragment of mAb 107 complexed to the low- and high-affinity states of CD11bA. Favored binding of the Ca2+ ion at MIDAS is caused by the unusual symmetric bidentate ligation of a Fab-derived ligand Asp to a heptacoordinated MIDAS Ca2+ ion. Binding of the Fab fragment of mAb 107 to CD11bA did not trigger the activating tertiary changes in the domain or in the full-length integrin. These data show that the denticity of the ligand Asp/Glu can modify the divalent cation selectivity at MIDAS and hence integrin function. Stabilizing the Ca2+ ion at MIDAS by bidentate ligation to a ligand Asp/Glu may provide one approach for designing pure integrin antagonists. The Journal of Immunology, 2011, 187: 000–000.

ntegrins are a/b heterodimeric adhesion receptors that antagonists developed based on the structures of natural integrin couple the extracellular matrix or counter-receptors on other ligands display agonist-like activities (5–7), which have contributed I cells with the contractile cytoskeleton, transducing mecha- to adverse autoimmune reactions and to paradoxical increased mor- nochemical signals across the plasma membrane that regulate tality in treated patients (4, 8, 9), limiting their use and reflecting the most cellular functions (1). Deregulation of integrin functions, how- need for a better understanding of the structure–activity relation- ever, plays critical roles in a diverse range of diseases, including in- ships in these conformationally dynamic receptors. flammatory and vascular diseases and tumor metastasis, establish- At the core of integrin interaction with physiologic ligands is ing as potential therapeutic targets (2–4). Small molecule a force-bearing Asp (or Glu)–Mg2+ ion bond (10), with Asp/Glu derived from the ligand and the metal ion from a GTPase-like von Willebrand factor type A domain present in the integrin a (aAor *Structural Biology Program, Division of Nephrology, Massachusetts General Hos- I domain) and/or b (bA or I-like domain) subunits (Fig. 1) (11). † pital, Harvard Medical School, Charlestown, MA, 02129; Leukocyte Biology and In solved structures of complexes of integrins with natural li- Inflammation Program, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129; and ‡Laboratory of Immunoge- gands, ligand mimetics, or pseudo-ligands (12–18), the metal ion netics, University of Torino Medical School, Torino, Italy is coordinated at the metal ion-dependent adhesion site (MIDAS), 1Current address: Department of Neurology and Clinical Neurosciences, Northwest- which replaces the catalytic site of GTPases. Side chain oxygen ern University Feinberg School of Medicine, Chicago, IL. atoms from three surface loops in the A domain coordinate the Received for publication August 18, 2011. Accepted for publication October 19, MIDAS metal ion, with the ligand-derived Asp/Glu binding mo- 2011. nodentately to complete the hexacoordinated Mg2+ ion (19–21); it This work was supported by grants from the National Institutes of Health (National is replaced by a water molecule in the unliganded structure (Fig. Institute of Diabetes and Digestive and Kidney Diseases). 1B,1C). Formation of the Asp/Glu–Mg2+ bond in aA domains is The atomic coordinates and structure factors presented in this article have been de- posited in the Data Bank (http://www.pdb.org) under accession codes 3Q3G coupled mechanically to a conformational switch of the domain for the low-affinity CD11bA/Fab 107 complex and 3QA3 for the high-affinity from the default low-affinity (closed) state to the high-affinity CD11bA/mAb107 complex. (open) state, which includes a 180˚ flip of a conserved Gly243, Address correspondence and reprint requests to Prof. M. Amin Arnaout, Division leading to the downward axial displacement of the C-terminal a7 of Nephrology, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129. E-mail address: [email protected] helix on the opposite pole of MIDAS (Fig. 1A). This movement Abbreviations used in this article: CD11bA, integrin CD11b/CD18 A domain; Fab enables aA to engage the bA MIDAS through an invariant Glu 107, Fab fragment of mAb 107; MIDAS, metal ion-dependent adhesion site; RMSD, at the C terminus of the a7 helix (22), thus translating ligand root-mean-square deviation; RU, resonance unit; scFv, single-chain variable frag- occupancy in aA into quaternary changes downstream, leading to ment; WT, wild-type. outside-in signaling and cell adhesion (23). In the aA-lacking 2+ Copyright 2011 by The American Association of Immunologists, Inc. 0022-1767/11/$16.00 integrin subgroup, extrinsic ligands directly bind the Mg ion

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1102394 2 ACa2+ AT MIDAS IN CD11bA/Fab COMPLEX at the bA MIDAS (19), initiating similar activating conforma- Formation of CD11bA/Fab 107 complexes tional changes. Human low-affinity (closed) CD11bA was mixed with Fab 107 in a 1.5:1.0 In addition to the role of the above conformational changes in molar ratio for 15 min on ice in 20 mM Tris buffer (pH 8.2) containing 2 mM integrin affinity modulation, it also is established that integrin– CaCl2. High-affinity CD11bA was incubated with Fab 107 in the same ligand interactions are critically dependent on the nature of the molar ratio for 60 min on ice in 20 mM Tris buffer (pH 8.2) containing 5 divalent cation at MIDAS. Solved crystal structures of closed (24) mM MgCl2. Each mixture then was applied to a Superdex 200 10/300 GL column (Pharmacia Biotech) using a BioLogic DuoFlow FPLC system and liganded (12–14) aA domains and of integrin ectodomains (Bio-Rad) at a flow rate of 0.4 ml/min at 4˚C. The elution profiles were complexed to natural ligands or ligand mimetics (16, 17, 19, 21) monitored in-line by UV absorbance at 280 nm. The eluted peaks were confirmed the presence of Mg2+ (or Mn2+) but not Ca2+ at MIDAS, analyzed by 12% SDS-PAGE under nonreducing conditions, followed by although Mg2+ and Ca2+ are present in equimolar concentrations in Coomassie staining, and fractions containing CD11bA/Fab 107 complex were pooled, concentrated, and used for protein crystallization. circulating plasma. This preference is related to the octahedral environment at MIDAS that favors Mg2+ over Ca2+ (25), ac- Crystallography, structure determination, and refinement counting for the critical dependence of integrin–ligand inter- 2+ The low-affinity CD11bA/Fab 107 complex (at 10 mg/ml) in 20 mM Tris actions on Mg at MIDAS (26–29). All of the previous studies in buffer (pH 8.2) containing 2 mM CaCl2 and the high-affinity CD11bA/Fab integrins have emphasized the charge of the ligand Asp/Glu as 107 complex (at 5 mg/ml) in 20 mM Tris buffer (pH 8.2) containing 5 mM a critical contributor in metal binding and selectivity at MIDAS. MgCl2 with 1 mM PMSF were crystallized at room temperature by vapor However, the Asp or Glu side chains are unique among the natural diffusion using the hanging drop method. Crystals used for data collection were grown from reservoir solutions containing 15% polyethylene glycol amino acids in possessing a carboxylate group that can ligate the 4000, 50 mM Tris buffer (pH 8.2), and 0.3 M NaCl (for the low-affinity metal ion via one or both of the carboxylate oxygen atoms. CD11bA/Fab 107 complex) and 13% polyethylene glycol 8000, 50 mM However, despite this unique feature, the possibility that the den- Tris buffer (pH 8.2), 0.25 M NaCl, and 10 mM CaCl2 (for the high-affinity ticity of the ligand Asp/Glu also may modulate metal ion selec- CD11bA/Fab 107 complex). Crystals were cryoprotected in reservoir so- lution containing 24% glycerol. Data were collected at the Advanced tivity and function in integrins has not been considered previously. Photon Source (Chicago, IL) at beam line 19ID and processed using The primate-specific and function-blocking mAb 107 binds HKL2000 (38). A low-resolution model of the low-affinity CD11bA/Fab with nanomolar affinity to isolated CD11bA in solution or in the 107 complex that was available in our laboratory was used for molecular context of the full-length CD11b/CD18 integrin in leukocytes replacement using Phaser (39). Rigid-body refinement was followed by the (30). Like ligand-mimetic antagonists, mAb 107 binds at MIDAS autobuild procedure in Phenix (40), with noncrystallographic symmetry 2+ 2+ averaging for the four molecules in the asymmetric unit. The final refined of CD11bA, but in contrast it favors Ca over Mg there (31). model was obtained after iterative rounds of model building in Coot (41), To elucidate the structural basis for this unusual metal ion se- followed by refinement in Buster (42). The refined low-affinity CD11bA/ lectivity and its potential impact on integrin conformation and Fab 107 complex was used as a starting model for the refinement of the function, we cocrystallized the Fab fragment of mAb 107 (Fab high-affinity CD11bA/Fab 107 complex. Each of the four molecules in this complex also contained a Ca2+ ion at MIDAS. Ca2+ rather than Mg2+ was 107) complexed with the low- (closed) (32) or high-affinity placed at MIDAS in this case, although both metal ions were present in (open) (12) forms of CD11bA. Both structures reveal stable co- the crystallization buffer, because placing Mg2+ instead of Ca2+ resulted in ordination of a Ca2+ ion at MIDAS, the result of an unexpected bidentate binding by a ligand-mimetic Asp derived from Fab 107, with the absence of ligand-induced conformational changes. Consistently, Fab 107 abolished agonist-induced conformational Table I. Statistics of X-ray diffraction data collection and structure refinement changes in native CD11b/CD18 in in physiologic metal ion conditions. The significance of these findings is dis- Data collection statistics cussed. 316 Protein WT Ile Gly Space group P212121 P212121 Unit cell (a, b, c)(A˚ ) 81.9, 157.2, 82.6, 158.3, Materials and Methods 232.1 233.3 Reagents, cells, and culture (a, b, g) (deg) 90, 90, 90 90, 90, 90 Redundancy 4.2/2.5a 5.9/5.1a Restriction and modification enzymes were obtained from New England Wavelength (A˚ ) 0.97918 0.97934 Biolabs, Life Technologies, or Fisher Scientific. K562 cells (American Type Resolution (A˚ ) 2.7 3.0 Culture Collection) stably expressing wild-type (WT) (low-affinity) or a a 316 Completeness (%) 97.2/80.8 99.1/97.9 high-affinity CD11b/CD18 [induced by an Ile Gly mutation in the a7 I/s(I) 11.3/1.2a 10.9/2.7a helix (31, 33)] were generated as described (34). All of the cell culture R (%)b 10.6/76.0a 15.6/81.4a reagents were from Invitrogen. Fab 107 was generated by digesting sym affinity-purified mAb 107 (IgG1) with immobilized papain, the immobi- Refinement statistics lized enzyme then was removed, and the Fab-containing supernatant was Molecule/asymmetric unit 4 4 applied to protein A to remove undigested material. The single-chain Protein atoms 19752 19631 variable fragment (scFv) form of mAb 107 was generated as described Water 320 75 (31). The CD11b/CD18 heterodimer-specific mAb IB4 (IgG2a) (35) was Ca2+ 44 obtained from the American Type Culture Collection, and the b2 integrin RMSD bond lengths (A˚ ) 0.009 0.009 activation reporter mAb 24 (IgG1), which binds to bA when ligated by aA RMSD bond angles (deg) 1.19 1.18 (36, 37), was from Abcam. Isotype control Abs MOPC-21 (IgG1) and Average B factor (A˚ 2) 54.0 49.8 MOPC-173 (IgG2a) were from BD Pharmingen, and allophycocyanin- R (%)c 21.1 19.4 conjugated goat anti-mouse Fc-specific Ab was from Jackson Immuno- work R (%)d 24.4 22.4 Research Laboratories. free Ramachandran plot Site-directed mutagenesis Favored (%) 96.3 95.3 Outliers (%) 0.6 0.9

Amino acid substitutions were generated using PCR-based mutagenesis, and a authenticity was confirmed by DNA sequencing. The rat-to-human knock- Numbers correspond to the last resolution shell. bR = S|I 2 ÆIæ|/SI, where I is the observed intensity and ÆIæ is the mean in (Asp178/Glu178,Gln279-Arg282/Lys279-Gln282,Arg203-Asn206-Lys210/Thr203- sym 206 210 178 203 206 210 178 203 206 210 intensity from multiple observations of symmetry-related reflections. Leu -His ,andAsp -Arg -Asn -Lys /Glu -Thr -Leu -His ) c Rwork = Shkl|Fo(hkl) 2 Fc(hkl)|/ShklFo(hkl). 107 d chimeras were generated in cDNA encoding WT rat CD11bA. Asp Gly and Rfree is the R value obtained for a test set of reflections consisting of a randomly Glu101Gly substitutions were generated in cDNA encoding WT scFv 107. selected 5% subset of the data set excluded from refinement. The Journal of Immunology 3 much lower B factors for Mg2+ than the surrounding atoms lying within fluorescence intensity was determined using the BD Cellquest program. a radius of 10 A˚ . Binding of mAb 24 was presented as a percentage of mAb IB4 binding. To assess the effect of mAb 107 on mAb 24 binding, K562 cells expressing WT (low-affinity) or constitutively active (high-affinity) CD11b/CD18 were Surface plasmon spectroscopy preincubated with a saturating concentration of the unlabeled Fab 107 (75 Surface plasmon resonance spectroscopy (BIAcore AB, Uppsala, Sweden) mg/ml) for 30 min at 37˚C followed by the addition of mAb 24 IgG for an was used to measure the binding parameters for the interaction of human or extra 20 min at the same temperature. Cells then were washed and stained rat WT (low-affinity) CD11bA, high-affinity CD11bA, and rat-to-human with the secondary Fc-specific fluorophore-labeled Ab, and fluorescence CD11bA chimeras with the IgG or scFv forms of mAb 107. mAb 107 was measured by a FACSCalibur flow cytometer. was coupled covalently via primary amines to the dextran matrix of a CM5 mAb binding to human neutrophils sensor chip as directed by the manufacturer. Chips treated in the same way with BSA (Sigma-Aldrich) or with no protein were used as negative Purified human neutrophils were isolated as described (30) from normal controls. CD11bA in various concentrations was passed over the CM5 volunteers through an Institutional Review Board-approved human sub- sensor chips at a flow rate of 5 ml/min at room temperature in TBS con- jects protocol. Neutrophils were resuspended at a concentration of 1 3 107 taining 1 mM CaCl2, 1 mM MgCl2, and 0.005% P20. The chip surface was cells/ml in HBSS medium (Life Technologies) containing 1% globulin- regenerated by 10 mM HCl. All of the data were analyzed using the free BSA (HBSS-BSA). A total of 1 3 106 cells in 100 ml HBSS-BSA BIAcore T100 evaluation program, after subtracting binding to the BSA- buffer containing 1 mM each of CaCl2 and MgCl2 were left untreated or coupled flow cell to discount the minimal nonspecific binding. For each treated with FMLF (5 3 1027 M final concentration) for 10 min at 37˚C. sensorgram, the peak response in the steady-state region was plotted Unlabeled Fab 107 was added to one half of the replicate tubes for an against the analyte concentration, and the plot was fitted to a single-site additional 30 min, followed by the addition of mAb 24 to all of the tubes binding equation (Langmuir isotherm) to determine the association and and incubation for an extra 20 min. Cells then were washed once, stained dissociation rate constants. with the Fc-specific fluorophore-labeled Ab (for 30 min at 0˚C), washed again, then fixed with 1% formaldehyde in PBS, and analyzed by flow cytometry as described above. mAb binding to recombinant CD11b/CD18 K562 cells stably expressing low- or high-affinity (Ile316Gly) CD11b/CD18 Results were maintained in IMDM plus 10% heat-inactivated FBS, 100 IU/ml Crystal structure of Fab 107 in complex with low-affinity penicillin, 100 mg/ml streptomycin, and 0.5–1.0 mg/ml G418. Low- or CD11bA high-affinity CD11b/CD18-expressing K562 cells (1 3 106 cells) were stained in suspension with mAbs IB4 or 24 (at 20 mg/ml) in 100 ml TBS The crystallographic data and refinement statistics for the low- containing 1 mM CaCl2 and 1 mM MgCl2 in 1% globulin-free BSA for 30 affinity CD11bA/Fab 107 complex are given in Table I. Each of min at room temperature or 37˚C. After being washed, cells were incu- the four molecules in the asymmetric unit of the complex con- bated with the fluorophore-conjugated anti-mouse Fc Ab (for 30 min at 2+ 0˚C), washed, and fixed with 1% formaldehyde, and fluorescence was tained a Ca ion at MIDAS. The positional root-mean-square measured by a FACSCalibur flow cytometer (BD Biosciences), and mean deviation (RMSD) between the main chains of all four CD11bA/

FIGURE 1. Structures of low- and high-affinity forms of human CD11bA and the corresponding changes in metal ion coordination at MIDAS. A, Ribbon diagram of the superimposed crystal structures of low- (cyan) (1jlm.pdb) and high-affinity (gray) (1ido.pdb) forms of CD11bA with the MIDAS metal ion shown in the respective colors. The conformational switch from the low- to high-affinity state, which reshapes MIDAS, involves a 2 A˚ inward movement of the a1 helix and the metal ion and a 180˚ flip of the Ca atom of Gly243 (in red) of MIDAS loop 3, forcing the solvent exposure of buried residues Phe275 and Phe302 and a 10 A˚ axial downward displacement of the C-terminal a7 helix (in red) (direction of these movements is shown by arrows). Buried and exposed side chains of F302 and F275 are in cyan and red, respectively. B and C, The three MIDAS loops (–L3) provide, respectively, the motif D140xS142xS144 (where x is any amino acid), T209, and D242. In the low-affinity CD11bA conformation (B), the two serines and D242 make direct bonds to the metal ion, while T209 makes indirect contact via a water molecule (v1) (in the T209-v1-metal-D242 closed MIDAS configuration); two other water molecules (v2 and v3) complete the octahedral coordination sphere of the MIDAS Mg2+ (or Mn2+) ion. In the high-affinity conformation (C), D242 moves from the primary to the secondary coordination sphere, increasing the net positive charge near the MIDAS ion, thus enhancing its electrostatic monodentate in- teraction with the ligand carboxylate, which replaces water molecule v3. The MIDAS metal coordination thus switches to the T209-metal-v1-D242 open MIDAS configuration. Coordinating residues and water molecules are shown as ball-and-stick representations with green carbon and red oxygen atoms. The ligand Glu is in gold. Hydrogen and metal ion bonds are represented with dashed red lines. All of the molecular graphics images were generated using the UCSF Chimera package. 4 ACa2+ AT MIDAS IN CD11bA/Fab COMPLEX

Fab complexes is small, being 0.14–0.18 A˚ for CD11bA, 0.45– 0.99 and 0.70–0.76 A˚ for the Fab L and H chains, respectively, and 0.59–1.34 A˚ for the complex. When compared with natural ligands and ligand mimetics (Fig. 1C), a novel feature of the interaction of Fab 107 with low-affinity CD11bA is that a Ca2+ ion is present at MIDAS, coordinated by seven oxygen atoms forming a pentagonal bipyramid, with five oxygen atoms lying in the equatorial plane and two oxygen atoms in the axial (apical) positions (Fig. 2). Two equatorial oxygen atoms belong to a bidentate carboxylate from Asp107 of the VH chain 3 (VH3) loop (Fig. 2B). This bidentate binding mode by the ligand Asp was not associated with the activating confor- mational changes normally triggered by the ligation of MIDAS in A domains (Fig. 2A) and contrasts with all of the previous in- tegrin structures where the ligand Asp/Glu binds MIDAS metal monodentately and elicits activating conformational changes in the integrin. In addition to the Asp107–MIDAS Ca2+ ion contact, the mAb epitope comprises residues in two MIDAS ion-coordinating loops and the short BC loop, with important contacts involving Arg208, Phe246,andGlu178-Glu179 in CD11bA (Fig. 3A). Arg208 from MIDAS loop L2 inserts into a cleft between the Fab variable H and L chains and makes a salt bridge with Glu101 of the VH3 loop and side chain and main chain contacts with Tyr109 (from VH3 loop) and Tyr97 (from VL3 loop). Phe246 from MIDAS loop L3 (which also contains the metal-coordinating residue Asp242) in- serts into a hydrophobic pocket walled off by three tyrosines from Fab 107 (Fig. 3A). Glu178-Glu179 in the BC loop of CD11bA contact Arg52 of the VH2 loop. Binding experiments have shown that the ligand Asp107 and Glu101 of the mAb (Fig. 3B) and Arg208, Phe246, and Glu178-Glu179 of CD11bA (30) are essential for the mAb 107–CD11bA interaction, in agreement with the structure and underscoring the role of these contacts in stabilizing the li- gand Asp–Ca2+ interaction. Further validation of the structure comes from the examination of the sequence of rat CD11bA (which does not bind mAb 107): with the exception of the human- to-rat substitutions Lys279Gln, Thr203Arg, and Glu178Asp, all of the other contact residues are conserved. Kinetic analysis of rat-to- human chimeras showed that swapping the rat residues Gln279, Arg282,Arg203,Asn206,andLys210 with the respective human 279 282 203 206 210 residues Lys , Gln , Thr , Leu , and His did not restore FIGURE 2. Crystal structure of Fab 107 in complex with low-affinity the binding of mAb 107 to the resulting rat-to-human chimeric CD11bA. A, Ribbon representation of the complex. CD11bA and the H 178 protein (data not shown). However, replacing rat Asp with and L chains of Fab 107 are cyan, pink, and yellow, respectively. The li- 178 human Glu in rat CD11bA largely reestablished binding to gand D107 side chain is shown, with oxygen atoms as red spheres, co- mAb 107 (Fig. 3C). This binding became almost identical to that ordinating the MIDAS Ca2+ ion (cyan sphere). Ligated CD11bA assumes with human WT CD11bA when Asp178Glu was combined with the low-affinity conformation (featured by burial of F302 and F275 and Arg203, Asn206,Lys210/Thr203, Leu206, and His210 substitution (Fig. packing of the C-terminal a7 helix against the body of the domain). The 3C). These studies demonstrate that Glu178 is mainly responsible three MIDAS loops (L1–L3) are labeled. Molecular graphics images were 2 for the primate specificity of the mAb. generated using the UCSF Chimera package. B,As-weighted 2Fo Fc density map (blue, contoured at 1.0s) of the MIDAS motif in the complex. Coordinating residues are shown as ball-and-stick representations with Crystal structure of Fab 107 in complex with high-affinity green carbon and red oxygen atoms. Water molecules (v) are depicted as CD11bA red spheres. Hydrogen and metal ion bonds are represented with dashed red lines. The ligand D107 (in gold) bidentately coordinates the MIDAS Structures of the monomeric high-affinity form of the aA domain Ca2+ ion (cyan sphere). The average ion-ligating group distance in this from several integrins in complex with ligands yielded an open con- structure (as well as in the high-affinity CD11bA/Fab 107 complex; see formation (12–14), where a ligand Asp/Glu side chain binds the below) is 2.42–2.51 A˚ . The estimated coordinate error for the models MIDAS Mg2+ (or Mn2+) ion monodentately, completing the octa- (derived from the refinement program Buster) is 0.39–0.41 A˚ , yielding a maximum distance of 2.8 A˚ , in agreement with the symmetric bidentate hedral coordination sphere around the metal and allowing the large 2+ conformational switch in the domain. The crystal structure of Ile316 carboxylate group–Ca distance reported by Harding (56). The orientation of this figure is as in Fig. 1B and 1C. Gly CD11bA in complex with Fab 107 (Fig. 4A, Table I) differs from the previously determined open structure of this domain (12) in two key features: each of the four CD11bA molecules in the asymmetric conformationally active a1anda7 helices (Fig. 1A)inbothlow-and unit adopts the low-affinity conformation (Fig. 4B)andaCa2+ ion is high-affinity CD11bA/Fab 107 complexes make discontinuous crys- again found at MIDAS, bound bidentately by the ligand Asp107.The tal contacts with symmetry-related molecules (combined respective The Journal of Immunology 5

FIGURE 3. The integrin/Fab 107 binding interface and its validation. A, Main interacting residues (shown as stick models) from Fab 107 and low-affinity CD11bA are colored as in Fig. 2A. The interacting MIDAS loops L2 and L3, the BC loop from CD11bA, and VL1, VL3, VH2, and VH3 loops from Fab 107 are labeled. Main chain oxygen and nitrogen atoms of E244 [a key physiologic ligand binding residue in the aA domain of integrin CD11a (14)] are red and blue, respectively. The conformationally sensitive G243 Ca atom is green. Hydrogen and metal ion bonds are represented with dashed red lines. B, Sensorgrams recording the interaction of immobilized WT (2771 resonance units, RU), D107G (5182 RU), or E101G (4344 RU) forms of scFv 107 with human low-affinity CD11bA (1.6 mM) in Ca2+ and Mg2+ buffer (1 mM each). C, Sensorgrams of interaction of immobilized Fab 107 (in 1 mM each of Ca2+ and Mg2+) with low-affinity human (hWT) and rat (rWT) CD11bA and rat-to-human D178E CD11bA (rD178/E) and D178, R203, N206, K210/E178,

T203, L206, and H210 (rDRNK/ETLH) chimeras (0.5 mM). Measurements from two independent experiments yielded Kd values (in nM) of 4.97 6 0.24, 56.46 6 17.2, and 7.57 6 0.22 for hWT, rD178/E, and rDRNK/ETLH, respectively. Fab 107 did not bind to low-affinity rWT. interface areas of 444 and 468 A˚ 2). It is unlikely that such contacts native receptor, binding of mAb 24 to native CD11b/CD18 was prevent the switch into the open conformation upon ligation of examined in the absence or presence of unlabeled Fab 107 after MIDAS by the ligand Asp107, because the CD11bA/Fab 107 com- the preactivation of human neutrophils with FMLF in buffer con- plexes have been obtained by cocrystallization. taining physiologic concentrations of Ca2+ and Mg2+. As shown Our published kinetic studies revealed ∼7-fold lower affinity of in Fig. 5G–I, unlabeled Fab 107 almost abolished the binding of mAb 107 to the open versus closed CD11bA conformation (30). mAb 24 to the native agonist-preactivated integrin. This difference may be explained now by the present structures: Should Fab 107 have approached open CD11bA, Ser144 from 107 Discussion MIDAS loop 1 likely would clash with the ligand Asp , and the The key finding in this report is that the function-blocking mAb 107 side chain of Glu244 (from MIDAS loop 3) would have to be 33 31 binds the MIDAS face of CD11bA in a manner similar to that of sandwiched between Ser and Tyr of the Fab 107 VL1 loop physiologic ligands and current ligand-mimetic antagonists but (Fig. 4C), which is energetically unfavorable. with two notable differences: 1) the mAb-derived ligand Asp107 stabilizes Ca2+ instead of Mg2+ at MIDAS by binding to the metal Effects of mAb 107 binding on conformational changes in the ion bidentately instead of the usual monodentate binding mode holoreceptor seen with ligand–Mg2+ binding, and 2) ligation of MIDAS by the The effect of mAb107 binding on conformational switching of mAb did not elicit the conformational changes in CD11bA or in the domain in the context of the full-length cellular integrin was the holoreceptor that normally lead to cell adhesion. examined next. We used the binding of mAb 24 as the reporter of Of the two divalent cations Mg2+ and Ca2+ that are abundant in the conformational switch from the low- to high-affinity state in peripheral blood, the octahedral environment at MIDAS is suited the A domain (36, 37). Binding of Fab 107 to recombinant WT ideally for binding Mg2+ (43). This selectivity is perhaps reflective (low-affinity) CD11b/CD18, stably expressed on K562 cells, in of the evolutionary origin of the extracellular A domains from an buffer containing physiologic concentrations of Ca2+ and Mg2+ ancient intracellular von Willebrand factor type A domain, where (1 mM each), failed to switch the integrin to the high-affinity Mg2+ is more abundant (44). Fitting the larger Ca2+ (ionic radius conformation; instead, baseline binding of mAb 24 to the integ- 1.0 A˚ ) (45) over Mg2+ (ionic radius 0.72 A˚ ) in the octahedral rin was reduced significantly (Fig. 5A–C). Thus, binding of mAb environment of MIDAS has been shown to be thermodynamically 107 to WT CD11bA MIDAS in the full-length integrin does unfavorable and would result in significant structural rearrange- not elicit the agonist-like activities observed upon the binding ment of the surroundings and in a decrease in the affinity for the of peptide or nonpeptide small molecule ligand mimetics to integ- natural ligand (25). rins (4). The bidentate coordination mode of the MIDAS metal explains We have shown previously that replacing the invariant Ile316 the higher affinity of Ca2+ over Mg2+ at MIDAS in low- or high in the C-terminal a7 helix with Gly in CD11bA generates a con- affinity CD11bA/Fab 107 complexes when measured by titration stitutively active integrin (31) by destabilizing the hydrophobic calorimetry [40 mM versus 1.0 mM in the low-affinity CD11bA/ packing of the a7 helix against the body of the domain. Binding of mAb107 complex and 25 versus 100 mM in the high-affinity Fab 107 to this Ile316Gly high-affinity receptor stably expressed on CD11bA/scFv107 complex (31)]. The derived energy difference K562 cells in similar buffer conditions produced a small but of 2.0 kcal/mol in metal ion binding to CD11bA in solution (31) significant reduction in mAb 24 binding to the integrin (Fig. 5D– also is in the range of the energy difference found between mono- F). To assess the effect of bound Fab 107 on the activation of the dentate and bidentate coordination modes in other metalloproteins 6 ACa2+ AT MIDAS IN CD11bA/Fab COMPLEX

receptor, as reported by mAb 24 (Fig. 5A–C). This finding is con- sistent with the crystal structure of the low-affinity CD11bA/Fab 107 complex (Fig. 2A,2B). Fab 107 also significantly inhibited binding of mAb 24 to the constitutive high-affinity receptor stably expressed on K562 cells, but the effect was rather small (Fig. 5D– F). This may be explained by the constitutive contacts that the mutationally stabilized high-affinity CD11bA is expected to make with bA in the context of the heterodimer (22), which are detected by mAb 24. These contacts are absent when high-affinity CD11bA is present in isolation in solution, where it can exist in an equilib- rium that allows some of the molecules to populate the low-affinity conformation (47, 48). In the more physiologic situation where the native integrin is activated by agonists such as FMLF, subsequent binding of Fab 107 prevented the conformational switch of CD11bA to the high-affinity state (Fig. 5G–I). This blockade may result from the conversion of the physiologically activated native receptor into the low-affinity conformation or from skewing of the conforma- tional equilibrium known to exist in the surface receptor between low- and high-affinity conformations (23) in favor of the former. In support of the latter scenario is the preferential binding of mAb 107 to the low-affinity state (30). Although the importance of heptacoordination in stabilizing Ca2+ at MIDAS is established from the structural and functional data presented, important interactions mainly involving residues Phe246,Arg208,andGlu178 are required for the ligand Asp107 to biden- tately ligate the MIDAS metal ion (Fig. 3B,3C) (30). It could be argued that these interactions account for locking CD11bA in the closed state. Alternatively, bidentate ligation by the ligand Asp to the MIDAS Ca2+ ion, once established, could be the primary factor preventing the activation of the conformational switch usually seen in the structures of monodentately ligated MIDAS metal cations. We favor the latter possibility for a couple of rea- sons. First, the heptacoordination of the MIDAS Ca2+ ion man- dates that Asp242 remains in the primary coordination sphere of the metal ion, preventing the critical activating 180˚ flip of con- served Gly243, which is necessary to switch the domain into the high-affinity state (Fig. 4C). In contrast, no notable change in the main chains of Phe246, Arg208, and Glu178 between inactive and active conformations is observed (Fig. 4C) to affect the confor- mational switch directly. Second, in studies of other metal ion FIGURE 4. Crystal structure of Fab 107 in complex with high-affinity binding sites that similarly contain an inner shell Asp/Glu, the CD11bA and comparison with structures of closed and open CD11bA carboxylate binding mode of the Asp/Glu has been shown to be forms. A, Ribbon representation of the high-affinity CD11bA/Fab 107 complex. D107 coordinates a MIDAS Ca2+ ion (cyan sphere) bidentately, critical not only in the binding affinity and selectivity of the metal but the high-affinity CD11bA assumes the low-affinity (closed) confor- cation but also in regulating the function of metalloproteins (43, mation. Orientation is as shown in Fig. 2A. The observed unraveling of the 49). For example, the switch from the monodentate to bidentate lower segment of the a7 helix (arrow) is caused by the helix-breaking Asp/Glu binding mode, which shifts the metal cation from Mg2+ I316G activating mutation. B, Superimposed crystal structures of low- and to Ca2+, plays an important role in catalysis by several metal- high-affinity CD11bA complexed with Fab 107, cyan and magenta, re- loenzymes (reviewed in Ref. 49). spectively. Orientation is similar to that in A. C, Major interacting residues The bidentate binding mode of the ligand Asp107, which stably on the MIDAS face (shown as stick models) from low-affinity CD11bA/ coordinates Ca2+ at MIDAS, favoring the closed conformation of Fab 107 (white), superimposed on the structure of closed CD11bA alone CD11bA, contrasts with the invariant monodentate coordination (orange) (1.jlm.pdb) and on that of open CD11bA alone (gray) (1ido.pdb). of the MIDAS Mg2+/Mn2+ ion in complexes of aA-containing in- The MIDAS ion is colored in the respective color of each domain. MIDAS loops L1–L3 are labeled. The orientation is the same as that in Fig. 3A. tegrins with physiologic ligands (13, 14, 16, 18). In these struc- Hydrogen and metal ion bonds are represented with dashed red lines. See tures where aA assumes the open conformation, the ligand Asp/ text for details. Glu originates from or at the end of a b strand or a rigid helix (Fig. 6), structural features that may favor the monodentate coordina- tion mode in the octahedral coordination preferred by Mg2+, with (46). Further, formation of the extra carboxylate bond–Ca2+ may Asp242 forced to move to the secondary metal coordination sphere, explain the measured negative binding enthalpy of Ca2+ versus facilitated by the 180˚ flip in the adjacent Gly243 main chain (Fig. Mg2+ to closed and open MIDAS (31). 4C). This integrin ligand adaptation thus may be necessary for Binding studies carried out on K562 cells stably expressing triggering outside-in signaling. Interestingly, examination of the WT full-length CD11b/CD18, which is expressed normally in the available crystal structures of the ligand-mimetic mAbs (20, 21) default low-affinity state on these cells, showed that binding of Fab that ligate the MIDAS ion monodentately and result in an open 107 inhibited rather than induced the activating tertiary changes in the MIDAS configuration reveals that the ligand Asp/Glu originates The Journal of Immunology 7

FIGURE 5. Effects of Fab 107 on binding of the activation reporter mAb 24 to full-length CD11b/CD18. A, Histograms (mean 6 SD, n = 3 independent experiments, each in triplicate) showing the effects of the absence and presence of unlabeled Fab 107 on binding of mAb 24 to the recombinant WT (low- affinity) integrin stably expressed on K562 cells. mAb 24 binding was expressed as a percentage of binding of the heterodimer-specific mAb IB4. The 45% decrease in mAb 24 binding in the presence of Fab 107 was significant at a p value of 0.018. B and C, Flow cytometry analysis of one of the experiments shown in A of K562 cells expressing WT CD11b/CD18 stained with mAb 24 in the absence (B) and presence (C) of Fab 107 or stained with mAb IB4 as a positive control. The reduction in mAb24 binding in the presence of Fab 107 is reflected by the left shift relative to mAb IB4-stained cells. D, Histograms (mean 6 SD of triplicate determinations) of a representative experiment, one of three independent experiments carried out, showing binding of mAb 24 in the absence or presence of Fab 107 to K562 cells stably expressing the constitutively active high-affinity receptor. The 11% decrease is significant (p = 0.025) and ranged from 11 to 16% in the three independent experiments. E and F, Flow cytometry analysis of the experiment shown in D. Note the high baseline binding of mAb 24 to the constitutively active Ile316Gly integrin (compare with that to WT, Fig. 5B). Binding of mAb 24 to K562 cells expressing Ile316Gly CD11b/CD18 is reduced in the presence of Fab 107 (F) when compared to its absence (E). mAb IB4-stained cells represent the internal positive control. G, Histograms (mean 6 SD of triplicate determinations) of a representative experiment, one of four carried out, showing FMLF-induced increased expression of mAb 24 epitope on agonist-activated native integrin on human neutrophils and its inhibition by Fab 107 binding to levels sig- nificantly lower than those found even in untreated neutrophils (p = 0.001, FMLF-treated versus untreated cells). H and I, Flow cytometry analysis of the experiment shown in G. Binding of mAb 24 to FMLF-treated polymorphonuclear neutrophils is reduced markedly in the presence of Fab 107 (I) when compared to its absence (H). mAb IB4-stained polymorphonuclear neutrophils are indicated. All of the binding reactions were carried out in buffer containing physiologic concentrations of Ca2+ and Mg2+ (1 mM each). from shorter VH3 loops (Fig. 6), which also may favor the mon- cally worse outcome owing to increased cardiovascular throm- odentate coordination mode, in comparison with the longer VH3 botic events (54). Such serious side effects underscore the need for loop in Fab 107 that favors bidentate ligation of the MIDAS metal. new approaches in designing integrin antagonists that do not elicit The hookworm-derived natural integrin antagonist in- the activating conformational shifts upon MIDAS ligation. One hibitory factor binds the MIDAS face of CD11bA with nanomolar approach used a small molecule that engages the ligand Arg affinity in the presence of Ca2+, and its binding is blocked by mAb binding site in the propeller in the aA-lacking integrin aIIbb3, 107 but not by other anti-CD11bA mAbs that do not ligate MIDAS thus impairing the ligation of the MIDAS metal by the ligand Asp (50–52). The Ca2+ dependency of this interaction suggests that the in the Arg-Gly-Asp motif (55). The present data suggest that bidentate ligation mode may extend to some natural neutrophil stabilizing the inhibitory Ca2+ ion at MIDAS by inducing a ligand inhibitory factor-like ligand-mimetic integrin antagonists as well. carboxylate monodentate to bidentate switch may provide a pro- Structural studies will be needed to test this prediction. mising novel solution for designing integrin antagonists devoid A major problem with existing ligand-mimetic integrin antag- of partial agonism. onists has been their potential to activate the integrin, thus induc- ing the harmful cell adhesion response that these drugs were Acknowledgments designed to prevent (4, 8, 53). For example, patients treated with We thank Dr. Frank J. Rotella and Dr. Rongguang Zhang for assistance with an oral ligand mimetic targeting platelet aIIbb3 had a paradoxi- data collection and Dr. Vineet Gupta for providing K562 stably expressing 8 ACa2+ AT MIDAS IN CD11bA/Fab COMPLEX

14. Shimaoka, M., T. Xiao, J. H. Liu, Y. Yang, Y. Dong, C. D. Jun, A. McCormack, R. Zhang, A. Joachimiak, J. Takagi, et al. 2003. Structures of the alpha L I domain and its complex with ICAM-1 reveal a shape-shifting pathway for integrin regulation. Cell 112: 99–111. 15. Xiao, T., J. Takagi, B. S. Coller, J. H. Wang, and T. A. Springer. 2004. Structural basis for allostery in integrins and binding to fibrinogen-mimetic therapeutics. Nature 432: 59–67. 16. Song, G., Y. Yang, J. H. Liu, J. M. Casasnovas, M. Shimaoka, T. A. Springer, and J. H. Wang. 2005. An atomic resolution view of ICAM recognition in a complex between the binding domains of ICAM-3 and integrin alphaLbeta2. Proc. Natl. Acad. Sci. USA 102: 3366–3371. 17. Springer, T. A., J. Zhu, and T. Xiao. 2008. Structural basis for distinctive rec- ognition of fibrinogen gammaC peptide by the platelet integrin alphaIIbbeta3. J. Cell Biol. 182: 791–800. 18. Zhang, H., J. M. Casasnovas, M. Jin, J. H. Liu, C. G. Gahmberg, T. A. Springer, and J. H. Wang. 2008. An unusual allosteric mobility of the C-terminal helix of a high-affinity alphaL integrin I domain variant bound to ICAM-5. Mol. Cell 31: 432–437. 19. Xiong, J. P., T. Stehle, R. Zhang, A. Joachimiak, M. Frech, S. L. Goodman, and M. A. Arnaout. 2002. Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Science 296: 151–155. 20. Karpusas, M., J. Ferrant, P. H. Weinreb, A. Carmillo, F. R. Taylor, and E. A. Garber. 2003. Crystal structure of the alpha1beta1 integrin I domain in complex with an FIGURE 6. Mode of interaction of ligand carboxylates in solved struc- antibody Fab fragment. J. Mol. Biol. 327: 1031–1041. tures of aA domains in complex with physiologic ligands or ligand-mi- 21. Zhang, H., J. H. Liu, W. Yang, T. Springer, M. Shimaoka, and J. H. Wang. 2009. metic mAbs. Ribbon diagrams of complexes of CD11bA/Fab 107 (cyan), Structural basis of activation-dependent binding of ligand-mimetic antibody CD49bA/collagen peptide (pink) (1dzi.pdb), CD11aA/ICAM1 (CD54) (yel- AL-57 to integrin LFA-1. Proc. Natl. Acad. Sci. USA 106: 18345–18350. 22. Alonso, J. L., M. Essafi, J. P. Xiong, T. Stehle, and M. A. Arnaout. 2002. Does low) (1mq8.pdb), CD49aA/AQC2 Fab complex (gray) (1mhp.pdb), and the integrin alphaA domain act as a ligand for its betaA domain? Curr. Biol. 12: CD11aA/AL-57 Fab complex (magenta) (3hi6.pdb). Structures of CD11aA R340–R342. complexes with ICAM3 (1top.pdb) and ICAM5 (3bn3.pdb) are super- 23. Arnaout, M. A., B. Mahalingam, and J. P. Xiong. 2005. Integrin structure, al- posable on those of CD11aA/ICAM1 and have not been included to reduce lostery, and bidirectional signaling. Annu. Rev. Cell Dev. Biol. 21: 381–410. 24. Qu, A., and D. J. Leahy. 1996. The role of the divalent cation in the structure of complexity of the figure. The MIDAS metal ions (spheres), the Ca’s the I domain from the CD11a/CD18 integrin. Structure 4: 931–942. (spheres), and side chains of ligand Asp/Glu are shown in their respective 25. San Sebastian, E., J. M. Mercero, R. H. Stote, A. Dejaegere, F. P. Cossı´o, and colors, with oxygen atoms in red. Structures were superimposed on the X. Lopez. 2006. On the affinity regulation of the metal-ion-dependent adhesion CD11bA structure using Matchmaker in Chimera. sites in integrins. J. Am. Chem. Soc. 128: 3554–3563. 26. Dransfield, I., C. Caban˜as, A. Craig, and N. Hogg. 1992. Divalent cation regu- lation of the function of the leukocyte integrin LFA-1. J. Cell Biol. 116: 219– high-affinity CD11b/CD18. The crystal structures shown in this report are 226. derived from work performed at Argonne National Laboratory, Structural 27. Michishita, M., V. Videm, and M. A. Arnaout. 1993. A novel divalent cation- binding site in the A domain of the beta 2 integrin CR3 (CD11b/CD18) is es- Biology Center at the Advanced Photon Source using the beam line 19ID. sential for ligand binding. Cell 72: 857–867. 28. Baldwin, E. T., R. W. Sarver, G. L. Bryant, Jr., K. A. Curry, M. B. Fairbanks, B. C. Finzel, R. L. Garlick, R. L. Heinrikson, N. C. Horton, L. L. Kelley, et al. Disclosures 1998. Cation binding to the integrin CD11b I domain and activation model as- The authors have no financial conflicts of interest. sessment. Structure 6: 923–935. 29. Calderwood, D. A., D. S. Tuckwell, J. Eble, K. Ku¨hn, and M. J. Humphries. 1997. The integrin alpha1 A-domain is a ligand binding site for collagens and laminin. J. Biol. Chem. 272: 12311–12317. References 30. Li, R., I. Haruta, P. Rieu, T. Sugimori, J. P. Xiong, and M. A. Arnaout. 2002. 1. Hynes, R. O. 2002. Integrins: bidirectional, allosteric signaling machines. Cell Characterization of a conformationally sensitive murine monoclonal antibody 110: 673–687. directed to the metal ion-dependent adhesion site face of integrin CD11b. J. 2. Liu, L., B. Schwartz, Y. Tsubota, E. Raines, H. Kiyokawa, K. Yonekawa, Immunol. 168: 1219–1225. J. M. Harlan, and L. M. Schnapp. 2008. Cyclin-dependent kinase inhibitors 31. Ajroud, K., T. Sugimori, W. H. Goldmann, D. M. Fathallah, J. P. Xiong, and block leukocyte adhesion and migration. J. Immunol. 180: 1808–1817. M. A. Arnaout. 2004. Binding Affinity of Metal Ions to the CD11b A-domain Is 3. Yonekawa, K., and J. M. Harlan. 2005. Targeting leukocyte integrins in human Regulated by Integrin Activation and Ligands. J. Biol. Chem. 279: 25483–25488. diseases. J. Leukoc. Biol. 77: 129–140. 32. Lee, J. O., L. A. Bankston, M. A. Arnaout, and R. C. Liddington. 1995. Two 4. Cox, D., M. Brennan, and N. Moran. 2010. Integrins as therapeutic targets: conformations of the integrin A-domain (I-domain): a pathway for activation? lessons and opportunities. Nat. Rev. Drug Discov. 9: 804–820. Structure 3: 1333–1340. 5. Peter, K., I. Ahrens, M. Schwarz, C. Bode, and J. Yla¨nne. 2004. Distinct roles of 33. Xiong, J. P., R. Li, M. Essafi, T. Stehle, and M. A. Arnaout. 2000. An isoleucine- ligand affinity and cytoskeletal anchorage in alphaIIbbeta3 (GP IIb/IIIa)- based allosteric switch controls affinity and shape shifting in integrin CD11b mediated cell aggregation and adhesion. Platelets 15: 427–438. A-domain. J. Biol. Chem. 275: 38762–38767. 6. Peter, K., M. Schwarz, J. Yla¨nne, B. Kohler, M. Moser, T. Nordt, P. Salbach, 34. Gupta, V., J. L. Alonso, T. Sugimori, M. Essafi, J. P. Xiong, and M. A. Arnaout. W. Ku¨bler, and C. Bode. 1998. Induction of fibrinogen binding and platelet 2008. Role of the beta-subunit arginine/lysine finger in integrin heterodimer aggregation as a potential intrinsic property of various glycoprotein IIb/IIIa formation and function. J. Immunol. 180: 1713–1718. (alphaIIbbeta3) inhibitors. Blood 92: 3240–3249. 35. Wright, S. D., P. E. Rao, W. C. Van Voorhis, L. S. Craigmyle, K. Iida, 7. Schwarz,M.,G.Meade,P.Stoll,J.Ylanne, N. Bassler, Y. C. Chen, C. E. Hagemeyer, M. A. Talle, E. F. Westberg, G. Goldstein, and S. C. Silverstein. 1983. Identi- I. Ahrens, N. Moran, D. Kenny, et al. 2006. Conformation-specific blockade of fication of the C3bi receptor of human monocytes and macrophages by using the integrin GPIIb/IIIa: a novel antiplatelet strategy that selectively targets ac- monoclonal antibodies. Proc. Natl. Acad. Sci. USA 80: 5699–5703. tivated platelets. Circ. Res. 99: 25–33. 36. Kamata, T., K. K. Tieu, T. Tarui, W. Puzon-McLaughlin, N. Hogg, and 8. Quinn, M. J., T. V. Byzova, J. Qin, E. J. Topol, and E. F. Plow. 2003. Integrin Y. Takada. 2002. The role of the CPNKEKEC sequence in the beta(2) subunit I alphaIIbbeta3 and its antagonism. Arterioscler. Thromb. Vasc. Biol. 23: 945–952. domain in regulation of integrin alpha(L)beta(2) (LFA-1). J. Immunol. 168: 9. Quinn, M. J., E. F. Plow, and E. J. Topol. 2002. Platelet glycoprotein IIb/IIIa 2296–2301. inhibitors: recognition of a two-edged sword? Circulation 106: 379–385. 37. Stanley, P., A. Smith, A. McDowall, A. Nicol, D. Zicha, and N. Hogg. 2008. 10. Craig, D., M. Gao, K. Schulten, and V. Vogel. 2004. Structural insights into how Intermediate-affinity LFA-1 binds alpha-actinin-1 to control migration at the the MIDAS ion stabilizes integrin binding to an RGD peptide under force. leading edge of the T cell. EMBO J. 27: 62–75. Structure 12: 2049–2058. 38. Otwinowski, Z., and W. Minor. 1997. Processing of X-ray Diffraction Data 11. Arnaout, M. A. 2002. Integrin structure: new twists and turns in dynamic cell Collected in Oscillation Mode. Academic, New York. adhesion. Immunol. Rev. 186: 125–140. 39. McCoy, A. J., R. W. Grosse-Kunstleve, P. D. Adams, M. D. Winn, L. C. Storoni, 12. Lee, J. O., P. Rieu, M. A. Arnaout, and R. Liddington. 1995. Crystal structure of and R. J. Read. 2007. Phaser crystallographic software. J. Appl. Crystallogr. 40: the A domain from the alpha subunit of integrin CR3 (CD11b/CD18). Cell 80: 658–674. 631–638. 40. Adams, P. D., P. V. Afonine, G. Bunko´czi, V. B. Chen, I. W. Davis, N. Echols, 13. Emsley, J., C. G. Knight, R. W. Farndale, M. J. Barnes, and R. C. Liddington. J. J. Headd, L. W. Hung, G. J. Kapral, R. W. Grosse-Kunstleve, et al. 2010. 2000. Structural basis of collagen recognition by integrin alpha2beta1. Cell 101: PHENIX: a comprehensive Python-based system for macromolecular structure 47–56. solution. Acta Crystallogr. D Biol. Crystallogr. 66: 213–221. The Journal of Immunology 9

41. Emsley, P., B. Lohkamp, W. G. Scott, and K. Cowtan. 2010. Features and de- 50. Rieu, P., T. Ueda, I. Haruta, C. P. Sharma, and M. A. Arnaout. 1994. The A- velopment of Coot. Acta Crystallogr. D Biol. Crystallogr. 66: 486–501. domain of beta 2 integrin CR3 (CD11b/CD18) is a receptor for the hookworm- 42. Bricogne, G., E. Blanc, M. Brandl, C. Flensburg, P. Keller, W. Paciorek, P. Roversi, derived neutrophil adhesion inhibitor NIF. J. Cell Biol. 127: 2081–2091. O. S. Smart, C. Vonrhein, and T. Womack. 2009. BUSTER, version 2.8.0. Global 51.Rieu,P.,T.Sugimori,D.L.Griffith,andM.A.Arnaout.1996.Solvent- Phasing Ltd., Cambridge, U.K. accessible residues on the metal ion-dependent adhesion site face of integrin 43. Dudev, T., and C. Lim. 2004. Monodentate versus bidentate carboxylate binding in magnesium and calcium : what are the basic principles? J. Phys. CR3 mediate its binding to the neutrophil inhibitory factor. J. Biol. Chem. 271: Chem. B 108: 4546–4557. 15858–15861. 44. Whittaker, C. A., and R. O. Hynes. 2002. Distribution and evolution of von 52. Zhang, L., and E. F. Plow. 1997. Identification and reconstruction of the binding Willebrand/integrin A domains: widely dispersed domains with roles in cell site within alphaMbeta2 for a specific and high affinity ligand, NIF. J. Biol. adhesion and elsewhere. Mol. Biol. Cell 13: 3369–3387. Chem. 272: 17558–17564. 45. Marcus, Y. 1988. Ionic radii in aqueous solutions. Chem. Rev. 88: 1475–1498. 53. Peter, K., M. Schwarz, T. Nordt, and C. Bode. 2001. Intrinsic activating prop- 46. Ryde, U. 1999. Carboxylate binding modes in zinc proteins: a theoretical study. erties of GP IIb/IIIa blockers. Thromb. Res. 103(Suppl. 1): S21–S27. Biophys. J. 77: 2777–2787. 54. Chew, D. P., D. L. Bhatt, S. Sapp, and E. J. Topol. 2001. Increased mortality with 47. Li, R., P. Rieu, D. L. Griffith, D. Scott, and M. A. Arnaout. 1998. Two functional oral platelet glycoprotein IIb/IIIa antagonists: a meta-analysis of phase III states of the CD11b A-domain: correlations with key features of two Mn2+- multicenter randomized trials. Circulation 103: 201–206. complexed crystal structures. J. Cell Biol. 143: 1523–1534. 55. Zhu, J., J. Zhu, A. Negri, D. Provasi, M. Filizola, B. S. Coller, and T. A. Springer. 48. McCleverty, C. J., and R. C. Liddington. 2003. Engineered allosteric mutants of the integrin alphaMbeta2 I domain: structural and functional studies. Biochem. J. 2010. Closed headpiece of integrin aIIbb3 and its complex with an aIIbb3- 372: 121–127. specific antagonist that does not induce opening. Blood 116: 5050–5059. 49. Dudev, T., and C. Lim. 2007. Effect of carboxylate-binding mode on metal 56. Harding, M. M. 2006. Small revisions to predicted distances around metal sites binding/selectivity and function in proteins. Acc. Chem. Res. 40: 85–93. in proteins. Acta Crystallogr. D Biol. Crystallogr. 62: 678–682.