Background: The activation-specific antibodies against integrins are powerful tools in integrin studies.
Results: An activation-specific antibody J19 against α4β7 was identified and characterized.
Conclusion: J19 specifically binds to the activated α4β7 by recognizing the epitope only exposed in extended conformation.
Significance: J19 is a potentially powerful tool for studying α4β7 function and treatment of α4β7-related inflammatory diseases.
Keywords: Antibody Engineering, Cell Adhesion, Epitope Mapping, Integrin, Phage Display, Activation-specific Antibody, scFv
Abstract
Integrin α4β7 is a lymphocyte homing receptor that mediates both rolling and firm adhesion of lymphocytes on vascular endothelium, two of the critical steps in lymphocyte migration and tissue-specific homing. The rolling and firm adhesions of lymphocytes rely on the dynamic shift between the inactive and active states of integrin α4β7, which is associated with the conformational rearrangement of integrin molecules. Activation-specific antibodies, which specifically recognize the activated integrins, have been used as powerful tools in integrin studies, whereas there is no well characterized activation-specific antibody to integrin α4β7. Here, we report the identification, characterization, and epitope mapping of an activation-specific human mAb J19 against integrin α4β7. J19 was discovered by screening a human single-chain variable fragment phage library using an activated α4β7 mutant as target. J19 IgG specifically bound to the high affinity α4β7 induced by Mn2+, DTT, ADP, or CXCL12, but not to the low affinity integrin. Moreover, J19 IgG did not interfere with α4β7-MAdCAM-1 interaction. The epitope of J19 IgG was mapped to Ser-331, Ala-332, and Ala-333 of β7 I domain and a seven-residue segment from 184 to 190 of α4 β-propeller domain, which are buried in low affinity integrin with bent conformation and only exposed in the high affinity extended conformation. Taken together, J19 is a potentially powerful tool for both studies on α4β7 activation mechanism and development of novel therapeutics targeting the activated lymphocyte expressing high affinity α4β7.
Introduction
Integrins are a family of α/β heterodimeric adhesion receptors that mediate cell-cell, cell-extracellular matrix, and cell-pathogen interactions and transmit signals bidirectionally across plasma membrane (1). Because of their unique function of integrating extracellular environment with cytoskeleton, integrins play important roles in adhesion-dependent cellular processes including cell migration, proliferation, survival, and differentiation (1–4). Integrin α4β7 is a lymphocyte homing receptor, which can mediate both rolling and firm adhesion of lymphocytes, two of the critical steps in lymphocytes homing to the intestine and gut-associated lymphoid tissues (5, 6). Its ligand, mucosal addressin cell adhesion molecule-1 (MAdCAM-1),3 is preferentially expressed on high endothelial venules of gut-associated lymphoid organs and on lamina propria venules, helping lymphocyte traffic to mucosal organs (7). The activation of integrin α4β7 is a critical step in the progression of inflammatory bowel disease (8, 9). Thus, α4β7 is a promising therapeutic target for the treatment of inflammatory bowel disease.
Different from most integrins supporting only firm adhesion of cells upon activation, integrin α4β7 can mediate both rolling and firm adhesion of lymphocytes (10, 11). The resting integrin α4β7 supports rolling adhesion of lymphocytes via its low affinity interaction with MAdCAM-1. Upon activation, α4β7 binds to MAdCAM-1 in high affinity, which results in firm cell adhesion. The transition from rolling adhesion to firm adhesion is regulated by the shift of integrin from low affinity to high affinity state (11, 12). Affinity regulation is associated with the conformational rearrangement of the integrin molecule (13, 14). Previous studies have shown that integrin extracellular domains exist in at least three distinct global conformational states that differ in affinity for ligand: low affinity bent conformation with a closed headpiece, intermediate affinity extended conformation with a closed headpiece, and high affinity extended conformation with an open headpiece (15–19). The equilibrium among these different states is regulated by integrin inside-out signaling and extracellular stimuli, such as divalent cations (20, 21). Compared with the low affinity state in Ca2+ + Mg2+, removal of Ca2+ or addition of Mn2+ strikingly increases ligand binding affinity of almost all integrins (11). Electron micrographic studies of integrins αVβ3 and α5β1 demonstrate that integrin activation is coupled with the switchblade-like extension of the extracellular domain and a change in angle between the βI and hybrid domains (18, 19). Crystal structures of integrin αIIbβ3 headpiece in the high affinity conformation demonstrate that the C-terminal α7-helix of the βI domain moves axially toward the hybrid domain, causing the β hybrid domain to swing outward by 60°, away from the α subunit (15, 22). The conformational rearrangement in the integrin headpiece destabilizes the bent conformation and induces integrin extension in which the headpiece extends and breaks free from an interface with the leg domains that connect it to the plasma membrane. This conversion from the low affinity to the high affinity conformation of integrin can be mimicked by the introduction of glycan wedges into the interface between the hybrid and the I domains of β7, β3, and β1 integrins, which activates integrins by stabilizing the outward swing of the hybrid domain and the high affinity headpiece conformation (12, 23). This wedge mutant integrin therefore can be used as a target for screening activation-specific antibodies that exclusively recognize the activated integrin. Up to date, there is no well characterized activation-specific antibody against integrin α4β7. Thus, a well characterized activation-specific antibody to α4β7 will be extremely useful for both studies on α4β7 activation mechanism and development of drug delivery system targeting the activated lymphocyte for the treatment of inflammatory bowel disease.
In this study, we discovered the activation-specific human mAb J19 against integrin α4β7 by screening from the human scFv phage library using the activated wedge mutant α4β7 as a target. J19 IgG specifically bound to the α4β7 activated by Mn2+, DTT, ADP, or CXCL12 but not to the low affinity integrin. Moreover, J19 IgG did not interfere with α4β7-MAdCAM-1 interaction, suggesting it was a mAb distinct from “ligand mimetic” group, and we demonstrate that J19 IgG recognizes an activation-dependent epitope on α4β7 consisting of residues from both α4 and β7 subunits. This epitope is buried in the low affinity integrin with bent conformation and only exposed in the extended conformation induced by integrin activation. These data also provide strong supporting evidence for the conformational rearrangement during integrin α4β7 activation.
EXPERIMENTAL PROCEDURES
Cell Lines and mAbs
K562 cells stably expressing human α4β7 integrin was cultured in RPMI 1640 medium (Invitrogen) and 10% FBS (Biocherom AG) supplemented with 0.2 mg/ml hygromycin and 0.5 mg/ml G418 (all from Amresco). 293T stable cell lines expressing human integrin α4β7 WT or wedge mutant were cultured in DMEM (Invitrogen) with 10% FBS supplemented with hygromycin (0.1 mg/ml).
The following integrin antibodies were used in this study: mAb TS2/16 (Santa Cruz) to human β1, murine mAb Ber-ACT8 (Santa Cruz) to human αE, murine mAb HP2/1 (Abcam) to human and rat α4, and rat mAb FIB27 (BD biosciences) to human and mouse β7. The murine mAb 9F10 against human α4 was prepared from hybridoma (Developmental Studies Hybridoma Bank, University of Iowa). The murine mAb Act-1 specific for human α4β7 was previously described (24, 25). Human IgG1 control was from Pierce.
Plasmid Construction and Transient Transfection into 293T Cells
J19 IgG expression constructs were built on the backbone of pIRES2-EGFP (Invitrogen). cDNAs encoding human IgG1 light chain and heavy chain constant regions were amplified by RT-PCR from human endothelial cell total mRNA.
cDNAs of human α4, αE, β7, and β1 subunits were inserted into vector pcDNA3.1/Hygro (−) (Invitrogen), respectively. Chimeric β1/β7 subunits were generated by overlap extension PCR (26, 27). Chimeric α4/αE subunits were generated by an improved PCR mutagenesis strategy for sequence swapping (28). Chimeras were named according to the origin of their segments. For example, β7542β1 indicates that residues 1–542 are from β7 subunit and residues 543 to the C terminus are from the β1 subunit. Amino acid sequence numbering was according to the mature α4 or β7 sequence. The β7 site-directed mutations were generated by using QuikChange (Stratagene). All of the constructs were confirmed by DNA sequencing. Transient transfection of 293T cells was performed as described (11).
Protein Purification and Analytical Gel Filtration
The J19 IgG was purified by protein A (Pierce) affinity chromatography. 293T cells were transiently transfected with J19 IgG expressing construct and cultured in DMEM supplemented with 10% “ultralow IgG” fetal bovine serum (Invitrogen).
The soluble integrin α4β7 with all ectodomains was purified as previously described (19, 22). Briefly, soluble integrin was purified from culture supernatant of 293T cells stably expressing soluble integrin α4β7 ectodomains with C-terminally fused His tag and Strep-tag II using nickel-nitrilotriacetic acid-agarose (Qiagen) followed by Strep-Tactin (IBA) affinity chromatography and gel filtration (Superdex 200; GE Healthcare).
Analytical gel filtrations were performed using precalibrated Superdex 200 on an ÄKTA purifier system running Unicorn 5.11 software at a flow rate of 0.5 ml/min at room temperature (29, 30). The elution profiles were monitored in-line by UV adsorption at 280 nm. Hepes-buffered saline (HBS) buffer (150 mm NaCl, 20 mm Hepes, pH 7.4) containing 1 mm Ca2+ and 1 mm Mg2+ or 0.5 mm Mn2+ was used throughout. 5.2 μg of purified WT or wedge mutant integrin α4β7 was loaded.
Selection of Integrin-binding Phages
Human scFv phage library was purchased from Geneservice. Antibody screening was done according to the protocol provided by Geneservice with minor modifications. Soluble integrin α4β7 was biotinylated and immobilized to streptavidin-coated Dynabeads (Invitrogen). Phage displaying scFv binding to α4β7 was captured by integrin-labeled beads (so called “panning”). In each round of panning, the binders to WT α4β7 were depleted first followed by screening the specific scFv fragments against wedge mutant. After three rounds of panning, specific binders were identified by monoclonal phage ELISA.
Flow Cytometry
Immunofluorescence flow cytometry was done as described (31). Before staining with antibody, 2.5 × 105 cells were washed with HBS containing 5 mm EDTA and then resuspended in either HBS containing 1 mm Ca2+ + 1 mm Mg2+ or activating HBS containing 2 mm Mn2+. For activation of α4β7 by other stimuli than divalent cations, 2.5 × 105 cells were resuspended and incubated at 37 °C for 15 min in HBS (1 mm Ca2+ + 1 mm Mg2+) with ADP and DTT at a final concentration of 10 and 500 μm, respectively. Stained cells were then measured using FACSCalibur (BD Biosciences) and analyzed using WinMDI 2.9 software.
Primary splenic lymphocytes (SPLs) were isolated as previously described (32) and suspended in HBS and stimulated by 0.2 μg/ml CXCL12 (R & D Systems) for 5 min at 37 °C. The cells were fixed with an equal volume of 2× formaldehyde (7.4%) before staining with 5 μg/ml J19 IgG.
Fluorescence Microscopy
Immunofluorescence staining assay was performed as reported (33). Cover glasses were coated with 10 μg/ml human MAdCAM-1 fused to the Fc1 and Fc2 regions of human IgG1 (huMAdCAM-1/Fc) in the presence or absence of 2 μg/ml CXCL12. The cells were incubated on the cover glass for 20 min at 37 °C, fixed with 3.7% polyformaldehyde, and blocked by 10% FBS. The cells were then stained with 5 μg/ml J19 IgG or FIB27, followed by Alexa Fluor® 488 goat anti-human IgG (H + L) and Cy3-conjugated goat anti-rat IgG (Invitrogen), respectively.
Flow Chamber Assay
The flow chamber assay was performed as described (11, 12). A polystyrene Petri dish to be used as the lower wall of the chamber was coated with a 5-mm-diameter, 20-μl spot of 10 μg/ml purified huMAdCAM-1/Fc in coating buffer (phosphate-buffered saline, 10 mm NaHCO3, pH 9.0) for 1 h at 37 °C, followed by 2% BSA in coating buffer for 1 h at 37 °C to block nonspecific binding sites. The cells were washed twice with HBS containing 5 mm EDTA and 0.5% BSA, resuspended at 1 × 107/ml in buffer A (HBS, 0.5% BSA), and kept at room temperature. The cells were diluted to 1 × 106/ml in buffer A containing different divalent cations immediately before infusion in the flow chamber using a Harvard apparatus programmable syringe pump. Then shear stress was increased from 0.3 dyn/cm2 up to 16 dyn/cm2. The number of cells remaining bound at the end of 1 dyn/cm2 was determined.
FRET
For detecting the orientation of integrin ectodomain relative to cell membrane, FRET was measured as described (30, 35). Briefly, 293T cells stably expressing integrin α4β7 treated with indicated cations were fixed by 3.7% paraformaldehyde and then stained with 20 μg/ml Alexa Fluor 488-Act-1 Fab, followed by staining with 10 μm FM4–64 FX (Invitrogen).
RESULTS
Generation of High Affinity Integrin α4β7 with Glycan Wedge Mutation
Previous studies have shown that the introduction of N-glycan at the integrin βI/hybrid domain interface will activate integrins and stabilize the high affinity conformation (12, 23). To obtain the high affinity human integrin α4β7, we introduced an N-glycosylation site at Asn-322 in the α4-β5 loop of β7 I domain by mutating Gln-324 to Thr as previously described (12). The WT and wedge mutant (Q324T) human α4β7 were transiently expressed in 293T cells, and the adhesive behavior in shear flow of those transfectants was characterized by allowing them to adhere to MAdCAM-1 in a parallel wall flow chamber. WT α4β7 293T transfectants behaved as previously described for lymphoid cells expressing α4β7 (36). In 1 mm Ca2+ + 1 mm Mg2+, more than 75% of adherent cells expressing WT α4β7 rolled on MAdCAM-1 substrates at the wall shear stress of 1 dyn/cm2 (Fig. 1A). In contrast, the cells were firmly adherent in 0.5 mm Mn2+ (Fig. 1A). Rolling and firm adhesions represent the low and high affinity interactions of integrin α4β7 with MAdCAM-1, respectively. By contrast with the rolling adhesion of WT α4β7 293T transfectants, 293T transfectants expressing α4β7 wedge mutant showed significantly increased firmly adherent cells in 1 mm Ca2+ + 1 mm Mg2+, which is similar to the adhesion behavior of WT α4β7 activated by 0.5 mm Mn2+ (Fig. 1A). α4β7 transfectants treated with the α4β7 blocking antibody Act-1 did not accumulate on MAdCAM-1 substrates. Thus, integrin α4β7 is constitutively activated by the glycan wedge introduced into the I/hybrid domain interface of β7 subunit.
FIGURE 1.
The activation and conformational change of α4β7 induced by wedge mutation. A, rolling and firm adhesions on MAdCAM-1 substrates of 293T transfectants. The number of rolling and firmly adherent WT and wedge mutant α4β7 transfectants was measured in the indicated divalent cations at a wall shear stress of 1 dyn/cm2. The error bars are ± S.D. (n = 3). B, purified WT α4β7 was analyzed on a Superdex 200 column in HBS, containing 1 mm Ca2+ + 1 mm Mg2+ (dashed line) or 0.5 mm Mn2+ (dotted line) and purified wedge mutant in HBS, containing 1 mm Ca2+ + 1 mm Mg2+ (solid line). The values next to the peaks indicate elution volumes (in milliliters). mAU, milliabsorbance unit.
Next, we introduced Q324T mutation into the human integrin α4β7 soluble construct with all ectodomains. To eliminate the heterogeneity resulting from partial cleavage of α4 subunit, a previously described Arg-558 to Ala mutation was introduced into α4 subunit to remove the protease cleavage site (37). Both WT and wedge mutant α4β7-soluble proteins were expressed in 293T cells and purified by nickel-nitrilotriacetic acid, Strep-Tactin affinity chromatography, and gel filtration. To study the conformational change of integrin in solution, the isocratic elution profiles of purified WT and wedge mutant α4β7 were compared using analytical gel filtration. Previous studies have shown that the shape change of integrin from the low affinity bent conformation to high affinity extended conformation could lead to the increase in hydrodynamic radius of integrin and the decrease of retention volume in gel filtration (19, 30). As expected, the low affinity WT α4β7 was eluted at 10.83 ml in 1 mm Ca2+ + 1 mm Mg2+, whereas the elution volume of high affinity WT α4β7 in 0.5 mm Mn2+ decreased to 10.67 ml, suggesting the more extended high affinity conformation of α4β7 activated by Mn2+ (Fig. 1B). Similar to WT α4β7 in Mn2+, the wedge mutant α4β7 showed decreased retention volume (10.72 ml) in Ca2+ + Mg2+ compared with WT α4β7, suggesting the extended conformation of wedge mutant α4β7. Thus, the wedge mutant can mimic the high affinity α4β7 by stabilizing the extended conformation of integrin.
Identification of Human Antibody to Activated Integrin α4β7
To identify an activation-specific antibody against α4β7, we performed human single fold scFv phage display library (Tomlinson I + J) selection using the high affinity wedge mutant α4β7 as target. The Tomlinson I + J libraries purchased from Geneservice contain over 100 million different human scFv fragments. The library was first depleted by the low affinity WT α4β7 soluble protein and then selected against the high affinity wedge mutant α4β7. In this way, binders specific for high affinity α4β7 were enriched after each round of selection. After three rounds of selections, the remaining binders were validated by monoclonal phage ELISA using purified WT and wedge mutant protein as target, respectively. Among a number of isolates that showed higher binding signals to wedge mutant than to WT α4β7 (data not shown), phage clone J19 bound specifically to wedge mutant and high affinity WT α4β7 activated by 2 mm Mn2+, but not to the low affinity WT α4β7 in 1 mm Ca2+ + 1 mm Mg2+ (Fig. 2A).
FIGURE 2.
The binding specificities of J19 phage and J19 scFv to integrin α4β7. A, monoclonal phage ELISAs were carried out with coated soluble WT α4β7, wedge mutant α4β7, and BSA control in the presence of indicated divalent cations. ELISA signals of J19 phage are shown. The error bars are ± S.D. (n = 3). B, 5 μg/ml J19 scFv or Act-1 binding to inactive (1 mm Ca2+ + 1 mm Mg2+) or active (2 mm Mn2+) α4β7 stably expressed on K562 cells were analyzed by flow cytometry. A representative experiment (of three) is shown as a histogram. The numbers within the panels show the specific mean fluorescence intensity of Act-1 and J19 scFv. The results are the means ± S.D. of three independent experiments. Open histogram, control mouse IgG or control scFv; filled histogram, Act-1 or J19 scFv.
To further characterize the J19 scFv phage isolate, the J19 scFv was expressed and purified. The binding of purified J19 scFv to integrin α4β7 was analyzed using flow cytometry with K562 cells stably expressing human α4β7 (Fig. 2B). Different from the specificity of J19 phage for high affinity α4β7, J19 scFv showed similar binding to the low affinity α4β7 in 1 mm Ca2+ + 1 mm Mg2+ and the high affinity α4β7 in 2 mm Mn2+. The binding of J19 scFv to α4β7 was comparable with that of α4β7 mAb Act-1 (Fig. 2B). Considering that scFv is of a smaller size than the phage particle, it is tempting to speculate that the binding sites of J19 in low affinity α4β7 can be accessed by smaller scFv but not the larger phage particle expressing the same scFv.
J19 IgG Specifically Binds to Activated Integrin α4β7
To further investigate the function of J19, we reformatted the J19 scFv to full-length human IgG1. The binding of J19 IgG to integrin α4β7 was determined using flow cytometric analysis with 293T cells stably expressing WT or wedge mutant α4β7 (Fig. 3A). Comparable with J19 phage, J19 IgG specifically bound to 293T cells expressing wedge mutant α4β7 in 1 mm Ca2+ + 1 mm Mg2+ and WT α4β7 293T transfectants in 2 mm Mn2+, but not WT α4β7 transfectants in 1 mm Ca2+ + 1 mm Mg2+, suggesting its specificity for the activated α4β7. As control, the binding activity of α4β7 mAb Act-1 was shown in parallel. In contrast to J19 IgG, Act-1 showed nonselective binding to both low and high affinity α4β7 (Fig. 3A). Both J19 IgG and Act-1 did not bind to mock 293T cells transfected with pcDNA3.1 vector alone (Fig. 3A). Similar results were obtained using K562 cell stably expressing integrin α4β7, suggesting that the specificity of J19 IgG for activated integrin α4β7 is not cell type-dependent (Fig. 3B). Moreover, J19 IgG bound to Mn2+-activated α4β7 in a dose-dependent manner (Fig. 3C). From the best fit curve generated with GraphPad Prism 5.01 software, the EC50 of J19 IgG for binding to activated α4β7 is 29.96 nm.
FIGURE 3.
J19 IgG specially recognizes the activated integrin α4β7. A, 293T cells stably expressing WT or wedge mutant α4β7 were stained with human IgG1 control, Act-1, or J19 IgG at 5 μg/ml in the presence of 1 mm Ca2+ + 1 mm Mg2+ or 2 mm Mn2+. Mock represents pcDNA3.1 vector 293T transfectants. B, K562 cells stably expressing α4β7 were stained with 5 μg/ml indicated antibodies in the presence of 1 mm Ca2+ + 1 mm Mg2+ or 2 mm Mn2+ and analyzed by flow cytometry. Mock represents K562 cells transfected with pcDNA3.1 vector. C, concentration-dependent binding of J19 IgG and human IgG1 control to K562 transfectants stably expressing WT α4β7 in the presence of indicated divalent cations. The cells were incubated with serially diluted J19 IgG or human IgG1 control. The error bars are ± S.D. (n = 3). D, binding of J19 IgG to K562 cells stably expressing α4β7 in the presence of DTT (500 μm) or ADP (10 μm) were determined by flow cytometric analysis as described under “Experimental Procedures.” A representative experiment (of three) is shown as a histogram. The numbers within the panels show the specific mean fluorescence intensity of human IgG1, Act-1, or J19 IgG. The results are the means ± S.D. of three independent experiments.
In addition to the strong activation by Mn2+, integrin can also be activated by other stimuli like DTT and ADP (38–40). DTT has been shown to activate integrin in a number of systems (38, 39). ADP was reported to induce integrin activation through inside-out signaling by activating PI3K pathway (41). To further study the binding specificity of J19 IgG to α4β7 activated by different stimuli, K562 cells stably expressing α4β7 were treated with Mn2+, DTT, or ADP and then followed by staining with 5 μg/ml J19 IgG. As shown in Fig. 3D, J19 IgG specifically bound to K562 α4β7 cells treated with these stimuli. By contrast, J19 IgG did not bind to the same cells without any stimulation (Fig. 3D). These results suggested that epitope recognized by J19 IgG in integrin α4β7 was only expressed after integrin activation. Moreover, the binding of J19 IgG to K562 α4β7 cells treated with Mn2+ is higher in comparison with those stimulated by DTT or ADP. The different expression level of J19 epitope induced by the above stimuli could be due to the different activation states and conformations of integrin α4β7.
J19 IgG Recognizes Integrin α4β7 Heterodimer from Human, Mouse, and Rat
Because integrin α4β7 shares α4 subunit with integrin α4β1 and β7 subunit with integrin αEβ7, J19 IgG may also bind to α4β1 or αEβ7 if it recognizes either subunit of α4β7 heterodimer. To evaluate the potential cross-reactivity of J19, its binding to human α4β1 and αEβ7 was determined using 293T cells transiently expressing α4β1 or αEβ7 (Fig. 4A). The expressions of α4β1 and αEβ7 were confirmed by mAbs HP2/1 (α4 mAb), TS2/16 (β1 mAb), Ber-ACT8 (αE mAb), and FIB27 (β7 mAb). The flow cytometric results showed no J19 IgG binding to α4β1 or αEβ7 expressing 293T cells in both Ca2+ + Mg2+ and Mn2+, indicating that J19 IgG does not recognize α4β1 or αEβ7 both pre- and postactivation (Fig. 4A). These results demonstrate that neither α4 nor β7 subunit alone is sufficient to support J19 binding and that this antibody recognizes α4β7 heterodimeric complex.
FIGURE 4.
J19 IgG recognizes α4β7 heterodimer and cross-reacted with mouse and rat α4β7. A, 293T cells were transfected with human α4β1 or αEβ7, and the integrin expression level was determined by indicated antibodies, respectively. The binding of J19 IgG to α4β1 or αEβ7 was analyzed by flow cytometry in the presence of 1 mm Ca2+ + 1 mm Mg2+ or 2 mm Mn2+. B, human, mouse, or rat α4β7 was transiently expressed in 293T cells, and the integrin expression level was determined by 5 μg/ml indicated antibodies, respectively. Reactivity of J19 IgG with α4β7 293T transfectants was determined by flow cytometry. A representative experiment (of three) is shown as a histogram. The numbers within the panels show the specific mean fluorescence intensity of indicated mAbs. The results are the means ± S.D. of three independent experiments.
We next test the cross-reactivity of J19 IgG with α4β7 from other species. The mouse and rat α4β7 were transiently expressed in 293T cells, respectively. The expression level of α4β7 was determined using mAb FIB27 against human and mouse β7 and mAb HP2/1 against rat α4. J19 IgG showed comparable binding to the activated human, mouse, and rat α4β7 but not to inactive ones (Fig. 4B).
J19 IgG Specifically Binds to Chemokine-activated Mouse SPLs
Having shown that J19 IgG was specific for the activated α4β7 expressed on K562 and 293T cell lines, we next tested whether J19 IgG also bound to the high affinity form of α4β7 expressed on primary lymphocytes. The mouse SPLs that highly express α4β7 were isolated, and the activation of integrin by CXCL12 stimulation was done as previously described (33, 42). In flow cytometric assay, binding of J19 IgG to SPLs was undetectable before CXCL12 treatment, whereas it significantly increased 5 min after adding 0.2 μg/ml CXCL12 (Fig. 5A). Similar results were obtained by immunostaining with J19 IgG and β7 mAb FIB27 (Fig. 5B). J19 IgG signal was only observed at the surface of SPLs after α4β7 was activated by CXCL12 or Mn2+, whereas FIB27 bound to α4β7 in an activation-independent manner. Thus, J19 IgG specifically recognizes the activated α4β7 of primary lymphocytes.
FIGURE 5.
J19 IgG binds to activated α4β7 on primary lymphocytes. A, binding of J19 IgG to mouse SPLs in the presence or absence of 0.2 μg/ml CXCL12 was analyzed by flow cytometry. A representative experiment (of three) is shown as a histogram. The numbers within the panels show the specific mean fluorescence intensity of J19 IgG. The results are the means ± S.D. of three independent experiments. Open histogram, isotype control IgG filled histogram, J19 IgG. B, mouse SPLs were plated on coverslips coated with 10 μg/ml MAdCAM-1 or 10 μg/ml MAdCAM-1 plus 2 μg/ml CXCL12 for 20 min and then stained with 5 μg/ml J19 IgG and FIB27 in the presence of 1 mm Ca2+ + 1 mm Mg2+ or 2 mm Mn2+ as indicated. Bar, 20 μm.
Effect of J19 IgG on α4β7-MAdCAM-1 Interaction
To further characterize J19 IgG, we studied the effect of this antibody on cell adhesion mediated by α4β7-MAdCAM-1 interaction in shear flow using K562 cells stably expressing human α4β7. The cells were preincubated with J19 IgG or α4β7 blocking mAb Act-1 and then infused into a parallel wall flow chamber with MAdCAM-1 immobilized on the lower wall. 20 μg/ml Act-1 almost completely abolished the cell adhesion at the wall shear stress of 1 dyn/cm2, whereas J19 IgG showed no effect on cell adhesion even at much higher concentration of 100 μg/ml (Fig. 6). Thus, J19 IgG does not affect the interaction between integrin α4β7 and MAdCAM-1, suggesting that it is distinct from the ligand-mimetic mAb.
FIGURE 6.
J19 IgG has no effect on α4β7-MAdCAM-1 interaction. K562 cells stably expressing human α4β7 were incubated with 100 μg/ml J19 IgG or 20 μg/ml mAb Act-1 in the presence of 1 mm Ca2+ + 1 mm Mg2+ or 2 mm Mn2+ for 15 min at 37 °C, followed by flow chamber assay as described under “Experimental Procedures.” The number of rolling and firmly adherent cells was measured under the wall shear stress of 1 dyn/cm2. The error bars are ± S.D. (n = 3).
Epitope Mapping of J19 IgG
Because of the lack of cross-reactivity with α4β1 by J19 IgG and the high homology between β1 and β7 subunits, we constructed a panel of β1/β7 chimeras to locate the epitope of J19 IgG in β7 subunit. A schematic of the constructed chimeras is shown in Fig. 7A. These chimeras were all transiently co-expressed with human α4 subunit in 293T cells, the expression level of which was confirmed by immunostaining with 9F10 against α4. Flow cytometric analysis of these 293T transfectants showed that the β7 segment 331–348 located in βI domain was absolutely required for the binding of J19 IgG to α4β7. All of the chimeras in which segment 331–348 was of β1 origin failed to be stained with 5 μg/ml J19 IgG, whereas all other chimeras in which this segment was of β7 origin were stained, as well as WT β7 (Fig. 7A). Within region 331–348, seven amino acids differ between human β1 and β7. Thereafter, we substituted the seven β7 residues with the corresponding β1 residues. Mutations of S331E, A332E, and A333F decreased ∼30–50% of J19 IgG binding in comparison with WT α4β7. By contrast, mutations of the other four residues, L334Q, Q338K, S341K/K342N, almost had no effect on the recognition of α4β7 by J19 IgG. Moreover, the β7 triple mutation S331E/A332E/A333F completely abolished the recognition of α4β7 by J19 IgG (Fig. 7B). These results strongly suggest that residues Ser-331, Ala-332, and Ala-333 in β7 I domain represent a direct binding site for J19 IgG.
FIGURE 7.
Epitope mapping of J19 IgG. A, mapping of J19 IgG epitope with β7 chimeras. mAb reactivity was determined with chimeric human β1/β7 subunits co-expressed with human α4 in 293T cells in the presence of 2 mm Mn2+ by using flow cytometry. J19 IgG recognition was measured as specific mean fluorescence intensity and quantitated as a percentage of total α4β7 expression defined by staining with mAb 9F10 to α4. B, fine mapping of J19 IgG epitope in β7 subunit. Human WT β7 or mutant β7 containing multiple or single β7 to β1 amino acid substitutions was co-expressed with human α4 subunit in 293T cells. J19 IgG recognition was quantified as in A. C, mapping of J19 IgG epitope with α4 chimeras. The human α4/αE chimeras were co-expressed with human β7 in 293T cells. The transfectants were stained with J19 IgG or mAb FIB27 to β7, followed by flow cytometry in the presence of 2 mm Mn2+. J19 IgG recognition was quantified as in Fig. 7A. The error bars are ± S.D. (n = 3).
The J19 IgG binding site in human α4 subunit was mapped by using α4/αE chimeras because α4 and αE share the same β7 subunit. Considering β-propeller domain in α subunit is close to the above mapped J19 epitope in βI domain, we first swapped β-propeller domain of α4 and αE subunits, whereas the swap of β-propeller domain of α4 and αE subunits resulted in no expression of both α4β7 and αEβ7 chimeric integrins. The abnormal expression of α4/αE chimeras is possibly due to the difference in structure between αI domain-less integrin α4 and αI domain-containing integrin αE. Thus, based on J19 binding sites in β7 subunit and the crystal structures of integrin αIIbβ3 and αVβ3, several segments in α4 β-propeller domain close to the epitope in β7 subunit were substituted with corresponding αE sequences, respectively. These chimeric cDNAs were all cloned into pcDNA 3.1 expression vectors and transiently co-expressed with human β7 subunit in 293T cells, then followed by immunostaining with 5 μg/ml J19 IgG. The expression of these chimeras was confirmed by immunostaining with mAb FIB27 against β7. Swapping of α4 segments 211–216 and 240–246 with those of αE had no effect on the binding of J19 IgG, whereas swapping of the α4 184–190 segment completely abolished J19 IgG binding to chimera α4184αE190α4 (Fig. 7C). These results demonstrate that the epitope of J19 IgG in α4 subunit locates in a seven-residue segment from 184 to 190 in β-propeller domain. However, J19 did not bind to chimeric αE (αE184α4190αE) when swapping the 184–190 segment of α4 into αE subunit (Fig. 7C), which might be due to the interference of J19 binding by the αI domain on the top of the β-propeller domain in αE subunit.
The Epitope Exposure of J19 IgG Is Coupled with Extension of Integrin α4β7 Ectodomain
The headpiece of integrin folds over its legs and faces down toward the membrane in the low affinity bend conformation and extends upward in a switchblade-like opening upon activation (2, 43). We next studied the relationship between J19 IgG epitope exposure and the conformational rearrangement of integrin α4β7 during its activation. To assess the orientation of integrin α4β7 ectodomain relative to the cell membrane using a FRET system, α4β7 was labeled with Alexa Fluor 488-Act-1 Fab fragment as donor, which binds to the top of α4β7 βI domain (44). The outer face of plasma membrane was labeled with FM4–64 FX (FM) as acceptor (30, 35). Compared with the inactive α4β7 in 1 mm Ca2+ + 1 mm Mg2+, the FRET efficiency of 293T transfectants bearing activated α4β7 in 2 mm Mn2+ was significantly decreased from 22 to 6%, indicating the extension of the α4β7 ectodomain (Fig. 8A). In parallel, immunostaining results showed that J19 IgG bound to activated α4β7 in 2 mm Mn2+ but not to inactive α4β7 in 1 mm Ca2+ + 1 mm Mg2+, suggesting that the epitope of J19 IgG was only exposed in the activated α4β7 with extended conformation in 2 mm Mn2+ (Fig. 8B). These data strongly indicate that the J19 IgG epitope is exposed when the integrin head domain moves away from the cell membrane in the extended conformation.
FIGURE 8.
The expression of J19 IgG epitope is associated with the conformational change of integrin α4β7 upon activation. A, FRET between integrin α4β7 βI domain and the plasma membrane. The error bars are ± S.D. (n = 10). B, the expression of J19 IgG epitope was measured as specific mean fluorescence intensity and quantitated as a percentage of total α4β7 expression defined by staining with mAb Act-1. 293T cells stably expressing α4β7 were stained with 5 μg/ml J19 IgG and Act-1 in the presence of 1 mm Ca2+ + 1 mm Mg2+ or 2 mm Mn2+ as indicated and followed by flow cytometry. The error bars are ± S.D. (n = 3).
DISCUSSION
In this study, we screened and characterized an activation-specific human monoclonal antibody to integrin α4β7 by panning a scFv-displaying phagemid library using an active form of α4β7 (wedge mutant) as target. The subtractive selection was performed by depleting the library on the isolated, inactive WT α4β7 protein first and then panning against the constitutively activated α4β7 wedge mutant. The specific scFv clone J19 was isolated from the library and reformatted to full-length human IgG1. The J19 IgG specifically binds to integrin α4β7 activated by different stimuli, other than the inactive α4β7. The binding epitope of J19 IgG was mapped to two small segments located in α4 β-propeller domain and β7 I domain, respectively (supplemental Fig. S1). The seven-residue segment from 184 to 190 in α4 subunit locates between β-strands 2 and 3 in β-sheet 3 of the β-propeller domain. The other segment consists of residues Ser-331, Ala-332, and Ala-333 located in a turn between β-strand 5 and α-helix 5 at the top of βI domain. In the low affinity bend conformation, the integrin β-propeller domain and the βI domain sit on their leg domains and face the cell membrane, leading to the epitope buried in the inactive integrin. Upon activation, integrin converted from the bent to extended conformation with either closed headpiece (intermediate affinity state) or open headpiece (high affinity state), which led to the exposure of J19 epitope (supplemental Fig. S1). Thus, J19 IgG is an activation-dependent mAb, which recognizes an epitope expressed only in integrin α4β7 with extended conformation.
The high affinity wedge mutant α4β7 used for J19 selection contains a mutation-introduced N-glycan at the integrin β7 I/hybrid domain interface and mimics the swing-out of hybrid domain in β7 subunit, which is predicted to activate integrins and stabilize the high affinity conformation. In our study, we showed that J19 IgG bound both wedge mutant and WT integrin α4β7 activated by physiological agonist, such as CXCL12. Thus, the epitope recognized by J19 IgG is expressed in both high affinity wedge mutant and the physiologically activated WT integrin α4β7. These results strongly suggest that the hybrid domain swing-out is the key conformational rearrangement during integrin activation and can induce a high affinity conformation mimicking the physiological activation of integrin α4β7.
Different from the J19 phage and J19 IgG, which specifically recognize the activated integrin α4β7, J19 scFv showed similar binding to both inactive and activated α4β7. The loss of specificity to activated α4β7 could be due to the smaller size of scFv compared with phage particle and full-length IgG1. The epitope of J19 in the inactive α4β7 with bend conformation is not accessible to large size J19 phage or IgG, whereas the space is large enough for smaller scFv to access the epitope. This result also provides supporting evidence for the induction of J19 epitope expression by the conformational rearrangements of integrin.
In αI domain-containing integrins, the αI domain is inserted into a loop between β-sheets 2 and 3 at the top of the β-propeller domain (45, 46). As part of the J19 epitope, the seven-residue segment from 184 to 190 locates in β-sheet 3 of the β-propeller domain, which will be masked by the αI domain in the αE subunit (supplemental Fig. S2). Thus, J19 IgG cannot bind to integrin αEβ7 even after replacement of the seven-residue segment in αE subunit with that from α4 (Fig. 7C).
Interestingly, the binding of J19 IgG to α4β7 could be enhanced by a number of stimuli, including Mn2+, DTT, and ADP, but to different levels. The binding of J19 IgG showed stronger binding to α4β7 expressing cells treated with Mn2+ than the same cells stimulated with DTT or ADP, suggesting that integrin activated by different stimuli could have different conformations and expression of J19 epitope. The different expression levels of J19 epitope were consistent with the detection of increased affinity for ligand after those stimulations, as measured by a sensitive flow chamber assay (34, 35, 47). Thus, J19 IgG could serve as a reporter for the activation extent of α4β7.
In addition to serving as a research tool for in vitro and in vivo studies of integrin activation, the J19 antibody also represents a therapeutic candidate for treatment of inflammatory bowel disease. J19 IgG selectively targets the activated lymphocytes and leaves the inactive ones intact, which may result in fewer potential side effects.
Supplementary Material
Acknowledgment
We thank Dr. Moonsoo Jin from Cornel University for instruction on IgG reformatting.
This work was supported by National Basic Research Program of China Grant 2010CB529703; National Natural Science Foundation of China Grants 31190061, 30970604, and 30700119; Science and Technology Commission of Shanghai Municipality Grant 11JC1414200; Novo Nordisk-CAS Research Foundation Grant NN-CAS 2008-1; and Shanghai Pujiang Program Grant 08PJ14106.

This article contains supplemental Figs. S1 and S2.
- MAdCAM-1
- mucosal addressin cell adhesion molecule-1
- scFv
- single-chain variable fragment
- HBS
- Hepes-buffered saline
- SPL
- splenic lymphocyte.
REFERENCES
- 1. Hynes R. O. (2002) Integrins. Bidirectional, allosteric signaling machines. Cell 110, 673–687 [DOI] [PubMed] [Google Scholar]
- 2. Takagi J., Springer T. A. (2002) Integrin activation and structural rearrangement. Immunol. Rev. 186, 141–163 [DOI] [PubMed] [Google Scholar]
- 3. DeMali K. A., Wennerberg K., Burridge K. (2003) Integrin signaling to the actin cytoskeleton. Curr. Opin. Cell Biol. 15, 572–582 [DOI] [PubMed] [Google Scholar]
- 4. Legate K. R., Fässler R. (2009) Mechanisms that regulate adaptor binding to β-integrin cytoplasmic tails. J. Cell Sci. 122, 187–198 [DOI] [PubMed] [Google Scholar]
- 5. Park E. J., Mora J. R., Carman C. V., Chen J., Sasaki Y., Cheng G., von Andrian U. H., Shimaoka M. (2007) Aberrant activation of integrin α4β7 suppresses lymphocyte migration to the gut. J. Clin. Invest. 117, 2526–2538 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Gorfu G., Rivera-Nieves J., Ley K. (2009) Role of β7 integrins in intestinal lymphocyte homing and retention. Curr. Mol. Med. 9, 836–850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Briskin M. J., McEvoy L. M., Butcher E. C. (1993) MAdCAM-1 has homology to immunoglobulin and mucin-like adhesion receptors and to IgA1. Nature 363, 461–464 [DOI] [PubMed] [Google Scholar]
- 8. Stefanelli T., Malesci A., De La Rue S. A., Danese S. (2008) Anti-adhesion molecule therapies in inflammatory bowel disease. Touch and go. Autoimmunity Reviews 7, 364–369 [DOI] [PubMed] [Google Scholar]
- 9. Rutgeerts P., Vermeire S., Van Assche G. (2009) Biological therapies for inflammatory bowel diseases. Gastroenterology 136, 1182–1197 [DOI] [PubMed] [Google Scholar]
- 10. Berlin C., Bargatze R. F., Campbell J. J., von Andrian U. H., Szabo M. C., Hasslen S. R., Nelson R. D., Berg E. L., Erlandsen S. L., Butcher E. C. (1995) α4 integrins mediate lymphocyte attachment and rolling under physiologic flow. Cell 80, 413–422 [DOI] [PubMed] [Google Scholar]
- 11. Chen J., Salas A., Springer T. A. (2003) Bistable regulation of integrin adhesiveness by a bipolar metal ion cluster. Nat. Struct. Biol. 10, 995–1001 [DOI] [PubMed] [Google Scholar]
- 12. Chen J., Takagi J., Xie C., Xiao T., Luo B. H., Springer T. A. (2004) The relative influence of metal ion binding sites in the I-like domain and the interface with the hybrid domain on rolling and firm adhesion by integrin α4β7. J. Biol. Chem. 279, 55556–55561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Luo B. H., Springer T. A. (2006) Integrin structures and conformational signaling. Curr. Opin. Cell Biol. 18, 579–586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Springer T. A., Dustin M. L. (2012) Integrin inside-out signaling and the immunological synapse. Curr. Opin. Cell Biol. 24, 107–115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Xiao T., Takagi J., Coller B. S., Wang J. H., Springer T. A. (2004) Structural basis for allostery in integrins and binding to fibrinogen-mimetic therapeutics. Nature 432, 59–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Luo B. H., Carman C. V., Springer T. A. (2007) Structural basis of integrin regulation and signaling. Annu. Rev. Immunol. 25, 619–647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Arnaout M. A., Goodman S. L., Xiong J. P. (2007) Structure and mechanics of integrin-based cell adhesion. Curr. Opin. Cell Biol. 19, 495–507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Takagi J., Strokovich K., Springer T. A., Walz T. (2003) Structure of integrin α5β1 in complex with fibronectin. EMBO J. 22, 4607–4615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Takagi J., Petre B. M., Walz T., Springer T. A. (2002) Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell 110, 599–611 [DOI] [PubMed] [Google Scholar]
- 20. Carman C. V., Springer T. A. (2003) Integrin avidity regulation. Are changes in affinity and conformation underemphasized? Curr. Opin. Cell Biol. 15, 547–556 [DOI] [PubMed] [Google Scholar]
- 21. Arnaout M. A., Mahalingam B., Xiong J. P. (2005) Integrin structure, allostery, and bidirectional signaling. Annu. Rev. Cell Dev. Biol. 21, 381–410 [DOI] [PubMed] [Google Scholar]
- 22. Zhu J., Luo B. H., Xiao T., Zhang C., Nishida N., Springer T. A. (2008) Structure of a complete integrin ectodomain in a physiologic resting state and activation and deactivation by applied forces. Mol. Cell 32, 849–861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Luo B. H., Springer T. A., Takagi J. (2003) Stabilizing the open conformation of the integrin headpiece with a glycan wedge increases affinity for ligand. Proc. Natl. Acad. Sci. U.S.A. 100, 2403–2408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Lazarovits A. I., Moscicki R. A., Kurnick J. T., Camerini D., Bhan A. K., Baird L. G., Erikson M., Colvin R. B. (1984) Lymphocyte activation antigens. I. A monoclonal antibody, anti-Act I, defines a new late lymphocyte activation antigen. J. Immunol. 133, 1857–1862 [PubMed] [Google Scholar]
- 25. Schweighoffer T., Tanaka Y., Tidswell M., Erle D. J., Horgan K. J., Luce G. E., Lazarovits A. I., Buck D., Shaw S. (1993) Selective expression of integrin α4β7 on a subset of human CD4+ memory T cells with Hallmarks of gut-trophism. J. Immunol. 151, 717–729 [PubMed] [Google Scholar]
- 26. Ho S. N., Hunt H. D., Horton R. M., Pullen J. K., Pease L. R. (1989) Site-directed mutagenesis by overlap extension using the polymerase chain reaction. Gene 77, 51–59 [DOI] [PubMed] [Google Scholar]
- 27. Horton R. M., Cai Z. L., Ho S. N., Pease L. R. (1990) Gene splicing by overlap extension. Tailor-made genes using the polymerase chain reaction. BioTechniques 8, 528–535 [PubMed] [Google Scholar]
- 28. Kirsch R. D., Joly E. (1998) An improved PCR-mutagenesis strategy for two-site mutagenesis or sequence swapping between related genes. Nucleic Acids Res. 26, 1848–1850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Adair B. D., Xiong J. P., Maddock C., Goodman S. L., Arnaout M. A., Yeager M. (2005) Three-dimensional EM structure of the ectodomain of integrin αVβ3 in a complex with fibronectin. J. Cell Biol. 168, 1109–1118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Xiong J. P., Mahalingham B., Alonso J. L., Borrelli L. A., Rui X., Anand S., Hyman B. T., Rysiok T., Müller-Pompalla D., Goodman S. L., Arnaout M. A. (2009) Crystal structure of the complete integrin alphaVbeta3 ectodomain plus an α/β transmembrane fragment. J. Cell Biol. 186, 589–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Chen J., Yang W., Kim M., Carman C. V., Springer T. A. (2006) Regulation of outside-in signaling and affinity by the β2 I domain of integrin αLβ2. Proc. Natl. Acad. Sci. U.S.A. 103, 13062–13067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Sun H., Wu Y., Qi J., Pan Y., Ge G., Chen J. (2011) The CC′ and DE loops in Ig domains 1 and 2 of MAdCAM-1 play different roles in MAdCAM-1 binding to low- and high-affinity integrin α4β7. J. Biol. Chem. 286, 12086–12092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Morin N. A., Oakes P. W., Hyun Y. M., Lee D., Chin Y. E., King M. R., Springer T. A., Shimaoka M., Tang J. X., Reichner J. S., Kim M. (2008) Nonmuscle myosin heavy chain IIA mediates integrin LFA-1 de-adhesion during T lymphocyte migration. J. Exp. Med. 205, 195–205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Shimaoka M., Kim M., Cohen E. H., Yang W., Astrof N., Peer D., Salas A., Ferrand A., Springer T. A. (2006) AL-57, a ligand-mimetic antibody to integrin LFA-1, reveals chemokine-induced affinity up-regulation in lymphocytes. Proc. Natl. Acad. Sci. U.S.A. 103, 13991–13996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Pan Y., Zhang K., Qi J., Yue J., Springer T. A., Chen J. (2010) Cation-π interaction regulates ligand-binding affinity and signaling of integrin α4β7. Proc. Natl. Acad. Sci. U.S.A. 107, 21388–21393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. de Château M., Chen S., Salas A., Springer T. A. (2001) Kinetic and mechanical basis of rolling through an integrin and novel Ca2+-dependent rolling and Mg2+-dependent firm adhesion modalities for the α4β7-MAdCAM-1 interaction. Biochemistry 40, 13972–13979 [DOI] [PubMed] [Google Scholar]
- 37. Teixidó J., Parker C. M., Kassner P. D., Hemler M. E. (1992) Functional and structural analysis of VLA-4 integrin α4 subunit cleavage. J. Biol. Chem. 267, 1786–1791 [PubMed] [Google Scholar]
- 38. Davis G. E., Camarillo C. W. (1993) Regulation of integrin-mediated myeloid cell adhesion to fibronectin. Influence of disulfide reducing agents, divalent cations and phorbol ester. J. Immunol. 151, 7138–7150 [PubMed] [Google Scholar]
- 39. Ni H., Li A., Simonsen N., Wilkins J. A. (1998) Integrin activation by dithiothreitol or Mn2+ induces a ligand-occupied conformation and exposure of a novel NH2-terminal regulatory site on the β1 integrin chain. J. Biol. Chem. 273, 7981–7987 [DOI] [PubMed] [Google Scholar]
- 40. Murgo A. J., Sistare F. D. (1992) K562 leukemia cells express P2T (adenosine diphosphate) purinergic receptors. J. Pharmacol. Exp. Ther. 261, 580–585 [PubMed] [Google Scholar]
- 41. Sun D. S., Lo S. J., Tsai W. J., Lin C. H., Yu M. S., Chen Y. F., Chang H. H. (2005) PI3-kinase is essential for ADP-stimulated integrin α(IIb)β3-mediated platelet calcium oscillation, implications for P2Y receptor pathways in integrin α(IIb)β3-initiated signaling cross-talks. J. Biomed. Sci. 12, 937–948 [DOI] [PubMed] [Google Scholar]
- 42. Yamamoto M., Fujihashi K., Kawabata K., McGhee J. R., Kiyono H. (1998) A mucosal intranet. Intestinal epithelial cells down-regulate intraepithelial, but not peripheral, T lymphocytes. J. Immunol. 160, 2188–2196 [PubMed] [Google Scholar]
- 43. Xie C., Shimaoka M., Xiao T., Schwab P., Klickstein L. B., Springer T. A. (2004) The integrin α-subunit leg extends at a Ca2+-dependent epitope in the thigh/genu interface upon activation. Proc. Natl. Acad. Sci. U.S.A. 101, 15422–15427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Tidswell M., Pachynski R., Wu S. W., Qiu S. Q., Dunham E., Cochran N., Briskin M. J., Kilshaw P. J., Lazarovits A. I., Andrew D. P., Butcher E. C., Yednock T. A., Erle D. J. (1997) Structure-function analysis of the integrin β7 subunit. Identification of domains involved in adhesion to MAdCAM-1. J. Immunol. 159, 1497–1505 [PubMed] [Google Scholar]
- 45. Weitz-Schmidt G., Schürpf T., Springer T. A. (2011) The C-terminal αI domain linker as a critical structural element in the conformational activation of αI integrins. J. Biol. Chem. 286, 42115–42122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Xie C., Zhu J., Chen X., Mi L., Nishida N., Springer T. A. (2010) Structure of an integrin with an αI domain, complement receptor type 4. EMBO J. 29, 666–679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Edwards B. S., Curry M. S., Southon E. A., Chong A. S., Graf L. H., Jr. (1995) Evidence for a dithiol-activated signaling pathway in natural killer cell avidity regulation of leukocyte function antigen-1. Structural requirements and relationship to phorbol ester- and CD16-triggered pathways. Blood 86, 2288–2301 [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.








