Disruption of disulfide restriction at integrin knees...

12
Journal of Cell Science Disruption of disulfide restriction at integrin knees induces activation and ligand-independent signaling of a 4 b 7 Kun Zhang 1, *, YouDong Pan 1, *, JunPeng Qi 1 , Jiao Yue 1 , MingBo Zhang 2,3 , ChenQi Xu 1 , GuoHui Li 2,` and JianFeng Chen 1,` 1 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China 2 Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China 3 College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, 116600, China *These authors contributed equally to this work ` Authors for correspondence ([email protected]; [email protected]) Accepted 12 August 2013 Journal of Cell Science 126, 5030–5041 ß 2013. Published by The Company of Biologists Ltd doi: 10.1242/jcs.134528 Summary Control of integrin activation and signaling plays crucial roles in cell adhesion, spreading and migration. Here, we report that selective breakage of two conserved disulfide bonds located at the knees of integrin a 4 C589–C594 and b 7 C494–C526 activated a 4 b 7 . This activated integrin had a unique structure that was different from the typical extended conformation of active integrin. In addition, these activated a 4 b 7 integrins spontaneously clustered on the cell membrane and triggered integrin downstream signaling independent of ligand binding. Although these disulfide bonds were not broken during a 4 b 7 activation by inside-out signaling or Mn 2+ , they could be specifically reduced by 0.1 mM dithiothreitol, a reducing strength that could be produced in vivo under certain conditions. Our findings reveal a novel mechanism of integrin activation under specific reducing conditions by which integrin can signal and promote cell spreading in the absence of ligand. Key words: Disulfide-restriction, Integrin clustering, Ligand-independent signaling Introduction Integrins are a family of a/b heterodimeric cell adhesion molecules that mediate cell–cell, cell–matrix, and cell– pathogen interactions, and signal bidirectionally across the plasma membrane (Ekblom et al., 1986; Luo et al., 2007). In vertebrates, 18 a-subunits and 8 b-subunits combine to yield 24 distinct integrins, which play vital roles in a wide range of biological and pathological events including immune responses, homeostasis, embryonic development, and diseases such as cancer and autoimmunity (Hynes, 2002). In contrast to most integrins, which only mediate firm cell adhesion upon activation, a 4 b 7 mediates both firm cell adhesion when activated and rolling adhesion before activation through its ligand mucosal addressing cell adhesion molecule-1 (MAdCAM-1), making it indispensable in the tissue-specific homing of lymphocytes to intestine and associated lymphoid tissues (Berlin et al., 1993). Dysregulation of a 4 b 7 -mediated recruitment of lymphocytes to inflamed intestinal tissues has been implicated in the pathogenesis of intestinal inflammatory disorders (Feagan et al., 2005). The biological functions of integrins rely on dynamic regulation of integrin affinity and signaling. Integrin affinity can be uniquely regulated through a process called inside-out signaling, in which stimuli received by other cell surface receptors initiate intracellular signals that impinge on integrin cytoplasmic domains and alter integrin conformation and ligand-binding affinity (Carman and Springer, 2003). In addition, binding of ligand to integrins can trigger the transduction of extracellular signals into the cytoplasm and activate downstream signals in the classic outside-in signaling (Litvinov et al., 2004; Luo et al., 2007). Moreover, lateral assembly of integrins (integrin clustering) is known to augment integrin-mediated adhesion by increasing the bond valency and also contributes to signal transduction (Miyamoto et al., 1995). Recent research advances have shed light on the structural basis for integrin affinity regulation and signaling. The entire ectodomain of integrin a V b 3 , a IIb b 3 and a X b 2 was crystallized in a bent conformation, which is believed to exist on the cell surface and to represent the inactive, low-affinity state (Xie et al., 2010; Xiong et al., 2001; Zhu et al., 2008). A number of negative staining electron microscopy and mutagenesis studies indicate that integrin activation is accompanied by a switchblade-like opening of the ectodomain during which the bent conformer undergoes a large angle rotation at the knee fulcrum and becomes extended to expose the ligand binding site on top of the integrin headpiece for high-affinity ligand binding (Takagi et al., 2002). However, other studies have indicated that such large-scale conformational change was not always required to render integrins competent to bind physiological ligands (Butta et al., 2003; Calzada et al., 2002; Chigaev et al., 2001; Xiong et al., 2003). Compelling evidence for this is the observation that the 5030 Research Article

Transcript of Disruption of disulfide restriction at integrin knees...

  • Journ

    alof

    Cell

    Scie

    nce

    Disruption of disulfide restriction at integrin kneesinduces activation and ligand-independent signalingof a4b7Kun Zhang1,*, YouDong Pan1,*, JunPeng Qi1, Jiao Yue1, MingBo Zhang2,3, ChenQi Xu1, GuoHui Li2,` andJianFeng Chen1,`

    1State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy ofSciences, Shanghai 200031, China2Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, ChineseAcademy of Sciences, Dalian 116023, China3College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, 116600, China

    *These authors contributed equally to this work`Authors for correspondence ([email protected]; [email protected])

    Accepted 12 August 2013Journal of Cell Science 126, 50305041 2013. Published by The Company of Biologists Ltddoi: 10.1242/jcs.134528

    SummaryControl of integrin activation and signaling plays crucial roles in cell adhesion, spreading and migration. Here, we report that selectivebreakage of two conserved disulfide bonds located at the knees of integrin a4C589C594 and b7C494C526 activated a4b7. Thisactivated integrin had a unique structure that was different from the typical extended conformation of active integrin. In addition, these

    activated a4b7 integrins spontaneously clustered on the cell membrane and triggered integrin downstream signaling independent ofligand binding. Although these disulfide bonds were not broken during a4b7 activation by inside-out signaling or Mn

    2+, they could bespecifically reduced by 0.1 mM dithiothreitol, a reducing strength that could be produced in vivo under certain conditions. Our findingsreveal a novel mechanism of integrin activation under specific reducing conditions by which integrin can signal and promote cell

    spreading in the absence of ligand.

    Key words: Disulfide-restriction, Integrin clustering, Ligand-independent signaling

    IntroductionIntegrins are a family of a/b heterodimeric cell adhesionmolecules that mediate cellcell, cellmatrix, and cell

    pathogen interactions, and signal bidirectionally across the

    plasma membrane (Ekblom et al., 1986; Luo et al., 2007). In

    vertebrates, 18 a-subunits and 8 b-subunits combine to yield 24distinct integrins, which play vital roles in a wide range of

    biological and pathological events including immune responses,

    homeostasis, embryonic development, and diseases such as

    cancer and autoimmunity (Hynes, 2002). In contrast to most

    integrins, which only mediate firm cell adhesion upon activation,

    a4b7 mediates both firm cell adhesion when activated and rollingadhesion before activation through its ligand mucosal addressing

    cell adhesion molecule-1 (MAdCAM-1), making it indispensable

    in the tissue-specific homing of lymphocytes to intestine and

    associated lymphoid tissues (Berlin et al., 1993). Dysregulation

    of a4b7-mediated recruitment of lymphocytes to inflamedintestinal tissues has been implicated in the pathogenesis of

    intestinal inflammatory disorders (Feagan et al., 2005).

    The biological functions of integrins rely on dynamic regulation

    of integrin affinity and signaling. Integrin affinity can be uniquely

    regulated through a process called inside-out signaling, in which

    stimuli received by other cell surface receptors initiate intracellular

    signals that impinge on integrin cytoplasmic domains and alter

    integrin conformation and ligand-binding affinity (Carman and

    Springer, 2003). In addition, binding of ligand to integrins can

    trigger the transduction of extracellular signals into the cytoplasm

    and activate downstream signals in the classic outside-in

    signaling (Litvinov et al., 2004; Luo et al., 2007). Moreover,

    lateral assembly of integrins (integrin clustering) is known to

    augment integrin-mediated adhesion by increasing the bond

    valency and also contributes to signal transduction (Miyamoto

    et al., 1995).

    Recent research advances have shed light on the structural

    basis for integrin affinity regulation and signaling. The entire

    ectodomain of integrin aVb3, aIIbb3 and aXb2 was crystallized ina bent conformation, which is believed to exist on the cell surface

    and to represent the inactive, low-affinity state (Xie et al., 2010;

    Xiong et al., 2001; Zhu et al., 2008). A number of negative

    staining electron microscopy and mutagenesis studies indicate

    that integrin activation is accompanied by a switchblade-like

    opening of the ectodomain during which the bent conformer

    undergoes a large angle rotation at the knee fulcrum and becomes

    extended to expose the ligand binding site on top of the integrin

    headpiece for high-affinity ligand binding (Takagi et al., 2002).

    However, other studies have indicated that such large-scale

    conformational change was not always required to render

    integrins competent to bind physiological ligands (Butta et al.,

    2003; Calzada et al., 2002; Chigaev et al., 2001; Xiong et al.,

    2003). Compelling evidence for this is the observation that the

    5030 Research Article

    mailto:[email protected]:[email protected]

  • Journ

    alof

    Cell

    Scie

    nce

    bent conformer of aIIbb3 ectodomain in solution is able to form astable complex with its physiological ligand (Adair et al., 2005).

    These studies suggest a level of complexity in the relationship

    between the conformation and affinity of integrins.

    All integrins contain a large number of cysteine residues in

    their ectodomains. Previous studies have shown that breakage of

    some disulfide bonds in integrins by reducing agents or

    mutagenesis can induce the activation of integrins (Mor-Cohen

    et al., 2012; Yan and Smith, 2001). However, the underlying

    mechanism remains elusive. Crystal structures of the ectodomain

    of integrins aIIbb3, aVb3 and aXb2 revealed two disulfide bondslocated at the apex of the knees, with one in the knee of the a-subunit and the other in the b-subunit I-EGF2 domain (Xie et al.,2010; Xiong et al., 2009; Zhu et al., 2008) (Fig. 1AC). Sequence

    alignment demonstrated that these two disulfide bonds (a4C589C594 and b7C494C526) are highly conserved between allintegrins (Fig. 1D,E). Considering that they are exposed to

    solution and are easily accessed by reducing agents, it is tempting

    to speculate that these two disulfide bonds could be disrupted

    under certain reducing conditions and that loss of the disulfide

    restriction might regulate integrin function.

    Here, we demonstrate that breaking either or both a4C589C594 and b7C494C526 induces the activation of a4b7 byincreasing integrin ligand-binding affinity. Surprisingly, the

    highly activated a4b7 that lacks the restriction of thesedisulfide bonds exhibits a unique conformation that is different

    from the characteristic extended conformation of integrin that is

    activated by inside-out signaling or Mn2+. In addition, breaking

    these disulfide bonds in a4b7 increases phosphorylation of focaladhesion kinase (FAK) and paxillin, and promotes cell spreading

    on immobilized poly-L-lysine, indicating the activation of

    integrin outside-in signaling independent of ligand binding,

    possibly as a result of enhanced integrin clustering before ligand

    binding. Moreover, these disulfide bonds can be specifically

    reduced by 0.1 mM dithiothreitol (DTT), although they were not

    found to be reduced during the activation of a4b7 by inside-out

    signaling or Mn2+. Thus, our findings suggest a novel mechanism

    of integrin affinity and signaling regulation through reduction of

    two conserved disulfide bonds at the knees of integrin. The

    resulting activated a4b7, with its unique conformation, canspontaneously cluster on the cell membrane and trigger integrin

    outside-in signaling and cell spreading independent of ligand

    binding.

    ResultsTwo disulfide bonds at the knees of integrin are crucial for

    keeping a4b7 inactive

    To date, there is no published experimental structure of the full-

    length integrin a4b7 ectodomain. Thus, we constructed the full-length ectodomain of human a4b7 in bent conformation by usinghomology modeling technique (Fig. 1A). The head domain was

    modeled after the crystal structure of human a4b7 headpiece(pdb3V4P), which consists of b-propeller and thigh domains ofthe a4 subunit and the bI and hybrid domains of the b7 subunit(Yu et al., 2012). The other domains were built based on their

    counterparts from the crystal structure of human aVb3 (pdb3IJE).Sequence alignments reveal that two disulfide bonds at the knee

    region of integrin a4b7 (a4C589C594 and b7C494C526) arehighly conserved among all integrins (Fig. 1D,E). The disulfide

    bond a4C589C594 occludes the a-subunit knee domain(Fig. 1B) and b7C494C526 is the first disulfide bond in thebI-EGF2 domain (Fig. 1C). To investigate the role of thesedisulfide bonds in the regulation of integrin a4b7 function, wesubstituted a4C589, a4C594, b7C494 and b7C526 with Ser andgenerated a4 mutations [a4C589S, a4C594S, a4C589SC594S(a4CC)], b7 mutations [b7C494S, b7C526S, b7C494C526S(b7CC)] and a a4b7 double mutation (a4CC/b7CC) to breakeach or both of the two disulfide bonds. CHO-K1 cell lines stably

    expressing either wide-type (WT) human integrin a4b7 or each ofthe above mutants were established (supplementary material

    Table S1). Adhesive behavior of these transfectants in shear flow

    was characterized in a parallel wall flow chamber by allowing the

    Fig. 1. The two conserved disulfide bonds located at the knees of

    integrin. (A) Model structure of full-length human integrin a4b7ectodomain in bent conformation. a4 subunit is shown in cyan with

    knee in red. b7 subunit is shown in pink with I-EGF2 C1C2 loop in

    blue. The two conserved disulfide bonds located at the knees of integrin

    are represented by spheres, with C and S atoms in green and yellow,

    respectively. (B,C) Enlarged view of a4 knee (B) and b7 I-EGF1/I-

    EGF2 (C). The disulfide bonds, a4C589C594 and b7C494C526, are

    shown as sticks in yellow. The coordinated Ca2+ at knee is shown as a

    sphere in orange. I-EGF1, I-EGF2 and the I-EGF2 C1C2 loop are

    shown in magenta, pink and blue, respectively. (D,E) Sequence

    alignment of human integrin a-subunit knee domains (D) and b-subunit

    I-EGF2 domains (E). Residues at the position of C589/C594 in a4subunit and C494/C526 in b7 subunit are shown in red.

    Regulation mechanism of a4b7 5031

  • Journ

    alof

    Cell

    Scie

    nce

    cells to adhere to human MAdCAM-1 adsorbed to the lower wall.

    The velocity of the cells remaining bound at a wall shear stress of

    1 dyn/cm2 was determined (Fig. 2A). Rolling and firm adhesion

    represents the low- and high-affinity interaction of MAdCAM-1

    with inactive and activated a4b7, respectively. WT a4b7 CHO-K1transfectants behaved as previously described for lymphoid cells

    expressing a4b7 (Chen et al., 2003). In 1 mM Ca2+ and 1 mM

    Mg2+ (1 mM Ca2+/Mg2+), ,80% of the bound a4b7 transfectantsrolled on MAdCAM-1 substrates (Fig. 2A). By contrast, the

    majority of cells were firmly adherent after activation of integrin

    by 0.5 mM Mn2+ (Fig. 2A). a4b7 transfectants treated with thea4b7 blocking antibody Act-1 or 5 mM EDTA did notaccumulate on MAdCAM-1 substrates (Fig. 2A), indicating the

    specificity of the interaction between a4b7 and MAdCAM-1. In

    contrast to the robust rolling cell adhesion mediated by WT a4b7in 1 mM Ca2+/Mg2+, all mutations that break the two disulfide

    bonds significantly increased the number of firmly adherent cells,demonstrating that a4b7 is activated by breaking either or both ofthe two disulfide bonds (Fig. 2A). Therefore, the two disulfidebonds at the knees of integrin are essential for keeping integrin

    a4b7 inactive.To further study the binding characteristics of WT a4b7 and

    a4CC/b7CC mutant to MAdCAM-1, we purified soluble a4b7proteins that contain the full-length ectodomain as describedpreviously (Qi et al., 2012; Zhu et al., 2008) and measured thebinding of WT a4b7 and a4CC/b7CC proteins to MAdCAM-1using surface plasmon resonance. In the presence of 1 mM Ca2+/Mg2+, soluble integrins bound to immobilized MAdCAM-1 in aconcentration-dependent manner (Fig. 2B). Compared with WTa4b7, a4CC/b7CC showed increased binding to MAdCAM-1(Fig. 2B), suggesting that breaking these disulfide bondsincreases integrin ligand-binding affinity. As a positive control,addition of 0.5 mM Mn2+ maximally increased the binding of

    WT a4b7 (Fig. 2B). The binding curves fitted well to a two-stepbinding model (supplementary material Table S2) as describedbefore (Takagi et al., 2002). For simplicity, we reported here

    apparent association and disassociation rate constants derivedfrom the initial phase of the association and dissociation curves,respectively. The kon values for WT a4b7 and a4CC/b7CCwere 0.8716105 M21 second21 and 3.136105 M21 second21,respectively, and the koff values were 5.31610

    23 second21 and1.4361023 second21. The calculated binding affinity of integrinto MAdCAM-1 is 2.71 nM for WT a4b7 and 0.628 nM for thea4CC/b7CC mutant. Thus, disruption of the two disulfide bondsaffects both association and dissociation rate, resulting in anoverall 4.32-fold increase in the binding affinity of a4b7 toMAdCAM-1.

    Metal-ion binding sites have been shown to play importantroles in the regulation of integrin affinity (Chen et al., 2003; Xie

    et al., 2004; Zhang and Chen, 2012). There is a conserved Ca2+

    binding site in the loop occluded by a4C589C594 in the a4 knee(Xiong et al., 2001); therefore, breaking this disulfide bond couldchange the orientation of the loop that contains the Ca2+-

    coordinating residues (C589, E592 and D635) and affect Ca2+

    binding. To exclude the possibility that integrin activationinduced by breaking the disulfide bond is a result of the loss of

    Ca2+ binding, we disrupted the Ca2+ binding site by mutatingE592 and D635 to Gly and Ala, respectively. CHO-K1 cell linesstably expressing a4E592G, a4D635A or a4E592GD635A(a4ED) mutants were established (supplementary materialTable S1). Cells expressing WT and mutant a4b7 integrinshowed similar adhesion behavior in shear flow on MAdCAM-1

    substrates (Fig. 2C), demonstrating that abolishment of Ca2+

    binding does not activate a4b7.The a4C589C594 and b7C494C526 disulfide bonds occlude

    4- and 13-amino-acid loops, respectively. It is tempting to

    speculate that disruption of the two disulfide bonds increases theflexibility of the integrin knee region, which might facilitate theextension of the integrin ectodomain and allosterically activate

    a4b7. To test our hypothesis, we individually deleted the twoloops to reduce flexibility at the knees of a4b7, which wasexpected to keep integrin inactive. Unexpectedly, deletion of

    either the a4 4-amino-acid loop (a4D4) or the b7 13-amino-acidloop (b7 I-EGF2 C1C2 loop; b7D13) markedly increased thenumber of firmly adherent cells on MAdCAM-1 substrates

    Fig. 2. Breakage of the two disulfide bonds at the knees of a4b7 activates

    integrin. (A,C) Adhesive modality of CHO-K1 cells stably expressing WT or

    mutant a4b7 on MAdCAM-1 substrates under flow conditions. Cells

    preincubated with the a4b7 blocking antibody Act-1 (5 mg/ml) for 5 minutesat 37 C or treated with 5 mM EDTA were used as controls. The number of

    rolling and firmly adherent cells was measured in the indicated divalent

    cations (1 mM Ca2+/Mg2+ or 0.5 mM Mn2+) at a wall shear stress of 1 dyn/

    cm2. Error bars represent s.d. (n53). ***P,0.001; NS, not significant.

    (B) Binding analysis of MAdCAM-1 to purified WT a4b7 and a4CC/b7CC

    mutant by surface plasmon resonance. MAdCAM-1 was immobilized on

    CM5 sensor chip at a density of 720 RU and integrins were injected at 30 ml/minute. Traces show increasing concentrations (2.5, 7 and 10 nM) of purified

    integrins. Arrows indicate the start and end points of injections.

    Journal of Cell Science 126 (21)5032

  • Journ

    alof

    Cell

    Scie

    nce

    (Fig. 2C), indicating the activation of a4b7. Thus, integrinactivation induced by breaking the two disulfide bonds at the

    knees of a4b7 is not simply due to the change of flexibility at theknee region.

    Breaking the two disulfide bonds induces a unique active

    conformation of a4b7Upon activation, integrin converts from the low-affinity bent

    conformation to the high-affinity extended conformation. A

    previous negative staining EM study indicated that the complete

    a4b7 ectodomain in 1 mM Ca2+/Mg2+ had a compact rather than

    extended conformation (Yu et al., 2012). Extension of the integrin

    ectodomain has been shown to decrease the retention volume of

    purified integrin proteins in gel filtration because of an increase in

    the hydrodynamic radius of integrin molecules (Takagi et al.,

    2002; Xiong et al., 2009). To investigate the conformational

    change induced by disruption of these disulfide bonds, we

    compared the isocratic elution profiles of purified WT a4b7 anda4CC/b7CC mutant with full-length ectodomain on a molecularsieve chromatography column. WT a4b7 was eluted at 10.84 ml in1 mM Ca2+/Mg2+, whereas its elution volume in 0.5 mM Mn2+

    decreased to 10.65 ml, which is consistent with the more extended

    conformation of Mn2+-activated a4b7 (Fig. 3A). Surprisingly, thea4CC/b7CC active mutant showed a larger retention volume of10.98 ml in Ca2+/Mg2+. Thus, our results suggest that breaking

    these disulfide bonds induces a relatively compact instead of the

    typical extended active ectodomain of a4b7 (Fig. 3A).To demonstrate that this unique conformation exists in a4CC/

    b7CC on the cell surface, we used fluorescence resonance energytransfer (FRET) to examine the conformational changes of a4b7. Toassess the orientation of a4b7 ectodomain relative to the plasmamembrane, b7 I domain was labeled with Alexa-Fluor-488-Act-1

    Fab as donor, and the outer leaflet of cell membrane was labeled with

    FM4-64-FX as acceptor (Pan et al., 2010). The FRET efficiency of

    a4CC/b7CC transfectants was significantly higher than that of WTa4b7 transfectants when cells adhered to poly-L-lysine (Fig. 3B),indicating that the b7 I domain in a4CC/b7CC was closer to the cellmembrane before ligand binding than that in WT a4b7. Binding ofMAdCAM-1 to a4b7 significantly decreased the FRET efficiency ofboth WT a4b7 and a4CC/b7CC transfectants, suggesting that theintegrin head domain stands away from the cell membrane (Fig. 3B).

    However, the FRET efficiency of a4CC/b7CC transfectants was stillsubstantially higher than that of WT a4b7 transfectants, even afterligand occupancy (Fig. 3B). Activation of integrin by 0.5 mM Mn2+

    maximally decreased the FRET efficiency of both WT a4b7 anda4CC/b7CC transfectants to a similar level (Fig. 3B), suggesting fullextension of a4b7 ectodomain. To further confirm that breaking thetwo disulfide bonds does not induce the typical conformational

    change in integrin a4b7 on the cell surface, we examined the epitopeexpression of J19, a monoclonal antibody (mAb) that recognizes an

    epitope located in the a4b7 head domain and only expressed in theextended a4b7 (Qi et al., 2012) (Fig. 3C). The conformationalchange in a4b7 upon activation by chemokine and Mn

    2+ could be

    well reported by J19 binding (Qi et al., 2012). Consistent with the

    FRET results, both WT a4b7 and a4CC/b7CC mutant transfectantswere stained with J19 only after stimulation with 0.5 mM Mn2+ but

    not in 1 mM Ca2+/Mg2+ (Fig. 3C), suggesting that the a4CC/b7CCmutant does not have a typical active integrin conformation in Ca2+/

    Mg2+. Collectively, these findings show that breaking the two

    disulfide bonds at the knees of a4b7 induces a unique activeconformation that is different from the typical extended

    conformation of active integrin.

    Integrin activation is reported to be coupled with the separation

    of the a- and b-cytoplasmic domains (Kim et al., 2003). We next

    Fig. 3. Conformational changes of a4b7 induced by

    breakage of the two disulfide bonds at the knees of

    integrin. (A) Hydrodynamic analyses of WT a4b7 and

    a4CC/b7CC mutant proteins by molecular sieve

    chromatography. A representative experiment (one of

    three) is shown. Purified WT and mutant integrin

    proteins in HBS containing 1 mM Ca2+/Mg2+ or

    0.5 mM Mn2+ were analyzed. Each integrin was

    applied onto a pre-calibrated Superdex 200 column.

    Values next to the major peaks indicate peak elution

    volumes (mean 6 s.d., n53) (in ml). (B) FRET

    between b7 I domain and the plasma membrane in

    CHO-K1 cells expressing WT a4b7 or a4CC/b7CC

    mutant. Error bars represent s.d. (n510). (C) Flow

    cytometry analyses of the binding of a4b7 extension-

    specific mAb J19 to CHO-K1 cells expressing WT

    a4b7 or a4CC/b7CC mutant. J19 IgG recognition was

    measured as specific mean fluorescence intensity and

    quantified as a percentage of total a4b7 expression

    defined by staining with mAb FIB504 to b7. Error bars

    represent s.d. (n53). (D) Inter-subunit FRET between

    integrin a4 and b7 cytoplasmic domains in 293T

    transfectants expressing WT or mutant a4-mCFP/b7-

    mYFP. Error bars represent s.d. (n510). **P,0.01;

    NS, not significant.

    Regulation mechanism of a4b7 5033

  • Journ

    alof

    Cell

    Scie

    nce

    used FRET to investigate whether the activation of integrin

    induced by breaking the disulfide bonds was accompanied by

    separation of integrin cytoplasmic tails. Monomeric mutants of

    cyan fluorescent protein (mCFP) and yellow fluorescent protein

    (mYFP) were fused to the C-termini of integrin a4 and b7subunits to act as donor and acceptor, respectively (Kim et al.,

    2003; Pan et al., 2010). Addition of C-terminal mCFP and mYFP

    did not affect the binding of WT and mutant a4b7 to MAdCAM-1. Interestingly, cells expressing a4CC/b7CC showed the sameFRET efficiency as WT a4b7-expressing cells in all conditions(Fig. 3D). Therefore, activation of a4b7 induced by breaking thetwo disulfide bonds at the knees of integrin is unique because it

    does not induce the separation of a4 and b7 cytoplasmic domains,a typical conformational change coupled with integrin activation.

    Breaking the two disulfide bonds triggers integrin outside-in signaling independent of ligand binding

    To examine the influence of the two disulfide bonds on integrin

    outside-in signaling, we studied a4b7-mediated cell spreading.a4b7 CHO-K1 stable transfectants were allowed to adhere toimmobilized poly-L-lysine or MAdCAM-1 in serum-free F12

    medium for 2 hours, followed by fixation and microscopic

    analysis (Fig. 4A). WT a4b7 transfectants spread substantially onMAdCAM-1 substrates. Interference reflection microscopy

    showed an irregular shape and extensive areas of cell-substrate

    contact (Fig. 4A,B). By contrast, the same cells did not spread on

    immobilized poly-L-lysine, and exhibited the same area of

    projection as cells in suspension (Fig. 4A,B). Interestingly,

    a4CC/b7CC mutant transfectants showed obvious cell spreading

    and irregular shape of cell-substrate contact even on poly-L-

    lysine (Fig. 4A,B), suggesting activation of integrin outside-in

    signaling. We further investigated the activation of FAK and

    paxillin, two downstream molecules of integrin outside-in

    signaling (Turner, 2000), by measuring their phosphorylation

    (Fig. 4CE). Compared with WT cells, the expression and

    phosphorylation of FAK and paxillin were significantly elevated

    in a4CC/b7CC mutant cells adhered to immobilized poly-L-lysine, suggesting the activation of integrin downstream signaling

    independent of ligand binding (Fig. 4CE). These data strongly

    suggest that breaking the two disulfide bonds can trigger integrin

    outside-in signaling in a manner that is independent of ligand

    binding. In addition, cells that adhered to MAdCAM-1 substrates

    showed further increased expression and phosphorylation of FAK

    and paxillin as a result of enhanced integrin outside-in signaling

    triggered by ligand binding (Fig. 4CE).

    Breaking the two disulfide bonds induces a4b7 clusteringbefore ligand occupancy

    Although it is believed that integrin outside-in signaling is

    dependent on its global conformational change and ligand

    binding (Legate et al., 2009; Takagi et al., 2002), the above

    results showed that breaking the two disulfide bonds at the knees

    of a4b7 could trigger outside-in signaling in the absence ofintegrin global conformational changes and ligand binding. To

    address the underlying molecular mechanisms, we investigated the

    overall capacity of WT a4b7 and a4CC/b7CC integrins to undergocell surface redistribution and clustering (Fig. 5), because lateral

    diffusion and clustering of integrins can contribute to rearrangement

    Fig. 4. Loss of the two disulfide bonds induces integrin-mediated outside-in signaling independent of ligand binding. (A) Differential interference contrast

    (DIC) and interference reflection microscopy (IRM) images of a4b7 CHO-K1 stable transfectants after adhering to immobilized poly-L-lysine or MAdCAM-1

    for 2 hours in serum-free F12 medium. Scale bar: 20 mm. (B) Quantification of cell area (projection on the substrates) based on DIC images from A. Errorbars represent s.d. (n550). (C) Phosphorylation levels of FAK (pY397) and paxillin (pY118) in a4b7 CHO-K1 stable cells adhered to immobilized poly-L-

    lysine or MAdCAM-1. Numbers to the left of the gel indicate position of molecular mass markers. (D,E) Quantitative analyses of pY397-FAK to total FAK

    (D) and pY118-paxillin to total paxillin (E). Error bars represent s.d. (n53). *P,0.05; **P,0.01; ***P,0.001; NS, not significant.

    Journal of Cell Science 126 (21)5034

  • Journ

    alof

    Cell

    Scie

    nce

    of the cytoskeleton and transduction of outside-in signaling (Cluzel

    et al., 2005). In a4CC/b7CC CHO-K1 transfectants adhered toimmobilized poly-L-lysine, numerous a4b7 microclusters wereobserved at the cell periphery (Fig. 5A), whereas no discernible

    clusters of a4b7 were observed in cells expressing WT a4b7 onimmobilized poly-L-lysine (Fig. 5A). In cells adhered to

    MAdCAM-1, both WT and mutant integrins formed clusters at

    the cell periphery (Fig. 5B). Moreover, a4CC/b7CC formed manymore integrin clusters underneath the main cell body than WT

    (Fig. 5B), which might account for the higher phosphorylation

    levels of FAK and paxillin in a4CC/b7CC transfectants than in WTa4b7-expressing cells adhered to MAdCAM-1 (Fig. 4CE).

    Next, we addressed whether a4CC/b7CC clustering is induced bylinkage of integrins to the cytoskeleton. Integrins have no intrinsic

    actin-binding sites, and their linkage to the cytoskeleton relies on the

    binding of scaffold proteins (Legate et al., 2009). Therefore, we

    generated a4b7 constructs that could not link to the cytoskeleton bydeleting regions of the cytoplasmic domains that contain binding sites

    for scaffold proteins in both a4 and b7 subunits of WT a4b7 (a4D976/b7D736) and the a4CC/b7CC mutant (a4CCD976/b7CCD736)(Fig. 5C) (Legate et al., 2009; OToole et al., 1994). a4D976/b7D736 and a4CCD976/b7CCD736 were transiently expressed inCHO-K1 cells and their distribution in cells adhered to immobilized

    poly-L-lysine was examined (Fig. 5C). Truncation of the cytoplasmic

    domains in WT a4b7 did not change the even distribution of a4b7 onthe cell surface. By contrast, although the capability to support cell

    spreading on poly-L-lysine was lost due to the disrupted linkage of

    integrin to the cytoskeleton, a4CC/b7CC with truncated cytoplasmic

    domains (a4CCD976/b7CCD736) still formed microclusters on thecell surface, suggesting that a4CC/b7CC integrins can spontaneouslycluster on the cell surface independent of linkage to the cytoskeleton.

    To further confirm that breaking the two disulfide bonds can induce

    integrin clustering, and to exclude any effect of poly-L-lysine, we

    investigated the distribution of a4b7 on the cell surface when cellswere in suspension (Fig. 5D). Consistently, integrin clusters were

    detected in a4CC/b7CC transfectants, but not in WT a4b7transfectants. Notably, CHO-K1 cells expressing a4CC/b7CCmutant are basically round in suspension but the shape is less

    regular than WT cells (Fig. 5D). We speculate that the clustered

    a4CC/b7CC might induce cytoskeleton rearrangement to some extent,resulting in slight cell shape change and triggering cell spreading that

    is independent of ligand binding. Collectively, these results clearly

    demonstrate that breaking these disulfide bonds can spontaneously

    induce a4b7 clustering independent of ligand binding, resulting inincreased integrin avidity and contributing to rearrangement of the

    cytoskeleton and transduction of outside-in signaling.

    Disulfide bonds are not reduced in a4b7 activated by inside-

    out signaling or Mn2+

    To study whether reduction of the two disulfide bonds at the

    knees of a4b7 is involved in a4b7 activation, we examined freecysteine residues in a4b7 before and after activation by differentstimuli, including talin overexpression, phorbol-12-myristate-13-

    acetate (PMA) stimulation and Mn2+ treatment. Talin has been

    reported to activate integrins by binding to integrin cytoplasmic

    domains (Critchley and Gingras, 2008). PMA is a phorbol ester

    Fig. 5. Breaking the two disulfide bonds

    induces integrin clustering independent of

    ligand binding and cytoskeleton linkage.

    Confocal images of a4b7 distribution in CHO-

    K1 transfectants after adhering to immobilized

    poly-L-lysine (A,C) or MAdCAM-1 (B) for

    2 hours in serum-free F12 medium. (C) The

    effect of a4b7 cytoplasmic domain deletion

    (a4D976 and b7D736) on a4b7 distribution in

    CHO-K1 transfectants that adhered to

    immobilized poly-L-lysine. The deleted regions

    of a4b7 cytoplasmic domains are shown in red.

    (D) Confocal images of integrin distribution in

    CHO-K1 transfectants in suspension. Cells

    were detached, starved for 4 hours and fixed

    with 3.7% paraformaldehyde before staining

    with mAb FIB27 against b7. The images are

    representative of three independent

    experiments. White arrowheads indicate the

    representative integrin clusters. Scale bars:

    10 mm.

    Regulation mechanism of a4b7 5035

  • Journ

    alof

    Cell

    Scie

    nce

    that can activate integrins through the protein kinase C (PKC)

    pathway (Banno and Ginsberg, 2008). Both agents activate

    integrins by inside-out signaling. By contrast, Mn2+ activates

    integrin independent of cytoplasmic signaling. a4b7 mutant (a4/b7C526S) that contains one free cysteine residue in the b7 subunitwas used as a sensitivity control for the detection of single free

    sulfhydryl. In addition, 5 mM DTT was used to maximally

    reduce the disulfide bonds in integrins.

    To detect free cysteine residues in a4b7, we stained freesulfhydryls and a4b7 on the surface of a4b7 and a4/b7C526S293T transient transfectants with Alexa-Fluor-488-maleimide

    and anti-b7 mAb FIB27, respectively. DTT treatment resulted inrobust Alexa Fluor 488 signals on the cell surface, some of which

    colocalized with integrin b7, suggesting the reduction of disulfidebonds in integrins after DTT treatment (Fig. 6A,B). a4/b7C526Stransfectants showed clear Alexa Fluor 488 signals with strong

    colocalization with b7, indicating that this system is sufficientlysensitive to detect a single free sulfhydryl (Fig. 6A,B). However,

    there were no detectable free sulfhydryls on WT a4b7

    transfectants on immobilized poly-L-lysine or MAdCAM-1

    before and after activation of integrin by PMA, talin or Mn2+

    (Fig. 6A,B). These data suggest that no disulfide bond is reduced

    in integrins activated by either inside-out signaling or Mn2+. To

    confirm the above results, we used biotin-BMCC to conjugate

    biotin to the exposed free sulfhydryls in proteins on the surface of

    WT and a4/b7C526S mutant a4b7 293T transfectants. Integrina4b7 was then immunoprecipitated and the biotinylatedsulfhydryls were visualized by probing with avidin-HRP

    (Fig. 6C). Consistently, only the b7 subunit from a4/b7C526Sand a4 and b7 subunits from DTT-treated a4b7 were biotinylatedand detected by avidin-HRP, suggesting that free cysteine

    residues only exist in these proteins, but not in WT a4b7 beforeand after activation by inside-out signaling or Mn2+ (Fig. 6).

    To confirm the above results in a more physiological system, three

    kinds of chemokines were used to activate a4b7 in freshly isolatedhuman peripheral blood lymphocytes (PBLs) (Laudanna et al.,

    2002). Consistently, free cysteine residues were detected in PBLs

    only after treatment with DTT, but not before or after activation by

    Fig. 6. Detection of the exposed free cysteine residues in a4b7 pre- and post-activation by inside-out signaling or Mn2+. WT a4b7 293T-transfectants

    were pre-treated with different stimuli to induce integrin activation, including 5 mM DTT at 37 C for 30 minutes, 10 ng/ml PMA at 37 C for 3 minutes, 0.5 mM

    Mn2+ for 5 minutes and talin overexpression. Cells expressing a4b7 mutant (a4/b7C526S), which contains one free cysteine residue in the b7 subunit, was

    used as a sensitivity control. (A,B) Confocal microscopy visualization of exposed free cysteine residues in cell surface proteins. Cells were allowed to adhere to

    immobilized poly-L-lysine (A) or MAdCAM-1 (B) for 5 minutes, and followed by staining with Alexa-Fluor-488-maleimide and mAb FIB27 to label free

    sulfhydryls in cell surface proteins and b7 integrin, respectively. Scale bars: 10 mm. (C) Detection of the exposed free cysteine residues in a4b7 byimmunoprecipitation and western blotting. Cells were treated with biotin-BMCC to label exposed free sulfhydryls in cell surface proteins. a4b7 integrin was

    then immunoprecipitated using mAb FIB504 and the biotin-labeled free sulfhydryls were detected by avidin-HRP. Numbers to the left of the gel indicate position

    of molecular mass markers. The results are representatives of three independent experiments.

    Journal of Cell Science 126 (21)5036

  • Journ

    alof

    Cell

    Scie

    nce

    SDF-1a, CCL21, CCL25 or Mn2+ (supplementary material Fig. S1).These results demonstrate that no integrin disulfide bonds are

    reduced before or after activation of a4b7 by either inside-outsignaling or Mn2+, suggesting that the two disulfide bonds at the

    knees of a4b7 are not reduced under normal activating conditions.

    a4C589C594 and b7C494C526 are selectively reduced by

    0.1 mM DTT

    Although the two disulfide bonds at the knees of a4b7 remainintact during integrin activation by inside-out signaling or Mn2+,

    they might be disrupted under certain reducing conditions that are

    produced in vivo during particular physiological and pathological

    processes (Alderete and Provenzano, 1997). To determine the

    minimal reducing strength required for the reduction of these two

    disulfide bonds in a4b7, we examined the free cysteine residuesin WT a4b7 and a4CC/b7CC mutant after treatment with serialconcentrations of DTT (Fig. 7A). After treatment with 0.1 and

    0.2 mM DTT, we detected biotinylated sulfhydryls only in a4 andb7 subunits from WT a4b7, but not in the a4CC/b7CC mutant thatlacks the two disulfide bonds at the knees of a4b7 (Fig. 7A).These data suggest that only these two disulfide bonds in a4b7 areselectively reduced by 0.1 and 0.2 mM DTT.

    Next, we examined whether treatment of WT a4b7 with DTTcould recapitulate the activation and outside-in signaling

    mediated by the a4CC/b7CC mutant. As expected, treatment ofWT a4b7 293T transfectants with 0.1 and 0.2 mM DTTsignificantly increased the number of cells firmly adhered to

    immobilized MAdCAM-1, indicating activation of integrin

    (Fig. 7B). In contrast to the decreased FRET between b7 Idomain and cell membrane (Fig. 7C) and between a4 and b7cytoplasmic tails (Fig. 7D) in a4b7 activated by Mn

    2+, a4b7activated by 0.1 and 0.2 mM DTT showed increased FRET

    between the b7 I domain and cell membrane and unchangedFRET between a4 and b7 cytoplasmic tails, suggesting that a4b7activated by 0.1 and 0.2 mM DTT has a compact conformation

    with clasped a4 and b7 cytoplasmic tails (Fig. 7C,D). Consistentwith the results obtained using a4CC/b7CC transfectants,treatment of WT a4b7 CHO-K1 transfectants with 0.1 and0.2 mM DTT resulted in cell spreading on immobilized poly-L-

    lysine with formation of remarkable a4b7 clusters (Fig. 7E).Moreover, phosphorylation levels of FAK and paxillin were

    elevated (Fig. 7F,G).

    To further confirm that a4C589C594 and b7C494C526 wereselectively reduced by 0.1 mM DTT and their biological roles are

    Fig. 7. Activation of integrin a4b7 by selectively reducing

    a4C589C594 and b7C494C526 with DTT. (A) Detection

    of the exposed free cysteine residues in a4b7. WT a4b7 or

    a4CC/b7CC 293T transfectants were treated with the

    indicated concentrations of DTT at 37 C for 10 minutes.

    Subsequently, the exposed free cysteine residues in WT

    a4b7 or a4CC/b7CC mutant were labeled with biotin-

    BMCC. a4b7 was then immunoprecipitated and the

    biotinylated sulfhydryls were detected by western blotting

    with avidin-HRP. Numbers to the left of the gel indicate

    position of molecular mass markers. (B) Adhesive modality

    of WT a4b7-expressing 293T cells before and after DTT

    treatment on MAdCAM-1 substrates under flow conditions.

    The number of rolling and firmly adherent cells was

    measured at a wall shear stress of 1 dyn/cm2. Error bars

    represent s.d. (n53). (C,D) FRET between b7 I domain and

    the plasma membrane in a4b7 293T transfectants (C) and

    inter-subunit FRET between integrin a4 and b7 cytoplasmic

    domains in a4-mCFP/b7-mYFP 293T transfectants (D)

    treated with different concentrations of DTT. Error bars

    represent s.d. (n510). (EG) WT a4b7 CHO-K1 stable

    transfectants were treated with the indicated concentrations

    of DTT at 37 C for 10 minutes, and then were allowed to

    adhere to immobilized poly-L-lysine for 2 hours in serum-

    free F12 medium. (E) b7 integrin was stained with mAb

    FIB27 and visualized with fluorescence confocal

    microscopy. The images are representatives of three

    independent experiments. White arrowheads indicate the

    representative integrin clusters. Scale bar: 10 mm. (F) Thephosphorylation levels of FAK (pY397) and paxillin

    (pY118) were examined by western blot. Numbers to the left

    of the gel indicate position of molecular size markers.

    (G) Quantification of pY397-FAK to total FAK and pY118-

    paxillin to total paxillin. Ratios of WT sample were

    normalized to those of the WT sample on poly-L-lysine in

    Fig. 4 and ratios of the other samples were normalized

    proportionally. Error bars represent s.d. (n53). **P,0.01;

    ***P,0.001; NS, not significant.

    Regulation mechanism of a4b7 5037

  • Journ

    alof

    Cell

    Scie

    nce

    unique, we disrupted each of the other eight disulfide bonds that

    could be exposed to solution and examined the effects of thesemutations on integrin a4b7-mediated cell adhesion under flowconditions (Fig. 8A). Among the eight disulfide bonds, only

    disruption of C459C478 or C469C481 in b7 subunitsignificantly increased the number of firmly adherent cells,indicating the activation of integrin a4b7 (Fig. 8A). However,neither of these two mutations could induce integrin clustering

    and cell spreading on immobilized poly-L-lysine as did the0.1 mM DTT treatment or disruption of a4C589C594 orb7C494C526 (Fig. 8B). Thus, only the two disulfide bonds atintegrin knees, a4C589C594 or b7C494C526, could beselectively reduced by 0.1 mM DTT. Taken together, thesedata suggest a novel mechanism for integrin activation and

    signaling by selectively reducing the two disulfide bonds at theknees of a4b7 under certain reducing environments.

    DiscussionA major finding of our present study is that integrin a4b7 can beactivated by selectively breaking two conserved disulfide bonds,a4C589C594 and b7C494C526, located at the knees ofintegrin. Interestingly, integrin activated by this mechanism hasa unique active conformation that is different from the globalconformation induced by Mn2+ stimulation. In addition, activated

    a4b7 integrin can spontaneously cluster on the cell membrane andtrigger integrin outside-in signaling independent of ligandbinding.

    All integrins contain a large number of disulfide bonds that aregenerally believed to facilitate protein folding and stabilize three-dimensional structures (Calvete et al., 1991). The two disulfide

    bonds at the knees of integrin a4b7 are exposed to solution,making them easily accessible to reducing agents. In this study,

    these two disulfide bonds were selectively reduced by 0.1 mMDTT, which induced the activation of integrin a4b7 and triggeredoutside-in signaling. Accumulating evidence indicates thatreducing environments are produced and play essential roles in

    some physiological and pathological processes. Dendritic cells,monocytes, macrophages and B lymphocytes have been shown torelease free thiols and oxidoreductase to generate and maintain

    reducing environments, which have been demonstrated to beessential for immune cell activation and successful antigenpresentation (Angelini et al., 2002; Castellani et al., 2008). In

    addition, a reducing condition equivalent to 40 mM to 1 mM DTThas been reported in pathogen invasion processes, such as vaginalinfection by Trichomonas vaginalis (Alderete and Provenzano,1997). Such conditions should be capable of inducing a4b7activation by reducing the two disulfide bonds at the knees ofintegrin. Thus, integrin a4b7 and its downstream signaling could beactivated by this mechanism in vivo to promote ligand-independent

    cell spreading under particular reducing environments.

    It is important to note that breaking the two conserveddisulfide bonds at knees of a4b7 triggered the activation ofintegrin downstream signaling (Fig. 4; Fig. 7EG) without theextension of a4b7 ectodomain and separation of a4 and b7cytoplasmic tails (Fig. 3BD and Fig. 7C,D), suggesting thatectodomain extension and tail separation are not absolutely

    required for the activation of integrin downstream signaling.Also, breakage of the two disulfide bonds induced spontaneousa4b7 clustering independent of ligand binding and cytoskeletonlinkage (Fig. 5). Thus, it is tempting to speculate that someparticular interfaces might be formed in this unique activateda4b7 with compact structure, which can mediate integrinclustering through interactions between integrin ectodomainsand subsequently trigger integrin downstream signaling.

    The importance of disordered regions in the regulation of

    protein allostery has been increasingly recognized (Hilser andThompson, 2007). Our study showed that deletion of either the a4knee (a4D4) or the b7 I-EGF2 C1C2 loop (b7D13) stronglyactivated integrin a4b7 (Fig. 2C). The head domains of a4D4b7and a4b7D13 mutants were much closer to the cell membranethan that of WT a4b7 both before and after ligand occupancy,suggested by the increased FRET efficiencies between mutant

    a4b7 head and plasma membrane compared with those of the WT(supplementary material Fig. S2A). Furthermore, the cytoplasmictails of a4D4b7 and a4b7D13 mutants were more difficult todissociate upon MAdCAM-1 binding, as reflected by the slightlyhigher intracellular FRET efficiencies relative to those of the WT(supplementary material Fig. S2B). Previous studies on aVb3crystals revealed that the b3 I-EGF2 C1C2 loop forms an importantinterface with the aV thigh domain to stabilize integrin in an inactivebent state, and that deletion of this loop induced constitutiveactivation of aVb3 (Xiong et al., 2009); Benoit and colleaguesdetected similar activation of both aIIbb3 and aVb3 by shortening theb3 I-EGF2 C1C2 loop (Smagghe et al., 2010) and advanced a newscenario whereby the b3 knee functions as an entropic spring toadjust integrin conformation equilibrium between bent andextended conformations at different set points. Partially consistentwith these studies, we observed robust activation of integrin a4b7upon deletion of the b7 I-EGF2 C1C2 loop (a4b7D13). However,b7 C1C2 loop deletion resulted in a compact conformation insteadof the extended conformations of aIIbb3 and aVb3 induced by

    Fig. 8. Disruption of the other solvent-accessible disulfide bonds does not

    regulate a4b7 function the same way as 0.1 mM DTT or a4CC/b7CC

    mutation. (A) Adhesive modality of 293T cells expressing WT or mutant

    a4b7 on MAdCAM-1 substrates under flow conditions. The number of rolling

    and firmly adherent cells was measured at a wall shear stress of 1 dyn/cm2.

    Error bars represent s.d. (n53). ***P,0.001; NS, not significant.

    (B) Confocal images of a4b7 distribution in CHO-K1 transfectants after

    adhering to immobilized poly-L-lysine for 2 hours in serum-free F12

    medium. Scale bar: 10 mm.

    Journal of Cell Science 126 (21)5038

  • Journ

    alof

    Cell

    Scie

    nce

    shortening the b3 C1C2 loop. Considering that a4b7 is uniqueamong most integrins for its ability to mediate both rolling and firm

    cell adhesion (Berlin et al., 1993; Hamann et al., 1994), our results

    suggest that a4b7 has a different regulatory mechanism for integrinconformation to support the unique two-phase cell adhesion.

    In conclusion, our findings reveal a novel mechanism of integrin

    activation under specific reducing conditions by which integrin can

    signal and promote cell spreading in the absence of ligand in someparticular physiological and pathological processes. Moreover, this

    study suggests the possibility of integrin activation and signaling in

    the absence of global conformational changes.

    Materials and MethodsStructural model construction

    The full-length human integrin a4b7 ectodomain in a bent conformation wasconstructed by modeling the crystal structure of a4b7 headpiece (pdb3V4P) andbuilding the legs homologically using the counterparts in the structure of aVb3(pdb3IJE) with Schrodinger software. This initial model was processed using thefollowing steps. Molecular dynamics simulation was performed with Gromacs4.5.3.To simulate the solvent environment, the system was placed in the center of arectangular water box. The closest distance from the protein to the water box was10 A. Na+ and Cl2 ions were added to neutralize the system. The resulting systemconsisted of ,240,000 atoms. A Gromos 0653A6 force field was used for a4b7(Oostenbrink et al., 2004). The simple point charge model was used for water. First,the system was energy-minimized for 5000 conjugate-gradient steps with all heavyatoms of the protein constrained with a force constant of 1000 kJ/mol.nm2. Then thesystem was equilibrated for 2 nseconds with all heavy atoms of the proteinconstrained followed for another 2 nsecond simulation, with Ca2+ constrained under1 atm at 300 K. Finally a 1.0 msecond equilibration MD was carried out. For thesimulation, periodic boundary condition was imposed. Bonds involving hydrogenwere set to be rigid with LINCS (Hess, 2008) algorithm for protein and SHAKE(Kollman, 1992) algorithm for water, thus permitting use of an integration step of2 fseconds. Non-bonded forces were cut off at 14 A and electrostatics were treated byutilizing the Particle Mesh Ewald method (Kollman, 1992). For the NPT simulation,the Berendsen weak coupling method (Berendsen, 1991) was applied with couplingconstants of 0.1 pseconds and 0.5 pseconds respectively, to maintain the system at300K and 1 atm. Images of trajectory were recorded every 200 picoseconds. The last2000 images were used to obtain the time-averaged model used in this paper.

    Antibodies and reagents

    mAb goat anti-rat IgG-Alexa-Fluor-488 was from Invitrogen. mAb against paxillinand b7 integrin (FIB27) were from BD Biosciences. pY118-paxillin antibody wasfrom Cell Signaling. mAb against FAK and pY397-FAK were from UpstateBiotechnology. mAb J19 human IgG was prepared as described (Qi et al., 2012).mAb FIB504 against human b7 was prepared from hybridoma (DevelopmentalStudies Hybridoma Bank). Human MAdCAM-1/Fc fusion protein (MAdCAM-1),and Act-1 mAb specific for a4b7 were as previously described (Tidswell et al.,1997). Complete protease inhibitor cocktail tablets and PhosSTOP phosphataseinhibitor cocktail tablets were from Roche. Chemokines were from R&D.

    Protein purification

    The full-length a4b7 ectodomain, consisting of b-propeller, thigh, knee, calf-1,calf-2 domains in a4 subunit and bI domain, hybrid, PSI, I-EGF1-4, b-Taildomains in b7 subunit, was purified as previously described (Qi et al., 2012; Yuet al., 2012; Zhu et al., 2008).

    Flow chamber assay

    The flow chamber assay was performed as described (Chen et al., 2003). Avelocity of 1 mm/second was the minimum velocity required to define a cell asrolling instead of firmly adherent.

    Flow cytometry

    Immunofluorescence flow cytometry was carried out as described (Chen et al.,2006). Expression levels of integrin a4b7 were determined by staining withFIB504, and followed by staining with goat anti-rat IgG-Alexa-Fluor-488. Beforestaining with J19 human IgG, cells were washed with HBS buffer with 5 mMEDTA, and then resuspended in HBS buffer containing either 1 mM Ca2+/Mg2+ or0.5 mM Mn2+. Stained cells were then measured using FACS Calibur (BDBiosciences) and analyzed using WinMDI 2.9 software.

    Molecular sieve chromatography of purified integrin a4b7All molecular sieve chromatography analyses were performed as previouslydescribed (Adair et al., 2005) using pre-calibrated Superdex 200 (10/300 GL)

    columns on an AKTA purifier system running Unicorn 5.01 software (GEHealthcare) at a flow rate of 0.5 ml/minute at room temperature. The elutionprofiles were monitored in-line by UV adsorption at 280 nm. HBS buffercontaining either 1 mM Ca2+/Mg2+ or 0.5 mM Mn2+ was used throughout. 6 mg ofpurified WT or mutant integrin a4b7 was injected.

    Surface plasmon resonance

    Experiments were performed using the BIAcore T100. Dilutions of WT or mutanta4b7 in HBS containing either 1 mM Ca

    2+/Mg2+ or 0.5 mM Mn2+ were injectedover 3 minutes at 30 ml/minute into the flow cell containing 720 RU ofMAdCAM-1 coupled to a sensor chip CM5. After each cycle of association anddissociation, the surface was regenerated by injecting 30 ml of regeneration buffercontaining 20 mM EDTA and 25 mM NaOH. This treatment did not affect thesubsequent binding reaction after up to 60 repetitions. All measurements werebaseline corrected by subtracting the sensorgram obtained with control surface andkinetic parameters were determined by fitting the data to two-step binding modelusing BIAevaluation software ver2.0.2.

    FRET assay

    FRET was measured as described (Kim et al., 2003; Pan et al., 2010; Xiong et al.,2009). For extracellular FRET, cells were seeded on poly-L-lysine (100 mg/ml) orMAdCAM-1 (10 mg/ml) substrates in serum-free medium, and incubated for10 minutes at 37 C. 0.5 mM Mn2+ was added to activate integrin where indicated.For DTT treatment, cells were pretreated with 0.1 or 0.2 mM DTT at 37 C for10 minutes. Adherent cells were fixed with 3.7% formaldehyde (PFA)/PBS for15 minutes at room temperature and nonspecific sites were blocked by incubationwith 10% serum-rich medium at room temperature for 10 minutes. Then cells werestained with 20 mg/ml Act-1 Fab conjugated with Alexa Fluor 488 for 20 minutesat 37 C. After two washes, cells were labeled with 10 mM FM-4-64 FX for4 minutes on ice, washed once, fixed and mounted with GVA mount (Invitrogen)under a coverslip.

    For detecting the association of integrin cytoplasmic tails, a4-mCFP/b7-mYFP293T transient transfectants were treated as above. Then cells were fixed with3.7% paraformaldehyde in PBS for 15 minutes at room temperature and subjectedto photobleach FRET imaging.

    FRET image acquisition, image registration, background subtraction and dataanalyses were performed with Leica TCS SP5 under a 636 oil objective. FRETefficiency (E) was calculated as E51[Fdonor(d)Pre/Fdonor(d)Post], whereFdonor(d)Pre and Fdonor(d)Post are the mean donor emission intensity of pre-and post-photobleaching.

    Cell spreading and western blotting

    Glass coverslips were coated with 100 mg/ml poly-L-lysine or 10 mg/mlMAdCAM-1 overnight at 4 C and blocked by 2% BSA at 37 C for 1 hour.Cells were seeded on coated coverslips in serum-free F12 medium at 37 C for2 hours, with 0.5 mM Mn2+ added to the medium during spreading whereindicated.

    Differential interference contrast and interference reflection microscopy wereconducted on the OLYMPUS IX71 microscope with a 636oil objective coupled toan Orca CCD camera (Q-IMAGING). For quantification of cell spreading, outlinesof 50 randomly selected adherent cells from three independent experiments weregenerated, and the number of pixels contained within each of these regions wasmeasured by using Image-Pro plus v6.0.

    For western blotting, after cell spreading, cells were washed and lysed with lysisbuffer (20 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% Triton X-100, 0.05% Tween-20, Complete protease inhibitor cocktail tablets and PhosSTOP phosphataseinhibitor cocktail tablets) on ice for 30 minutes. Cell lysates were then analyzed byblotting for FAK, pY397-FAK, paxillin, pY118-paxillin and b-actin. Intensityanalyses were conducted using Image J software.

    Integrin clustering

    After cell spreading, cells were fixed with 3.7% paraformaldehyde in PBS at roomtemperature for 10 minutes. Anti-b7 mAb FIB27 (5 mg/ml) was used to stain b7 atroom temperature for 2 hours, followed by staining with goat anti-rat IgG-Cy3(5 mg/ml) at room temperature for 1 hour. Finally, coverslips were mounted withGVA mount and images were obtained with Leica TCS SP5 under a 636 oilobjective.

    Free cysteine detection

    All the buffers used in these assays were degassed for at least 30 minutes. Cellswere pretreated with 5 mM DTT at 37 C for 30 minutes where indicated. PMA(10 ng/ml) stimulation was applied at 37 C for 3 minutes. To visualize freecysteine residues on cell surfaces using immunofluorescence staining, 293Ttransfectants with or without stimulation were allowed to adhere to poly-L-lysineor MAdCAM-1 surfaces for 5 minutes before staining. For human peripheral bloodlymphocytes (PBLs), the indicated chemokines were coated together with poly-L-lysine or MAdCAM-1 at 2 mg/ml and cells were allowed to adhere for 3 minutes

    Regulation mechanism of a4b7 5039

  • Journ

    alof

    Cell

    Scie

    nce

    before staining. Alexa-Fluor-488-maleimide (Invitrogen) was applied for45 minutes to label free cysteine residues, and then cells were washed andfixed. b7 detection was the same as for the integrin clustering test. DAPI (1 mg/ml)was applied for 15 minutes at room temperature to detect cell nuclei. Finally,coverslips were mounted with GVA mount and images were obtained with LeicaTCS SP5 under a 636 oil objective.

    In the immunoprecipitation assay, chemokines used to activate integrin a4b7 onPBLs were applied at 500 ng/ml for 3 minutes at room temperature. For DTTtitration, 293T transfectants were treated with different concentrations of DTT at37 C for 10 minutes. Thereafter cells were labeled with 400 mM 1-biotinamido-4(49-[maleimodoethyl-cyclohexane]-carboxamido) butane (biotin-BMCC) (Pierce)at room temperature for 30 minutes and subsequently washed and lysed. a4b7immunoprecipitates were analyzed by blotting biotinylated sulfhydryls withhorseradish peroxidase-conjugated avidin (avidin-HRP).

    Statistical analysis

    The paired Students t-test was used for statistical analyses of experiments withtwo conditions. In the cases of three or more conditions, analysis of variance(ANOVA) was used with Bonferroni post-tests (two factors) or Dunnett post-tests(one factor).

    AcknowledgementsWe thank Dr DianQing Wu and Dr JunLin Guan for advice onexperiments and data.

    Author contributionsK.Z., Y.D.P., C.Q.X., G.H.L. and J.F.C. designed experiments; K.Z.,Y.D.P., J.P.Q., M.B.Z. and J.Y. performed experiments and analyzeddata; K.Z., Y.D.P., G.H.L. and J.F.C. interpreted results; themanuscript was drafted by K.Z. and Y.D.P. and edited by J.F.C.

    FundingThis work was supported by grants from the National Basic ResearchProgram of China [grant numbers 2010CB529703, 2014CB541905,2012CB721002]; the National Natural Science Foundation of China[grant numbers 31190061, 31271487, 30970604, 31070641]; theScience and Technology Commission of Shanghai Municipality[grant number 11JC1414200]; the National 863 program of China[grant number 2012AA01A305]; the China Postdoctoral ScienceFoundation [grant number 2012T50445; and the PostdoctoralResearch Program from SIBS [grant number 2012KIP503]. Theauthors gratefully acknowledge the support of SA-SIBS scholarshipprogram.

    Supplementary material available online at

    http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.134528/-/DC1

    ReferencesAdair, B. D., Xiong, J. P., Maddock, C., Goodman, S. L., Arnaout, M. A. and

    Yeager, M. (2005). Three-dimensional EM structure of the ectodomain of integrinalphaVbeta3 in a complex with fibronectin. J. Cell Biol. 168, 1109-1118.

    Alderete, J. F. and Provenzano, D. (1997). The vagina has reducing environmentsufficient for activation of Trichomonas vaginalis cysteine proteinases. Genitourin.Med. 73, 291-296.

    Angelini, G., Gardella, S., Ardy, M., Ciriolo, M. R., Filomeni, G., Di Trapani, G.,

    Clarke, F., Sitia, R. and Rubartelli, A. (2002). Antigen-presenting dendritic cellsprovide the reducing extracellular microenvironment required for T lymphocyteactivation. Proc. Natl. Acad. Sci. USA 99, 1491-1496.

    Banno, A. and Ginsberg, M. H. (2008). Integrin activation. Biochem. Soc. Trans. 36,229-234.

    Berendsen, H. J. C. (1991). Transport properties computed by linear response throughweak coupling to a bath. In Computer Simulations in Material Science (ed. M. Meyerand V. Pontikis), pp. 139-155. Dordrecht, The Netherlands: Kluwer AcademicPublishers.

    Berlin, C., Berg, E. L., Briskin, M. J., Andrew, D. P., Kilshaw, P. J., Holzmann, B.,

    Weissman, I. L., Hamann, A. and Butcher, E. C. (1993). a 4 b 7 integrin mediateslymphocyte binding to the mucosal vascular addressin MAdCAM-1. Cell 74, 185-195.

    Butta, N., Arias-Salgado, E. G., Gonzalez-Manchon, C., Ferrer, M., Larrucea, S.,

    Ayuso, M. S. and Parrilla, R. (2003). Disruption of the beta3 663-687 disulfidebridge confers constitutive activity to beta3 integrins. Blood 102, 2491-2497.

    Calvete, J. J., Henschen, A. and Gonzalez-Rodrguez, J. (1991). Assignment ofdisulphide bonds in human platelet GPIIIa. A disulphide pattern for the beta-subunitsof the integrin family. Biochem. J. 274, 63-71.

    Calzada, M. J., Alvarez, M. V. and Gonzalez-Rodriguez, J. (2002). Agonist-specificstructural rearrangements of integrin alpha IIbbeta 3. Confirmation of the bentconformation in platelets at rest and after activation. J. Biol. Chem. 277, 39899-39908.

    Carman, C. V. and Springer, T. A. (2003). Integrin avidity regulation: are changes inaffinity and conformation underemphasized? Curr. Opin. Cell Biol. 15, 547-556.

    Castellani, P., Angelini, G., Delfino, L., Matucci, A. and Rubartelli, A. (2008). Thethiol redox state of lymphoid organs is modified by immunization: role of differentimmune cell populations. Eur. J. Immunol. 38, 2419-2425.

    Chen, J. F., Salas, A. and Springer, T. A. (2003). Bistable regulation of integrinadhesiveness by a bipolar metal ion cluster. Nat. Struct. Biol. 10, 995-1001.

    Chen, J., Yang, W., Kim, M., Carman, C. V. and Springer, T. A. (2006). Regulationof outside-in signaling and affinity by the beta2 I domain of integrin alphaLbeta2.Proc. Natl. Acad. Sci. USA 103, 13062-13067.

    Chigaev, A., Blenc, A. M., Braaten, J. V., Kumaraswamy, N., Kepley, C. L.,

    Andrews, R. P., Oliver, J. M., Edwards, B. S., Prossnitz, E. R., Larson, R. S. et al.(2001). Real time analysis of the affinity regulation of alpha 4-integrin. Thephysiologically activated receptor is intermediate in affinity between resting andMn(2+) or antibody activation. J. Biol. Chem. 276, 48670-48678.

    Cluzel, C., Saltel, F., Lussi, J., Paulhe, F., Imhof, B. A. and Wehrle-Haller,B. (2005). The mechanisms and dynamics of (alpha)v(beta)3 integrin clustering inliving cells. J. Cell Biol. 171, 383-392.

    Critchley, D. R. and Gingras, A. R. (2008). Talin at a glance. J. Cell Sci. 121, 1345-1347.

    Ekblom, P., Vestweber, D. and Kemler, R. (1986). Cell-matrix interactions and celladhesion during development. Annu. Rev. Cell Biol. 2, 27-47.

    Feagan, B. G., Greenberg, G. R., Wild, G., Fedorak, R. N., Pare, P., McDonald,J. W., Dube, R., Cohen, A., Steinhart, A. H., Landau, S. et al. (2005). Treatment ofulcerative colitis with a humanized antibody to the alpha4beta7 integrin. N. Engl.J. Med. 352, 2499-2507.

    Hamann, A., Andrew, D. P., Jablonski-Westrich, D., Holzmann, B. and Butcher,E. C. (1994). Role of alpha 4-integrins in lymphocyte homing to mucosal tissues invivo. J. Immunol. 152, 3282-3293.

    Hess, B. (2008). P-LINCS: A parallel linear constraint solver for molecular simulation.J. Chem. Theory Comput. 4, 116-122.

    Hilser, V. J. and Thompson, E. B. (2007). Intrinsic disorder as a mechanism tooptimize allosteric coupling in proteins. Proc. Natl. Acad. Sci. USA 104, 8311-8315.

    Hynes, R. O. (2002). Integrins: bidirectional, allosteric signaling machines. Cell 110,673-687.

    Kim, M., Carman, C. V. and Springer, T. A. (2003). Bidirectional transmembranesignaling by cytoplasmic domain separation in integrins. Science 301, 1720-1725.

    Kollman, S. M. P. A. (1992). An analytical version of the SHAKE and RATTLEalgorithms for rigid water models. J. Comput. Chem. 3, 952-962.

    Laudanna, C., Kim, J. Y., Constantin, G. and Butcher, E. (2002). Rapid leukocyteintegrin activation by chemokines. Immunol. Rev. 186, 37-46.

    Legate, K. R., Wickstrom, S. A. and Fassler, R. (2009). Genetic and cell biologicalanalysis of integrin outside-in signaling. Genes Dev. 23, 397-418.

    Litvinov, R. I., Nagaswami, C., Vilaire, G., Shuman, H., Bennett, J. S. and Weisel,

    J. W. (2004). Functional and structural correlations of individual alphaIIbbeta3molecules. Blood 104, 3979-3985.

    Luo, B.-H., Carman, C. V. and Springer, T. A. (2007). Structural basis of integrinregulation and signaling. Annu. Rev. Immunol. 25, 619-647.

    Miyamoto, S., Akiyama, S. K. and Yamada, K. M. (1995). Synergistic roles forreceptor occupancy and aggregation in integrin transmembrane function. Science 267,883-885.

    Mor-Cohen, R., Rosenberg, N., Einav, Y., Zelzion, E., Landau, M., Mansour, W.,Averbukh, Y. and Seligsohn, U. (2012). Unique disulfide bonds in epidermal growthfactor (EGF) domains of b3 affect structure and function of aIIbb3 and avb3 integrinsin different manner. J. Biol. Chem. 287, 8879-8891.

    OToole, T. E., Katagiri, Y., Faull, R. J., Peter, K., Tamura, R., Quaranta, V.,Loftus, J. C., Shattil, S. J. and Ginsberg, M. H. (1994). Integrin cytoplasmicdomains mediate inside-out signal transduction. J. Cell Biol. 124, 1047-1059.

    Oostenbrink, C., Villa, A., Mark, A. E. and van Gunsteren, W. F. (2004). Abiomolecular force field based on the free enthalpy of hydration and solvation: theGROMOS force-field parameter sets 53A5 and 53A6. J. Comput. Chem. 25, 1656-1676.

    Pan, Y., Zhang, K., Qi, J., Yue, J., Springer, T. A. and Chen, J. (2010). Cation-piinteraction regulates ligand-binding affinity and signaling of integrin alpha4beta7.Proc. Natl. Acad. Sci. USA 107, 21388-21393.

    Qi, J., Zhang, K., Zhang, Q., Sun, Y., Fu, T., Li, G. and Chen, J. (2012).Identification, characterization and epitope mapping of a human monoclonal antibodyJ19 that specifically recognizes the activated integrin alpha4 beta7. J. Biol. Chem.287, 15749-15759.

    Smagghe, B. J., Huang, P. S., Ban, Y. E., Baker, D. and Springer, T. A. (2010).Modulation of integrin activation by an entropic spring in the beta-knee. J. Biol.Chem. 285, 32954-32966.

    Takagi, J., Petre, B. M., Walz, T. and Springer, T. A. (2002). Global conformationalrearrangements in integrin extracellular domains in outside-in and inside-outsignaling. Cell 110, 599-11.

    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. et al. (1997).Structure-function analysis of the integrin beta 7 subunit: identification of domainsinvolved in adhesion to MAdCAM-1. J. Immunol. 159, 1497-1505.

    Journal of Cell Science 126 (21)5040

    http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.134528/-/DC1http://dx.doi.org/10.1083/jcb.200410068http://dx.doi.org/10.1083/jcb.200410068http://dx.doi.org/10.1083/jcb.200410068http://dx.doi.org/10.1073/pnas.022630299http://dx.doi.org/10.1073/pnas.022630299http://dx.doi.org/10.1073/pnas.022630299http://dx.doi.org/10.1073/pnas.022630299http://dx.doi.org/10.1042/BST0360229http://dx.doi.org/10.1042/BST0360229http://dx.doi.org/10.1016/0092-8674(93)90305-Ahttp://dx.doi.org/10.1016/0092-8674(93)90305-Ahttp://dx.doi.org/10.1016/0092-8674(93)90305-Ahttp://dx.doi.org/10.1016/0092-8674(93)90305-Ahttp://dx.doi.org/10.1182/blood-2003-01-0213http://dx.doi.org/10.1182/blood-2003-01-0213http://dx.doi.org/10.1182/blood-2003-01-0213http://dx.doi.org/10.1074/jbc.M205886200http://dx.doi.org/10.1074/jbc.M205886200http://dx.doi.org/10.1074/jbc.M205886200http://dx.doi.org/10.1074/jbc.M205886200http://dx.doi.org/10.1016/j.ceb.2003.08.003http://dx.doi.org/10.1016/j.ceb.2003.08.003http://dx.doi.org/10.1002/eji.200838439http://dx.doi.org/10.1002/eji.200838439http://dx.doi.org/10.1002/eji.200838439http://dx.doi.org/10.1038/nsb1011http://dx.doi.org/10.1038/nsb1011http://dx.doi.org/10.1073/pnas.0605666103http://dx.doi.org/10.1073/pnas.0605666103http://dx.doi.org/10.1073/pnas.0605666103http://dx.doi.org/10.1074/jbc.M103194200http://dx.doi.org/10.1074/jbc.M103194200http://dx.doi.org/10.1074/jbc.M103194200http://dx.doi.org/10.1074/jbc.M103194200http://dx.doi.org/10.1074/jbc.M103194200http://dx.doi.org/10.1083/jcb.200503017http://dx.doi.org/10.1083/jcb.200503017http://dx.doi.org/10.1083/jcb.200503017http://dx.doi.org/10.1242/jcs.018085http://dx.doi.org/10.1242/jcs.018085http://dx.doi.org/10.1146/annurev.cb.02.110186.000331http://dx.doi.org/10.1146/annurev.cb.02.110186.000331http://dx.doi.org/10.1056/NEJMoa042982http://dx.doi.org/10.1056/NEJMoa042982http://dx.doi.org/10.1056/NEJMoa042982http://dx.doi.org/10.1056/NEJMoa042982http://dx.doi.org/10.1021/ct700200bhttp://dx.doi.org/10.1021/ct700200bhttp://dx.doi.org/10.1073/pnas.0700329104http://dx.doi.org/10.1073/pnas.0700329104http://dx.doi.org/10.1016/S0092-8674(02)00971-6http://dx.doi.org/10.1016/S0092-8674(02)00971-6http://dx.doi.org/10.1126/science.1084174http://dx.doi.org/10.1126/science.1084174http://dx.doi.org/10.1034/j.1600-065X.2002.18604.xhttp://dx.doi.org/10.1034/j.1600-065X.2002.18604.xhttp://dx.doi.org/10.1101/gad.1758709http://dx.doi.org/10.1101/gad.1758709http://dx.doi.org/10.1182/blood-2004-04-1411http://dx.doi.org/10.1182/blood-2004-04-1411http://dx.doi.org/10.1182/blood-2004-04-1411http://dx.doi.org/10.1146/annurev.immunol.25.022106.141618http://dx.doi.org/10.1146/annurev.immunol.25.022106.141618http://dx.doi.org/10.1126/science.7846531http://dx.doi.org/10.1126/science.7846531http://dx.doi.org/10.1126/science.7846531http://dx.doi.org/10.1074/jbc.M111.311043http://dx.doi.org/10.1074/jbc.M111.311043http://dx.doi.org/10.1074/jbc.M111.311043http://dx.doi.org/10.1074/jbc.M111.311043http://dx.doi.org/10.1083/jcb.124.6.1047http://dx.doi.org/10.1083/jcb.124.6.1047http://dx.doi.org/10.1083/jcb.124.6.1047http://dx.doi.org/10.1002/jcc.20090http://dx.doi.org/10.1002/jcc.20090http://dx.doi.org/10.1002/jcc.20090http://dx.doi.org/10.1002/jcc.20090http://dx.doi.org/10.1073/pnas.1015487107http://dx.doi.org/10.1073/pnas.1015487107http://dx.doi.org/10.1073/pnas.1015487107http://doi:10.1074/jbc.M112.341263http://doi:10.1074/jbc.M112.341263http://doi:10.1074/jbc.M112.341263http://doi:10.1074/jbc.M112.341263http://dx.doi.org/10.1074/jbc.M110.145177http://dx.doi.org/10.1074/jbc.M110.145177http://dx.doi.org/10.1074/jbc.M110.145177http://dx.doi.org/10.1016/S0092-8674(02)00935-2http://dx.doi.org/10.1016/S0092-8674(02)00935-2http://dx.doi.org/10.1016/S0092-8674(02)00935-2

  • Journ

    alof

    Cell

    Scie

    nce

    Turner, C. E. (2000). Paxillin and focal adhesion signalling. Nat. Cell Biol. 2, E231-E236.

    Xie, C., Shimaoka, M., Xiao, T., Schwab, P., Klickstein, L. B. and Springer, T. A.(2004). The integrin alpha-subunit leg extends at a Ca2+-dependent epitope inthe thigh/genu interface upon activation. Proc. Natl. Acad. Sci. USA 101, 15422-15427.

    Xie, C., Zhu, J., Chen, X., Mi, L., Nishida, N. and Springer, T. A. (2010). Structureof an integrin with an alphaI domain, complement receptor type 4. EMBO J. 29, 666-679.

    Xiong, J. P., Stehle, T., Diefenbach, B., Zhang, R., Dunker, R., Scott, D. L.,

    Joachimiak, A., Goodman, S. L. and Arnaout, M. A. (2001). Crystal structure ofthe extracellular segment of integrin alpha Vbeta3. Science 294, 339-345.

    Xiong, J. P., Stehle, T., Goodman, S. L. and Arnaout, M. A. (2003). New insights intothe structural basis of integrin activation. Blood 102, 1155-1159.

    Xiong, J. P., Mahalingham, B., Alonso, J. L., Borrelli, L. A., Rui, X., Anand, S.,Hyman, B. T., Rysiok, T., Muller-Pompalla, D., Goodman, S. L. et al. (2009).Crystal structure of the complete integrin alphaVbeta3 ectodomain plus an alpha/betatransmembrane fragment. J. Cell Biol. 186, 589-600.

    Yan, B. and Smith, J. W. (2001). Mechanism of integrin activation by disulfide bondreduction. Biochemistry 40, 8861-8867.

    Yu, Y., Zhu, J., Mi, L. Z., Walz, T., Sun, H., Chen, J. and Springer, T. A. (2012).Structural specializations of a(4)b(7), an integrin that mediates rolling adhesion.J. Cell Biol. 196, 131-146.

    Zhang, K. and Chen, J. (2012). The regulation of integrin function by divalent cations.Cell Adh. Migr. 6, 20-29.

    Zhu, J., Luo, B. H., Xiao, T., Zhang, C., Nishida, N. and Springer, T. A. (2008).Structure of a complete integrin ectodomain in a physiologic resting state andactivation and deactivation by applied forces. Mol. Cell 32, 849-861.

    Regulation mechanism of a4b7 5041

    http://dx.doi.org/10.1038/35046659http://dx.doi.org/10.1038/35046659http://dx.doi.org/10.1073/pnas.0406680101http://dx.doi.org/10.1073/pnas.0406680101http://dx.doi.org/10.1073/pnas.0406680101http://dx.doi.org/10.1073/pnas.0406680101http://dx.doi.org/10.1038/emboj.2009.367http://dx.doi.org/10.1038/emboj.2009.367http://dx.doi.org/10.1038/emboj.2009.367http://dx.doi.org/10.1126/science.1064535http://dx.doi.org/10.1126/science.1064535http://dx.doi.org/10.1126/science.1064535http://dx.doi.org/10.1182/blood-2003-01-0334http://dx.doi.org/10.1182/blood-2003-01-0334http://dx.doi.org/10.1083/jcb.200905085http://dx.doi.org/10.1083/jcb.200905085http://dx.doi.org/10.1083/jcb.200905085http://dx.doi.org/10.1083/jcb.200905085http://dx.doi.org/10.1021/bi002902ihttp://dx.doi.org/10.1021/bi002902ihttp://dx.doi.org/10.1083/jcb.201110023http://dx.doi.org/10.1083/jcb.201110023http://dx.doi.org/10.1083/jcb.201110023http://dx.doi.org/10.4161/cam.18702http://dx.doi.org/10.4161/cam.18702http://dx.doi.org/10.1016/j.molcel.2008.11.018http://dx.doi.org/10.1016/j.molcel.2008.11.018http://dx.doi.org/10.1016/j.molcel.2008.11.018

    Fig 1Fig 2Fig 3Fig 4Fig 5Fig 6Fig 7Fig 8Ref 1Ref 2Ref 3Ref 4Ref 5Ref 6Ref 7Ref 8Ref 9Ref 10Ref 11Ref 12Ref 13Ref 14Ref 15Ref 16Ref 17Ref 18Ref 19Ref 20Ref 21Ref 22Ref 23Ref 24Ref 25Ref 26Ref 27Ref 28Ref 29Ref 30Ref 31Ref 32Ref 33Ref 34Ref 35Ref 36Ref 37Ref 38Ref 39Ref 40Ref 41Ref 42Ref 43Ref 44Ref 45Ref 46Ref 47

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages true /GrayImageMinResolution 150 /GrayImageMinResolutionPolicy /OK /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 200 /GrayImageDepth 8 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /FlateEncode /AutoFilterGrayImages false /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages true /MonoImageMinResolution 1200 /MonoImageMinResolutionPolicy /OK /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 600 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly true /PDFXNoTrimBoxError false /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox false /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (Euroscale Coated v2) /PDFXOutputConditionIdentifier (FOGRA1) /PDFXOutputCondition () /PDFXRegistryName (http://www.color.org) /PDFXTrapped /False

    /CreateJDFFile false /SyntheticBoldness 1.000000 /Description >>> setdistillerparams> setpagedevice