当前位置: 首页 > 期刊 > 《动脉硬化血栓血管生物学》 > 2004年第5期 > 正文
编号:11330687
Signaling in Leukocyte Transendothelial Migration
http://www.100md.com 《动脉硬化血栓血管生物学》
     From Sanquin Research at CLB and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands.

    ABSTRACT

    Under a variety of (patho) physiological conditions, leukocytes will leave the bloodstream by crossing the endothelial monolayer that lines the vessels and migrate into the underlying tissues. It is now clear that the process of extravasation involves a range of adhesion molecules on both leukocytes and endothelial cells, as well as extensive intracellular signaling that drives adhesion and chemotaxis on the one hand and controls a transient modulation of endothelial integrity on the other. We review here the current knowledge of the intracellular signaling pathways that are activated in the context of transendothelial migration in leukocytes and in endothelial cells. In leukocytes, polarization of receptors and of the signaling machinery is of key importance to drive adhesion and directional migration. Subsequent adhesion-induced signaling in endothelial cells, mediated by Rho-like GTPases and reactive oxygen species, induces a transient and focal loss of endothelial cell–cell adhesion to allow transmigration of the leukocyte. This review underscores the notion that we have likely just scratched the surface in revealing the complexity of the signaling that controls leukocyte transendothelial migration.

    Key Words: endothelial cells ? migration ? Rho GTPases ? adhesion ? reactive oxygen species

    Introduction

    The migratory properties of white blood cells are indispensable to drive immune responses throughout the body. To ensure migration to the proper locations, the trafficking of leukocytes is tightly regulated. The migration and extravasation of leukocytes across the endothelium that lines the vessel wall occurs in several distinct steps, referred to as the multi-step paradigm, originally introduced by Butcher1 and extended by Springer.2 The first step comprises the rolling of the leukocytes over the endothelial cells, mediated by transient weak interactions between adhesion molecules. Subsequently, loosely attached leukocytes are in such close proximity of the endothelium that they can be activated by chemotactic cytokines, presented on the apical surface of the endothelium. As a consequence, the activated leukocytes will spread and firmly adhere to the endothelium and finally migrate through the intercellular clefts between the endothelial cells to the underlying tissue. An additional level of complexity has been added by work that showed that in addition to chemokines, shear stress can also act as a transmigration-promoting stimulus.3 Figure 1 shows a schematic overview of the different stages that occur during transendothelial migration (TEM) of leukocytes.

    Figure 1. Schematic overview of the distinct steps that comprise leukocyte transendothelial migration. The first step (I) represents the rolling of leukocytes and involves adhesion molecules such as selectins on the leukocytes or endothelial cells and their ligands on the cellular counterparts. The second step (II) represents firm adhesion, mediated through immobilized chemoattractants on the endothelium and adhesion molecules such as integrins on the leukocytes and CAMs on the endothelium. The third step (III) represents the diapedesis of the leukocytes through the endothelial cell-cell junctions, in which homophilic binding molecules such as the JAMs, PECAM-1, CD99, and VE-cadherin are involved.

    The extravasation of leukocytes is essential for many (patho) physiological processes, including migration of T-lymphocytes for immune surveillance, recruitment of activated lymphocytes and granulocytes during acute and chronic inflammatory responses, and homing and mobilization of hematopoietic progenitor cells. In the past decade, knowledge of the molecules and the signaling events, both in the leukocytes and in the endothelial cells, that control transendothelial migration of leukocytes has increased significantly and is discussed here. We have divided this overview into 3 parts: the first part deals with chemokine-induced leukocyte activation; the second part describes the signaling events that occur in endothelial cells after leukocyte adhesion; and the final part discusses our current knowledge about the role of the endothelial cell-cell junctions in the final stage of the transmigration process.

    Chemokine-Induced Leukocyte Activation

    The migration of leukocytes across the endothelium is mainly driven by a large family of extracellular ligands: the chemotactic cytokines, better known as chemokines. Chemokines are small (8 to 14 kDa) structurally related molecules that interact with 7-transmembrane-spanning G-protein-coupled receptors and regulate leukocyte trafficking.4 Today, >40 different chemokines have been identified, and this number is still growing. The nomenclature of chemokines is based on the arrangement of the N-terminal cysteine residues. These residues can be adjacent (CC chemokines) or have one or more extra amino acids in between them (CXC or CX3C chemokines). Two chemokines have only one cysteine at the N-terminus, ie, lymphotactin/SCM-1 and SCM-1?. In the current nomenclature, an "L" (as in CXCL12) refers to a ligand, whereas an "R" (as in CXCR4) refers to a chemokine receptor.5 The main topics of this review are discussed with reference to stromal cell-derived factor-1 (SDF-1 or, according to the current nomenclature, CXCL12), because SDF-1/CXCL12 is involved in the migration of many, if not all, different types of leukocytes and various types of tumor cells6,7 and is expressed in virtually all human tissues.8,9

    Chemokines and their receptors are one of the many levels that coordinate the migration of leukocytes and leukocyte subsets at various levels, resulting in a tightly controlled and very complex system of cell trafficking.8,10,11 Various studies have shown that leukocyte transendothelial migration is induced by soluble chemokine gradients in vitro.8,12 However, it is unlikely that soluble chemokine gradients persist in the bloodstream, because the gradient will be rapidly washed away.3,13 It is therefore generally accepted that leukocytes respond to chemokines that are immobilized on the surface of the endothelium, which is underscored by detailed studies showing that chemokines, such as IL-8/CXCL8 and SDF-1/CXCL12, are indeed present on the vascular endothelium in vivo.13–16 These immobilized chemokines are presented to nearby rolling leukocytes by heparin-sulfate proteoglycans, highly glycosylated proteins that are expressed on the surface of endothelial cells.17–19 Middleton et al have shown that IL-8, a potent chemokine for neutrophils, is presented on the microvillous-like extensions on the luminal surface of the endothelium. Moreover, these investigators showed that after injection of IL-8 into the skin of a rabbit, IL-8 is transported through the endothelial cells from the basolateral side to the luminal side.13 For SDF-1/CXCL12, it has been shown extensively that immobilization of this chemokine leads to increased adhesion and TEM of leukocytes under physiological flow.3,16,20,21

    Chemokine Receptors

    Chemokines transmit their pro-migratory signals through G-protein-coupled, 7-times-spanning membrane receptors. These receptors initiate adhesion and motility via (pertussis toxin-sensitive) G-proteins, leading to integrin activation via inside-out signaling, followed by coordinated actin polymerization, spreading at the leading edge of the cell and contraction at the back.22,23 Directional migration of leukocytes is accompanied by polarization of the cell body, of the actin and tubulin cytoskeleton, and of a wide range of intracellular signaling proteins, such as PI-3K, PTEN, Rho-like GTPases, and G?-subunits. Their differential distribution mediates the amplification of the chemokine gradient on the outside into a (steep) signaling gradient inside the cell.24,25 Concomitant polarization of the chemokine receptors at the cell surface is subject to conflicting reports. Studies with C5a-receptor-GFP fusion proteins in neutrophils or cAMP-receptor-GFP fusion proteins in Dictyostelium cells showed uniform distribution of these receptors over the plasma membrane in cells that migrate toward chemoattractants.26 Other (chemokine) receptors, such as CCR2 and CCR5 on T-lymphocytes and the fMLP receptor on neutrophils, are distributed to the leading edge on exposure to their cognate ligands.27,28 In B-lymphocytes, SDF-1 induces polarization of CXCR4 to the leading edge of the cell.29 In addition, it has been reported that SDF-1/CXCL12 can bind to fibronectin and subsequently induces a polarized distribution of CXCR4 on adherent T-lymphocytes.30 We recently showed that immobilized SDF-1 on TNF-activated endothelium rapidly induces polarization of CXCR4 to the leading edge of the migrating cell and that CXCR4 subsequently co-localizes with cholesterol lipid rafts (Figure 2A).20 Disruption of lipid rafts by cyclodextrin inhibited SDF-1/CXCL12-induced CXCR4 internalization and cell migration, indicating that internalization and proper SDF-1-mediated signaling requires the presence of lipid rafts.20,31

    Figure 2. A, Schematic overview of the signaling pathways at the leading edge of a migrating cell. A leukocyte adheres through L-selectin to cellular adhesion molecules (CAM) or glycoproteins such as CD34 on the endothelial cells. L-selectin induces CXCR4 upregulation from intracellular granules on the leukocyte surface, resulting in increased activation of Rap1. Rap1 induces 4-integrin activation, possibly through phosphorylation. These actions on the leukocyte surface appear to take place in a lipid raft. B, Schematic overview of the signaling pathways at the trailing edge of a migrating cell. An adhered leukocyte moves forward by leading-edge protrusion and retraction of its tail. Therefore, L?2-integrin activates through Vav1, RhoA, which subsequently activates the kinase p160ROCK. ROCK activation leads, through MLCK activation, to MLC phosphorylation, resulting in retraction of the actin cytoskeleton. The actin cytoskeleton is linked via the ERM proteins to the adhesion molecules ICAM-3, PSGL-1, CD43, and CD44, which are expressed at the rear end of the cell and might serve as an anchor for other migrating cells.

    Adhesion Molecules

    In the first phase of extravasation, leukocytes roll over the endothelium, a process that is mediated by a family of adhesion molecules called selectins.32,33 Recently, it became clear that stimulation of L-selectin by antibody-specific cross-linking induced CXCR4 expression in lymphocytes.34 Moreover, these authors showed that CXCR4 is stored in intracellular vesicles and found that L-selectin, just as CXCR4, localizes to lipid rafts.35 These findings suggest that selectin-mediated rolling of leukocytes over the endothelium already primes the leukocytes to target CXCR4 to lipid rafts. In addition, Shamri et al showed that the integrin VLA-4 (4?1) requires cholesterol rafts for adhesive activity but that leukocyte function-associated antigen-1 (LFA-1; L?2) acts independent from lipid rafts to mediate adhesion.21 These data suggest the presence of independent clusters of adhesion molecules on the plasma membrane at the leading edge of migrating leukocytes, before chemokine stimulation (Figure 2A).

    Integrins play a major role in TEM of leukocytes and are, in fact, indispensable for proper extravasation.36 The main integrins that regulate SDF-1-induced adhesion and spreading of leukocytes are LFA-1 (L?2), Mac-1 (M?2), VLA-4 (4?1), and VLA-5 (5?1).37,38 LFA-1, Mac-1, and VLA-4 bind to the adhesion molecules intercellular cell adhesion molecule-1 (ICAM-1) and ICAM-2 (for LFA-1 and Mac-1), and vascular cell adhesion molecule-1 (VCAM-1) (for VLA-4), present on the apical surface of the endothelium.39,40 For the integrins VLA-4 and VLA-5, it has been shown that they bind to fibronectin.41 Studies in mice that lack ?2-integrins or studies that used blocking antibodies against ?2-integrins showed that neutrophil traffic was dependent on this integrin.42,43 Similarly, mice that lacked the 4-integrin showed impaired T-lymphocyte homing to Peyer patches, although not to other secondary lymphoid organs.44 In vivo studies with blocking antibodies to the 4-integrin VLA-4 also showed that this integrin was involved in migration of CD34+ cells.37 These data point to a prominent role for these integrins in the control of trafficking of leukocytes in general.45

    Binding of the integrins to their ligands induces multiple signaling cascades in the leukocytes.46,47 This outside-in signaling is translated by the leukocyte into cell polarization and forward movement. This movement is generally assumed to be driven by the combination of leading edge protrusion and contractility at both the front and the back of the cell, which allows displacement of the cell body.48–51 Some of these studies have used Dictyostelium discoidea amoebae as a model, because the ability to sense and to respond to extracellular signals is remarkably similar to that of mammalian leukocytes.49

    A large number of structural and regulatory proteins are located at the leading edge of the migrating cell, including proteins that regulate the dynamics of the actin cytoskeleton, such as the small GTPase Rac1, its effector PAK1, and the Arp2/3 complex.52,53 Also, focal adhesion and focal contact proteins such as adapter proteins, tyrosine kinases, and integrins are found at the leading edge.52,53 These studies mainly focused on fibroblasts; therefore, one should take in account that different cell types might use different signaling pathways to achieve the same goals.

    At the back of the migrating cell, proteins such as ICAM-3, PSGL-1, CD43, and CD44 are localized.54,55 ERM proteins act as adapter proteins and link these transmembrane proteins to the actin cytoskeleton.56,57 To move forward, cells have to form new adhesion sites and break-down old ones, both in the protruding and in the retracting part of the cell. The turnover of focal adhesions is very complex and involves integrins58 and microtubules.59 Because of space limitations, we cannot discuss the complex mechanisms that drive of focal adhesion turnover.60 Recently, it has been shown that the 4-integrin is phosphorylated on serine residues at the leading edge of polarized cells and is required for lamellipodia stability, although 4 is also present at the trailing edge.61 In addition, these authors showed that paxillin, a protein present in focal adhesions,62 co-localizes with 4 at the trailing edge but not at the leading edge with the phosphorylated 4. Moreover, enforced association of paxillin with 4 inhibits migration, indicating that the 4-paxillin interaction mediates attachment and detachment of membrane protrusions, which is required for migrating cells to move forward.

    Next to the 4-integrin, LFA-1 (L?2) signaling is required for proper leukocyte migration. Whereas the 4-integrin uses paxillin to regulate the attachment and detachment of the cells’ leading edge, LFA-1 seems to signal through myosin light chain kinase and through p160ROCK, which regulates cell contraction and detachment from the matrix (Figure 2B).63 Moreover, LFA-1 engagement induces actin polymerization at the leading edge of the cell,64 possibly through the Rho family exchange factor Vav1.65 Thus, both 4 integrins and LFA-1 are required for leukocyte migration, acting through distinct signaling cascades to drive efficient chemotaxis.

    Small GTPases

    As is well known from studies in fibroblasts as well as in leukocytes, Rho-like small GTPases (in particular Rho, Rac, and Cdc42) are crucial for coordinated cell adhesion, spreading, and migration.66 Rac1, being a critical regulator of cell spreading, actin polymerization, and membrane ruffling in any type of cell, is found throughout the cytoplasm under resting conditions. However, in its active, GTP-bound form, Rac1 localizes to growth factor-induced membrane ruffles and to the leading edge of migrating cells.67 These authors showed additionally that ?1-integrins induce the interaction of active Rac1 with its downstream effectors, such as PAK1.67,68 Using C-terminal cell-permeable peptides as selective inhibitors, we recently showed that Rac1 (and not Rac2 or CDC42) plays an important role in SDF-1-induced actin polymerization in HL-60 cells.69

    Next to the involvement of Rac1 in driving membrane protrusion, the small GTPase Rap1 was shown to be involved in the control of the adhesive capacities of LFA-1 and VLA-4.70,71 Moreover, Rap1 can be activated by SDF-1, and in its active form induces cell polarization. In addition, active Rap1 promotes transendothelial migration of T-lymphocytes.72 Recently, RAPL, a Rap1-binding protein, was shown to associate with Rap1 after SDF-1 stimulation. RAPL induces polarization of T-lymphocytes and distribution of LFA-1 to the leading edge of the cell.73 These data suggest that Rap1 can signal between chemokine-activated G-protein-coupled receptors, such as CXCR4, and integrin activation (Figure 2A).

    As described, lamellipodia and membrane protrusions at the front of the cell are required to move the cell forward. Consequently, the rear of the cell has to contract and to detach from the underlying matrix, ie, from the endothelial monolayer. RhoA and one of its effectors, the serine/threonine kinase p160ROCK, have been found to mediate tail retraction of migrating leukocytes.74,75 Inhibition of RhoA activity by C3 transferase in monocytes prevents migration through an endothelial monolayer, although overall attachment of monocytes to activated endothelial monolayers was not affected. Interestingly, the adhesion to either ICAM-1 or VCAM-1 was largely promoted on inhibition of p160ROCK.75 In addition, the ?2-integrin mislocalized in these cells from the leading edge to the trailing edge, indicating that RhoA and p160ROCK signaling mediates integrin localization at the leading edge. The results suggest a model in which RhoA activity is inhibited at the front of the cell to allow the induction of protrusions, in concert with enhanced Rac1 activity at the leading edge. Conversely, RhoA is activated at the rear of the cell to retract and detach the uropod and Rac1 may become inhibited at sites of tail retraction. The events that occur in chemokine-mediated leukocytes and are involved in migration are summarized in Figure 2A and 2B.

    Consequences of Leukocyte-Endothelium Interactions

    Huang et al were the first to show that intracellular calcium levels in endothelial cells were increased on adhesion and transmigration of neutrophils.4 Moreover, they showed that this was also required for efficient TEM. In addition, Hixenbaugh et al showed that chemoattractant-stimulated neutrophils induce an increase in the phosphorylation of myosin light chain (MLC) in endothelial cells.76 Increased MLC phosphorylation accompanies RhoA activation, subsequent cell contraction, and a loss of endothelial cell-cell junctions, which then leads to increased permeability of the endothelial barrier.77,78 Using an inhibitor to MLC kinase (MLCK) on endothelial cells, Saito et al showed that transmigration of neutrophils was blocked and MLC phosphorylation was inhibited.79 Adhesion molecules such as P-selectin and E-selectin and VCAM-1 on the endothelium might be responsible for transmitting the signals, induced by adhered neutrophils into the endothelium, because activation of these adhesion molecules through antibody-specific cross-linking results in a transient increase of intracellular calcium.80 These studies put forward the notion that leukocyte adhesion affects the endothelial cells in a specific fashion and that endothelial cell function contributes importantly to TEM. Today, the involvement of endothelial signaling in transmigration of leukocytes is an accepted phenomenon, and various signaling events in endothelial cells have been implicated in the process of leukocyte TEM, although the exact mechanisms by which the endothelial cells facilitate the passage of leukocytes is still not entirely clear.81,82

    As noted, leukocytes use mainly ?1 (VLA-4/4?1) and ?2 (LFA-1/L?2) integrins to firmly adhere to the endothelium. The ligands for these integrins on the endothelium are VCAM-1 and ICAM-1, respectively. More recently, a novel family of integrin ligands, called junctional adhesion molecules (JAM), has been discovered.83 The role of JAMs in transendothelial migration will be discussed. Signaling through ICAM-1, induced by antibody-mediated cross-linking, activates RhoA.84 Moreover, other proteins that regulate the actin cytoskeleton, such as FAK, Cas, and cortactin, are phosphorylated on tyrosine on antibody-induced cross-linking of ICAM-1.85 Thus, engagement of ICAM-1 induces signaling in endothelial cells. However, one should take into account that receptor cross-linking is a rather global stimulus compared with leukocyte binding to the endothelium. The short (28 amino acids) intracellular tail of ICAM-1 interacts with ezrin, an adapter protein that links ICAM-1 with the actin cytoskeleton.86 Ezrin is a member of the ERM protein family and plays an important role in lamellipodia induction at the leading edge of fibroblasts.87,88 Moreover, it appears that ezrin links the proteoglycan syndecan-2, which might possibly bind SDF-1, to the actin cytoskeleton.89 Detailed analysis of monocytes adherent to activated endothelium or cross-linking of the adhesion receptors ICAM-1 and VCAM-1 showed clustering of the ERM proteins near those sites of activation.90 Next to its function as an adapter protein, there is evidence that shows ezrin to act as a signaling molecule, because active ezrin is reported to be responsible for the formation of lamellipodia.91 In addition, these authors showed that Rac1, but not RhoA or Cdc42, is activated in cells that express the active form of ezrin. Other previous work suggested that the ERM proteins are rapidly phosphorylated in a Rho-dependent manner.92 Barreiro et al recently showed that the endothelial cells form a so-called docking structure to "catch" the migrating leukocyte, and they subsequently showed that moesin and ezrin localized to those docking structures, together with ICAM-1 and VCAM-1.93

    We recently showed that clustering of VCAM-1 activates Rac1.94 Inhibition of downstream signaling by Rac1 with an inhibitory peptide, encoding the effector loop of Rac1 (aa17-32;95), reduced the migration of leukocytes across endothelial monolayers.94 Because transduction of an active mutant of Rac1, RacV12, reduces endothelial cell-cell contacts,96–98 blocking Rac activity might prevent opening of endothelial cell-cell contacts, thereby preventing leukocytes from migrating. Moreover, ligation of VCAM-1 activates phosphatidylinositol 3-kinase (PI3-K), implicated as an activator of Rac1.99,100

    Studies performed by our group already showed that inhibition of endothelial RhoA by C3 transferase decreased monocyte migration across human umbilical vein endothelial cells, although the C3 pretreatment had little or no effect on the migration of primary CD34+ cells across bone marrow endothelial cells.12,94 Also, inhibition of p160ROCK signaling in endothelial cells by Y-27632 decreases not only migration of lymphocytes across endothelium but also adhesion.93 The latter results indicate that RhoA activity is linked to adhesion molecules on the endothelium and that blockade of RhoA activity or downstream effectors prevents leukocyte adhesion as well as endothelial cell contraction and subsequent loss of cell-cell contacts. Thus, blocking signaling through Rac1 or RhoA in endothelial cells inhibits transendothelial migration of monocytes. It is therefore likely that both RhoA and Rac1 in endothelial cells are involved in leukocyte-induced endothelial signaling: RhoA through ligation of ICAM-1 and Rac1 through the ligation of VCAM-1.

    For many years, it has been known that reactive oxygen species (ROS) play an important role in the maintenance of the endothelial monolayer integrity.101,102 The generated ROS are spontaneously or enzymatically converted to hydrogen peroxide (H2O2), which may disrupt the integrity of the endothelial monolayer, possibly through inhibition of tyrosine phosphatases.103,104 Some studies already reported the presence of components of the neutrophil-NADPH-oxidase (gp91) complex in endothelial cells.105,106 Other studies suggested the expression of various NADPH-oxidase homologues in endothelial cells.107 Upregulation of Nox-based NADPH-oxidases after vascular damage have been reported.108,109 Nevertheless, how the activity or intracellular localization of these protein complexes is regulated remains unclear and warrants further study. Our group has previously shown that active Rac1 induces ROS production in endothelial cells and that endothelial ROS are actively involved in TEM, because leukocyte migration across endothelium is inhibited when the endothelial monolayer is pretreated with oxygen-radical scavengers.97 Similar findings have been reported by Wang and Doerschuk, who showed that engagement of ICAM-1 induces ROS production in endothelial cells.110 In line with these findings, we and others have found that activation of VCAM-1 and PECAM-1, by antibody-mediated cross-linking, results in increased levels of ROS production.12,111,112 These findings suggest that leukocyte adhesion to endothelium induces ROS production, which in turn might transmit signals into the endothelium to facilitate leukocyte passage.

    Activation of ICAM-1, by cross-linking, remodels the actin cytoskeleton through activation of p38 mitogen-activated protein kinase.113 Also, VCAM-1 is able to activate p38 mitogen-activated protein kinase in a ROS-dependent manner.94 Extracellular signal-regulated kinase-2, which is also involved in motility and in proliferation, is also activated by VCAM-1.99 Lorenzon et al showed previously that VCAM-1 can act as a signaling receptor.80 In their studies, VCAM-1 cross-linking was found to induce a transient increase in endothelial calcium concentration, which is required for efficient TEM.

    These data suggest that engagement of ICAM-1 and VCAM-1, through the integrin-mediated adhesion of leukocytes, induces intracellular signaling in the endothelial cells that finally results in the opening of the endothelial cell-to-cell junctions. This hypothesis is supported by studies that showed that active Rac1 induces loss of cell-to-cell adhesion in endothelial cells.96–98 This contrasts with epithelial cells, in which active Rac1 promotes cadherin-based cell-to-cell adhesion.114 Although a lot is known about epithelial regulation of cell-to-cell junctions, relatively little is known about the control of endothelial cell-to-cell junctions with respect to the opening and resealing of these junctions during the passage of leukocytes. The last part of this review discusses our current knowledge about this topic.

    Endothelial Cell-to-Cell Junctions in Leukocyte Transendothelial Migration

    Leukocytes need to migrate through the endothelial cell-cell junctions to cross the endothelial monolayer in the final stage of extravasation.2 To successfully cross the junctions, the leukocytes have to bypass or block the adhesion, which is mediated by junctional proteins. The endothelial cell-to-cell junctions contain a large number of (adhesion) proteins, including vascular endothelial (VE)-cadherin (CD144, cadherin-5), a homophilic calcium-dependent transmembrane adhesion molecule, which is localized at the adherens junctions. In addition, the endothelial junctions comprise tight junction proteins, eg, occludin, JAM family members, and claudins, as well as proteins such as CD99 and PECAM-1 (CD31), which are more diffusely distributed at the cell-cell junctions.

    Many studies have described PECAM-1 as an important player in the process of transendothelial migration based on the effects of blocking PECAM-1 in vitro115–118 and in vivo.119–127 However, PECAM-1 knockout mice do not show a significant loss of inflammatory response, demonstrating that PECAM-1 is not indispensable for transmigration.128 The mechanisms that compensate for the loss of PECAM-1 are not clear; however, Mamdouh et al recently found that in the absence of leukocytes, PECAM-1 is present in a compartment directly under the junctional border membrane.129 These investigators suggest a new mechanism in which passing leukocytes trigger these compartments to fuse with the junctional border membrane. Because PECAM-1 is present on the leukocyte as well as on the endothelium, this mechanism might facilitate leukocytes to cross the endothelial cell-to-cell junction through homophilic PECAM-1 binding.129 This mechanism also implies that leukocytes could be engulfed by endothelial cells, for instance, through the aforementioned docking structure.93 Intriguingly, it has been described that neutrophils are also able to cross the endothelial layer by a transcellular route, particularly in vivo.130 How this process is controlled is unknown.

    PECAM-1 is phosphorylated on tyrosine residues after endothelial cell stimulation by mechanical force and shear stress by means of the Fer tyrosine kinase, which is localized on microtubules.131,132 Moreover, Ilan et al showed that PECAM-1 can function as a reservoir for tyrosine-phosphorylated ?-catenin.133 These data suggest that PECAM-1 binding to ?-catenin prevents cytosolic ?-catenin from degradation by the proteosomes. Subsequently, PECAM-1 engagement induces SHP-2 phosphorylation through Fer, which then dephosphorylates PECAM-1-bound ?-catenin. It has been shown that increased tyrosine phosphorylation of junctional proteins leads to loss of cell-to-cell adhesion and that subconfluent endothelial monolayers have increased levels of tyrosine phosphorylation of the junctional proteins VE-cadherin, ?-catenin, and -catenin.134,135 Moreover, SHP-2 and ?-catenin are both present in the VE-cadherin complex at cell-to-cell junctions.136 In addition, inhibition of protein tyrosine phosphatase activity promotes the flux of albumin across an endothelial monolayer and the extravasation of neutrophils, indicating that phosphorylation plays an essential role in the control of endothelial integrity and of the process of TEM.137 Phosphatase activity can be inhibited by H2O2, because H2O2 mediates the oxidation of critical cysteine residues in tyrosine phosphatases.138 As a result, tyrosine phosphorylation levels are increased, triggering loss of endothelial cell-to-cell junctions. This suggestion links VCAM-1-induced ROS production and phosphatase/kinase activity to the transendothelial migration of leukocytes.

    Another family of junctional proteins that has attracted a lot of attention are the JAMs. Recently, new nomenclature for the JAM family has been put forward by Muller and is used in this review.139 JAM-A has been cloned and identified recently and appears to mediate homophilic and heterophilic interactions.140 JAM localizes at the apical regions of cell-to-cell junctions, not only of endothelial cells but also of epithelial cells, and regulates the electrical resistance across monolayers of these cells.141,142 Antibodies to JAM cause decreased transendothelial migration of leukocytes in vitro and in vivo, strongly suggesting that this molecule might be involved in TEM.140,143 Ostermann et al have shown that JAM-A on endothelial cells is a ligand for the ?2-integrin LFA-1, and as such is involved in transendothelial migration of leukocytes.144 The JAM protein family now comprises 3 members: JAM-A (approximately 32 kDa), present on endothelial cells, epithelial cells, and on all leukocytes, functions as a regulator of TEM and of electrical resistance. JAM-B (or VE-JAM), which is 40 kDa in size, is expressed on vascular and lymphatic endothelium and has been proposed to play a role in lymphocyte homing.145,146 JAM-B also interacts with the ?1-integrin receptor VLA-4.147 Most recently, a third family member has been identified as JAM-C ( 43 kDa), present on endothelial cells, T-lymphocytes, platelets, and NK-cells.147,148 JAM-C also functions as a regulator of TEM of leukocytes and electrical resistance. It interacts with JAM-B and Mac-1 (M?2-integrin, present on leukocytes) but not with LFA-1 or VLA-4.149,150 These findings indicate that all 3 JAM family members could potentially be involved in leukocyte transendothelial migration but that they are independently involved in specific parts of the TEM process.83

    The integral transmembrane protein CD99 regulates VLA-4-dependent adhesion of T-lymphocytes to endothelial cells under physiological shear stress.151 In contrast to VLA-4, ?2-integrins (ie, LFA-1) were not influenced by CD99 stimulation. Recently, CD99 was found to be present on endothelial cells as a homophilic binding molecule at cell-to-cell junctions.152 Antibodies to CD99 were found to block transmigration of monocytes, particularly during diapedesis. Blocking antibodies to PECAM-1 inhibit monocyte migration before diapedesis started, indicative for a sequential engagement of these proteins during transmigration across the junction.152 Thus, whereas JAM and PECAM-1 are involved in the first stages of transmigration across endothelium, CD99 seems to regulate further passage through the endothelial junctions. A note should be made that, as mentioned, PECAM-1 is required for migration through the basement membrane but is apparently also involved in early steps of diapedesis.

    Previous reports have shown that adhesion of neutrophils to endothelium disrupts the VE-cadherin complex at cell-cell junctions.153,154 However, it was subsequently shown that preparation of cell lysates under relatively mild conditions allows neutrophil proteases to cause breakdown of the VE-cadherin-catenin complex.155 However, the latter study by Moll et al did not indicate whether proteases are involved in the physiological process; it only clarified that such experiments could not demonstrate regulated proteolysis of VE-cadherin on neutrophil attachment. Nevertheless, it has been observed with GFP fusion proteins and live-cell imaging that VE-cadherin and the associated catenins are indeed dislocalized from cell-cell junctions on passage of monocytes or CD34+ cells through the endothelium.12,156,157 Recent evidence showed that elastase, a protease present in neutrophil granules and released on activation, is able to degrade the extracellular part of VE-cadherin, keeping the hypothesis alive that neutrophils disrupt endothelial cell-cell junctions by releasing elastase and possibly other proteases to migrate through the endothelial cleft.158 However, studies with neutrophils from elastase-deficient or MMP-9-deficient mice have shown that these proteases are not required for rolling over, adhesion to, or migration across endothelium, although these neutrophils were still able to disrupt VE-cadherin function.159 Together, these studies indicate that proteases might be involved in transendothelial migration, but that the role for elastase and MMP-9 seems to be limited. Figure 3 summarizes schematically the endothelial signaling that accompanies and drives TEM.

    Figure 3. Schematic overview of the signaling pathways that occur during TEM in endothelial cells. Adherent leukocytes activate endothelial cells through binding to adhesion molecules such as ICAM-1 and VCAM-1. The small GTPase Rac1 induces ruffles, possibly leading to docking structures and ROS production. ROS inhibit phosphatase activity, which results in increased kinase activity and loss of VE-cadherin-mediated cell-cell junctions. Subsequently, the small GTPase RhoA induces activation of MLC and cell contraction. Together with junctional molecules such as PECAM-1, JAM, and CD99, these actions are likely to facilitate transmigration of leukocytes across an endothelial monolayer.

    Conclusions

    Little more than one decade since the seminal studies that started this field of research, it has become increasingly clear that the emigration of leukocytes from the bloodstream involves complex signaling in the migrating leukocyte and in the endothelium. This signaling needs to be carefully controlled, not only to ensure proper and efficient extravasation but also to limit leukocyte transendothelial migration to those sites where the cells are required. Mislocalized or excessive leukocyte exit damages the endothelium and underlying tissues. Therefore, leukocyte migration and adhesion represent important areas for drug targeting. This is even more underscored by the realization that tumor cell metastasis resembles leukocyte TEM and also that some tumor cells even rely on specific chemokines, such as SDF-1, for their spreading through the body. The events that take place in the endothelium and accompany or even allow leukocyte diapedesis have everything to do with control of endothelial integrity. Given the essential role of the endothelium and its barrier function for normal physiology, detailed knowledge of the intracellular signaling and adhesion molecules involved is of key importance. The unique interactions between activated leukocytes and endothelial cells will likely, despite their complexity, help us to understand the interplay between cell-surface molecules, intracellular signaling cascades, and the control of motility and cell-cell adhesion.

    References

    Butcher EC. Leukocyte-endothelial cell recognition: three (or more) steps to specificity and diversity. Cell. 1991; 67: 1033–1036.

    Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell. 1994; 76: 301–314.

    Cinamon G, Shinder V, Alon R. Shear forces promote lymphocyte migration across vascular endothelium bearing apical chemokines. Nat Immunol. 2001; 2: 515–522.

    Huang AJ, Manning JE, Bandak TM, Ratau MC, Hanser KR, Silverstein SC. Endothelial cell cytosolic free calcium regulates neutrophil migration across monolayers of endothelial cells. J Cell Biol. 1993; 120: 1371–1380.

    Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity. 2000; 12: 121–127.

    Zeelenberg IS, Ruuls-Van Stalle L, Roos E. Retention of CXCR4 in the endoplasmic reticulum blocks dissemination of a T cell hybridoma. J Clin Invest. 2001; 108: 269–277.

    Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001; 410: 50–56.

    Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997Jan 6. 185: 111–120.

    Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1). J Exp Med. 1996; 184: 1101–1109.

    Kitayama J, Carr MW, Roth SJ, Buccola J, Springer TA. Contrasting responses to multiple chemotactic stimuli in transendothelial migration: heterologous desensitization in neutrophils and augmentation of migration in eosinophils. J Immunol. 1997; 158: 2340–2349.

    Siveke JT, Hamann A. T helper 1 and T helper 2 cells respond differentially to chemokines. J Immunol. 1998; 160: 550–554.

    van Buul JD, Voermans C, van den Berg, V, Anthony EC, Mul FP, van Wetering S, van der Schoot CE, Hordijk PL. Migration of human hematopoietic progenitor cells across bone marrow endothelium is regulated by vascular endothelial cadherin. J Immunol. 2002; 168: 588–596.

    Middleton J, Neil S, Wintle J, Clark-Lewis I, Moore H, Lam C, Auer M, Hub E, Rot A. Transcytosis and surface presentation of IL-8 by venular endothelial cells. Cell. 1997; 91: 385–395.

    Marshall LJ, Ramdin LS, Brooks T, DPhil PC, Shute JK. Plasminogen activator inhibitor-1 supports IL-8-mediated neutrophil transendothelial migration by inhibition of the constitutive shedding of endothelial IL-8/heparan sulfate/syndecan-1 complexes. J Immunol. 2003; 171: 2057–2065.

    Pablos JL, Santiago B, Galindo M, Torres C, Brehmer MT, Blanco FJ, Garcia-Lazaro FJ. Synoviocyte-derived CXCL12 is displayed on endothelium and induces angiogenesis in rheumatoid arthritis. J Immunol. 2003; 170: 2147–2152.

    Peled A, Grabovsky V, Habler L, Sandbank J, Arenzana-Seisdedos E, Petit I, Ben-Hur H, Lapidot T, Alon R. The chemokine SDF-1 stimulates integrin-mediated arrest of CD34(+) cells on vascular endothelium under shear flow. J Clin Invest. 1999; 104: 1199–1211.

    Kainulainen V, Nelimarkka L, Jarvelainen H, Laato M, Jalkanen M, Elenius K. Suppression of syndecan-1 expression in endothelial cells by tumor necrosis factor-alpha. J Biol Chem. 1996; 271: 18759–18766.

    Lopez-Casillas F, Cheifetz S, Doody J, Andres JL, Lane WS, Massague J. Structure and expression of the membrane proteoglycan beta glycan, a component of the TGF-beta receptor system. Cell. 1991; 67: 785–795.

    Netelenbos T, Drager AM, van het Hof B, Kessler FL, Delouis C, Huijgens PC, van den Born J, van Dijk W. Differences in sulfation patterns of heparan sulfate derived from human bone marrow and umbilical vein endothelial cells. Exp Hematol. 2001; 29: 884–893.

    van Buul JD, Voermans C, van Gelderen J, Anthony EC, van der Schoot CE, Hordijk PL. Leukocyte-endothelium interaction promotes SDF-1-dependent polarization of CXCR4. J Biol Chem. 2003; 278: 30302–30310.

    Shamri R, Grabovsky V, Feigelson SW, Dwir O, van Kooyk Y, Alon R. Chemokine stimulation of lymphocyte alpha 4 integrin avidity but not of leukocyte function-associated antigen-1 avidity to endothelial ligands under shear flow requires cholesterol membrane rafts. J Biol Chem. 2002; 277: 40027–40035.

    Samstag Y, Eibert SM, Klemke M, Wabnitz GH. Actin cytoskeletal dynamics in T lymphocyte activation and migration. J Leukoc Biol. 2003; 73: 30–48.

    Murdoch C, Finn A. Chemokine receptors and their role in inflammation and infectious diseases. Blood. 2000; 95: 3032–3043.

    Srinivasan S, Wang F, Glavas S, Ott A, Hofmann F, Aktories K, Kalman D, Bourne HR. Rac and Cdc42 play distinct roles in regulating PI(3,4,5)P3 and polarity during neutrophil chemotaxis. J Cell Biol. 2003; 160: 375–385.

    Jin T, Zhang N, Long Y, Parent CA, Devreotes PN. Localization of the G protein beta-gamma complex in living cells during chemotaxis. Science. 2000; 287: 1034–1036.

    Servant G, Weiner OD, Neptune ER, Sedat JW, Bourne HR. Dynamics of a chemoattractant receptor in living neutrophils during chemotaxis. Mol Biol Cell. 1999; 10: 1163–1178.

    Nieto M, Frade JM, Sancho D, Mellado M, Martinez A, Sanchez-Madrid F. Polarization of chemokine receptors to the leading edge during lymphocyte chemotaxis. J Exp Med. 1997; 186: 153–158.

    McKay DA, Kusel JR, Wilkinson PC. Studies of chemotactic factor-induced polarity in human neutrophils. Lipid mobility, receptor distribution and the time-sequence of polarization. J Cell Sci. 1991; 100: 473–479.

    Vicente-Manzanares M, Rey M, Jones DR, Sancho D, Mellado M, Rodriguez-Frade JM, del Pozo MA, Yanez-Mo M, de Ana AM, Martinez AC, Merida I, Sanchez-Madrid F. Involvement of phosphatidylinositol 3-kinase in stromal cell-derived factor-1 alpha-induced lymphocyte polarization and chemotaxis. J Immunol. 1999; 163: 4001–4012.

    Pelletier AJ, Der Laan LJ, Hildbrand P, Siana MA, Thompson DA, Dawson PE, Torbett BE, Saloman DR. Presentation of chemokine SDF-1alpha by fibronectin mediates directed migration of T cells . Blood. 2000; 96: 2682–2690.

    Nguyen DH, Taub D. CXCR4 function requires membrane cholesterol: implications for HIV infection. J Immunol. 2002; 168: 4121–4126.

    Crockett-Torabi E. Selectins and mechanisms of signal transduction. J Leukoc Biol. 1998; 63: 1–14.

    Vestweber D, Blanks JE. Mechanisms that regulate the function of the selectins and their ligands. Physiol Rev. 1999; 79: 181–213.

    Ding Z, Issekutz TB, Downey GP, Waddell TK. L-selectin stimulation enhances functional expression of surface CXCR4 in lymphocytes: implications for cellular activation during adhesion and migration. Blood. 2003; 101: 4245–4252.

    Sitrin RG, Johnson DR, Pan PM, Harsh DM, Huang J, Petty HR, Blackwood RA. Lipid raft compartmentalization of urokinase receptor signaling in human neutrophils. Am J Respir Cell Mol Biol. 2004; 30: 233–241.

    Worthylake RA, Burridge K. Leukocyte transendothelial migration: orchestrating the underlying molecular machinery. Curr Opin Cell Biol. 2001; 13: 569–577.

    Peled A, Kollet O, Ponomaryov T, Petit I, Franitza S, Grabovsky V, Slav MM, Nagles A, Lider O, Alon R, Zipori D, Lapidot T. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood. 2000; 95: 3289–3296.

    Voermans C, Rood PM, Hordijk PL, Gerritsen WR, van der Schoot CE. Adhesion molecules involved in transendothelial migration of human hematopoietic progenitor cells. Stem Cells. 2000; 18: 435–443.

    Wang J, Springer TA. Structural specializations of immunoglobulin superfamily members for adhesion to integrins and viruses. Immunol Rev. 1998; 163: 197–215.

    Dejana E, Breviario F, Caveda L. Leukocyte-endothelial cell adhesive receptors. Clin Exp Rheumatol. 1994; 12: S25–S28.

    Weber C, Alon R, Moser B, Springer TA. Sequential regulation of alpha 4 beta 1 and alpha 5 beta 1 integrin avidity by CC chemokines in monocytes: implications for transendothelial chemotaxis. J Cell Biol. 1996; 134: 1063–1073.

    Borjesson DL, Simon SI, Hodzic E, DeCock HE, Ballantyne CM, Barthold SW. Roles of neutrophil beta 2 integrins in kinetics of bacteremia, extravasation, and tick acquisition of Anaplasma phagocytophila in mice. Blood. 2003; 101: 3257–3264.

    Issekutz AC, Rowter D, Springer TA. Role of ICAM-1 and ICAM-2 and alternate CD11/CD18 ligands in neutrophil transendothelial migration. J Leukoc Biol. 1999; 65: 117–126.

    Arroyo AG, Yang JT, Rayburn H, Hynes RO. Differential requirements for alpha4 integrins during fetal and adult hematopoiesis. Cell. 1996; 85: 997–1008.

    Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules. Blood. 199; 844: 2068–2101.

    van Kooyk Y, Figdor CG. Avidity regulation of integrins: the driving force in leukocyte adhesion. Curr Opin Cell Biol. 2000; 12: 542–547.

    Hughes PE, Pfaff M. Integrin affinity modulation. Trends Cell Biol. 1998; 8: 359–364.

    Devreotes P, Janetopoulos C. Eukaryotic chemotaxis: distinctions between directional sensing and polarization. J Biol Chem. 2003; 278: 20445–20448.

    Iijima M, Huang YE, Devreotes P. Temporal and spatial regulation of chemotaxis. Dev Cell. 2002; 3: 469–478.

    Rickert P, Weiner OD, Wang F, Bourne HR, Servant G. Leukocytes navigate by compass: roles of PI3Kgamma and its lipid products. Trends Cell Biol. 2000; 10: 466–473.

    Weiner OD. Regulation of cell polarity during eukaryotic chemotaxis: the chemotactic compass. Curr Opin Cell Biol. 2002; 14: 196–202.

    Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci. 2003; 116: 1409–1416.

    Webb DJ, Parsons JT, Horwitz AF. Adhesion assembly, disassembly and turnover in migrating cells - over and over and over again. Nat Cell Biol. 2002; 4: E97–100.

    Alonso-Lebrero JL, Serrador JM, Dominguez-Jimenez C, Barreiro O, Luque A, del Pozo MA, Snapp K, Kansas G, Schwartz-Albiez R, Furthmayr H, Lozano F, Sanchez-Madrid F. Polarization and interaction of adhesion molecules P-selectin glycoprotein ligand 1 and intercellular adhesion molecule 3 with moesin and ezrin in myeloid cells. Blood. 2000; 95: 2413–2419.

    Del Pozo MA, Sanchez-Mateos P, Nieto M, Sanchez-Madrid F. Chemokines regulate cellular polarization and adhesion receptor redistribution during lymphocyte interaction with endothelium and extracellular matrix. Involvement of cAMP signaling pathway. J Cell Biol. 1995; 131: 495–508.

    Serrador JM, Alonso-Lebrero JL, del Pozo MA, Furthmayr H, Schwartz-Albiez R, Calvo J, Lozano F, Sanchez-Madrid F. Moesin interacts with the cytoplasmic region of intercellular adhesion molecule-3 and is redistributed to the uropod of T lymphocytes during cell polarization. J Cell Biol. 1997; 138: 1409–1423.

    Del Pozo MA, Nieto M, Serrador JM, Sancho D, Vicente-Manzanares M, Martinez C, Sanchez-Madrid F. The two poles of the lymphocyte: specialized cell compartments for migration and recruitment. Cell Adhes Commun. 1998; 6: 125–133.

    Liu L, Schwartz BR, Lin N, Winn RK, Harlan JM. Requirement for RhoA kinase activation in leukocyte de-adhesion. J Immunol. 2002; 169: 2330–2336.

    Krylyshkina O, Kaverina I, Kranewitter W, Steffen W, Alonso MC, Cross RA, Small JV. Modulation of substrate adhesion dynamics via microtubule targeting requires kinesin-1. J Cell Biol. 2002; 156: 349–359.

    Small JV, Geiger B, Kaverina I, Bershadsky A. How do microtubules guide migrating cells? Nat Rev Mol Cell Biol. 2002; 3: 957–964.

    Goldfinger LE, Han J, Kiosses WB, Howe AK, Ginsberg MH. Spatial restriction of {alpha}4 integrin phosphorylation regulates lamellipodia stability and {alpha}4{beta}1-dependent cell migration. J Cell Biol. 2003; 162: 731–741.

    Turner CE. Paxillin and focal adhesion signalling. Nat Cell Biol. 2000; 2: E231–E236.

    Smith A, Bracke M, Leitinger B, Porter JC, Hogg N. LFA-1-induced T cell migration on ICAM-1 involves regulation of MLCK-mediated attachment and ROCK-dependent detachment. J Cell Sci. 2003; 116: 3123–3133.

    Porter JC, Bracke M, Smith A, Davies D, Hogg N. Signaling through integrin LFA-1 leads to filamentous actin polymerization and remodeling, resulting in enhanced T cell adhesion. J Immunol. 2002; 168: 6330–6335.

    Riteau B, Barber DF, Long EO. Vav1 phosphorylation is induced by beta2 integrin engagement on natural killer cells upstream of actin cytoskeleton and lipid raft reorganization. J Exp Med. 2003; 198: 469–474.

    Hall A. Rho GTPases and the actin cytoskeleton. Science. 1998; 279: 509–514.

    Kraynov VS, Chamberlain C, Bokoch GM, Schwartz MA, Slabaugh S, Hahn KM. Localized Rac activation dynamics visualized in living cells. Science. 2000; 290: 333–337.

    Del Pozo MA, Kiosses WB, Alderson NB, Meller N, Hahn KM, Schwartz MA. Integrins regulate GTP-Rac localized effector interactions through dissociation of Rho-GDI. Nat Cell Biol. 2002; 4: 232–239.

    van Hennik PB, ten Klooster JP, Halstead JR, Voermans C, Anthony EC, Divecha N, Hordijk PL. The C-terminal domain of Rac1 contains two motifs that control targeting and signaling specificity. J Biol Chem. 2003; 278: 39166–39175.

    de Bruyn KM, Rangarajan S, Reedquist KA, Figdor CG, Bos JL. The small GTPase Rap1 is required for Mn(2+)- and antibody-induced LFA-1- and VLA-4-mediated cell adhesion. J Biol Chem. 2002; 277: 29468–29476.

    Reedquist KA, Ross E, Koop EA, Wolthuis RM, Zwartkruis FJ, van Kooyk Y, Salmon M, Buckley CD, Bos JL. The small GTPase, Rap1, mediates CD31-induced integrin adhesion. J Cell Biol. 2000; 148: 1151–1158.

    Shimonaka M, Katagiri K, Nakayama T, Fujita N, Tsuruo T, Yoshie O, Kinashi T. Rap1 translates chemokine signals to integrin activation, cell polarization, and motility across vascular endothelium under flow. J Cell Biol. 2003; 161: 417–427.

    Katagiri K, Maeda A, Shimonaka M, Kinashi T. RAPL, a Rap1-binding molecule that mediates Rap1-induced adhesion through spatial regulation of LFA-1. Nat Immunol. 2003; 4: 741–748.

    Alblas J, Ulfman L, Hordijk P, Koenderman L. Activation of Rhoa and ROCK are essential for detachment of migrating leukocytes. Mol Biol Cell. 2001; 12: 2137–2145.

    Worthylake RA, Lemoine S, Watson JM, Burridge K. RhoA is required for monocyte tail retraction during transendothelial migration. J Cell Biol. 2001; 154: 147–160.

    Hixenbaugh EA, Goeckeler ZM, Papaiya NN, Wysolmerski RB, Silverstein SC, Huang AJ. Stimulated neutrophils induce myosin light chain phosphorylation and isometric tension in endothelial cells. Am J Physiol. 1997; 273: H981–H988.

    Nieuw Amerongen GP, van Delft S, Vermeer MA, Collard JG, van Hinsbergh VW. Activation of RhoA by thrombin in endothelial hyperpermeability: role of Rho kinase and protein tyrosine kinases. Circ Res. 2000; 87: 335–340.

    Nieuw Amerongen GP, Vermeer MA, van Hinsbergh VW. Role of RhoA and Rho kinase in lysophosphatidic acid-induced endothelial barrier dysfunction. Arterioscler Thromb Vasc Biol. 2000; 20: E127–E133.

    Saito H, Minamiya Y, Kitamura M, Saito S, Enomoto K, Terada K, Ogawa J. Endothelial myosin light chain kinase regulates neutrophil migration across human umbilical vein endothelial cell monolayer. J Immunol. 1998; 161: 1533–1540.

    Lorenzon P, Vecile E, Nardon E, Ferrero E, Harlan JM, Tedesco F, Dobrina A. Endothelial cell E- and P-selectin and vascular cell adhesion molecule- 1 function as signaling receptors. J Cell Biol. 1998; 142: 1381–1391.

    Hordijk P. Endothelial signaling in leukocyte transmigration. Cell Biochem Biophys. 2003; 38: 305–322.

    Wang Q, Doerschuk CM. The signaling pathways induced by neutrophil-endothelial cell adhesion. Antioxid Redox Signal. 2002; 4: 39–47.

    Chavakis T, Preissner KT, Santoso S. Leukocyte trans-endothelial migration: JAMs add new pieces to the puzzle. Thromb Haemost. 2003; 89: 13–17.

    Etienne S, Adamson P, Greenwood J, Strosberg AD, Cazaubon S, Couraud PO. ICAM-1 signaling pathways associated with Rho activation in microvascular brain endothelial cells. J Immunol. 1998; 161: 5755–5761.

    Durieu-Trautmann O, Chaverot N, Cazaubon S, Strosberg AD, Couraud PO. Intercellular adhesion molecule 1 activation induces tyrosine phosphorylation of the cytoskeleton-associated protein cortactin in brain microvessel endothelial cells. J Biol Chem. 1994; 269: 12536–12540.

    Heiska L, Alfthan K, Gronholm M, Vilja P, Vaheri A, Carpen O. Association of ezrin with intercellular adhesion molecule-1 and -2 (ICAM-1 and ICAM-2). Regulation by phosphatidylinositol 4, 5- bisphosphate. J Biol Chem. 1998; 273: 21893–21900.

    Bretscher A. Regulation of cortical structure by the ezrin-radixin-moesin protein family. Curr Opin Cell Biol. 1999; 11: 109–116.

    Lamb RF, Ozanne BW, Roy C, McGarry L, Stipp C, Mangeat P, Jay DG. Essential functions of ezrin in maintenance of cell shape and lamellipodial extension in normal and transformed fibroblasts. Curr Biol. 1997; 7: 682–688.

    Granes F, Urena JM, Rocamora N, Vilaro S. Ezrin links syndecan-2 to the cytoskeleton. J Cell Sci. 2000; 113: 1267–1276.

    Wojciak-Stothard B, Williams L, Ridley AJ. Monocyte adhesion and spreading on human endothelial cells is dependent on Rho-regulated receptor clustering. J Cell Biol. 1999; 145: 1293–1307.

    Pujuguet P, Del Maestro L, Gautreau A, Louvard D, Arpin M. Ezrin Regulates E-Cadherin-dependent Adherens Junction Assembly through Rac1 Activation. Mol Biol Cell. 2003; 14: 2181–2191.

    Matsui T, Maeda M, Doi Y, Yonemura S, Amano M, Kaibuchi K, Tsukita S, Tsukita S. Rho-kinase phosphorylates COOH-terminal threonines of ezrin/radixin/moesin (ERM) proteins and regulates their head-to-tail association. J Cell Biol. 1998; 140: 647–657.

    Barreiro O, Yanez-Mo M, Serrador JM, Montoya MC, Vicente-Manzanares M, Tejedor R, Furthmayr H, Sanchez-Madrid F. Dynamic interaction of VCAM-1 and ICAM-1 with moesin and ezrin in a novel endothelial docking structure for adherent leukocytes. J Cell Biol. 2002; 157: 1233–1245.

    van Wetering S, van den Berk N, van Buul JD, Mul FP, Lommerse I, Mous R, ten Klooster JP, Zwaginga JI, Hordijk PL. VCAM-1-mediated Rac signaling controls endothelial cell-cell contacts and leukocyte transmigration. Am J Physiol. 2003; 285: C343–C352.

    Vastrik I, Eickholt BJ, Walsh FS, Ridley A, Doherty P. Sema3A-induced growth-cone collapse is mediated by Rac1 amino acids 17–32. Curr Biol. 1999; 9: 991–998.

    Braga VM, Del Maschio A, Machesky L, Dejana E. Regulation of cadherin function by Rho and Rac: modulation by junction maturation and cellular context. Mol Biol Cell. 1999; 10: 9–22.

    van Wetering S, van Buul JD, Quik S, Mul FP, Anthony EC, ten Klooster JP, Collard JG, Hordijk PL. Reactive oxygen species mediate Rac-induced loss of cell-cell adhesion in primary human endothelial cells. J Cell Sci. 2002; 115: 1837–1846.

    Vouret-Craviari V, Boquet P, Pouyssegur J, Obberghen-Schilling E. Regulation of the actin cytoskeleton by thrombin in human endothelial cells: role of Rho proteins in endothelial barrier function. Mol Biol Cell. 1998; 9: 2639–2653.

    Lazaar AL, Krymskaya VP, Das SK. VCAM-1 activates phosphatidylinositol 3-kinase and induces p120Cbl phosphorylation in human airway smooth muscle cells. J Immunol. 2001; 166: 155–161.

    Sander EE, van Delft S, ten Klooster JP, Reid T, van der Kammen RA, Michiels F, Collard JG. Matrix-dependent Tiam1/Rac signaling in epithelial cells promotes either cell-cell adhesion or cell migration and is regulated by phosphatidylinositol 3-kinase. J Cell Biol. 1998; 143: 1385–1398.

    Lum H, Roebuck KA. Oxidant stress and endothelial cell dysfunction. Am J Physiol. 2001; 280: C719–C741.

    Wolin MS. Interactions of oxidants with vascular signaling systems. Arterioscler Thromb Vasc Biol. 2000; 20: 1430–1442.

    Zhao X, Alexander JS, Zhang S, Zhu Y, Sieber NJ, Aw TY, Carden DL. Redox regulation of endothelial barrier integrity. Am J Physiol. 2001; 281: L879–L886.

    Chiarugi P, Pani G, Giannoni E, Taddei L, Colavitti R, Raugei G, Symons M, Borrello S, Galeotti T, Ramponi G. Reactive oxygen species as essential mediators of cell adhesion: the oxidative inhibition of a FAK tyrosine phosphatase is required for cell adhesion. J Cell Biol. 2003; 161: 933–944.

    Jones SA, O’Donnell VB, Wood JD, Broughton JP, Hughes EJ, Jones OT. Expression of phagocyte NADPH oxidase components in human endothelial cells. Am J Physiol. 1996; 271: H1626–H1634.

    Li JM, Shah AM. Intracellular localization and preassembly of the NADPH oxidase complex in cultured endothelial cells. J Biol Chem. 2002; 277: 19952–19960.

    Cheng G, Cao Z, Xu X, van Meir EG, Lambeth JD. Homologs of gp91phox: cloning and tissue expression of Nox3, Nox4, and Nox5. Gene. 2001; 269: 131–140.

    Sorescu D, Weiss D, Lassegue B, Clempus RE, Szocs K, Sorescu GP, Valppu J, Quinn MT, Lambeth JD, Vega JD, Taylor WR, Griendling KK. Superoxide production and expression of nox family proteins in human atherosclerosis. Circulation. 2002; 105: 1429–1435.

    Szocs K, Lassegue B, Sorescu D, Hilenski LL, Valppu L, Couse TL, Wilcox JN, Quinn MT, Lambeth JD, Griendling KK. Upregulation of Nox-based NAD(P)H oxidases in restenosis after carotid injury. Arterioscler Thromb Vasc Biol. 2002; 22: 21–27.

    Wang Q, Doerschuk CM. Neutrophil-induced changes in the biomechanical properties of endothelial cells: roles of ICAM-1 and reactive oxygen species. J Immunol. 2000; 164: 6487–6494.

    Matheny HE, Deem TL, Cook-Mills JM. Lymphocyte migration through monolayers of endothelial cell lines involves VCAM-1 signaling via endothelial cell NADPH oxidase. J Immunol. 2000; 164: 6550–6559.

    Wang Q, Chiang ET, Lim M, Lai J, Rogers R, Janney PA, Shepro D, Doerschuk CM. Changes in the biomechanical properties of neutrophils and endothelial cells during adhesion. Blood. 2001; 97: 660–668.

    Wang Q, Doerschuk CM. The p38 Mitogen-Activated Protein Kinase Mediates Cytoskeletal Remodeling in Pulmonary Microvascular Endothelial Cells Upon Intracellular Adhesion Molecule-1 Ligation. J Immunol. 2001; 166: 6877–6884.

    Hordijk PL, ten Klooster JP, van der Kammen RA, Michiels F, Oomen LC, Collard JG. Inhibition of invasion of epithelial cells by Tiam1-Rac signaling. Science. 1997; 278: 1464–1466.

    Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required for transendothelial migration of leukocytes. J Exp Med. 1993; 178: 449–460.

    Liao F, Huynh HK, Eiroa A, Greene T, Polizzi E, Muller WA. Migration of monocytes across endothelium and passage through extracellular matrix involve separate molecular domains of PECAM-1. J Exp Med. 1995; 182: 1337–1343.

    Christofidou-Solomidou M, Nakada MT, Williams J, Muller WA, DeLisser HM. Neutrophil platelet endothelial cell adhesion molecule-1 participates in neutrophil recruitment at inflammatory sites and is down-regulated after leukocyte extravasation. J Immunol. 1997; 158: 4872–4878.

    Thompson RD, Noble KE, Larbi KY, Dewar A, Duncan GS, Mak TW, Nourshargh S. Platelet-endothelial cell adhesion molecule-1 (PECAM-1)-deficient mice demonstrate a transient and cytokine-specific role for PECAM-1 in leukocyte migration through the perivascular basement membrane. Blood. 2001; 97: 1854–1860.

    Vaporciyan AA, DeLisser HM, Yan HC, Mendiguren II, Thom SR, Jones ML, Ward PA, Albelba SM. Involvement of platelet-endothelial cell adhesion molecule-1 in neutrophil recruitment in vivo. Science. 1993; 262: 1580–1582.

    Bogen S, Pak J, Garifallou M, Deng X, Muller WA. Monoclonal antibody to murine PECAM-1 (CD31) blocks acute inflammation in vivo. J Exp Med. 1994; 179: 1059–1064.

    Wakelin MW, Sanz MJ, Dewar A, Albelba SM, Larkin SW, Boughton-Smith N, Williams TJ, Nourshargh S. An anti-platelet-endothelial cell adhesion molecule-1 antibody inhibits leukocyte extravasation from mesenteric microvessels in vivo by blocking the passage through the basement membrane. J Exp Med. 1996; 184: 229–239.

    Gumina RJ, el Schultz J, Yao Z, Kenny D, Warltier DC, Newman PJ, Gross GL. Antibody to platelet/endothelial cell adhesion molecule-1 reduces myocardial infarct size in a rat model of ischemia-reperfusion injury. Circulation. 1996; 94: 3327–3333.

    Murohara T, Delyani JA, Albelda SM, Lefer AM. Blockade of platelet endothelial cell adhesion molecule-1 protects against myocardial ischemia and reperfusion injury in cats. J Immunol. 1996; 156: 3550–3557.

    Nakada MT, Amin K, Christofidou-Solomidou M, O’Brien CD, Sun J, Gurubhagavatula I, Heavner GA, Taylor AH, Paddock C, Sun QH, Zehnder JL, Newman PJ, Albelda SM, DeLisser HM. Antibodies against the first Ig-like domain of human platelet endothelial cell adhesion molecule-1 (PECAM-1) that inhibit PECAM-1-dependent homophilic adhesion block in vivo neutrophil recruitment. J Immunol. 2000; 164: 452–462.

    Liao F, Schenkel AR, Muller WA. Transgenic mice expressing different levels of soluble platelet/endothelial cell adhesion molecule-IgG display distinct inflammatory phenotypes. J Immunol. 1999; 163: 5640–5648.

    Thompson RD, Wakelin MW, Larbi KY, Dewar A, Asimakopoulos G, Horton MA, Nakada MT, Nourshargh S. Divergent effects of platelet-endothelial cell adhesion molecule-1 and beta 3 integrin blockade on leukocyte transmigration in vivo. J Immunol. 2000; 165: 426–434.

    Liao F, Ali J, Greene T, Muller WA. Soluble domain 1 of platelet-endothelial cell adhesion molecule (PECAM) is sufficient to block transendothelial migration in vitro and in vivo. J Exp Med. 1997; 185: 1349–1357.

    Duncan GS, Andrew DP, Takimoto H, Kaufman SA, Yoshida H, Spellberg J, Luis de la Porpa J, Elia A, Wakeham A, Karan-Tamir B, Muller WA, Senadli G, Zukowski MM, Mak TW. Genetic evidence for functional redundancy of Platelet/Endothelial cell adhesion molecule-1 (PECAM-1): CD31-deficient mice reveal PECAM-1-dependent and PECAM-1-independent functions. J Immunol. 1999; 162: 3022–3030.

    Mamdouh Z, Chen X, Pierini LM, Maxfield FR, Muller WA. Targeted recycling of PECAM from endothelial surface-connected compartments during diapedesis. Nature. 2003; 421: 748–753.

    Feng D, Nagy JA, Pyne K, Dvorak HF, Dvorak AM. Neutrophils emigrate from venules by a transendothelial cell pathway in response to FMLP. J Exp Med. 1998; 187: 903–915.

    Osawa M, Masuda M, Kusano K, Fujiwara K. Evidence for a role of platelet endothelial cell adhesion molecule-1 in endothelial cell mechanosignal transduction: is it a mechanoresponsive molecule? J Cell Biol. 2002; 158: 773–785.

    Kogata N, Masuda M, Kamioka Y, Yamagishi A, Endo A, Okadaq M, Mochizuki N. Identification of fer tyrosine kinase localized on microtubules as a platelet endothelial cell adhesion molecule-1 phosphorylating kinase in vascular endothelial cells. Mol Biol Cell. 2003; 14: 3553–3564.

    Ilan N, Mahooti S, Rimm DL, Madri JA. PECAM-1 (CD31) functions as a reservoir for and a modulator of tyrosine-phosphorylated (?)-catenin. J Cell Sci. 1999; 112: 3005–3014.

    Ayalon O, Geiger B. Cyclic changes in the organization of cell adhesions and the associated cytoskeleton, induced by stimulation of tyrosine phosphorylation in bovine aortic endothelial cells. J Cell Sci. 1997; 110: 547–556.

    Lampugnani MG, Corada M, Andriopoulou P, Esser S, Risau W, Dejana E. Cell confluence regulates tyrosine phosphorylation of adherens junction components in endothelial cells. J Cell Sci. 1997; 110: 2065–2077.

    Ukropec JA, Hollinger MK, Salva SM, Woolkalis MJ. SHP2 association with VE-cadherin complexes in human endothelial cells is regulated by thrombin. J Biol Chem. 2000; 275: 5983–5986.

    Young BA, Sui X, Kiser TD, Hyun SW, Wang P, Sakarya S, Angelini DJ, Schaphorst KL, Hasday JD, Cross AS, Romer LH, Passaniti A, Goldblum SE. Protein tyrosine phosphatase activity regulates endothelial cell-cell interactions, the paracellular pathway, and capillary tube stability. Am J Physiol. 2003; 285: L63–L75.

    Rhee SG, Bae YS, Lee SR, Kwon J. Hydrogen peroxide: a key messenger that modulates protein phosphorylation through cysteine oxidation. Sci STKE. 2000; 2000: E1.

    Muller WA. Leukocyte-endothelial-cell interactions in leukocyte transmigration and the inflammatory response. Trends Immunol. 2003; 24: 327–334.

    Martin-Padura I, Lostaglio S, Schneemann M, Williams L, Romano M, Fruscella P, Panzeri C, Stoppacciaro A, Ruco L, Villa A, Simmons D, Dejana E. Junctional adhesion molecule, a novel member of the immunoglobulin superfamily that distributes at intercellular junctions and modulates monocyte transmigration. J Cell Biol. 1998; 142: 117–127.

    Liu Y, Nusrat A, Schnell FJ, Reaves TA, Walsh S, Pochet M, Parkos CA. Human junction adhesion molecule regulates tight junction resealing in epithelia. J Cell Sci. 2000; 113: 2363–2374.

    Aurrand-Lions M, Johnson-Leger C, Wong C, Du PL, Imhof BA. Heterogeneity of endothelial junctions is reflected by differential expression and specific subcellular localization of the three JAM family members. Blood. 2001; 98: 3699–3707.

    Del Maschio A, De Luigi A, Martin-Padura I, Brockhaus M, Bartfai T, Fruscella P, Adorini L, Martino G, Furlan R, De Simoni MG, Dejana E. Leukocyte recruitment in the cerebrospinal fluid of mice with experimental meningitis is inhibited by an antibody to junctional adhesion molecule (JAM). J Exp Med. 1999; 190: 1351–1356.

    Ostermann G, Weber KS, Zernecke A, Schroder A, Weber C. JAM-1 is a ligand of the beta(2) integrin LFA-1 involved in transendothelial migration of leukocytes. Nat Immunol. 2002; 3: 151–158.

    Johnson-Leger CA, Aurrand-Lions M, Beltraminelli N, Fasel N, Imhof BA. Junctional adhesion molecule-2 (JAM-2) promotes lymphocyte transendothelial migration. Blood. 2002; 100: 2479–2486.

    Palmeri D, van Zante A, Huang CC, Hemmerich S, Rosen SD. VE-JAM, a novel member of the immunoglobulin superfamily, is localized to intercellular boundaries of endothelial cells. J Biol Chem. 2000; 275: 19139–19145.

    Cunningham SA, Rodriguez JM, Arrate MP, Tran TM, Brock TA. JAM2 interacts with alpha4beta1. Facilitation by JAM3. J Biol Chem. 2002; 277: 27589–27592.

    Liang TW, Chiu HH, Gurney A, Sidle A, Tumas DB, Schow P, Foster J, Klassen T, Demis K, DeMarco RA, Pham T, Frantz G, Fong S. Vascular endothelial-junctional adhesion molecule (VE-JAM)/JAM 2 interacts with T, NK, and dendritic cells through JAM 3. J Immunol. 2002; 168: 1618–1626.

    Arrate MP, Rodriguez JM, Tran TM, Brock TA, Cunningham SA. Cloning of human junctional adhesion molecule 3 (JAM3) and its identification as the JAM2 counter-receptor. J Biol Chem. 2001; 276: 45826–45832.

    Santoso S, Sachs UJ, Kroll H, Lindner M, Ruf A, Preissner KT, Chavakis T. The junctional adhesion molecule 3 (JAM-3) on human platelets is a counterreceptor for the leukocyte integrin Mac-1. J Exp Med. 2002; 196: 679–691.

    Bernard G, Raimondi V, Alberti I, Pourtein M, Widjenes J, Ticchioni M, Bernard A. CD99 (E2) up-regulates alpha4beta1-dependent T cell adhesion to inflamed vascular endothelium under flow conditions. Eur J Immunol. 2000; 30: 3061–3065.

    Schenkel AR, Mamdouh Z, Chen X, Liebman RM, Muller WA. CD99 plays a major role in the migration of monocytes through endothelial junctions. Nat Immunol. 2002; 3: 143–150.

    Allport JR, Ding H, Collins T, Gerritsen ME, Luscinskas FW. Endothelial-dependent mechanisms regulate leukocyte transmigration: a process involving the proteasome and disruption of the vascular endothelial-cadherin complex at endothelial cell-to-cell junctions. J Exp Med. 1997; 186: 517–527.

    Del Maschio A, Zanetti A, Corada M, Rival Y, Ruco L, Lampugnani MG, Dejana E. Polymorphonuclear leukocyte adhesion triggers the disorganization of endothelial cell-to-cell adherens junctions. J Cell Biol. 1996; 135: 497–510.

    Moll T, Dejana E, Vestweber D. In vitro degradation of endothelial catenins by a neutrophil protease. J Cell Biol. 1998; 140: 403–407.

    Allport JR, Muller WA, Luscinskas FW. Monocytes induce reversible focal changes in vascular endothelial cadherin complex during transendothelial migration under flow. J Cell Biol. 2000; 148: 203–216.

    Sandig M, Negrou E, Rogers KA. Changes in the distribution of LFA-1, catenins, and F-actin during transendothelial migration of monocytes in culture. J Cell Sci. 1997; 110: 2807–2818.

    Hermant B, Bibert S, Concord E, Dublet B, Weidenhaupt M, Vernet T, Gulino-Debrac D. Identification of proteases involved in the proteolysis of vascular endothelium cadherin during neutrophil transmigration. J Biol Chem. 2003; 278: 14002–14012.

    Allport JR, Lim YC, Shipley JM, Senior RM, Shapiro SD, Matsuyoshi N, Vestweber D, Luscinskas FW. Neutrophils from MMP-9- or neutrophil elastase-deficient mice show no defect in transendothelial migration under flow in vitro. J Leukoc Biol. 2002; 71: 821–828.(Jaap D. van Buul; Peter L)