当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 循环研究杂志 > 2005年 > 第3期 > 正文
编号:11257837
New Kids on the Signaling Block
     the Angiogenesis Research Center, Section of Cardiology

    Departments of Medicine and Pharmacology and Toxicology, Dartmouth Medical School, Lebanon, N.H.

    Abstract

    Cell-associated proteoglycans provide highly complex and sophisticated systems to control interactions of extracellular cell matrix components and soluble ligands with the cell surface. Syndecans, a conserved family of heparan- and chondroitin-sulfate carrying transmembrane proteins, are emerging as central players in these interactions. Recent studies have demonstrated the essential role of syndecans in modulating cellular signaling in embryonic development, tumorigenesis, and angiogenesis. In this review, we focus on new advances in our understanding of syndecan-mediated cell signaling.

    Key Words: proteoglycan angiogenesis signal transduction cytoskeleton migration

    Introduction

    Cell interactions with their environment are critical to a large number of processes including growth, migration, adhesion, and apoptosis, among many others. In addition to a variety of specialized receptor systems that have evolved to transmit signals from specific ligands, other systems have also evolved to inform cells of the broader extracellular context. Thus, adhesion receptors such as integrins can be activated by a number of extracellular matrix proteins, whereas selectins and various cellular adhesion molecules participate in celleCcell interactions.

    Over the last few years, it has become increasingly clear that cells possess yet another unique system that integrates signaling from circulating ligands such as growth factors and extracellular matrix proteins with other cellular receptor systems such as integrins. This unique function, performed by the syndecan family of proteins, places them at the center of signal integration in the cell and has earned them the name of "tuners of transmembrane signaling."1

    Syndecan Structure

    Syndecans are a family of transmembrane core proteins capable of carrying heparan sulfate (HS) and chondroitin sulfate (CS) chains. Whereas invertebrates have only one syndecan, four syndecan genes (syndecan-1, -2, -3, and -4) are present in vertebrates. Each syndecan has a short cytoplasmic domain, a single-span transmembrane domain (TM), and an extracellular domain with attachment sites for three to five HS or CS chains (Figure 1). The presence of HS chains allows interactions with a large number of proteins, including heparin-binding growth factors such as fibroblast growth factors (FGFs), vascular endothelial growth factors (VEGFs), transforming growth factor- (TGF-), and platelet-derived growth factors. Furthermore, HSs facilitate interactions with various extracellular matrix proteins, including fibronectin and plasma proteins such as antithrombin-1. The role of CS chains is less clear. A recent study has suggested that syndecan-1 and syndecan-4 CS chains cooperate with HS chains in binding to the heparin-binding growth factors midkine and pleitrophin2 and to the extracellular matrix protein laminin.3

    Extracellular Domain

    The structural diversity of HS and CS chains results from a series of post-translational modifications beginning with the attachment of the first hexosamine to the linkage region tetrasaccharide, which is O-linked to serine or threonine in the core transmembrane protein committing the oligosaccharide chain to either HS (N-acetylglucosamine, uronic acid) or CS (N-acetylgalactosamine, uronic acid) disaccharide polymer.4 Then HS and CS glycosaminoglycan (GAG) backbones are extended by different sets of polymerizing glycosyltransferases, resulting in HS and CS chains of 20 to 80 disaccharides in average length. Then these newly synthesized chains are modified by epimerization of glucuronic to iduronic acid residues and sulfation of the C2 position of glucuronic and iduronic acid. A myriad of specific CS (4-O and 6-O N-acetylgalactosaminyl) and HS (2-O, 3-O, and 6-O N-acetylglucosaminyl and N-deacetylase/N-) sulfotransferases complete the decoration of these GAG chains.5

    Various combinations of enzymes involved in post-translational HS chain modifications produce unique binding motifs that selectively recognize different proteins. Thus, a specific combination of 2-O and 6-O sulfation is necessary for synthesis of the FGF2-binding site, whereas 3-O-sulfotransferase 1 activity is needed for generation of the antithrombin-1eCbinding site. Modifications of sulfotransferase expression and activity can significantly modulate syndecan functions as demonstrated, for example, in the case of 2-O-sulfotransferase (2-OST) deficiency that results in marked abnormalities of FGF signaling.6 On the other hand, augmentation of 2-OST expression, as occurs in the setting of hypoxia, leads to enhanced FGF responsiveness.7

    The recent discovery of specific mammalian sulfatases (Sulf 1 and Sulf2), which can modify GAG chains extracellularly, adds another layer of complexity to the system. Sulf1 and Sulf2 are secreted, HS-specific 6-O-sulfatases,8,9 and their activity can modify heparin-binding growth factor signaling.10 In quail, Qsulf1, the avian homolog of Sulf1 and Sulf2, has been shown to remodel HS on the cell surface to promote Wnt-1 signaling;11 whereas in human cancer cell lines, Hsulf1 expression inhibited FGF2 and hepatocyte growth factor (HGF) stimulation of cell growth.12

    It has been generally assumed that all syndecan HS and CS chains are created equal; that is, there is no preference for specific HS/CS sequences on specific syndecans.13 However, a recent study has suggested that CS and HS chains on syndecan-1 and syndecan-4 are structurally different.2 If confirmed, this will add yet more complexity to syndecan biology. In addition to interacting with extracellular protein via their HS and CS chains, the syndecan-4 core can directly engage in proteineCprotein interactions.14 The extracellular domain of syndecan-4 binds specifically to human foreskin fibroblasts (IC50=10eC8 M) and mouse aortic endothelial cells, as well as other cell types, whereas same-species syndecan family members did not efficiently compete with these interactions.14

    Transmembrane and Intracellular Domains

    Although the degree of conservation in the extracellular domain of syndecans is fairly low, the TM is highly conserved. As with classic type I membrane protein architecture, syndecans have a single-pass TM. Uniquely though, the TM and a small sequence adjacent to it have a high affinity for self-association, which, in the case of syndecan-4, has been shown to be required for protein kinase C- (PKC) activation.15

    The cytoplasmic domain, despite being relatively short, has a number of important regions. The domain is divided into three regions: conserved regions 1 and 2 (C1 and C2) and a variable (V) region (Figure 1). The C1 domain, immediately adjacent to the plasma cell membrane, is virtually identical in all four mammalian syndecans. It is thought to be involved in syndecan dimerization (all syndecans probably exist as homodimers and higher-order oligomers) and in binding of several intracellular proteins, including ezrin, tubulin, Src kinase, and cortactin.16eC18 The universally conserved C-terminal C2 domain contains a postsynaptic density-95/disc large protein/zonula occludens-1 (PDZ2)-binding site at the C-terminal end and two tyrosine residues. There are four identified syndecan-interacting PDZ domaineCcontaining proteins: syntenin, synectin, synbindin, and calcium/calmodulin-dependent serine protein kinase.19eC23

    The V domain is highly heterogeneous among the four mammalian syndecans. This region has been studied most extensively in syndecan-4 because of its unique features. The syndecan-4 V-region sequence includes a phosphatidylinositol-4,5-bisphosphate (PIP2) binding site that is involved in syndecan-4 dimerization, in which two syndecan-4 cytoplasmic domains are linked by two PIP2 molecules in antiparallel fashion (Figure 2).24

    In addition, PIP2 binding to the V region plays a critical role in binding and activation of PKC, an enzyme that plays a key role in syndecan-4 signaling (see below).15,25eC29 Indeed, this feature places PKC in the class of receptor-activated kinases and allows it to play a role not dissimilar from that of integrin-linked kinases. -Actinin and PKC compete for syndecan-4 binding, possibly providing a mechanism for regulation of syndecan-4eCcytoskeletal interaction.30 Another syndecan-4eCspecific binding protein is syndesmos.31 Although the precise nature of its interaction with syndecan-4 has not been defined, it requires C1 and V syndecan-4 domains for binding. Syndesmos binding to focal adhesion adaptor proteins paxillin and Hic-5 may be an alternative to an -actinin mechanism, linking syndecan-4 to focal adhesions.32

    Nuclear magnetic resonance structural analysis demonstrates that the C-terminal orientation of syndecan-4 changes on PIP2 binding, implying a possible regulation of PDZ interactions either by PIP2 or the PDZ-binding partners (Figure 2).24,33

    Regulation of Syndecan Expression

    Each syndecan family member has a distinct temporal and spatial pattern of expression, with every mammalian cell expressing at least one syndecan that is highly regulated during development.34 Syndecan-1 is expressed predominantly in epithelial and mesenchymal tissues, syndecan-2 in cells of mesenchymal origin and neuronal and epithelial cells, and syndecan-3 almost exclusively in neuronal and musculoskeletal tissue, whereas syndecan-4 is found in virtually every cell type.35

    Given their role in tuning numerous signaling events, it is not surprising that syndecan levels are tightly regulated. Growth factors play an important role in regulation of syndecan expression. Thus, tumor necrosis factor- (TNF-) upregulates syndecan-2 and downregulates syndecan-1 in endothelial cells.36 Similarly, TGF-2 upregulates syndecan-4 and downregulates syndecan-1 in epithelial cells.37 In aortic smooth muscle cells, FGF2 induces syndecan-4 but not syndecan-1 or syndecan-2 expression.38 Mechanical stress is also a prominent inducer of syndecan-4 expression in smooth muscle during arteriogenesis.39 Syndecan-4 levels are markedly upregulated in several disease states, including arterial injury40 and acute myocardial infarction.41 Another condition associated with a pronounced increase in syndecan-1 and syndecan-4 expression is wound healing.42,43 Although there are probably numerous factors responsible for this increase, an interesting mediator is an inflammatory cell-derived peptide, PR39, which has a unique ability to markedly upregulate syndecan-1 and syndecan-4 expression in vitro and in vivo.44,45

    Very little is known about regulation of syndecan-2 and syndecan-3 expression. Syndecan-2 levels increase during transformation of fat cells into myofibroblasts, whereas the level of syndecan-1, syndecan-3, and syndecan-4 remains constant.46 Syndecan-3 expression is highly regulated during development. In rodents, its level in central nervous system rises at birth, peaks on day 7, and then declines to the level of adult.47 This period of high level of syndecan-3 expression corresponds with oligodendrocyte differentiation and myelin formation in the central nervous system. The temporal dermal expression of syndecan-3 suggests the possibility of its involvement in feather development.48 During chick embryo limb development, syndecan-3 is transiently expressed during the period of mesenchymal condensation.49

    Interestingly, changes in the gene expression of one syndecan family member may affect others. For example, increased syndecan-3 expression in satellite cells leads to the downregulation of syndecan-4eCtransduced FGF2 and HGF signaling,50 whereas syndecan-3 expression is reduced in syndecan-4eC/eC satellite cells.

    Syndecan Function in Development

    Recent studies have begun clarifying the roles played by various syndecans during the developmental process (Table). Homozygous disruption of the syndecan-1 gene in mice leads to viable offspring. The mice are grossly normal but demonstrate abnormally slow re-epithelialization after injury51 and increased leukocyte adhesion.52 Syndecan-1 appears to be involved in modulation of Wnt-1 signaling because mammary glandeCspecific expression of Wnt-1 leads to development of tumors in wild-type but not in syndecan-1 knockout mice.53

    Studies in zebrafish using anti-syndecan-2 morpholino oligonucleotides demonstrated that this syndecan plays an important role in vascular development involving modulation of VEGF signaling. Syndecan-2 knockdown resulted in suppression of intersegmental vessels, whereas formation of dorsal vessels (aorta, cardinal vein) was not affected.54 Overexpression of VEGF165 in syndecan-2 morphans was not able to induce ectopic vessel formation, whereas coexpression of syndecan-2 and VEGF165 resulted in an increase of vessel formation compared with VEGF alone. Application of moderate doses of syndecan-2 and VEGF morpholinos demonstrated synergistic inhibition of angiogenesis. Expression of cytoplasmic domain-truncated syndecan-2 mimics the phenotype produced by syndecan-2 knockdown. On the basis of these data, syndecan-2 appears to potentiate VEGF-induced capillary sprouting.

    Syndecan-2 is also involved in lefteCright asymmetry regulation.35 In Xenopus, syndecan-2 is phosphorylated by PKC only in right animal cap ectodermal cells.55 Overexpression of the cytoplasmic domain-truncated form of syndecan-2 results in perturbed determination of lefteCright asymmetry of embryo and also randomizes expression of lefty, a lefteCright symmetry regulator.55

    Syndecan-3 levels in hypothalamus have been demonstrated to physiologically regulate feeding behavior, and mice with homozygous disruption of both syndecan-3 alleles displayed reduced feeding behavior in response to food deprivation56 as well as impaired performance in tasks using hippocampal functioning, indicating learning and memory abnormalities.57 They develop muscular dystrophy characterized by fibrosis, deteriorated locomotion, and hyperplasia of myonuclei and satellite cells.50

    Syndecan-4 deficiency has generated much interest. Null mice are viable and fertile but display a number of subtle defects. However, syndecan-4eC/eC embryos demonstrate much more frequent thrombi formation in the vessels of the placental labyrinth than the littermate controls, resulting in much higher embryo loss and implicating syndecan-4 in regulation of blood clotting.58 The adult syndecan-4eC/eC mice have increased mortality after lipopolysaccharide injection, suggesting their inability to clear pathogens59 and to downregulate TNF-—induced suppression of interleukin-1 (IL-1) expression in macrophages.57 The syndecan-4eC/eC mice also have increased susceptibility to -carrageenaneCinduced renal damage, presumably because of increased deposition in renal collecting ducts.60

    Other interesting observations include impaired skin wound healing that is thought to be secondary to defective angiogenesis.61 However, no other angiogenesis deficiency phenotypes have been reported to date. Finally, satellite cells from syndecan-4eC/eC mice fail to reconstitute damaged muscles, suggesting that syndecan-4 presence is required for migration of these skeletal muscle progenitor cells.50 This may in part be attributable to the defective FGF2- and HGF-induced activation of extracellular signal-regulated kinase (Erk)-1/2.50

    Modulation of Outside-In Signaling

    The preceding section demonstrates that disruption of various syndecan genes leads to phenotypes consistent with the "tuning" role of syndecans in signal transduction. The precise mechanism of this tuning has generated much interest and appears to be different for different syndecans. In the discussion that follows, although we consider signaling events associated with individual syndecans, it should be kept in mind that considerable overlap between various syndecan-mediated events probably exists and that the "specific" signaling events described for an individual syndecan may just as well reflect the limitation of our knowledge as the real biological specificities. Nevertheless, there is also a considerable degree of specificity. In particular, it is useful to think in the signaling context of syndecan-1 and syndecan-3 as one subfamily and syndecan-2 and syndecan-4 as another. This classification reflects homology of V domain between corresponding syndecans as well as functional similarities. For example, syndecan-1 and syndecan-3 expression is mostly associated with inhibition of cell growth, whereas syndecan-2 and syndecan-4 expression leads to its stimulation.

    HS and CS Chains

    The way in which syndecans engage in signal transduction remains a matter of active research and controversy. The first and the oldest hypothesis postulates that syndecans serve as coreceptors for various heparin-binding growth factors. This is thought to occur because of GAG chains binding growth factors, thereby restricting their presence to the membrane surface and facilitating their subsequent interactions with corresponding high-affinity receptors.62 Numerous studies have shown that inhibition of HS chain synthesis grossly affects signaling. For example, heparinase treatment of cultured smooth muscle cells decreases their responsiveness to FGF2,63 whereas heparinase treatment in vivo inhibits angiogenesis.64

    Genetic data are equally compelling. Inhibition of HS formation attributable to mutations of EXT1 and EXT2 genes, which catalyze polymerization of glucuronic acid and N-acetylglucosamine, the crucial step in HS synthesis, results in hereditary multiple exostoses, an autosomal skeletal disorder characterized by inappropriate chondrocyte proliferation and bone growth.65 The defect is thought to be attributable to abnormal diffusion of hedgehog (Hh) proteins.66 A homozygous disruption of Ext-1 expression in mice results in gastrulation defects and early embryonic lethality.67

    More subtle changes in the HS chain composition also have profound effects. Sulfation of 2-O and 6-O HS sites is thought to be required for formation of FGF-binding sites. A homozygous deletion of 2-OST results in mice that survive until birth but die perinatally because of the complete failure of kidney formation.6 Similarly, a knockdown of the 6-O-sulfotransferase in Drosophila severely perturbs tracheal development, an FGF-dependent process.68 Deletions of other genes involved in HS biosynthesis have equally profound phenotypic effects.69

    In addition to HS-dependent signaling, syndecans also clearly can engage in proteineCprotein interactions via their protein core ectodomains, an event that can also initiate intracellular signaling, as is discussed below. Furthermore, syndecaneCgrowth factor interaction, whether accomplished via the chain or the core-based interaction, does not simply serve to present the factor to its high-affinity receptor. Rather, such binding can initiate signaling events via the cytoplasmic syndecan domains that have their own unique aspects, as is further discussed. Finally, each of the syndecans has its own characteristic biology that we briefly review.

    Syndecan-1 and CelleCMatrix Interaction

    Syndecan-1 is an important regulator of celleCcell and celleCextracellular matrix interactions. Downregulation of its expression in epithelial cells by antisense mRNA results in loss of cell polarity associated with a reduced level of E-cadherin on the cell surface, suggesting involvement in the epithelialeCmesenchymal switch during development and in wound healing.70 Overexpression of syndecan-1 or shedding of its ectodomain inhibits FGF2-induced cell proliferation.71 Mice overexpressing syndecan-1 have delayed dermal wound repair because of the inhibitory effect of soluble syndecan-1 ectodomain.72 However, under certain circumstances, heparinases can convert syndecan-1 ectodomain from an inhibitor to an activator of FGF2.73 This reaction enables FGF2 interaction with syndecan-4 (see below), although a specific protein sequence in the syndecan-1 ectodomain may also play a role.74 The ability of syndecan-1 overexpression to inhibit cell growth and migration could be an explanation of the aggressive invasive behavior of tumor cells lacking syndecan-1.75,76

    Interestingly, downregulation of syndecan-1 expression in carcinoma cells results in impaired cell spreading on vitronectin but not on fibronectin.77 This seems to be mediated by the ability of syndecan-1 to modulate vitronectin interaction with its 3 integrin receptor because glycosylphosphatidylinositol (GPI)eCsyndecan-1 extracellular domain construct expression in syndecan-1eC/eC cells converts their "no spreading phenotype" to normal spreading.

    Syndecan-1 plays an important role in regulation of inflammation,78 as suggested by increased leukocyteeCendothelial interactions in syndecan-1 null mice.79 It also plays an important role in chemokine gradient formation for trans-endothelial and trans-epithelial migration of neutrophils.78 The former process is facilitated by an IL-8/syndecan-1 complex and regulated by shedding of syndecan-1.80 Trans-epithelial neutrophil migration is dependent on matrilysin-mediated shedding of syndecan-1 from the mucosal surface of epithelium.81 As would be expected, matrix-metalloproteinase matrilysineCdeficient mice demonstrate impaired trans-epithelial migration of neutrophils.

    Finally, syndecan-1 may also play a role in regulation of ephrin signaling that is involved in venoarterial differentiation. Ephrin-B4 (EphB4) is expressed in venule endothelium. Homozygous disruption of EphB4 or one of its ligands, ephrin-B2, results in fatal vascular defects.82,83 Activation of EphB4-positive endothelial cells causes upregulation of syndecan-1,84 resulting in suppression of FGF2 signaling. However, in certain settings, the role of syndecan-1 could be the opposite, depending on the presence of heparinases. Degradation of heparan by platelet heparanase, for example, produces heparin-like HS fragments, capable of promoting FGF2 mitogenicity.73

    Syndecan-2 and TGF- Signaling

    Syndecan-2 is the predominant syndecan expressed during embryonic development. Although its role in adult cells has not been well established, recent studies suggest its involvement in regulating TGF- signaling. Whereas TGF- can bind to HS chains, syndecan-2, but not other syndecans, binds TGF- directly via a proteineCprotein interaction. Furthermore, syndecan-2 coimmunoprecipitates with the TGF- as well as with the type III TGF- receptor (betaglycan),85 and expression of a syndecan-2 mutant with a truncated cytoplasmic domain results in impaired response to TGF-.85 These data correlate with a previously reported inability of cells overexpressing this construct to assemble laminin or fibronectin into a fibrillar matrix.86

    The mechanism of syndecan-2eCTGF- interactions is complex and not entirely clear (Figure 3). TGF- binding to betaglycan transfers it to the type II receptor, which undergoes autophosphorylation and then trans-phosphorylates betaglycan and the type I receptor. The phosphorylated type I/type II receptor complex then engages in downstream signaling. Betaglycan binds to synectin (GAIP-C terminus protein) that retains betaglycan on the cell surface, thus preventing its degradation.87 However, synectin also binds syndecan-2. As already mentioned, expression of a syndecan-2 mutant with a truncated cytoplasmic domain increases plasma membrane betaglycan levels.85 Whereas an increase in syndecan-2 protein increases type I and type II TGF- receptor expression and decreases betaglycan, probably as the consequence of competition for synectin binding,85 this decrease in betaglycan expression possibly leads to disregulated activity of the type-I/type-II TGF- receptor complex.

    Similar to syndecan-1, syndecan-2 may also play a role in ephrin signaling. Cell surface ephrineCEph signaling induces clustering of syndecan-2 and recruitment of cytoplasmic molecules, which leads to localized actin polymerization via Rho family GTPases, N-Wiscott-Aldrich syndrome protein, and the Arp2/3 complex.88

    Syndecan-3 and Growth Factor Signaling

    Syndecan-3 plays an important role in regulation of skeletal muscle differentiation and development. Its levels are transiently elevated in the developing limb bud but are absent in adult skeletal muscle. Skeletal muscle myoblasts are held in an undifferentiated state until they receive signals to further differentiate. The maintenance of these cells in their undifferentiated state has been shown to be controlled by specific growth factors, including FGF2, HGF/scatter factor, and TGF-.89 Syndecan-3 inhibition results in expression of myogenin, a master transcription factor for muscle differentiation, and in accelerated skeletal muscle differentiation and myoblast fusion.90

    In the model of skeletal muscle regeneration induced by barium chloride injection, syndecan-3 showed the earliest and largest increase in satellite cells.91 Myoblast grafting of C2C12 cells transfected with antisense syndecan-3 resulted in cells with a normal proliferation rate but defective in fusion and formation of skeletal muscle fibers. Further, examination of syndecan-3 null mice demonstrates mislocalization of MyoD (a myocyte differentiation transcription factor) and an aberrant differentiation of skeletal muscle.50

    Syndecan-3 also appears to be involved in regulation of Hh signaling. Hh is a family of secreted signaling proteins that play an important role in embryogenesis and differentiation. One of the mammalian family of Hh members, indian Hh (iHh), is produced by prehypertrophic chondrocytes and regulates chondrocyte proliferation. Proper spatial and temporal regulation of iHh is regulated by syndecan-3.92

    Syndecan-4, FGF Signaling, and the Extracellular Matrix

    Syndecan-4 has become the most extensively studied member of the syndecan family. The proteoglycan has a number of activities, including modulation of FGF2 signaling, regulation of cell migration via cross-talk with 1 integrin, and control of adhesion via cytoskeletal modifications. All these various events are achieved via actions of a 33-aa cytoplasmic tail, one of the most overworked small proteins among the signaling giants.

    FGF Signaling and Endocytosis

    In addition to being able to bind FGF2 to HS chains and to present it to FGF tyrosine kinase receptors, syndecan-4 directly initiates a number of intracellular signaling events. The most direct demonstration of syndecan-4 signaling came from studies of syndecan-4/glypican chimeras in endothelial cells. Expression of a full-length syndecan-4 (S4), syndecan-1 (S1), glypican-1 (G1), or of chimera constructs consisting of the ectoplasmic domain of G1 linked to the transmembrane/cytoplasmic domain of syndecan-4 (G1-S4c) or of the ectoplasmic domain of syndecan-4 linked to the G1 GPI anchor (S4-GPI), significantly increased cell-associated HS mass and the number of low-affinity FGF2-binding sites. However, only cells expressing S4 and G1-S4c constructs but not G1, S1, or S4-GPI cells demonstrated enhanced responsiveness to FGF2. Thus, the presence of syndecan-4 cytoplasmic domain and not simply an increase in the cell surface HS mass with a corresponding increase in the number of the low-affinity FGF2-binding sites is required for signaling. However, removal of the syndecan-4 HS chains also blocked enhanced FGF2 responsiveness, demonstrating that syndecan-4 HS chains and the syndecan-4 cytoplasmic domain are required for FGF2 signaling.93 Syndecan-3 cannot replace syndecan-4 to promote FGF2- and HGF-induced cell migration.50

    Additional evidence of direct syndecan-4 signaling ability comes from studies in vascular smooth muscle cells expressing dominant-negative FGF receptor. FGF2 was able to transiently stimulate Erk 1/2 activation and induce cell migration, activities that were inhibited by removal of HS chains.94 It should be noted that FGF2-induced Erk activation in the absence of functional FGF receptor 1 is transient and does not lead to cell proliferation.

    The final piece of evidence demonstrating syndecan-4 modulation of FGF2 signaling comes from studies that explored the effect of dominant-negative syndecan-4 construct expression on endothelial cell responses to FGF2 and other growth factors. Introduction of syndecan-4 constructs with the mutated PDZ- or PIP2-binding regions that respectively eliminated syndecan-4 ability to bind PDZ proteins or PIP2 inhibited FGF2- but not serum-induced endothelial cell growth, migration, and the ability to form vascular structures on Matrigel.27

    Although it is clear that syndecan-4 can modulate FGF2 signaling, the precise mechanism of this modulation is uncertain. A sustained activation of FGF2 signaling requires internalization and perhaps nuclear transport of the ligand.95 Syndecan-4 is directly involved in FGF2 endocytosis that proceeds in a clathrin- and dynamin-independent fashion and requires syndecan-4eCdependent activation of Rac1.96 The sensitivity of this process to amiloride as well as colocalization of internalized FGF2 with dextran suggests that FGF2 uptake proceeds via macropinocytosis.96 An interesting feature of this process is the fact that FGF2 uptake proceeds from lipid rafts, a specialized plasma membrane domain thought to be involved in signaling.

    Endocytosis, Lipid Rafts, and Caveolae

    Plasma membrane location of syndecan-4 and its participation in FGF2 endocytosis raises the issue of its relation to lipid rafts, plasma membrane domains that provide fluid platforms to segregate membrane components and dynamically compartmentalize membranes.97 Those phospholipid- and cholesterol-rich domains contain specific sets of proteins that include GPI-anchored proteins, doubly acetylated proteins (Src family tyrosine kinases), G subunit of heterotrimeric G-proteins, and palmitylated proteins including endothelial NO synthase.98,99

    Whereas in unstimulated cells, syndecan-4 is predominantly present in the nonraft compartments, clustering with FGF2 or antieCsyndecan-4 antibody induces its shift to lipid rafts.100 This shift is necessary for the initiation of FGF2 endocytosis because its blockade by cholesterol depletion from the plasma cell membrane completely blocks FGF2 uptake.96 Similarly, low-density lipoprotein (LDL) causes clustering and clathrin-independent uptake of syndecan-1, syndecan-2, and syndecan-4.101 In the model in which syndecan-1 was used, LDL clustering also induced a shift of syndecan into the lipid rafts.102

    Caveolae are a subset of lipid rafts. They are flask-shaped invaginations of the plasma membrane103 that play a role in regulation of certain endocytic pathways and regulation of enzymes such as endothelial NO synthase. A variety of protein- and lipid-signaling molecules are concentrated in caveolae. These include PKC, Rho GTPases, and nonreceptor tyrosine kinases such as c-Src, Yes, Fyn, phosphatidylinositol 3-kinase, and phosphatidylinositol kinases, among others.103eC105 Caveolins are the principal building blocks of caveolae and are usually accepted as its marker. Caveolin-1 oligomeric complex creates a stable membrane structure that transiently interacts with plasma membrane and endosomes.106

    On initiation of cell migration, pools of syndecan-1, syndecan-4, and caveolin-1 are directed into the same retracting region of the moving cell.107eC109 The caveolin property of inhibiting kinase activity of a variety of proteins involved in leading edge formation was speculated to play a key role in cell migration.110 In contrast to the deactivating role of caveolin, syndecan-4 is needed for activation of several kinases, including focal adhesion kinase (FAK), which results in increased turnover of focal adhesions.111eC113 Silencing of caveolin-1 gene expression or introduction of a dominant-negative mutant of syndecan-4 leads to slow migration and impaired tube formation of endothelial cells on Matrigel.27,107 Syndecan-4 and caveolin-1 knockout mice demonstrate reduced postnatal angiogenesis that may be related to the impairment of endothelial cell migration.61,110

    Whereas syndecan-4 does not colocalize with caveolin-1 at the cell surface, syndecan-4eC and caveolin-1eCcontaining vesicles frequently move in tandem, presumably along microtubules.100 Remarkably, internalization of caveolae depends on Src and PKC activation.114 The ability of syndecan-4 to activate PKC is well established and is discussed in detail below. Similar to syndecan-3, it can also activate Src.17 Thus, syndecan-4 may be responsible for modulation of caveolae, pinching from the plasma membrane in response to heparin-binding growth factors.115,116

    The association of syndecan-4 with fast-moving caveolin-1eCpositive vesicles could also be used for a fast delivery of syndecan-4 pinosomal cargo to intracellular destinations. This process might be needed for proper distribution of macropinosome-associated small GTPases, leading to migratory polarization of the cell.117,118 This interaction could also be involved in extracellular matrix turnover. Downregulation of caveolin-1 results in inhibition of fibronectin internalization and degradation,119 suggesting the participation of fibronectin receptors such as integrins or syndecans in caveolae-dependent uptake of fibronectin.

    Activation of PKC

    Another feature of syndecan-4 signaling is activation of PKC in the absence of Ca2+. This unique ability of syndecan-4 to activate a calcium-dependent PKC was first shown by Oh et al to require core protein multimerization and the presence of PIP2.15,120 Syndecan-4 oligomerization in the presence of PIP2 depends on the phosphorylation status of the Ser183 site in its cytoplasmic domain.27 Phosphorylation of this site markedly reduces its affinity for PIP226 and prevents PKC activation in vitro and in vivo.26,121 Interestingly, the Ser183 site is phosphorylated by PKC.28 This sets up a system that allows one PKC to regulate activity of another (Figure 4). The Ser dephosphorylation is performed by as yet undefined type I/IIa serine phosphatase.121

    The molecular details of syndecan-4eCdependent PKC activation have not been established. The results of yeast two hybrid screening demonstrated that although a full-length PKC weakly binds the V domain of syndecan-4, the PKC constructs that lack the pseudosubstrate region or that encode only the whole catalytic domain interacted more strongly.15,122 A mutation of Tyr192 in the syndecan-4 cytoplasmic domain abolished this interaction.122 Interestingly, the corresponding binding site in the catalytic domain of PKC (amino acid sequence 513 to 672) encompasses the regulatory autophosphorylation sites implicated in control of PKC activation and stability.122 On the other hand, in vitro surface plasmon resonance studies suggested that binding affinity of a full-length PKC to the syndecan-4 cytoplasmic domain is very low in the absence of PIP2.26

    Although the precise role of syndecan-4eCdependent PKC activation has not been defined, colocalization of the two proteins in focal adhesions120 suggests a role in cell adhesion, spreading, and stress fiber formation. Studies in cultured fibroblasts have shown that overexpression of syndecan-4, but not a mutant lacking its cytoplasmic domain, specifically increases the level of endogenous PKC and enhances the translocation of PKC in the plasma cell membrane in general and in the membrane raft fractions in particular, and increases the activity of membrane PKC.123

    Cell Adhesion and the Cytoskeleton

    The involvement of members of the syndecan family in regulation of cell adhesion suggests that these proteins likely modulate cytoskeletal rearrangements. Indeed, fibronectin binds to syndecan-4 HS chains, and this interaction plays an important role in regulation of cell adhesion and spreading.124 Fibroblasts use integrin receptors and syndecan-4 to induce Rho-dependent spreading in fibronectin.125,126 Overexpression of syndecan-4 results in the flattening of Chinese hamster ovary K1 (CHO-K1) cells, promotion of focal adhesion formation, and a decrease in cell migration.127 Syndecan-4 null fibroblasts plated on fibronectin exhibit enhanced lamellipodia formation and increased level of Rac1128 and low Rho111 activities compared with wild-type cells. Expression of syndecan-4 in these knockout cells downregulates Rac1 activity.128 In endothelial cells also plated on fibronectin, clustering of syndecan-4 construct by antibody results in upregulation of Rac1 activity.96 Those seemingly contrary results might be attributable to the differences in localization changes followed by binding to matrix versus soluble ligand. In this way, growth factors might modulate celleCmatrix interactions by removal of the syndecan component via endocytosis.96

    In vascular smooth muscle cells, shear stress causes syndecan-4 dissociation from the focal adhesions.39 Overexpression of syndecan-4 blocks this dissociation and also results in reduced mechanical stress-induced cell migration. This observation suggests that syndecan-4 might be involved in regulation of smooth muscle migration during arteriogenesis.

    Another role of syndecan-4 involves regulation of fibronectin signaling and matrix contraction together with tenascin-C. Tenascin-C is an extracellular matrix protein involved in regulation of cellular response to fibronectin by preventing cell spreading.129 Syndecan-4 knockout fibroblasts, while spreading normally on a 2D matrix but failing to spread in a 3D fibronectin matrix, do not activate RhoA and, consequently, do not form focal adhesions and stress fibers. AntieCsyndecan-4 antibody treatment of wild-type fibroblasts similarly prevents spreading on fibronectin, decreases RhoA, and activates FAK. Stimulation of RhoA by lysophosphatidic acid in syndecan-4eC/eC cells rescues syndecan functions by activating RhoA and inducting cell spreading and matrix contraction. Addition of tenascin-C blocks spreading of control but not syndecan-4eC/eC fibroblasts on a 2D fibronectin matrix, whereas syndecan-4 overexpression overcomes this tenascin-C effect.130

    Activated B lymphocytes, when seeded on syndecan-4 antibodies, exhibit dramatic morphological changes,131 the most profound being filopodial extensions, implying the possibility of CDC42 activation. Overexpression of a truncated form of syndecan-4 lacking an intracellular domain demonstrated that the extracellular domain is sufficient to generate a response, indicating the need for a transmembrane partner to transmit signal. One such potential partner is the chemokine receptor 4 (CXCR4). Syndecan-4, but neither syndecan-1 nor syndecan-2, coimmunoprecipitate with CXCR4.132 This syndecan-4/CXCR4 complex is likely a functional unit involved in chemokine stromal celleCderived factor-1 (SDF-1) binding that may play a role in SDF-1eCdependent stem cell recruitment.

    Syndecan-2 and syndecan-3 overexpression also induce filopodia formation in COS-1 and CHO-K1 cells.133,134 In the case of syndecan-2, this process is CDC42 dependent.133,134 The deletion of V or C2 domains, but not entire cytoplasmic part of syndecan-3, results in more filopodia formation, whereas extensive membrane blebbing is observed in cells expressing a C2-truncated mutant.134 The blebs are distributed uniformly over the cell surface rather than next to the leading edge, suggesting improper targeting or activation of Rac1 resulted to bleb formations away from cell edges as a possible mechanism.

    Members of the ezrin-radixin-moesin family have NH2- and C-terminal domains that associate with the plasma membrane and the actin cytoskeleton, respectively. After overexpression of a constitutively active form of RhoA, association of syndecan-2 with actin cytoskeleton through ezrin binding was observed in COS-1 cells.16,18 Thus, ezrin provides Rho GTPases a regulated link between syndecan-2 and actin. The ezrin-binding motif is identical between syndecan-2 and syndecan-4, implying the possibility of syndecan-4eCezrin interactions (Figure 1).18 Such regulation of cytoskeleton by Rho GTPases promises to be one of the most important tuning functions of syndecans.

    Summary

    As we hope this review demonstrates, syndecans have been shown to function as potent and specific regulators of cell signaling in a variety of settings. This regulatory function is very complex because of the involvement of different family members, differences in level and cellular location of expression, GAG chain modifications, and extracellular domain shedding. Overall, given their ability to participate in signal transduction involving the extracellular matrix, cell surface molecules, and soluble ligands, syndecans are emerging as conductors of the entire signaling orchestra.

    Acknowledgments

    This work was supported in part by National Institutes of Health grants HL62289 and HL63609 (M.S.). We would like to thank members of the Simons’ laboratory and the Angiogenesis Research Center colleagues for helpful discussions and incisive comments. Certain areas of syndecans signaling dealing with their role in the CNS have been omitted in this review. We also would like to apologize to authors whose primary articles were not included because of space limitations.

    References

    Zimmermann P, David G. The syndecans, tuners of transmembrane signaling. FASEB J. 1999; 13: S91eCS100.

    Deepa SS, Yamada S, Zako M, Goldberger O, Sugahara K. Chondroitin sulfate chains on syndecan-1 and syndecan-4 from normal murine mammary gland epithelial cells are structurally and functionally distinct and cooperate with heparan sulfate chains to bind growth factors. A novel function to control binding of midkine, pleiotrophin, and basic fibroblast growth factor. J Biol Chem. 2004; 279: 37368eC37376.

    Okamoto O, Bachy S, Odenthal U, Bernaud J, Rigal D, Lortat-Jacob H, Smyth N, Rousselle P. Normal human keratinocytes bind to the alpha3LG4/5 domain of unprocessed laminin-5 through the receptor syndecan-1. J Biol Chem. 2003; 278: 44168eC44177.

    Varki A, Cummings R, Esko JD, Freeze H, Hart G, Marth J, eds. Essentials of Glycobiology. 1st ed. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press; 1999.

    Esko JD, Selleck SB. Order out of chaos: assembly of ligand binding sites in heparan sulfate. Annu Rev Biochem. 2002; 71: 435eC471.

    Wilson VA, Gallagher JT, Merry CL. Heparan sulfate 2-O-sulfotransferase (Hs2st) and mouse development. Glycoconj J. 2002; 19: 347eC354.

    Li J, Shworak NW, Simons M. Increased responsiveness of hypoxic endothelial cells to FGF2 is mediated by HIF-1alpha-dependent regulation of enzymes involved in synthesis of heparan sulfate FGF2-binding sites. J Cell Sci. 2002; 115: 1951eC1959.

    Morimoto-Tomita M, Uchimura K, Werb Z, Hemmerich S, Rosen SD. Cloning and characterization of two extracellular heparin-degrading endosulfatases in mice and humans. J Biol Chem. 2002; 277: 49175eC49185.

    Ohto T, Uchida H, Yamazaki H, Keino-Masu K, Matsui A, Masu M. Identification of a novel nonlysosomal sulphatase expressed in the floor plate, choroid plexus and cartilage. Genes Cells. 2002; 7: 173eC185.

    Lai J, Chien J, Staub J, Avula R, Greene EL, Matthews TA, Smith DI, Kaufmann SH, Roberts LR, Shridhar V. Loss of HSulf-1 up-regulates heparin-binding growth factor signaling in cancer. J Biol Chem. 2003; 278: 23107eC23117.

    Ai X, Do AT, Lozynska O, Kusche-Gullberg M, Lindahl U, Emerson CP Jr. QSulf1 remodels the 6-O sulfation states of cell surface heparan sulfate proteoglycans to promote Wnt signaling. J Cell Biol. 2003; 162: 341eC351.

    Lai JP, Chien J, Strome SE, Staub J, Montoya DP, Greene EL, Smith DI, Roberts LR, Shridhar V. HSulf-1 modulates HGF-mediated tumor cell invasion and signaling in head and neck squamous carcinoma. Oncogene. 2004; 23: 1439eC1447.

    Shworak NW, Rosenberg RD. Heparan sulfate proteoglycans. In: Ware JA, Simons M, eds. Angiogenesis and Cardiovascular Disease. New York, NY: Oxford University Press; 1999: 60eC78.

    McFall AJ, Rapraeger AC. Identification of an adhesion site within the syndecan-4 extracellular protein domain. J Biol Chem. 1997; 272: 12901eC12904.

    Oh ES, Woods A, Couchman JR. Multimerization of the cytoplasmic domain of syndecan-4 is required for its ability to activate protein kinase C. J Biol Chem. 1997; 272: 11805eC11811.

    Granes F, Urena J, Rocamora N, Vilaro S. Ezrin links syndecan-2 to the cytoskeleton. J Cell Sci. 2000; 113: 1267eC1276.

    Kinnunen T, Kaksonen M, Saarinen J, Kalkkinen N, Peng HB, Rauvala H. Cortactin-Src kinase signaling pathway is involved in N-syndecan- dependent neurite outgrowth. J Biol Chem. 1998; 273: 10702eC10708.

    Granes F, Berndt C, Roy C, Mangeat P, Reina M, Vilaro S. Identification of a novel Ezrin-binding site in syndecan-2 cytoplasmic domain. FEBS Lett. 2003; 547: 212eC216.

    Gao Y, Li M, Chen W, Simons M. Synectin, syndecan-4 cytoplasmic domain binding PDZ protein, inhibits cell migration. J Cell Physiol. 2000; 184: 373eC379.

    Grootjans JJ, Zimmermann P, Reekmans G, Smets A, Degeest G, Durr J, David G. Syntenin, a PDZ protein that binds syndecan cytoplasmic domains. Proc Natl Acad Sci U S A. 1997; 94: 13683eC13688.

    Cohen AR, Wood DF, Marfatia SM, Walther Z, Chishti AH, Anderson JM. Human CASK/LIN-2 Binds Syndecan-2 and Protein 4.1 and Localizes to the Basolateral Membrane of Epithelial Cells. J Cell Biol. 1998; 142: 129eC138.

    Hsueh YP, Yang FC, Kharazia V, Naisbitt S, Cohen AR, Weinberg RJ, Sheng M. Direct interaction of CASK/LIN-2 and syndecan heparan sulfate proteoglycan and their overlapping distribution in neuronal synapses. J Cell Biol. 1998; 142: 139eC151.

    Ethell IM, Hagihara K, Miura Y, Irie F, Yamaguchi Y. Synbindin, a novel syndecan-2-binding protein in neuronal dendritic spines. J Cell Biol. 2000; 151: 53eC68.

    Shin J, Lee W, Lee D, Koo BK, Han I, Lim Y, Woods A, Couchman JR, Oh ES. Solution structure of the dimeric cytoplasmic domain of syndecan-4. Biochemistry. 2001; 40: 8471eC8478.

    Horowitz A, Simons M. Phosphorylation of the cytoplasmic tail of syndecan-4 regulates activation of protein kinase C alpha. J Biol Chem. 1998; 273: 25548eC25551.

    Horowitz A, Murakami M, Gao Y, Simons M. Phosphatidylinositol-4,5-bisphosphate mediates the interaction of syndecan-4 with protein kinase C. Biochemistry. 1999; 38: 15871eC15877.

    Horowitz A, Tkachenko E, Simons M. Fibroblast growth factor-specific modulation of cellular response by syndecan-4. J Cell Biol. 2002; 157: 715eC725.

    Murakami M, Horowitz A, Tang S, Ware JA, Simons M. PKC-delta regulates PKC-alpha activity In a syndecan-4 dependent manner. J Biol Chem. 2002; 277: 20367eC20371.

    Oh ES, Woods A, Lim ST, Theibert AW, Couchman JR. Syndecan-4 proteoglycan cytoplasmic domain and phosphatidylinositol 4,5- bisphosphate coordinately regulate protein kinase C activity. J Biol Chem. 1998; 273: 10624eC10629.

    Greene DK, Tumova S, Couchman JR, Woods A. Syndecan-4 associates with alpha-actinin. J Biol Chem. 2003; 278: 7617eC7623.

    Baciu PC, Saoncella S, Lee SH, Denhez F, Leuthardt D, Goetinck PF. Syndesmos, a protein that interacts with the cytoplasmic domain of syndecan-4, mediates cell spreading and actin cytoskeletal organization. J Cell Sci. 2000; 113: 315eC324.

    Denhez F, Wilcox-Adelman SA, Baciu PC, Saoncella S, Lee S, French B, Neveu W, Goetinck PF. Syndesmos, a syndecan-4 cytoplasmic domain interactor, binds to the focal adhesion adaptor proteins paxillin and Hic-5. J Biol Chem. 2002.

    Lee D, Oh ES, Woods A, Couchman JR, Lee W. Solution structure of a syndecan-4 cytoplasmic domain and its interaction with phosphatidylinositol 4,5-bisphosphate. J Biol Chem. 1998; 273: 13022eC13029.

    Bernfield M, Kokenyesi R, Kato M, Hinkes MT, Spring J, Gallo RL, Lose EJ. Biology of the syndecans: a family of transmembrane heparan sulfate proteoglycans. Annu Rev Cell Biol. 1992; 8: 365eC393.

    Couchman JR. Syndecans: proteoglycan regulators of cell-surface microdomains Nat Rev Mol Cell Biol. 2003; 4: 926eC937.

    Halden Y, Rek A, Atzenhofer W, Szilak L, Wabnig A, Kungl AJ. Interleukin-8 binds to syndecan-2 on human endothelial cells. Biochem J. 2004; 377: 533eC538.

    Dobra K, Nurminen M, Hjerpe A. Growth factors regulate the expression profile of their syndecan co-receptors and the differentiation of mesothelioma cells. Anticancer Res. 2003; 23: 2435eC2444.

    Cizmeci-Smith G, Langan E, Youkey J, Showalter LJ, Carey DJ. Syndecan-4 is a primary-response gene induced by basic fibroblast growth factor and arterial injury in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 1997; 17: 172eC180.

    Li L, Chaikof EL. Mechanical stress regulates syndecan-4 expression and redistribution in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2002; 22: 61eC68.

    Geary RL, Koyama N, Wang TW, Vergel S, Clowes AW. Failure of heparin to inhibit intimal hyperplasia in injured baboon arteries. The role of heparin-sensitive and -insensitive pathways in the stimulation of smooth muscle cell migration and proliferation. Circulation. 1995; 91: 2972eC2981.

    Li J, Brown LF, Laham RJ, Volk R, Simons M. Macrophage-dependent regulation of syndecan gene expression. Circ Res. 1997; 81: 785eC796.

    Gallo R, Kim C, Kokenyesi R, Adzick NS, Bernfield M. Syndecans-1 and -4 are induced during wound repair of neonatal but not fetal skin. J Invest Dermatol. 1996; 107: 676eC683.

    Oksala O, Salo T, Tammi R, Hakkinen L, Jalkanen M, Inki P, Larjava H. Expression of proteoglycans and hyaluronan during wound healing. J Histochem Cytochem. 1995; 43: 125eC135.

    Gallo RL, Ono M, Povsic T, Page C, Eriksson E, Klagsburn M, Bernfield M. Syndecans, cell surface heparan sulfate proteoglycans, are induced by a proline-rich antimicrobial peptide from wounds. Proc Natl Acad Sci U S A. 1994; 91: 11035eC11039.

    Li J, Post M, Volk R, Gao Y, Li M, Metais C, Sato K, Tsai J, Aird W, Rosenberg RD, Hampton TG, Sellke F, Carmeliet P, Simons M. PR39, a peptide regulator of angiogenesis. Nat Med. 2000; 6: 49eC55.

    Weiner OH, Zoremba M, Gressner AM. Gene expression of syndecans and betaglycan in isolated rat liver cells. Cell Tissue Res. 1996; 285: 11eC16.

    Carey DJ, Conner K, Asundi VK, O‘Mahony DJ, Stahl RC, Showalter L, Cizmeci-Smith G, Hartman J, Rothblum LI. cDNA cloning, genomic organization, and in vivo expression of rat N-syndecan. J Biol Chem. 1997; 272: 2873eC2879.

    Song HK, Lee SH, Goetinck PF. FGF-2 signaling is sufficient to induce dermal condensations during feather development. Dev Dyn. 2004; 231: 741eC749.

    Gould SE, Upholt WB, Kosher RA. Characterization of chicken syndecan-3 as a heparan sulfate proteoglycan and its expression during embryogenesis. Dev Biol. 1995; 168: 438eC451.

    Cornelison DD, Wilcox-Adelman SA, Goetinck PF, Rauvala H, Rapraeger AC, Olwin BB. Essential and separable roles for syndecan-3 and syndecan-4 in skeletal muscle development and regeneration. Genes Dev. 2004; 18: 2231eC2236.

    Stepp MA, Gibson HE, Gala PH, Iglesia DD, Pajoohesh-Ganji A, Pal-Ghosh S, Brown M, Aquino C, Schwartz AM, Goldberger O, Hinkes MT, Bernfield M. Defects in keratinocyte activation during wound healing in the syndecan-1-deficient mouse. J Cell Sci. 2002; 115: 4517eC4531.

    Reizes O, Benoit SC, Strader AD, Clegg DJ, Akunuru S, Seeley RJ. Syndecan-3 modulates food intake by interacting with the melanocortin/AgRP pathway. Ann N Y Acad Sci. 2003; 994: 66eC73.

    Alexander CM, Reichsman F, Hinkes MT, Lincecum J, Becker KA, Cumberledge S, Bernfield M. Syndecan-1 is required for Wnt-1-induced mammary tumorigenesis in mice. Nat Genet. 2000; 25: 329eC332.

    Chen E, Hermanson S, Ekker SC. Syndecan-2 is essential for angiogenic sprouting during zebrafish development. Blood. 2004; 103: 1710eC1719.

    Kramer KL, Barnette JE, Yost HJ. PKC-gamma regulates syndecan-2 inside-out signaling during xenopus left-right development. Cell. 2002; 111: 981eC990.

    Reizes O, Lincecum J, Wang Z, Goldberger O, Huang L, Kaksonen M, Ahima R, Hinkes MT, Barsh GS, Rauvala H, Bernfield M. Transgenic expression of syndecan-1 uncovers a physiological control of feeding behavior by syndecan-3. Cell. 2001; 106: 105eC116.

    Ishiguro K, Kojima T, Muramatsu T. Syndecan-4 as a molecule involved in defense mechanisms. Glycoconj J. 2002; 19: 315eC318.

    Ishiguro K, Kadomatsu K, Kojima T, Muramatsu H, Nakamura E, Ito M, Nagasaka T, Kobayashi H, Kusugami K, Saito H, Muramatsu T. Syndecan-4 deficiency impairs the fetal vessels in the placental labyrinth. Dev Dyn. 2000; 219: 539eC544.

    Ishiguro K, Kadomatsu K, Kojima T, Muramatsu H, Iwase M, Yoshikai Y, Yanada M, Yamamoto K, Matsushita T, Nishimura M, Kusugami K, Saito H, Muramatsu T. Syndecan-4 deficiency leads to high mortality of lipopolysaccharide-injected mice. J Biol Chem. 2001; 276: 47483eC47488.

    Ishiguro K, Kadomatsu K, Kojima T, Muramatsu H, Matsuo S, Kusugami K, Saito H, Muramatsu T. Syndecan-4 deficiency increases susceptibility to kappa-carrageenan- induced renal damage. Lab Invest. 2001; 81: 509eC516.

    Echtermeyer F, Streit M, Wilcox-Adelman S, Saoncella S, Denhez F, Detmar M, Goetinck P. Delayed wound repair and impaired angiogenesis in mice lacking syndecan- 4. J Clin Invest. 2001; 107: R9eCR14.

    Nugent MA, Edelman ER. Kinetics of basic fibroblast growth factor binding to its receptor and heparan sulfate proteoglycan: a mechanism for cooperactivity. Biochemistry. 1992; 31: 8876eC8883.

    Chu CL, Buczek-Thomas JA, Nugent MA. Heparan sulphate proteoglycans modulate fibroblast growth factor-2 binding through a lipid raft-mediated mechanism. Biochem J. 2004; 379: 331eC341.

    Sasisekharan R, Moses MA, Nugent MA, Cooney CL, Langer R. Heparinase inhibits neovascularization. Proc Natl Acad Sci U S A. 1994; 91: 1524eC1528.

    Duncan G, McCormick C, Tufaro F. The link between heparan sulfate and hereditary bone disease: finding a function for the EXT family of putative tumor suppressor proteins. J Clin Invest. 2001; 108: 511eC516.

    Bellaiche Y, The I, Perrimon N. Tout-velu is a Drosophila homologue of the putative tumor suppressor EXT-1 and is needed for Hh diffusion. Nature. 1998; 394: 85eC88.

    Lin X, Wei G, Shi Z, Dryer L, Esko JD, Wells DE, Matzuk MM. Disruption of gastrulation and heparan sulfate biosynthesis in EXT1-deficient mice. Dev Biol. 2000; 224: 299eC311.

    Kamimura K, Fujise M, Villa F, Izumi S, Habuchi H, Kimata K, Nakato H. Drosophila heparan sulfate 6-O-sulfotransferase (dHS6ST) gene. Structure, expression, and function in the formation of the tracheal system. J Biol Chem. 2001; 276: 17014eC17021.

    Lee JS, Chien CB. When sugars guide axons: insights from heparan sulphate proteoglycan mutants. Nat Rev Genet. 2004; 5: 923eC935.

    Couchman JR, Chen L, Woods A. Syndecans and cell adhesion. Int Rev Cytol. 2001; 207: 113eC150.

    Mali M, Elenius K, Miettinen HM, Jalkanen M. Inhibition of basic fibroblast growth factor-induced growth promotion by overexpression of syndecan-1. J Biol Chem. 1993; 268: 24215eC24222.

    Elenius V, Gotte M, Reizes O, Elenius K, Bernfield M. Inhibition by the soluble syndecan-1 ectodomains delays wound repair in mice overexpressing syndecan-1. J Biol Chem. 2004; 279: 41928eC41935.

    Kato M, Wang H, Kainulainen V, Fitzgerald ML, Ledbetter S, Ornitz DM, Bernfield M. Physiological degradation converts the soluble syndecan-1 ectodomain from an inhibitor to a potent activator of FGF-2. Nat Med. 1998; 4: 691eC697.

    Langford JK, Yang Y, Kieber-Emmons T, Sanderson RD. Identification of an invasion regulatory domain within the core protein of syndecan-1. J Biol Chem. 2004: M412451200.

    Harada K, Masuda S, Hirano M, Nakanuma Y. Reduced expression of syndecan-1 correlates with histologic dedifferentiation, lymph node metastasis, and poor prognosis in intrahepatic cholangiocarcinoma. Hum Pathol. 2003; 34: 857eC863.

    Reiland J, Sanderson RD, Waguespack M, Barker SA, Long R, Carson DD, Marchetti D. Heparanase degrades syndecan-1 and perlecan heparan sulfate: functional implications for tumor cell invasion. J Biol Chem. 2004; 279: 8047eC8055.

    Beauvais DM, Burbach BJ, Rapraeger AC. The syndecan-1 ectodomain regulates alphavbeta3 integrin activity in human mammary carcinoma cells. J Cell Biol. 2004; 167: 171eC181.

    Gotte M. Syndecans in inflammation. FASEB J. 2003; 17: 575eC591.

    Gotte M, Joussen AM, Klein C, Andre P, Wagner DD, Hinkes MT, Kirchhof B, Adamis AP, Bernfield M. Role of syndecan-1 in leukocyte-endothelial interactions in the ocular vasculature. Invest Ophthalmol Vis Sci. 2002; 43: 1135eC1141.

    Marshall LJ, Ramdin LS, Brooks T, PC DP, 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: 2057eC2065.

    Li Q, Park PW, Wilson CL, Parks WC. Matrilysin shedding of syndecan-1 regulates chemokine mobilization and transepithelial efflux of neutrophils in acute lung injury. Cell. 2002; 111: 635eC646.

    Gerety SS, Wang HU, Chen ZF, Anderson DJ. Symmetrical mutant phenotypes of the receptor EphB4 and its specific transmembrane ligand ephrin-B2 in cardiovascular development. Mol Cell. 1999; 4: 403eC414.

    Wang HU, Chen ZF, Anderson DJ. Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell. 1998; 93: 741eC753.

    Yuan K, Hong TM, Chen JJ, Tsai WH, Lin MT. Syndecan-1 up-regulated by ephrinB2/EphB4 plays dual roles in inflammatory angiogenesis. Blood. 2004; 104: 1025eC1033.

    Chen L, Klass C, Woods A. Syndecan-2 regulates transforming growth factor-beta signaling. J Biol Chem. 2004; 279: 15715eC15718.

    Klass CM, Couchman JR, Woods A. Control of extracellular matrix assembly by syndecan-2 proteoglycan. J Cell Sci. 2000; 113: 493eC506.

    Blobe GC, Liu X, Fang SJ, How T, Lodish HF. A novel mechanism for regulating transforming growth factor beta (TGF-beta) signaling. Functional modulation of type III TGF-beta receptor expression through interaction with the PDZ domain protein, GIPC. J Biol Chem. 2001; 276: 39608eC39617.

    Irie F, Yamaguchi Y. EPHB receptor signaling in dendritic spine development. Front Biosci. 2004; 9: 1365eC1373.

    Brunetti A, Goldfine ID. Role of myogenin in myoblast differentiation and its regulation by fibroblast growth factor. J Biol Chem. 1990; 265: 5960eC5963.

    Fuentealba L, Carey DJ, Brandan E. Antisense inhibition of syndecan-3 expression during skeletal muscle differentiation accelerates myogenesis through a basic fibroblast growth factor-dependent mechanism. J Biol Chem. 1999; 274: 37876eC37884.

    Casar JC, Cabello-Verrugio C, Olguin H, Aldunate R, Inestrosa NC, Brandan E. Heparan sulfate proteoglycans are increased during skeletal muscle regeneration: requirement of syndecan-3 for successful fiber formation. J Cell Sci. 2004; 117: 73eC84.

    Shimo T, Gentili C, Iwamoto M, Wu C, Koyama E, Pacifici M. Indian hedgehog and syndecans-3 coregulate chondrocyte proliferation and function during chick limb skeletogenesis. Dev Dyn. 2004; 229: 607eC617.

    Volk R, Schwartz JJ, Li J, Rosenberg RD, Simons M. The role of syndecan cytoplasmic domain in basic fibroblast growth factor-dependent signal transduction. J Biol Chem. 1999; 274: 24417eC24424.

    Chua CC, Rahimi N, Forsten-Williams K, Nugent MA. Heparan sulfate proteoglycans function as receptors for fibroblast growth factor-2 activation of extracellular signal-regulated kinases 1 and 2. Circ Res. 2004; 94: 316eC323.

    Bryant DM, Wylie FG, Stow JL. Regulation of endocytosis, nuclear translocation, and signaling of FGFR1 by E-cadherin. Mol Biol Cell. 2005; 16: 14eC23.

    Tkachenko E, Lutgens E, Stan RV, Simons M. Fibroblast growth factor 2 endocytosis in endothelial cells proceed via syndecan-4-dependent activation of Rac1 and a Cdc42-dependent macropinocytic pathway. J Cell Sci. 2004; 117: 3189eC3199.

    Simons K, Vaz WL. Model systems, lipid rafts, and cell membranes. Annu Rev Biophys Biomol Struct. 2004; 33: 269eC295.

    Brown DA, London E. Structure and function of sphingolipid- and cholesterol-rich membrane rafts. J Biol Chem. 2000; 275: 17221eC17224.

    Simons K, Toomre D. Lipid rafts and signal transduction. Nat Rev Mol Cell Biol. 2000; 1: 31eC39.

    Tkachenko E, Simons M. Clustering induces redistribution of syndecan-4 core protein into raft membrane domains. J Biol Chem. 2002; 277: 19946eC19951.

    Fuki IV, Kuhn KM, Lomazov IR, Rothman VL, Tuszynski GP, Iozzo RV, Swenson TL, Fisher EA, Williams KJ. The syndecan family of proteoglycans. Novel receptors mediating internalization of atherogenic lipoproteins in vitro. J Clin Invest. 1997; 100: 1611eC1622.

    Fuki IV, Meyer ME, Williams KJ. Transmembrane and cytoplasmic domains of syndecan mediate a multi-step endocytic pathway involving detergent-insoluble membrane rafts. Biochem J. 2000; 351: 607eC612.

    Stan RV. Structure and function of endothelial caveolae. Microsc Res Tech. 2002; 57: 350eC364.

    Liu J, Oh P, Horner T, Rogers RA, Schnitzer JE. Organized endothelial cell surface signal transduction in caveolae distinct from glycosylphosphatidylinositol-anchored protein microdomains. J Biol Chem. 1997; 272: 7211eC7222.

    Kawamura S, Miyamoto S, Brown JH. Initiation and transduction of stretch-induced RhoA and Rac1 activation through caveolae: cytoskeletal regulation of ERK translocation. J Biol Chem. 2003; 278: 31111eC31117.

    Pelkmans L, Burli T, Zerial M, Helenius A. Caveolin-stabilized membrane domains as multifunctional transport and sorting devices in endocytic membrane traffic. Cell. 2004; 118: 767eC780.

    Beardsley A, Fang K, Mertz H, Castranova V, Friend S, Liu J. Loss of caveolin-1 polarity impedes endothelial cell polarization and directional movement. J Biol Chem. 2004.

    Borset M, Hjertner O, Yaccoby S, Epstein J, Sanderson RD. Syndecan-1 is targeted to the uropods of polarized myeloma cells where it promotes adhesion and sequesters heparin-binding proteins. Blood. 2000; 96: 2528eC2536.

    Baciu PC, Goetinck PF. Protein kinase C regulates the recruitment of syndecan-4 into focal contacts. Mol Biol Cell. 1995; 6: 1503eC1513.

    Woodman SE, Ashton AW, Schubert W, Lee H, Williams TM, Medina FA, Wyckoff JB, Combs TP, Lisanti MP. Caveolin-1 knockout mice show an impaired angiogenic response to exogenous stimuli. Am J Pathol. 2003; 162: 2059eC2068.

    Wilcox-Adelman SA, Denhez F, Goetinck PF. Syndecan-4 modulates focal adhesion kinase phosphorylation. J Biol Chem. 2002; 277: 32970eC32977.

    Mukai M, Togawa A, Imamura F, Iwasaki T, Ayaki M, Mammoto T, Nakamura H, Tatsuta M, Inoue M. Sustained tyrosine-phosphorylation of FAK through Rho-dependent adhesion to fibronectin is essential for cancer cell migration. Anticancer Res. 2002; 22: 3175eC3184.

    Hsia DA, Mitra SK, Hauck CR, Streblow DN, Nelson JA, Ilic D, Huang S, Li E, Nemerow GR, Leng J, Spencer KS, Cheresh DA, Schlaepfer DD. Differential regulation of cell motility and invasion by FAK. J Cell Biol. 2003; 160: 753eC767.

    Shajahan AN, Timblin BK, Sandoval R, Tiruppathi C, Malik AB, Minshall RD. Role of Src-induced dynamin-2 phosphorylation in caveolae-mediated endocytosis in endothelial cells. J Biol Chem. 2004; 279: 20392eC20400.

    Chen J, Braet F, Brodsky S, Weinstein T, Romanov V, Noiri E, Goligorsky MS. VEGF-induced mobilization of caveolae and increase in permeability of endothelial cells. Am J Physiol Cell Physiol. 2002; 282: C1053eCC1063.

    Gleizes PE, Noaillac-Depeyre J, Amalric F, Gas N. Basic fibroblast growth factor (FGF-2) internalization through the heparan sulfate proteoglycans-mediated pathway: an ultrastructural approach. Eur J Cell Biol. 1995; 66: 47eC59.

    Schlunck G, Damke H, Kiosses WB, Rusk N, Symons MH, Waterman-Storer CM, Schmid SL, Schwartz MA. Modulation of rac localization and function by dynamin. Mol Biol Cell. 2004; 15: 256eC267.

    del Pozo MA, Alderson NB, Kiosses WB, Chiang H-H, Anderson RGW, Schwartz MA. Integrins regulate rac targeting by internalization of membrane domains. Science. 2004; 303: 839eC842.

    Sottile J, Chandler J. Fibronectin matrix turnover occurs through a caveolin-1-dependent process. Mol Biol Cell. 2005; 16: 757eC768.

    Oh ES, Woods A, Couchman JR. Syndecan-4 proteoglycan regulates the distribution and activity of protein kinase C. J Biol Chem. 1997; 272: 8133eC8136.

    Horowitz A, Simons M. Regulation of syndecan-4 phosphorylation in vivo. J Biol Chem. 1998; 273: 10914eC10918.

    Lim ST, Longley RL, Couchman JR, Woods A. Direct binding of syndecan-4 cytoplasmic domain to the catalytic domain of protein kinase C alpha (PKC alpha) increases focal adhesion localization of PKC alpha. J Biol Chem. 2003; 278: 13795eC13802.

    Keum E, Kim Y, Kim J, Kwon S, Lim Y, Han I, Oh ES. Syndecan-4 regulates localization, activity and stability of protein kinase C-alpha. Biochem J. 2004; 378: 1007eC1014.

    Woods A, Longley RL, Tumova S, Couchman JR. Syndecan-4 binding to the high affinity heparin-binding domain of fibronectin drives focal adhesion formation in fibroblasts. Arch Biochem Biophys. 2000; 374: 66eC72.

    Saoncella S, Echtermeyer F, Denhez F, Nowlen JK, Mosher DF, Robinson SD, Hynes RO, Goetinck PF. Syndecan-4 signals cooperatively with integrins in a Rho-dependent manner in the assembly of focal adhesions and actin stress fibers. Proc Natl Acad Sci U S A. 1999; 96: 2805eC2810.

    Bass MD, Humphries MJ. Cytoplasmic interactions of syndecan-4 orchestrate adhesion receptor and growth factor receptor signaling. Biochem J. 2002; 368: 1eC15.

    Longley RL, Woods A, Fleetwood A, Cowling GJ, Gallagher JT, Couchman JR. Control of morphology, cytoskeleton and migration by syndecan-4. J Cell Sci. 1999; 112: 3421eC3431.

    Saoncella S, Calautti E, Neveu W, Goetinck PF. Syndecan-4 regulates ATF-2 transcriptional activity in a Rac1-dependent manner. J Biol Chem. 2004; 279: 47172eC47176.

    Midwood KS, Williams LV, Schwarzbauer JE. Tissue repair and the dynamics of the extracellular matrix. Int J Biochem Cell Biol. 2004; 36: 1031eC1037.

    Midwood KS, Valenick LV, Hsia H, Schwarzbauer JE. Co-regulation of fibronectin signaling and matrix contraction by tenascin-C and syndecan-4. Mol Biol Cell. 2004; 15: 5670eC5677.

    Yamashita Y, Oritani K, Miyoshi EK, Wall R, Bernfield M, Kincade PW. Syndecan-4 is expressed by B lineage lymphocytes and can transmit a signal for formation of dendritic processes. J Immunol. 1999; 162: 5940eC5948.

    Hamon M, Mbemba E, Charnaux N, Slimani H, Brule S, Saffar L, Vassy R, Prost C, Lievre N, Starzec A, Gattegno L. A syndecan-4/CXCR4 complex expressed on human primary lymphocytes and macrophages and HeLa cell line binds the CXC chemokine stromal cell-derived factor-1. Glycobiology. 2004; 14: 311eC323.

    Granes F, Garcia R, Casaroli-Marano RP, Castel S, Rocamora N, Reina M, Urena JM, Vilaro S. Syndecan-2 induces filopodia by active cdc42Hs. Exp Cell Res. 1999; 248: 439eC456.

    Berndt C, Montanez E, Villena J, Fabre M, Vilaro S, Reina M. Influence of cytoplasmic deletions on the filopodia-inducing effect of syndecan-3. Cell Biol Int. 2004; 28: 829eC833.

    Steigemann P, Molitor A, Fellert S, Jackle H, Vorbruggen G. Heparan sulfate proteoglycan syndecan promotes axonal and myotube guidance by slit/robo signaling. Curr Biol. 2004; 14: 225eC230.

    Minniti AN, Labarca M, Hurtado C, Brandan E. Caenorhabditis elegans syndecan (SDN-1) is required for normal egg laying and associates with the nervous system and the vulva. J Cell Sci. 2004; 117: 5179eC5190.

    Kaksonen M, Pavlov I, Voikar V, Lauri SE, Hienola A, Riekki R, Lakso M, Taira T, Rauvala H. Syndecan-3-deficient mice exhibit enhanced LTP and impaired hippocampus-dependent memory. Mol Cell Neurosci. 2002; 21: 158eC172.

    Li J, Parovian C, Hampton TG, Metais C, Tkachenko E, Sellke FW, Simons M. Modulation of microvascular signaling by heparan sulfate matrix: studies in syndecan-4 transgenic mice. Microvasc Res. 2002; 64: 38eC46.

    Zhang Y, Li J, Partovian C, Sellke FW, Simons M. Syndecan-4 modulates basic fibroblast growth factor 2 signaling in vivo. Am J Physiol Heart Circ Physiol. 2003; 284: H2078eCH2082.(Eugene Tkachenko, John M.)