Next Article in Journal
Disease Transmission by Misfolded Prion-Protein Isoforms, Prion-Like Amyloids, Functional Amyloids and the Central Dogma
Previous Article in Journal
Phagocytosis in Teleosts. Implications of the New Cells Involved
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Adhesion Molecules and Ubiquitination—Functions and Significance

Department of Human Metabolomics, Institute of Nutrition and Food Sciences, University of Bonn, Katzenburgweg 9a, Bonn 53115, Germany
*
Author to whom correspondence should be addressed.
Submission received: 28 October 2015 / Revised: 2 December 2015 / Accepted: 15 December 2015 / Published: 23 December 2015

Abstract

:
Cell adhesion molecules of the immunoglobulin (Ig) superfamily represent the biggest group of cell adhesion molecules. They have been analyzed since approximately 40 years ago and most of them have been shown to play a role in tumor progression and in the nervous system. All members of the Ig superfamily are intensively posttranslationally modified. However, many aspects of their cellular functions are not yet known. Since a few years ago it is known that some of the Ig superfamily members are modified by ubiquitin. Ubiquitination has classically been described as a proteasomal degradation signal but during the last years it became obvious that it can regulate many other processes including internalization of cell surface molecules and lysosomal sorting. The purpose of this review is to summarize the current knowledge about the ubiquitination of cell adhesion molecules of the Ig superfamily and to discuss its potential physiological roles in tumorigenesis and in the nervous system.

1. Introduction

Cell adhesion molecules are membrane-associated cell surface glycoproteins playing important roles in cell recognition, adhesion, migration and differentiation [1,2]. They can be subdivided into four different groups, which are defined by different structures and functional characteristics: the cadherins, integrins, selectins and immunoglobulin (Ig)-like proteins [3,4,5,6].
In this review we will focus on members of the Ig superfamily. They mediate calcium-independent cell adhesion and represent the biggest and structural most versatile group of cell adhesion molecules. One common component of all Ig superfamily members is the existence of at least one Ig-like domain in their extracellular region.
Usually, the members of this family contain big extracellular regions by which their adhesive function is mediated. Additionally, many of them contain a transmembrane region and a cytosolic domain which transmits signals into the cell thus providing intensive exchange of information [7]. Some Ig family members are attached to the plasma membrane via a glycosylphosphatidyl inositol (GPI) anchor. The extracellular fraction also contains secreted isoforms of Ig superfamily cell adhesion molecules that can be generated either by alternative splicing or cleavage by extracellular proteases. Some family members have additionally fibronectin (FN) type III domains in their extracellular region. In this review, we will describe in more detail the cell adhesion molecules NCAM, L1, MCAM and ALCAM since they play a role in the nervous system and in cancer and have been described to be ubiquitinated and discuss possible roles of this posttranslational modification (Figure 1). SHP substrate-1 (SHPS-1), another member of the Ig superfamily, was first shown to become ubiquitinated, however, since it additionally belongs to the signal-regulatory protein (SIRP) family, it will not be discussed in this context [8].
Figure 1. Overview about Immunoglobulin (Ig) superfamily members that are modified by ubiquitin. Ig superfamily members contain in their amino terminal extracellular domain Ig-like domains. Additionally, some of them have fibronectin (FN) type III domains (NCAM and L1). The ubiquitinated Ig superfamily members contain a transmembrane region and a cytoplasmic domain. NCAM: neural cell adhesion molecule; ALCAM: activated leukocyte cell adhesion molecule; MCAM: melanoma cell adhesion molecule; V: variable.
Figure 1. Overview about Immunoglobulin (Ig) superfamily members that are modified by ubiquitin. Ig superfamily members contain in their amino terminal extracellular domain Ig-like domains. Additionally, some of them have fibronectin (FN) type III domains (NCAM and L1). The ubiquitinated Ig superfamily members contain a transmembrane region and a cytoplasmic domain. NCAM: neural cell adhesion molecule; ALCAM: activated leukocyte cell adhesion molecule; MCAM: melanoma cell adhesion molecule; V: variable.
Biology 05 00001 g001

2. Structure and Functions of Different Cell Adhesion Molecules of the Ig Superfamily

2.1. The Neural Cell Adhesion Molecule NCAM

2.1.1. Expression and Functions

The neural cell adhesion molecule NCAM was the first Ig superfamily member to be identified [9,10]. NCAM is widely expressed in the central and peripheral nervous system and in many other tissues but its function has mainly been investigated in the nervous system. Here, it is involved in processes like cell migration, synaptic plasticity, axonal growth and fasciculation [11,12,13,14,15,16,17,18,19]. A number of studies support a role of NCAM in the development of schizophrenia [20,21,22,23,24,25,26,27].
Several isoforms of NCAM are generated by alternative splicing with the three major isoforms being NCAM120, NCAM140 and NCAM180. NCAM120 is a GPI-anchored isoform, whereas NCAM140 and NCAM180 are transmembrane proteins. They are identical in their extracellular domains containing five Ig-like domains and two FN type III domains. NCAM140 and NCAM180 differ only in an alternatively spliced exon in their cytoplasmic tails leading to additional 261 amino acids in the NCAM180 isoform [28].
In the nervous system, the different isoforms exhibit different expression patterns with NCAM140 being mainly expressed on migratory growth cones and axon shafts of developing neurons but also on glia cells, whereas NCAM180 is enriched at sites of cell-cell contact and in particular at postsynaptic densities of mature neurons. In contrast, NCAM120 is preferentially expressed on glia cells [29,30,31].
In addition to the nervous system, NCAM is expressed in many other tissues and cell types like skeletal muscle cells, heart muscle cells, pancreatic endocrine cells, adult intestine, on natural killer cells and on peripheral blood T lymphocytes. It has also been reported to be expressed in lung epithelium in chicken [32,33,34,35,36,37,38,39].
NCAM also plays an important role in tumor progression. In some cancer types NCAM exerts a tumor-suppressive role (e.g., glial tumors), whereas, e.g. neuroblastoma and tumors of neuroendocrine origin, highly express NCAM, pointing to a tumorigenic effect [40,41,42,43,44,45]. Furthermore, abnormal NCAM expression has been shown in myeloma, gastrointestinal, thyroid, small lung cancer, epithelial ovarian and renal cancer [46,47,48]. Interestingly, in some tumors, NCAM switches from the NCAM120 isoform to NCAM140 or NCAM180 isoforms during tumor development [49,50,51,52,53]. The polysialylation (PSA), an extracellular posttranslational modification of NCAM (see below) also seems to play a role in tumorigenesis although its role seems to differ in different cancer types [42,54,55].

2.1.2. Cellular Mechanisms

NCAM can interact with several molecules through its extracellular domain and its cytoplasmic tail [28]. Extracellular homophilic NCAM interactions can occur between NCAM molecules present on the same (cis-interaction) or on opposing cell surfaces (trans-interaction). On the cell surface, NCAM is present in a cis-dimeric form which is mediated by the first two Ig-like domains. These cis-dimers, in turn, mediate trans interactions. This trans interaction of NCAM cis-dimers is probably mediated by an interaction either between the second and third Ig-like domains or between all three N-terminal Ig-like domains [56]. Homophilic NCAM-NCAM interactions are important for cell adhesion, axonal fasciculation and NCAM-dependent neurite growth [57].
NCAM binds to several other molecules in a heterophilic mode including the Ig family members axonin-1/TAG-1 and L1. Binding to L1 induces phosphorylation of tyrosine and serine residues in L1 and stimulates neurite outgrowth [58,59,60]. Another important interaction partner is the fibroblast growth factor receptor (FGFR) whose direct binding to both FN modules of NCAM can activate FGFR signaling cascades thereby influencing neurite growth or tumor progression [61,62]. Additionally, it has been demonstrated that NCAM can act as a non-canonical ligand of FGFR1 and induce FGFR-dependent cell migration by promoting FGFR internalization with subsequent recycling of the FGFR to the cell surface [63,64]. The recently described interaction of NCAM with EphA3 regulates synapse formation [65,66]. NCAM also binds directly to glial derived neurotrophic factor (GDNF) and GDNF family receptor α-1 (GFRα-1), thereby functioning as an alternative signaling receptor for neurotrophic factors [67]. Further extracellular interaction partners of NCAM are components of the extracellular matrix like heparin, heparin-sulfate proteoglycans (HSPGs) and chondroitin-sulfate proteoglycans (CSPG) [68,69,70]. Interestingly, NCAM also binds prion protein (PrP) which recruits NCAM to lipid rafts and activates NCAM-dependent signal transduction via the non-receptor tyrosine kinase p59fyn [71]. This interaction is required for neuronal differentiation of neural precursor cells of the subventricular zone (SVZ) [72]. A direct interaction with adenosine triphosphate (ATP) was also demonstrated [73].
Intracellular binding partners of NCAM include cytoskeletally associated proteins and signaling molecules. The first intracellular interaction partner to be identified was the cytoskeletal linker protein spectrin [74]. It binds to NCAM180 and NCAM140 and even indirectly to the GPI-linked NCAM120 isoform which is mainly confined to lipid rafts [75]. Spectrin builds a molecular bridge between NCAM and protein kinase C 2-β (PKCβ2). This indirect binding of PKCβ2 via spectrin to NCAM140 and NCAM180 also plays a role in NCAM-mediated neurite outgrowth [75,76,77]. Furthermore, GAP-43 (growth associated protein) could be co-precipitated with NCAM providing another connection to the cytoskeleton [78]. Further cytoskeletal interaction partners of NCAM140 and NCAM180 are α- and β-tubulin and α-actinin. The cytoplasmic proteins leucine-rich acidic nuclear protein (LANP), syndapin, the protein phosphatases PP1 and PP2A and phospholipase C-γ (PLC-γ) were also identified as interaction partners of NCAM140 and NCAM180 [79,80]. Specifically NCAM180 binds to microtubule associated protein 1A (MAP1A), β-actin, tropomyosin, RhoA-binding kinase-α and turned on after division-64 (TOAD-64). Recently, a direct interaction with the motor protein kinesin-1 could be demonstrated [79,80,81].
Since approximately twenty years it is known that NCAM can also act as a signaling receptor [82,83,84]. NCAM-dependent signaling can be initiated by homo- or heterophilic extracellular interactions (see above). Outside of lipid rafts NCAM activates the cAMP-dependent kinase (PKA) by yet unknown mechanisms [85]. Additionally, interactions with the FGFR activate PLC-γ leading to diacylglycerol (DAG) formation and subsequently to PKCβ2 activation, generation of arachidonic acid and elevated intracellular Ca2+ (calcium ions) [75]. In contrast, in lipid rafts NCAM binds constitutively receptor protein tyrosine phosphatase α (RPTPα) and the non-receptor tyrosine kinase p59fyn [86,87]. Activation of p59fyn by homophilic or heterophilic NCAM interactions leads to recruitment of focal adhesion kinase (FAK) to the p59fyn-NCAM complex and subsequently to activation of the ras-mitogen activated protein kinase (MAPK) pathway [77,88]. It seems that convergence of signaling from both NCAM fractions—raft and non-raft-associated—is required to allow cytoskeletal rearrangement and gene transcription cumulating in neurite outgrowth and cell migration [89]. All three major isoforms of NCAM are extensively posttranslationally modified. Both transmembrane isoforms NCAM140 and NCAM180 can be palmitoylated at cytosolic cysteine residues close to the transmembrane domain which is necessary for the correct distribution of NCAM in the plasma membrane and for correct intracellular signaling [89,90]. Different serine and threonine residues within the cytoplasmic domain can be phosphorylated [91,92]. Two so far identified kinases being responsible for NCAM phosphorylation are GSK-3 (glycogen synthase kinase-3) and casein kinase (CK) I [93]. NCAM contains one tyrosine residue in its cytoplasmic domain which can also be phosphorylated. This phosphorylation can be enhanced by direct association of NCAM with the receptor tyrosine kinase B (TrkB) and plays a role in NCAM-dependent neurite outgrowth [94,95]. However, NCAM is probably modified by several other, yet unknown kinases.
Additionally, the ubiquitin-fold modifier-conjugating enzyme-1 (Ufc1) interacts with the intracellular domain of NCAM140 [96]. Ufc1 is involved in the modification of proteins with the ubiquitin-like molecule ubiquitin-fold modifier-1 (Ufm1) and might therefore stimulate the ufmylation of NCAM. However, the exact mechanisms remain to be investigated.
A further important posttranslational modification is the extensive N-glycosylation in NCAM’s extracellular domain. Six potential glycosylation sites have been identified so far [97]. Most importantly NCAM is the major carrier for a unique modification, PSA which is linked N-glycosidically to two glycosylation sites in the Ig5 domain. In the brain, PSA is mainly expressed during development, reaches its maximum expression perinatally and is then drastically downregulated. In later developmental stages the PSA modification only remains in regions of the brain that maintain neurogenesis, including the SVZ, the granule cell layer of the hippocampus, particular regions of the hypothalamus and regions undergoing structural plasticity [98,99]. Several in vitro and in vivo studies suggest that PSA expression on NCAM converts NCAM from a molecule that promotes stability to one that promotes plasticity [100,101]. The PSA modification is also involved in NCAM’s effect on tumorigenesis but its role is discussed controversially. Depending on the tumor type, PSA seems either to reduce or to increase the tumorigenic potential [42,54,55].
Soluble NCAM forms are generated by different members of the disintegrin and metalloprotease (ADAM) family cleaving close to the plasma membrane resulting in an approximately 115 kDa fragment [18,19,102,103]. Shedding can be induced by tyrosine kinase and MAP kinase activity and has been implicated in neurite branching, outgrowth and cell migration [18,19,102]. Depending on the cell type, NCAM shedding either reduces or increases neurite outgrowth [19,102]. After induction of NCAM internalization another short extracellular 55 kDa fragment without any known function was observed, probably generated by a serin protease [104].

2.2. The Cell Adhesion Molecule L1

2.2.1. Expression and Functions

Since its discovery in 1984 L1 has been established as a key player throughout the development of the nervous system [105]. In the developing nervous system it is widely expressed on postmitotic neurons, on astrocytes and on Schwann cells, in the adulthood on neurons and on cells of other tissues. L1 consists of six Ig-like domains, five FN type III domains, one transmembrane domain and a cytoplasmic tail and has a molecular mass of approximately 200 kDa. The molecular weight varies in different cell types dependent on different and extensive glycosylation at 22 potential N-glycosylation sites in L1’s extracellular domain. Different isoforms of L1 are generated by alternative splicing in a cell-specific manner. The isoform containing exons 2 and 27 represents the neuronal form of L1. Presence of exon 2 results in enhanced homophilic binding whereas amino acids RSLE encoded by exon 27 together with a preceding tyrosine result in a tyrosine-dependent sorting signal (YRSL) leading to clathrin-dependent endocytosis of L1 by interaction with the μ2-subunit of the adaptor protein 2 (AP-2) complex [106,107,108]. The non-neuronal isoforms without exons 2 and 27 are expressed in cell types like Schwann cells, hematopoetic cells and epithelial cells [109,110].
In addition to its function in neural development, L1 is also important for synaptic plasticity and regeneration in adult brain [111,112,113,114]. The importance of L1 is underlined by the occurrence of severe neurological disorders resulting from mutations in the human L1 gene collectively referred to as L1 syndrome [115,116,117,118,119].
Outside of the nervous system L1 has a major function during cancer progression. It has first been described in ovarian and endometrial carcinoma where its expression correlates with a poor prognosis [120]. Many subsequent studies revealed L1 expression in several tumor tissues. The function of L1 in tumorigenesis is also based on its ability to increase cell growth, motility, invasion and chemoresistance [121].

2.2.2. Cellular Mechanisms

L1 interacts—like NCAM—with several molecules through its extracellular and cytoplasmic domain. L1 is likely to be part of a complex that involves cis and trans interactions at the cell surface thereby modulating L1 binding or activity [122]. In the nervous system, homophilic trans-interactions are important for axonal fasciculation and have been shown to have neurite growth promoting effects [123].
Heterophilic extracellular interaction partners include integrins, axonin-1/TAG-1, neurocan and phosphocan, neuropilin-1, F3/F11/contactin, ALCAM/DM-GRASP, CD24, NCAM, laminin and the FGFR [58,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138]. The functions of heterophilic interactions of L1 have not yet fully been clarified. They have been suggested to enhance effects of homophilic L1 interactions. In this context, axonin-1/TAG-1 seems to play a role in L1-dependent neurite growth [130,131]. The cis-interaction of L1 with NCAM enhances L1 trans-interaction and concomitantly L1-stimulated cell aggregation, cell migration and neurite growth [58,60,139,140].
Additionally, the cytoplasmic tail of L1 provides linkages to the cytoskeleton and is associated with several kinases. One connection to the cytoskeleton is mediated by two binding sites in L1 for ERM family members which include the proteins ezrin, radixin and moesin (ERM). These molecules are crosslinkers between the membrane and the cytoskeleton. The first binding site for ezrin encompasses the Y1176RSLE sequence which is also involved in binding of the AP-2 complex (see above) [141]. The sequence KGGKY1151 located close to the transmembrane domain represents the second binding site to ezrin and has earlier been described to act as a linker to actin [142,143]. The phosphorylation status of the respective tyrosine residue of L1 (Y1151 by a src family member or Y1176 by pp60c-src) seems to be relevant for the interaction with ezrin [143,144]. This interaction may decrease lateral movement of L1 in the plasma membrane and thus regulate L1-dependent neurite outgrowth and branching [141,143,144]. Phosphorylation of S1152 by p90rsk is important for axon outgrowth mediated by L1 and might also be involved in the regulation of ezrin binding [144,145].
As mentioned above, phosphorylation of Y1176 tightly regulates AP-2 binding in addition to regulation of ezrin binding. In the phosphorylated form, AP-2 cannot bind to the consensus sequence YRSL whereas in the non-phosphorylated form a binding site for AP-2 is created and clathrin-dependent endocytosis is initiated [146,147]. Endocytosis and cell migration also seem to be influenced by phosphorylation at S1181 which represents a casein kinase II consensus motif [148]. After endocytosis the major part of L1 is recycled to the cell surface. The endocytosis and recycling take place in the migratory growth cone where it is implicated in neurite outgrowth and polarized adhesion [106,149,150,151]. Endosomal trafficking of L1 is also required to target L1 from the somatodendritic compartment to the growing axon by transcytosis [152]. Recently, Rabex-5, a multidomain protein that has guanine nucleotide exchange factor (GEF) activity for Rab5, has been shown to bind to L1 and to regulate L1 internalization [153,154].
L1 also binds reversibly to ankyrin, an actin/spectrin adapter protein through a highly conserved FIGQY1229 motif in the cytoplasmic domain of L1 [155,156,157,158]. This interaction promotes stationary behavior of cells in culture and neurite initiation by inhibiting retrograde actin flow [159,160]. Phosphorylation of Y1229 by a yet unknown kinase disrupts L1-ankyrin binding resulting in enhanced neurite outgrowth in vitro and altered neuronal branching which leads to a decrease in perisomatic synapses of inhibitory GABAergic interneurons during cortex development [160,161,162,163,164]. This conserved motif also mediates the binding of L1 to the microtubule-associated protein doublecortin in the phosphorylated form [165].
These data show that phosphorylation of L1 by several kinases regulates intracellular binding. As for several other cell adhesion molecules, the involvement of L1 in signaling pathways is extremely complex. L1 has been shown to be phosphorylated in vitro and in vivo at several sites and these interactions are essential for L1 function. L1 crosslinking at the cell surface activates the MAP kinase extracellular signal-regulated kinase 2 (ERK2) which in turn phosphorylates S1204 and S1248 and goes along with L1 endocytosis [146]. Sustained activation of ERK2 by L1 crosslinking leads to increased motility and invasion into the surrounding matrix [166]. ERK activation is mediated by pp60c-src, phosphoinositide 3 kinase (PI3K), the Vav2 guanine nucleotide exchange factor, Rac1 GTPase and p21 activated kinase (PAK1) [146,167]. A fragment of L1 becomes additionally posttranslationally modified by small ubiquitin-like modifier (SUMO), which is necessary for its nuclear import [168].
The extracellular interaction of L1 with the FGFR is implicated in activation of FGFR signaling pathways and leads to L1-dependent neurite outgrowth via activation of PLC-γ, release of arachidonic acid and subsequent opening of voltage-gated Ca2+ channels as also shown for NCAM [169,170,171,172,173]. Ran binding protein in the microtubule-organizing center (RanBPM) was also identified as an L1 interacting protein and seems to serve as an adaptor in L1-mediated signaling in neurite growth [174,175]. Another mechanism of L1 signaling depends on its extracellular interaction with neuropilin-1 and semaphorin 3A (Sema3A), which induce recruitment of FAK to L1 and subsequent ERK activation resulting in growth cone collapse [176]. Finally, CK II co-precipitates with L1 and phosphorylates L1 constitutively at S1181 in vitro [177]. Since S1181 is located directly behind the YRSL motif an implication in L1 intracellular trafficking has early been suggested and its implication in endocytosis shown later on [148,178].
More complexity to L1 function is added by its extracellular and intramembranous cleavage by different proteases releasing soluble L1 fragments into the extracellular space thereby modulating cell migration of tumor cell lines and neurite outgrowth of neurons. Constitutive and induced cleavage of L1 generate fragments of 200, 140, 135, 80, 70, 32 and 28 kDa molecular weight, respectively [134,168,179,180,181,182,183,184,185].

2.3. The Melanoma Cell Adhesion Molecule MCAM

2.3.1. Expression and Functions

The melanoma cell adhesion molecule MCAM has first been described in 1987 by Lehmann et al., as a cell surface marker specific on melanoma cells but not on benign melanocytes [186]. Later on, it was classified as a cell adhesion molecule with a postulated tumorigenic potential [187]. MCAM is broadly and highly expressed in embryonic tissue but more limited in adult tissue where it is present, e.g., in smooth muscle cells, hair follicular cells, activated T cells, intermediate trophoblasts, mesenchymal stem cells and dental pulp [186,188,189,190,191,192,193]. Its expression can be induced by environmental signals thus initiating appropriate MCAM-specific reactions [194]. Its overexpression in several carcinoma tissues, e.g., prostate carcinoma, choriocarcinoma, angiosarcoma, Kaposi’s sarcoma, and leiomyosarcoma suggested a potential role in tumorigenic processes [195,196].
MCAM consists of a short cytoplasmic domain, a single transmembrane domain, and an extracellular domain containing five Ig-like units (V-V-C2-C2-C2) [186,197]. In its extracellular domain it contains eight putative N-glycosylation sites [198]. Different isoforms of MCAM have been described, a long form and a short form with molecular weights between 113 and 119 kDa, and a soluble form [198,199,200,201]. The long and short forms are identical in their extracellular domains and the transmembrane region, but differ greatly in the amino acid composition in their cytoplasmic tail. These isoforms are generated by alternative splicing whereas the soluble form seems to be either generated by alternative splicing or membrane shedding by a metalloprotease [198,202,203].
MCAM has been functionally implicated in cell adhesion, cell migration, proliferation, differentiation, signaling and immune response [194]. Since MCAM is mainly expressed in developmental stages it has been suggested to play a role in developmental processes. Consistently, it has been described to support adherence between neurons and glia cells with endothelial cells since it is mainly expressed on blood vessels but not on neural cells. Thus, by increasing vascularization of the tissue it facilitates development of the cerebellum and the peripheral nervous system [204,205,206,207]. It is also involved in organogenesis and maintenance of organ function like in the kidney and thymus and has been shown to be important for retina development in quail [208,209,210,211,212,213,214].
Furthermore, it has been implicated in inflammatory processes since it seems to play a role in recruitment of activated T cells to inflammatory sites and is upregulated in various inflammatory diseases [194,200].
An early study reported that in human tissues more than 70% of melanoma metastases express MCAM and that its expression correlates with increasing tumor thickness [215]. Later studies found that MCAM is overexpressed in most malignant and metastatic cancer and plays a significant role in tumor progression [194,216,217,218]. The function of MCAM is based on its ability to modulate cell motility and adhesion. In this context, it has been shown to increase motility and invasiveness of many tumor cells in vitro and metastasis in vivo by altering several apoptotic proteins involved in cell survival, proliferation and angiogenesis. Specifically, it was recently demonstrated that MCAM is involved in reduction of caspase-activation probably via its interplay with ALCAM [219]. Additionally, it elevates levels of the vascular endothelial growth factor, vascular endothelial growth factor receptor 2 (VEGF, VEGFR2) and CD31 as alternative ways to affect tumor metastasis [220]. Another study reported that inhibiting MCAM expression in melanoma cells may lead to loss of gap-junction communication, as evidenced by reduced invasion in a three-dimensional skin reconstruct model [221]. Furthermore, an MCAM antibody decreases melanoma cell invasion, tumor vascularization, matrix metalloproteinase-2 (MMP-2) expression and reduces formation of lung metastasis in nude mice [222].

2.3.2. Cellular Mechanisms

MCAM interacts heterophilically with some recently identified molecules like laminin-411, Galectin-1, Wnt5a, the VEGFR2 and the neurite outgrowth factor [223,224]. It also mediates homophilic interaction that is involved in neurite extension and neuron development probably by modulating cell–cell adhesion [201,224,225,226,227,228,229,230,231]. However, in contrast to other well analyzed cell adhesion molecules not much is known about interaction partners of MCAM.
In the cytoplasmic domain MCAM contains a conserved positively charged KKGK motif mediating interaction with proteins of the ERM family. This interaction leads to recruitment of Rho GDP-dissociation inhibitor (RhoGDI) 1, activation of RhoA and results in increased cell migration [232]. MCAM induces microvilli formation in lymphocytes and increases the rolling and adhesion of lymphocytes on endothelial cells. The microvilli formation depends on the interaction of the long MCAM isoform with the actin cytoskeleton and requires a PKC phosphorylation site in the cytoplasmic tail [200]. A double leucine motif in the cytoplasmic tail is responsible for basolateral targeting in epithelia [194].
Additionally, MCAM can—like other cell adhesion molecules—activate signal transduction cascades. A reciprocal relationship between MCAM expression and AKT phosphorylation in melanoma cells has been suggested. Blocking AKT activation results in decreased MCAM expression, while overexpression of MCAM, likewise, increases phosphorylation of AKT and of the pro-apoptotic protein Bad thus becoming inactive [233,234]. In addition, engagement of MCAM in endothelial cells leads to activation of p59fyn and subsequent tyrosine phosphorylation and activation of FAK. Paxillin can also bind to activated FAK and thus increase cell motility [235]. Additionally, MCAM triggering initiates PLCγ-mediated Ca2+-influx resulting in activation of PYK2 and p130CAS [236]. The p38 kinase can be activated as a response to MCAM triggering leading to activation of the NFκB cascade which is critical in cell migration, angiogenesis and tumor metastasis [237,238]. MCAM has also been suggested to stimulate the transcription factors c-fos and c-jun [239]. Overall, these results demonstrate that MCAM plays a role in both cell–cell interactions and signal transduction in tumor cells.

2.4. The Activated Leukocyte Cell Adhesion Molecule ALCAM

2.4.1. Expression and Functions

Another member of the Ig superfamily is the cell adhesion molecule ALCAM/DM-GRASP, which was first described by Burns et al. [240]. The name DM-GRASP was originally derived from its detection by antibodies recognizing an immunoglobulin-like restricted axonal surface protein. The prefix DM originates from expression of DM-GRASP in the dorsal funiculus and ventral midline of the chick spinal cord. DM-GRASP has several synonyms including SC1, BEN, ALCAM and CD166 [241,242,243]. In this review, the term ALCAM will further be used for all homologues in all species. ALCAM is a glycoprotein with eight putative N-glycosylation sites [244]. The molecular weight of the mature form has been described between 95 and 110 kDa [240,244]. In its extracellular domain ALCAM contains five Ig-like domains (V-V-C2-C2-C2), a transmembrane region and a short cytoplasmic domain [243,245]. A soluble splice variant of ALCAM consisting of the single amino-terminal V-type l Ig-like domain which is required for cell–cell-adhesive interactions was detected in endothelial cells [246]. It can also be cleaved in its extracellular domain close to the plasma membrane by ADAM17 resulting in a secreted form which has been suggested to reduce cell–cell adhesion in cancer [247,248]. ALCAM has first been detected in chicken on a restricted population of axons and was later found on many other cell types like activated leucocytes, hematopoetic stem cells and myeloid progenitors [240,249]. It has also been detected on neuronal cells, mesenchymal stem cells and bone marrow stromal cells and is expressed in most developing tissues like the central and peripheral nervous system, sensory organs, during hematopoiesis and in endothelial and epithelial lineage pointing to a function of ALCAM in many developmental processes [245,249,250,251,252,253]. Indeed, loss of ALCAM results in loss of cell adhesion and in developmental defects [254]. Although it is additionally expressed in several multipotent cells including cells of umbilical cord blood, bone marrow, testes, fetal lung, intervertebral disc and dental pulp [255,256,257,258,259,260]. It is not yet known if ALCAM contributes functionally to the multipotent capacity of these cell types [254].
ALCAM is also expressed in almost all cancers. In vivo mouse studies suggest a role of ALCAM in cancer progression [261,262,263]. However, depending on the original tissue of the cancer cells ALCAM becomes either upregulated or downregulated compared to healthy tissue, complicating the understanding of its role in cancer [264]. Due to its expression in cancer cells it is used more and more frequently as a biomarker of cancer progression in several tumor types like prostate cancer, colorectal cancer, breast cancer, oral cancers, pancreatice cancer, neuroblastoma, ovarian cancer and melanoma [265,266,267,268,269,270,271,272,273,274,275].
Several studies investigated ALCAM’s role in the nervous system. In neurons it plays a role in cell adhesion, axonal growth and navigation, in neuronal cell migration, differentiation and synapse formation and specifically serves as a guidance molecule for cellular migration and neuronal outgrowth during development. Consistently, ALCAM-deficient mice exhibit delayed maturation of neuromuscular junctions and defects in axon fasciculation [240,250,276,277,278,279,280,281,282,283,284,285]. Interestingly, ALCAM is expressed on growing retinal ganglion cell (RGC) axons and provides a permissive signal for axon guidance of RGCs [249,278,279]. ALCAM’s functional significance in RGCs is highlighted by the fact that it is translated in growth cones which depends on ERK and target of rapamycin (TOR) activation and that this local translation is crucial for the preference of RGC axons on ALCAM substrate [286]. Recently, ALCAM has also been shown to function in axonal growth of dorsal root ganglion neurons and to modulate neurotrophin signaling thereby regulating neurotrophin-dependent neurite outgrowth [287,288]. ALCAM has also been associated with multiple sclerosis susceptibility although the mechanism is controversially discussed [289,290].

2.4.2. Cellular Mechanisms

In addition to homophilic interactions via the amino-terminal V-type Ig-like domain ALCAM is able to mediate heterophilic bindings with L1/NgCAM and CD6 [129,276,291,292]. The heterophilic binding to CD6 is more robust and persistent than the homophilic ALCAM interaction and plays a role in activation of T cells and transmigration of leukocytes across the blood brain barrier. Another important prerequisite for this mechanism seems to be its concentration in cholesterol-enriched membrane microdomains of leukocytes [293,294,295,296,297,298,299]. In neurons the heterophilic interaction with L1 seems to be more important than other extracellular interactions and has been suggested to play a role in guidance of retinal axons during development [129,279,285]. Both, the heterophilic and homophilic ALCAM interactions are mediated by the N-terminal Ig-like domains of ALCAM (trans interactions) whereas lateral oligomerization of ALCAM at the cell surface (cis oligomerization) is mediated by the three membrane proximal C-type Ig-like domains [291,300]. Together, cis-oligomerization at the cell surface and trans interactions between adjacent cells would synergistically promote ALCAM recruitment and network formation at sites of cell-cell contact [253].
Homo- and heterophilic binding of ALCAM and hence ALCAM-dependent cell adhesion depends on PKCα [301]. In this context it is interesting that the short cytoplasmic domain of ALCAM does neither contain any known adaptor-binding sites nor consensus sequences for potential interaction partners. Nevertheless, a potential interaction and functional involvement of ALCAM and α-catenin have also been suggested [302].
The Rac-specific guanine nucleotide exchange factor Tiam1 which is one important regulator of Rho GTPase functions in tumor cells is also functionally involved in ALCAM-dependent cell adhesion and reduces cell migration in metastatic melanoma cells [303,304]. ALCAM can—like other members of the Ig superfamily—be internalized via a clathrin-dependent mechanism after ligand binding in the extracellular region followed by its recycling to the cell surface. This process has been discussed as a potential therapeutic mechanism to deliver immunotoxins into tumor cells [305].
Table 1 and Table 2 give an overview about the mentioned posttranslational modifications and interaction partners of the described cell adhesion molecules.
Table 1. Overview about the above mentioned posttranslational modifications of the described cell adhesion molecules. EC, extracellular; IC, intracellular; * form of N-glycosylation.
Table 1. Overview about the above mentioned posttranslational modifications of the described cell adhesion molecules. EC, extracellular; IC, intracellular; * form of N-glycosylation.
Cell Adhesion MoleculeModificationReferences
NCAMPolysialylation * (EC)[306,307]
N-glycosylation (EC)[97]
Palmitoylation (IC)[90]
Phosphorylation (IC)[91,92,93,94,95]
Ufmylation (IC)[96]
L1N-glycosylation (EC)[308,309,310,311]
Phosphorylation (IC)[144,145,146,147,148,161,162,163,308,312]
Sumoylation (IC)[168]
MCAMN-glycosylation (EC)[198]
Phosphorylation (IC)[200]
ALCAMN-glycosylation (EC)[243,244]
Table 2. Overview about the above mentioned interaction partners of the described cell adhesion molecules. EC, extracellular; IC intracellular.
Table 2. Overview about the above mentioned interaction partners of the described cell adhesion molecules. EC, extracellular; IC intracellular.
Cell Adhesion MoleculeInteraction PartnersReferences
NCAMβ-actin (IC)[80]
α-actinin (IC)[80]
ADAM (EC)[19,102,103]
ATP (EC)[73]
Axonin-1/TAG-1 (EC)[59]
CKI (IC)[93]
EphA3 (EC)[65,66]
FGFR (EC)[61,63,64]
GAP-43 (IC)[78]
GFR-α1, GDNF (EC)[67]
GSK-3 (IC)[93]
Heparin, HSPGs, CSPGs (EC)[68,69,70]
Kinesin-1 (IC)[81]
L1 (EC)[58]
LANP (IC)[79]
NCAM (EC)[313]
MAP1A (IC)[80]
p59fyn (IC)[86]
PLC-γ (IC)[79]
PP1, PP2A (IC)[79]
PrP (EC)[71,72]
RhoA-binding kinase-α (IC)[80]
RPTPα (IC)[87]
Spectrin (IC)[74]
ST8SiaII, ST8SiaIV[314]
Syndapin (IC)[79]
Tropomyosin (IC)[80]
TOAD-64 (IC)[79]
TRKB (IC)[95]
α- and β-tubulin (IC)[80]
Ufc-1 (IC)[96]
L1ADAM (EC)[134,179,181,183]
AP-2 (μ-subunit) (IC)[178]
ALCAM/DM-GRASP (EC)[129]
Ankyrin (IC)[156]
Axonin-1/TAG-1 (EC)[130,131]
CD24 (EC)[128]
CK II (IC)[177]
Doublecortin[165]
Erk2[146]
ERM proteins (IC)[141,142]
F3/F11/contactin (EC)[124]
FGFR (EC)[138]
Integrins (EC)[315]
L1 (EC)[123]
Laminin (EC)[125]
NCAM (EC)[58]
Neurocan (EC)[126]
Neuropilin-1 (EC)[136]
P90rsk (IC)[145]
Phosphocan (EC)[127]
Rabex-5 (IC)[154]
(RanBPM)[175]
MCAMActin (IC)[200]
ERM family[232]
Galectin-1 (EC)[316]
Laminin-411 (EC)[317]
MCAM (EC)[224]
Neurite outgrowth factor (EC)[318]
VEGFR2 (EC)[319]
Wnt5a (EC)[320]
ALCAMADAM17 (EC)[247]
ALCAM (EC)[276,321]
CD6 (EC)[292]
L1 (EC)[129]

3. Structure and Function of Ubiquitin

Ubiquitin is a polypeptide consisting of 76 amino acids. It exists in all eukaryotic cells and is highly conserved from yeast to humans. It is covalently attached to the ε-amino group of lysine (K) residues of target proteins. Alternatively, it can also be attached to the amino-terminal region of the target protein [322,323,324]. The attachment of ubiquitin is an ATP-dependent, three-step process: in the first step ubiquitin is activated by forming a thioester bond with a ubiquitin-activating enzyme (E1). Next, activated ubiquitin is transferred to a ubiquitin-conjugating enzyme (E2). Finally, ubiquitin-ligases (E3 enzymes) catalyze the transfer of ubiquitin with its carboxy-terminal glycine to a lysine residue of the target protein [325,326]. The E3 enzymes are one of the most important components in the ubiquitination process since they interact directly with the substrate and mediate in large part the ubiquitination specificity [327]. They can be classified into four different groups: the HECT (Homologous to the E6-AP Carboxyl Terminus) type, the RING (Really Interesting New Gene) type, the U-box type and the PHD (plant homeodomain)-finger type ligases. Thus far, 500–1000 different ubiquitin ligases are known [328,329]. Some ligases contain a phosphotyrosine-binding domain therefore only transferring ubiquitin to the responsible target protein after its tyrosine phosphorylation. This has intensively been studied for receptor tyrosine kinases which can be ubiquitinated by one of the Casitas b-lineage lymphoma (cbl) RING-type ubiquitin ligases [330]. Alternatively, phosphorylation activates binding of WW domain-containing ligases to target proteins as e.g. shown for CXCR-4 that binds AIP4/Itch ligase after phosphorylation [331]. Other E3 ligases do not bind directly to the target protein but rather need adapter proteins like members of the arrestin family that bind directly the cell surface protein. This has been shown, e.g., for neural precursor cell expressed, developmentally down-regulated 4 (Nedd4) ligase family members [332]. However, adding more complexity to this system, some E2 enzymes can transfer ubiquitin without interaction with an E3 ligase [333].
Ubiquitin contains seven lysine residues to which other ubiquitin molecules can be attached thereby generating ubiquitin chains of different length, structure and exhibiting different functions [334]. Therefore different types of ubiquitination are possible: mono-ubiquitination when only a single ubiquitin molecule is attached to a target protein; multiple mono-ubiquitination when ubiquitin is attached to several lysine residues of the target protein; and poly-ubiquitination when a ubiquitin chain is covalently linked to the target protein [335]. Classically, ubiquitin chains that are linked by K48 were considered as a signal for proteasomal degradation. Protein degradation by the proteasome is virtually involved in any cellular process. The proteasome is composed of a core particle (20S) containing multiple proteolytic sites and a regulatory 19S particle that is responsible for access of the substrates to the 20S core. After entrance into an internal chamber of the 20S particle ubiquitin is cleaved from the substrate to enter the cellular ubiquitin pool and the substrates are hydrolyzed [336]. The importance of proteasomal degradation is underlined by the development of several diseases attributable to genetic mutations in one of the components of the ubiquitin-proteasome system. Protein accumulation results in many diseases like several neurodegenerative disorders, cystic fibrosis, Liddle syndrome and many cancers [337]. Additional to its classical function, several studies discovered that K48-poly-ubiquitin chains also mediate non-proteolytical functions and further that all seven lysine residues of ubiquitin can be involved in building ubiquitin chains thus generating chains of different length and structure. It is now also known that the proteasome accepts other ubiquitin chains than K48-linked chains [338,339].
K63-linked ubiquitin chains are involved in many processes: protein trafficking by ubiquitination of cell surface proteins thereby directing them to the lysosomal degradation pathway [340]; DNA repair by ubiquitination of histone proteins resulting in non-homologues end joining and homologue recombination, respectively, after DNA damage [341,342]; inflammation e.g., by activation of the NFκB pathway by attachment of ubiquitin chains to different effectors within this cascade [343]; and regulation of ribosomal protein synthesis by attachment of ubiquitin to one of the ribosome subunits, L28 [344].
The functions of other ubiquitin-linked chains are not well understood. Some results suggested K11-linked ubiquitin chains preferentially in endoplasmic reticulum-associated degradation (ERAD), Tumor Necrosis Factor signaling and Anaphase-Promoting Complex-mediated proteolysis [345,346,347]. Additionally, K27- and K33-linked ubiquitin chains have been implicated in stress response and finally K29-linked chains seem to play a role in lysosomal degradation and ubiquitin fusion protein degradation [348,349,350]. Additionally, multiple mono-ubiquitination has been shown to be involved in protein trafficking whereas single mono-ubiquitination is needed for sorting membrane proteins to the endosome and lysosome [351,352,353,354].
Specifically, there is increasing evidence that ubiquitin can act either as an endocytosis or sorting signal for cell surface proteins. For its action as an endocytosis signal usually multiple ubiquitin molecules or K63-linked poly-ubiquitin chains are necessary. This is attributable to the weak affinity of proteins of the endocytic machinery to a single ubiquitin tag. Usually ubiquitin signals are recognized by proteins containing ubiquitin-binding domains (UBDs). Thus far, approximately 300 proteins belonging to 20 families have been identified and more and more UBD-domain-containing proteins are being discovered [354,355,356]. UBDs bind to hydrophobic patches of ubiquitin and—depending on the type of ubiquitination—the hydrophobic patches are more (K63-linked ubiquitin chains) or less (K48-linked ubiquitin chains) exposed [332]. Binding of adjacent ubiquitin molecules within a ubiquitin chain might increase the affinity to UBDs. Additionally, the cells use other supporting mechanisms for efficient binding of ubiquitinated cargo proteins [332]. The best characterized adapter molecules that link ubiquitinated proteins to the clathrin-dependent endocytosis are Eps15 and Epsin which prefer K63-linked chains although it has recently been shown that one ubiquitin molecule is sufficient to recruit Eps15 to endosomes [357,358,359,360,361]. Altogether, to act as a sorting signal multiple mono-ubiquitination is favored over mono-ubiquitination. Proteins that appear after their internalization in endosomes may contain ubiquitin chains which may function either as a better binding motif for sorting molecules or as a buffer against the function of de-ubiquitinating enzymes (DUBs) [362]. In this regard, it has also been hypothesized that ubiquitination of cell surface receptors and subsequent lysosomal degradation may also play an essential role in the prevention of recycling of non-functional proteins to the cell surface [363].
Furthermore, it has been observed that ubiquitin can be attached to substrate proteins at different steps of endocytosis. It may, for example, regulate the first endocytic steps if it is attached to proteins that are still present at the plasma membrane, whereas it can serve as a lysosomal degradation signal if attached after internalization [364].
Like other posttranslational modifications ubiquitination is a reversible process. While attachment of ubiquitin is mediated by E1, E2 and E3 enzymes the specific removal of ubiquitin is catalyzed by DUBs. Nearly 100 DUBs are encoded in the human genome and they belong to five different families [365]. In humans four members of the ubiquitin C-terminal hydrolases (UCH), 55 active members of ubiquitin-specific hydrolases (USP), four members of the Machado Joseph disease domain proteins (MJD), 14 members of the ovarian tumor proteins (OTU) and seven active members of the JAB1, MPN, MOV34 metalloenzymes (JAMM) have been identified [340]. One important function of DUBs is to prevent cell surface molecules from degradation by removing ubiquitin. After de-ubiquitination they will instead be recycled to the cell surface [366,367,368,369].
Altogether, these facts demonstrate that ubiquitination is a highly versatile and complex posttranslational modification whose function has not yet completely been understood. Much more research is necessary to clarify the details of the already known functions and biochemical characteristics of ubiquitin and to identify possibly further functions.

4. Ubiquitination of Ig Superfamily Members

4.1. Ubiquitination of NCAM

It was first described in 2007 that NCAM becomes ubiquitinated and that ubiquitination regulates NCAM’s endocytosis [370].
The first evidence for internalization of cell adhesion molecules was published by the group of Eric Kandel dealing with the NCAM homologue ApCAM in Aplysia californica [371]. They investigated mechanisms of long-term facilitation in Aplysia sensory neurons which can be modulated by repeated application of serotonin [372,373]. It has been shown that after treatment with serotonin the amount of ApCAM at the cell surface was decreased by approximately 50% and that the internalized ApCAM was sorted into prelysosomal-endosomal compartments, probably resulting in its lysosomal degradation. This result proved that a learning relevant neurotransmitter is able to stimulate receptor-mediated endocytosis of ApCAM. Concomitant with the downregulation of ApCAM, an increased expression of clathrin light chain was observed suggesting a possible clathrin-mediated endocytosis [374]. Clathrin-dependent but also lipid-raft-dependent endocytosis of NCAM140 and NCAM180 have been confirmed in later studies [370,375]. A constitutive internalization of ApCAM which like NCAM lacks a typical internalization sequence, was excluded [371,376]. After stimulation with serotonin ApCAM internalization appeared most prominently at sites of membrane apposition. The decrease of cell adhesion at these sites by removal of cell adhesion molecules is most likely important for defasciculation of the axonal processes potentially to favor synaptogenesis and therewith restructuring of the neuron architecture and memory formation [371,377,378].
In accordance with these early results, a reduced amount of NCAM180 was observed in the rat dentate gyrus 3–4 h after passive avoidance training which could be attributable to increased internalization and a possible subsequent degradation of NCAM as shown for ApCAM [371,376,379]. Minana et al., demonstrated later on that NCAM140 and NCAM180 are inducibly internalized in astrocytes by a clathrin-dependent mechanism [375]. In 2007 the developmentally regulated internalization of NCAM140 and NCAM180 was shown in neuronal cells. Interestingly, NCAM140 endocytosis seems to be relevant in immature neurons, whereas endocytosis of NCAM180 seems to be important when axon-dendrite interactions become established indicating a regulatory role of NCAM in neurite outgrowth or synapse stability, respectively. However, in contrast to ApCAM most of the internalized NCAM is recycled to the plasma membrane and only a small amount is degraded in lysosomes [370]. This discrepancy may be explained by the different systems in which the internalization was shown or by the different stimulation methods of ApCAM’s/NCAM’s endocytosis. Furthermore, NCAM internalization was detected in the soma, neurites and growth cones suggesting a role of its endocytosis in the different compartments. It is conceivable that NCAM internalization in neurites and growth cones might facilitate neurite growth in brain development and/or enhance synaptic plasticity in adult brain by translocalization of NCAM to other cell surface sites.
Additionally, it was shown that NCAM is constitutively ubiquitinated with a drastic increase of ubiquitination after induction of NCAM endocytosis. Overexpression of ubiquitin resulted in increased endocytosis, whereas NCAM degradation was unaffected [370]. These results gave the first direct evidence that ubiquitination of NCAM acts as a signal for its endocytosis, although the amount of cell surface NCAM was not significantly reduced. This could be attributable to the intensive recycling of NCAM [370]. Therefore the function of NCAM internalization is probably not its downregulation but could rather result firstly in the removal of NCAM interacting molecules from the cell surface or secondly in its redistribution to other cell surface sites. Supporting the first hypothesis, PSA downregulation seems to depend on clathrin-mediated NCAM endocytosis in a rhabdomyosarcoma cell line [380]. Whether this is also true for neuronal cells needs further investigation. Otherwise, a translocalization to other cell surface sites has been demonstrated for L1, supporting the latter hypothesis [152]. In a previous study, it appeared that all three main NCAM isoforms, NCAM180, NCAM140 and the GPI-anchored NCAM120, could be modified by ubiquitin 3–4 h after passive avoidance training [379]. Ubiquitination occurs typically in the cytosol, although extracellular ubiquitination has also been described which could explain a ubiquitination of NCAM120 [381]. However, in neuronal cells a deletion construct of NCAM missing the entire cytoplasmic domain (ΔCT) is not ubiquitinated (Figure 2) excluding a possible extracellular ubiquitination of NCAM. Interestingly, in Aplysia only the membrane spanning isoform of ApCAM is internalized but not the GPI-anchored isoform [376]. Whether NCAM120 is internalized in neurons still needs to be investigated. However, ubiquitination of an interaction partner of NCAM120 which could be co-precipitated with NCAM could explain this result.
Analysis of the ubiquitination type of NCAM revealed an exclusive multiple mono-ubiquitination of NCAM at the plasma membrane [370]. In yeast, mono-ubiquitination was shown to be sufficient to target membrane proteins for internalization [382,383,384,385]. However, in more complex eukaryotes a single ubiquitin represents a very weak endocytosis signal [386,387]. For this reason, multiple mono-ubiquitination of NCAM fits perfectly into the described models. K63-linked ubiquitin chains can also act as endocytosis signals, however, since NCAM is not poly-ubiquitinated, this could not be the responsible mechanism for NCAM internalization [370].
Interestingly, ApCAM endocytosis is regulated by the phosphorylation of a MAP kinase consensus motif in its cytoplasmic tail. The same motif is present in NCAM and may also regulate NCAM’s endocytosis. Therefore, it has been speculated that phosphorylation of NCAM precedes its ubiquitination by creating a binding site for (a) ubiquitin ligase(s) leading to subsequent ubiquitination and endocytosis [370]. After its internalization the ubiquitin residues are most likely removed from NCAM in sorting endosomes by the DUB activity of the ubiquitin C-terminal hydrolase L1 (UCHL1) resulting in recycling of NCAM to the cell surface whereas a small portion of NCAM—which probably still contains covalently attached ubiquitin—is lysosomally degraded [388]. The action of UCHL1 has earlier been suggested to play a role in NCAM expression. The decrease of NCAM180 after passive avoidance training is accompanied by an increased expression of UCHL1, suggesting that ubiquitination is involved in memory consolidation. However, the results of this study proposed a ubiquitin ligase function of UCHL1 [379]. UCHL1 can act as a ubiquitin ligase in its dimeric form but interestingly, monomeric UCHL1 is well known as DUB that hydrolyses and detaches ubiquitin from proteins, which was confirmed for NCAM [388,389,390]. The different activities on NCAM in both studies may be explained by the dimerization state of UCHL1, the mono-ubiquitin stabilizing function of UCHL1 given by its ability to bind ubiquitin non-covalently and save NCAM from degradation or by the different approaches [391]. Interestingly, only the constitutive ubiquitination of NCAM was regulated by UCHL1 DUB activity, whereas the induced ubiquitination after activation of NCAM’s endocytosis was not altered. In line with this observation is that the endocytosis rate of NCAM remains unchanged after overexpression of UCHL1. Nevertheless, lysosomal degradation of NCAM was reduced after overexpression of UCHL1 supporting a role of UCHL1 in NCAM functions after its internalization. Since the triggering with NCAM antibodies leads to NCAM clustering at the cell surface and a possible conformational change in the cytoplasmic tail of NCAM, the accessibility of NCAM for UCHL1 could be reduced [88]. This would explain that only the constitutively ubiquitinated cell surface NCAM could be de-ubiquitinated by UCHL1. After internalization of NCAM an UCHL1 binding site might be exposed allowing its binding and therewith rescueing NCAM from lysosomal degradation. An observed partial colocalization of internalized NCAM and UCHL1 strengthens this hypothesis [388]. However, it cannot be excluded that other DUBs are involved in the regulation of NCAM’s intracellular trafficking. More research needs to be conducted to reveal other regulative factors for the regulation of NCAM ubiquitination and endocytosis and the physiological role of these processes in brain development and synaptic plasticity in adult brain.
Figure 2. NCAM is ubiquitinated in the cytoplasmic region. B35 cells transfected with NCAM140 wild type (wt) or NCAM140 missing the entire cytoplasmic region (ΔCT), respectively, were lysed and subjected to immunoprecipitation (IP) with NCAM-specific antibodies. Immunoblot analysis (IB) was performed using ubiquitin-specific antibodies, recognizing mono- and polyubiquitinated proteins. The blot was reprobed with NCAM-specific antibodies as control.
Figure 2. NCAM is ubiquitinated in the cytoplasmic region. B35 cells transfected with NCAM140 wild type (wt) or NCAM140 missing the entire cytoplasmic region (ΔCT), respectively, were lysed and subjected to immunoprecipitation (IP) with NCAM-specific antibodies. Immunoblot analysis (IB) was performed using ubiquitin-specific antibodies, recognizing mono- and polyubiquitinated proteins. The blot was reprobed with NCAM-specific antibodies as control.
Biology 05 00001 g002

4.2. Ubiquitination of L1

The ubiquitination of L1 was shown independently by different groups in primary neurons and different cell lines [154,392,393]. L1 is mainly mono- or multiple mono-ubiquitinated at the cell surface whereas only a small part seems to be poly-ubiquitinated [154,392]. The function of L1’s poly-ubiquitination is not yet known. Whereas the multiple mono-ubiquitination of NCAM is important for its endocytosis and does not play a role in its degradation, in L1 the mono- and/or multiple mono-ubiquitination seems rather to play a role in its lysosomal degradation as shown by co-localization and chemical inhibition studies [154,370,392]. The lysosomal degradation of L1 was mainly observed in the cell somata compared to neurites. Therefore, it has been speculated that the ubiquitination regulates the appearance of L1 in axons by either degrading it after attachment of ubiquitin or transporting it by transcytosis to the correct place [152,392]. Thus, ubiquitination of L1 might represent a fine regulation of L1-dependent growth cone motility and collapse [151,394]. In this context it would be interesting to identify responsible E3 ligase(s) and DUB(s) that are involved in reversible ubiquitination of L1 and to analyze the expression of these enzymes during growth cone motility and after synapse formation.
The effect of ubiquitination on L1 endocytosis seems to be less clear. Although it has been shown that overexpression of ubiquitin did not change the internalization rate of L1, it might be possible that the overexpression of ubiquitin cannot be effective since the other enzymatic components of the ubiquitin system (i.e., E1, E2, and E3) are not overexpressed and may limit the entire process [370,392]. Otherwise, it has also been published that efficient internalization of L1 depends—at least partially—on its ubiquitination as demonstrated by using a ubiquitination-deficient L1 mutant [154]. However, the L1 mutant K11R missing all lysine residues used in this study may exhibit an altered three-dimensional structure of the cytoplasmic tail thereby changing other interactions necessary for optimal internalization. This aspect needs to be clarified in future studies.
Ubiquitination often depends on preceding phosphorylation thereby creating a binding motif for the respective E3 ligase(s) [395,396,397]. Consistent with this, phosphorylation of Y1176 in L1’s cytoplasmic tail plays a role in its ubiquitination. However, it is discussed controversially whether phosphorylation of Y1176 increases or decreases the ubiquitination of L1. Two different L1 mutants abolishing phosphorylation at position 1176 have been used to study the impact of Y1176 phosphorylation on ubiquitination level. The mutation Y1176 to phenylalanine exhibited reduced ubiquitination whereas mutation to alanine resulted in increased ubiquitination [154,392]. Both effects are conceivable: If phosphorylation precedes ubiquitination of L1 it is logical that a non-phosphorylated mutant exhibits lower ubiquitination levels than the wild type L1. This can only be clarified after identification of the responsible ubiquitin ligase. Another possibility is that AP-2 binding which is only possible in the non-phosphorylated form of Y1176 interferes with binding of the corresponding ubiquitin ligase thus explaining the reduced ubiquitination [147]. However, ubiquitination seems rather to interfere with ezrin-binding since ezrin does not co-precipitate with L1 after overexpression of ubiquitin [154]. A subsequent study showed that ubiquitination takes indeed place at the ERM binding site. Mutation analysis revealed that K1147 and/or K1150 of the cytosolic tail of L1 are the main ubiquitination sites of L1 [398]. It is furthermore imaginable that ubiquitin is attached as a result of endocytosis induction of L1 which would explain higher ubiquitination levels of L1Y1176A mutant. This would also explain the dependency between ubiquitination and the internalization rate as shown by Aikawa [154]. In this case, it is tempting to speculate that L1 ubiquitination might regulate lateral movement of L1 in the plasma membrane regulating neurite outgrowth and branching [141,142,143,144]. In agreement, it has been shown that Rabex-5 co-precipitates with L1, with higher affinity after induction of L1 endocytosis and with low affinity to a ubiquitin-deficient mutant. The interaction positively regulates L1’s internalization and seems to increase its ubiquitination thereby regulating endocytosis and proper postendocytic trafficking to lysosomes making the possible regulatory mechanisms more complex [154].
Altogether, subsequent studies are needed to investigate the cellular mechanisms of L1 ubiquitination and the role in L1-dependent functions in the nervous system and cancer development.

4.3. Ubiquitination of MCAM

Recently, it could be demonstrated that also MCAM can be ubiquitinated [219]. MCAM has classically been described in melanoma cells but it also seems to play a role in hepatocellular carcinoma via its interplay with ALCAM which can itself be ubiquitinated (see also Section 4.4) [399]. Earlier studies have shown that ALCAM is a valuable cancer stem cell marker in various cancer types and plays a role in tumor progression, e.g., in breast cancer [400,401,402,403]. ALCAM and MCAM in combination seem to be promising candidates as diagnostic markers for hepatocellular carcinoma and their crosstalk has intensively been investigated in a hepatocellular carcinoma cell culture model, the Bel-7402 cells [219,239,404]. In this cell culture model, ALCAM can regulate expression of MCAM at the level of degradation. It activates Akt which leads to subsequent phosphorylation and activation of c-Raf, MEK1 and ERK1 causing increased ubiquitination and hence downregulation of the two E3 ligases Smurf1 and βTrCP. These enzymes are responsible for ubiquitination of MCAM [219]. Therefore, by enhancing degradation of Smurf1 and βTrCP MCAM becomes less ubiquitinated and is stabilized at the cell surface explaining why downregulation of ALCAM decreases MCAM levels in Bel-7402 cells [219]. Interestingly, MCAM is the only cell adhesion molecule of the Ig superfamily for which ubiquitin ligases have been identified so far. In the same cell culture system expression of ALCAM negatively correlated with COP1, a RING type ubiquitin ligase. Therefore, COP1 could be a potential ubiquitin ligase regulating ALCAM degradation and hence its tumorigenic potential although further studies need to verify the involvement of COP1 in ALCAM degradation [405]. ALCAM is also ubiquitinated and subsequently downregulated in retinal ganglion cells from chicken (see also Section 4.4) indicating that ALCAM is degraded after ubiquitination independent from the species and cell culture system [399]. It has been shown for MCAM that it can be internalized after stimulation with its ligand Wnt5a resulting in its translocation to a polarized structure in a melanoma cell culture model. This process is necessary for cell orientation, polarity and directed migration [406]. It would be interesting to analyze whether MCAM endocytosis is regulated by ubiquitin. It has also not yet been investigated by which type of ubiquitination MCAM is modified or whether it is degraded lysosomally or by the proteasomal pathway. Nevertheless, the results showing a tightly regulated expression of MCAM by ALCAM and the ubiquitination of MCAM provides novel and highly interesting insights into the cellular regulation of MCAM and hence into hepatocellular carcinoma and potential future therapeutic approaches.

4.4. Ubiquitination of ALCAM

The ubiquitination of membrane-integrated ALCAM was first described in 2008 in retinal ganglion cells isolated from chicken [399]. In its cytoplasmic domain ALCAM contains seven lysine residues providing potential docking sites for ubiquitin. However, since ALCAM is probably mono- and/or di-ubiquitinated only one and/or two of the lysine residues seem to be used [399].
ALCAM is internalized via the clathrin-dependent pathway followed by recycling of ALCAM to the cell surface [305,399]. In cancer cells internalization of ALCAM seems to be necessary for rearrangement of cell–cell contacts since the maximum of internalized ALCAM has been observed at the cleavage furrow during cytokinesis [305]. In neurons, the endocytosis has been detected in the central domain of growth cones. Inhibition of clathrin-dependent endocytosis resulted in random axon growth in contrast to a control condition where axons grew preferentially on an ALCAM substrate proving that endocytosis of ALCAM is essential for axon navigation [399]. It has been hypothesized that also other members of the Ig superfamily use endocytosis to regulate their amount and localization at the cell surface in order to modulate neurite outgrowth and cell adhesion e.g., ApCAM, L1, and NCAM [149,150,152,370,376]. Therefore, it is conceivable that varying concentrations of ALCAM could represent one important mechanism in the development of the nervous system to regulate axonal outgrowth and pathfinding.
Interestingly, in cancer cells internalization of ALCAM happens much more slowly than in neurons. This fact supports a role of ALCAM internalization in rapid regulation of growth cone navigation in response to environmental changes. However, a small part of ALCAM in neurons seems to be internalized clathrin-independently and more slowly than the clathrin-dependent part of ALCAM. In leukocytes ALCAM is almost exclusively present in cholesterol-enriched microdomains and it is most likely that it exhibits the same distribution in other cell types [299]. However, it has been described that lipid rafts are excluded from the endocytic vesicles during clathrin-mediated endocytosis indicating that clathrin-mediated endocytosis takes place independent of lipid rafts [407]. On the other hand, it has been reported that clathrin-mediated endocytosis can be sensitive to cholesterol depletion indicating that the cellular mechanisms are not yet clear [408,409]. In this context, it would be interesting to investigate whether ALCAM undergoes clathrin-dependent or -independent endocytosis in the lipid-raft fraction.
Ubiquitination of ALCAM regulates the amount at the cell surface by enhancing its degradation probably by the lysosomal pathway as determined by analyzing the number and diameter of ALCAM-positive vesicles representing multivesicular bodies [399,410]. Overexpression of ubiquitin has no effect on the endocytosis of ALCAM although the authors discuss the possibility that an increased ubiquitination may not affect its internalization rate since it has been shown earlier that an enhanced ubiquitin pool does not affect internalization of (overexpressed) trans-membrane proteins. This could probably be due to sufficient endogenous ubiquitin levels already allowing optimal internalization. Mutation of corresponding lysine residues might solve this question.
Furthermore, it still needs to be investigated whether ubiquitination might affect the slow endocytosis of ALCAM which seems to be clathrin-independent [399]. Concomitantly, ubiquitin overexpression results in a higher percentage of cells with internalized ALCAM pointing to a potential role of ubiquitin in ALCAM’s slow endocytosis. Several reports support the view that ubiquitination might regulate either lipid-raft-dependent or clathrin-dependent endocytic pathways [360,386,410,411,412]. The authors discuss also that the kind of ubiquitination might influence the fate of ALCAM after its endocytosis: Di-ubiquitinated ALCAM would be more efficiently sorted and hence degraded than mono-ubiquitinated ALCAM since the affinity between ubiquitin-sorting receptor and ubiquitinated ALCAM is higher between two ubiquitin molecules and adapter proteins than one ubiquitin molecule and the respective adapter protein [413,414].
Altogether, more studies are needed to investigate the cellular mechanisms of ALCAM endocytosis and ubiquitination and the functional role of these processes. This aspect is also especially very interesting in cancer cells since ALCAM has been suggested to play a crucial role in cancer progression.

5. Conclusions and Perspectives

Some of the here described cell adhesion molecules have been known for a long time and have been analyzed in detail. However, their ubiquitination has only been shown in the last decade. One reason is most likely the ongoing ubiquitin research giving continuously more insight into the complexity and function of ubiquitin modifications.
For the here described cell adhesion molecules of the Ig superfamily, ubiquitination plays a role in internalization and/or intracellular trafficking, i.e., lysosomal degradation, transcytosis or recycling (Figure 3). The discovery that cell adhesion molecules can be posttranslationally modified by ubiquitin adds an additional regulatory factor for their functions. The here mentioned cell adhesion molecules play a role in the nervous system and in tumor development. In the nervous system, their ubiquitination might be implicated in the fine regulation of their presence at the cell surface and therefore allow rapid adaptation to environmental influences that occur during brain development or synaptic plasticity during learning processes. Therefore, it might be highly interesting to investigate other CAMs for their internalization and ubiquitination. In this context, SynCAM could be a favorable candidate for further analysis. It is expressed at synaptic connections and is critical for assembly, organization and maintenance of synapses [415,416,417]. Hence, it is tempting to speculate that a possible internalization and regulation by ubiquitin might regulate synapse development and stability.
In cancer cells, the ubiquitination might be responsible for the fine regulation of cell adhesion and migration of cancer cells thus being potentially involved in their metastatic potential. Further studies are needed to clarify these aspects. It would be further interesting to analyze whether different cellular processes are active in tumorigenic cells than in neural cells. An example of a different cellular mechanism in cancer is likely provided by the cell adhesion molecule “roundabout” (Robo). The Robo family represents, together with its ligands, the Slits, essential repellent axon guidance molecules during embryonic development and has several other functions [418,419,420]. Removal of Robo from the cell surface by clathrin-dependent endocytosis and subsequent lysosomal targeting results in inappropriate midline crossing [421]. Otherwise, proteasomal degradation of Robo after its ubiquitination regulates breast cancer cell migration indicating the involvement of a different cellular mechanism in cancer cells. Therefore, it would be interesting to analyze whether Robo ubiquitination also plays a role in embryonic neural development and which process is regulated by its possible ubiquitination [422]. Similarly, for the cell adhesion molecule “deleted in colon carcinoma” (DCC), a proteasomal degradation has been shown after ubiquitination; however, a role of ubiquitination in axonal pathfinding and/or cancer has not been investigated yet [423]. Even less is known for other cell adhesion molecules, like CHL1, an L1 related cell adhesion molecule. CHL1 undergoes endocytosis which is required for CHL1-dependent neuritogenesis. A possible regulation by ubiquitination has not been analyzed so far [424].
Figure 3. Overview about the function of ubiquitination for different Ig superfamily cell adhesion molecules. All depicted molecules are internalized after a specific stimulus. Ubiquitination (U) of NCAM increases its endocytosis (↑) but has no effect on its lysosomal degradation after overexpression of ubiquitin. However, cleavage of ubiquitin by the de-ubiquitinating enzyme UCHL1 results in decreased lysosomal degradation of NCAM thus favoring its recycling to the cell surface (↑). For L1 it could be shown that ubiquitination increases its lysosomal degradation (↑) whereas it is not yet clear whether it also upregulates its internalization (↑?). ALCAM’s endocytosis does not seem to be regulated by its ubiquitination although ubiquitin overexpression decreases ALCAM expression. This is most likely attributable to increased lysosomal degradation (↑). MCAM ubiquitination is mediated by the E3 ligases Smurf1 and βTrCP by a tight crosstalk with ALCAM-mediated signal transduction and leads to MCAM downregulation. The detailed mechanism of its degradation has not yet been investigated (?).
Figure 3. Overview about the function of ubiquitination for different Ig superfamily cell adhesion molecules. All depicted molecules are internalized after a specific stimulus. Ubiquitination (U) of NCAM increases its endocytosis (↑) but has no effect on its lysosomal degradation after overexpression of ubiquitin. However, cleavage of ubiquitin by the de-ubiquitinating enzyme UCHL1 results in decreased lysosomal degradation of NCAM thus favoring its recycling to the cell surface (↑). For L1 it could be shown that ubiquitination increases its lysosomal degradation (↑) whereas it is not yet clear whether it also upregulates its internalization (↑?). ALCAM’s endocytosis does not seem to be regulated by its ubiquitination although ubiquitin overexpression decreases ALCAM expression. This is most likely attributable to increased lysosomal degradation (↑). MCAM ubiquitination is mediated by the E3 ligases Smurf1 and βTrCP by a tight crosstalk with ALCAM-mediated signal transduction and leads to MCAM downregulation. The detailed mechanism of its degradation has not yet been investigated (?).
Biology 05 00001 g003
Altogether, although cell adhesion molecules have been analyzed since the 1970s the molecular mechanisms regulating the cellular functions of cell adhesion molecules are so far not well understood. Their ubiquitination represents a novel approach for understanding their function in more detail. Especially in tumorigenesis, their ubiquitination could represent a new avenue for potential therapies since the ubiquitination is highly specifically regulated by the ubiquitin ligases. In this context, some ubiquitin ligase inhibitors have already been developed, however, they target mainly proteins that are proteasomally degraded, e.g., p53, and there was not much clinical advance in treating selected tumors [327]. Furthermore, MCAM is the only Ig superfamily member for which ubiquitin ligases have been identified so far. Therefore, much more research is needed to identify the regulation of their ubiquitination (i.e., identification of ubiquitin ligases (DUBs)) and to clarify the physiological role of ubiquitination of Ig family cell adhesion molecules in different cells and conditions.

Acknowledgments

We thank Sarah Förster for critical reading of the manuscript.

Author Contributions

Mirka Homrich, Hilke Wobst and Simone Diestel participated in the conception, literature review and writing of the manuscript; and Ingo Gotthard participated in the writing of the manuscript, literature review and design of the figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Edelman, G.M. Cell adhesion molecules. Science 1983, 219, 450–457. [Google Scholar] [CrossRef] [PubMed]
  2. Edelman, G.M.; Crossin, K.L. Cell adhesion molecules: Implications for a molecular histology. Annu. Rev. Biochem. 1991, 60, 155–190. [Google Scholar] [CrossRef] [PubMed]
  3. Takeichi, M. Cadherins: A molecular family important in selective cell-cell adhesion. Annu. Rev. Biochem. 1990, 59, 237–252. [Google Scholar] [CrossRef] [PubMed]
  4. Reichardt, L.F.; Tomaselli, K.J. Extracellular matrix molecules and their receptors: Functions in neural development. Annu. Rev. Neurosci. 1991, 14, 531–570. [Google Scholar] [CrossRef] [PubMed]
  5. Lasky, L.A. Selectin-carbohydrate interactions and the initiation of the inflammatory response. Annu. Rev. Biochem. 1995, 64, 113–139. [Google Scholar] [CrossRef] [PubMed]
  6. Brümmendorf, T.; Rathjen, F.G. Structure/function relationships of axon-associated adhesion receptors of the immunoglobulin superfamily. Curr. Opin. Neurobiol. 1996, 6, 584–593. [Google Scholar] [CrossRef]
  7. Gumbiner, B.M. Cell adhesion: The molecular basis of tissue architecture and morphogenesis. Cell 1996, 84, 345–357. [Google Scholar] [CrossRef]
  8. Murai-Takebe, R.; Noguchi, T.; Ogura, T.; Mikami, T.; Yanagi, K.; Inagaki, K.; Ohnishi, H.; Matozaki, T.; Kasuga, M. Ubiquitination-mediated regulation of biosynthesis of the adhesion receptor SHPS-1 in response to endoplasmic reticulum stress. J. Biol. Chem. 2004, 279, 11616–11625. [Google Scholar] [CrossRef] [PubMed]
  9. Jørgensen, O.S.; Bock, E. Brain specific synaptosomal membrane proteins demonstrated by crossed immunoelectrophoresis. J. Neurochem. 1974, 23, 879–880. [Google Scholar] [CrossRef] [PubMed]
  10. Rutishauser, U.; Thiery, J.P.; Brackenbury, R.; Sela, B.A.; Edelman, G.M. Mechanisms of adhesion among cells from neural tissues of the chick embryo. Proc. Natl. Acad. Sci. USA 1976, 73, 577–581. [Google Scholar] [CrossRef] [PubMed]
  11. Doherty, P.; Fruns, M.; Seaton, P.; Dickson, G.; Barton, C.H.; Sears, T.A.; Walsh, F.S. A threshold effect of the major isoforms of NCAM on neurite outgrowth. Nature 1990, 343, 464–466. [Google Scholar] [CrossRef] [PubMed]
  12. Cremer, H.; Lange, R.; Christoph, A.; Plomann, M.; Vopper, G.; Roes, J.; Brown, R.; Baldwin, S.; Kraemer, P.; Scheff, S.; et al. Inactivation of the N-CAM gene in mice results in size reduction of the olfactory bulb and deficits in spatial learning. Nature 1994, 367, 455–459. [Google Scholar] [CrossRef] [PubMed]
  13. Cremer, H.; Chazal, G.; Goridis, C.; Represa, A. NCAM is essential for axonal growth and fasciculation in the hippocampus. Mol. Cell. Neurosci. 1997, 8, 323–335. [Google Scholar] [CrossRef] [PubMed]
  14. Yin, X.; Watanabe, M.; Rutishauser, U. Effect of polysialic acid on the behavior of retinal ganglion cell axons during growth into the optic tract and tectum. Development 1995, 121, 3439–3446. [Google Scholar] [PubMed]
  15. Müller, D.; Wang, C.; Skibo, G.; Toni, N.; Cremer, H.; Calaora, V.; Rougon, G.; Kiss, J.Z. PSA-NCAM is required for activity-induced synaptic plasticity. Neuron 1996, 17, 413–422. [Google Scholar] [CrossRef]
  16. Hu, H.; Tomasiewicz, H.; Magnuson, T.; Rutishauser, U. The role of polysialic acid in migration of olfactory bulb interneuron precursors in the subventricular zone. Neuron 1996, 16, 735–743. [Google Scholar] [CrossRef]
  17. Chazal, G.; Durbec, P.; Jankovski, A.; Rougon, G.; Cremer, H. Consequences of neural cell adhesion molecule deficiency on cell migration in the rostral migratory stream of the mouse. J. Neurosci. 2000, 20, 1446–1457. [Google Scholar] [PubMed]
  18. Diestel, S.; Hinkle, C.L.; Schmitz, B.; Maness, P.F. NCAM140 stimulates integrin-dependent cell migration by ectodomain shedding. J. Neurochem. 2005, 95, 1777–1784. [Google Scholar] [CrossRef] [PubMed]
  19. Hinkle, C.L.; Diestel, S.; Lieberman, J.; Maness, P.F. Metalloprotease-induced ectodomain shedding of neural cell adhesion molecule (NCAM). J. Neurobiol. 2006, 66, 1378–1395. [Google Scholar] [CrossRef] [PubMed]
  20. Poltorak, M.; Khoja, I.; Hemperly, J.J.; Williams, J.R.; El-Mallakh, R.; Freed, W.J. Disturbances in cell recognition molecules (N-CAM and L1 antigen) in the CSF of patients with schizophrenia. Exp. Neurol. 1995, 131, 266–272. [Google Scholar] [CrossRef]
  21. Honer, W.G.; Falkai, P.; Young, C.; Wang, T.; Xie, J.; Bonner, J.; Hu, L.; Boulianne, G.L.; Luo, Z.; Trimble, W.S. Cingulate cortex synaptic terminal proteins and neural cell adhesion molecule in schizophrenia. Neuroscience 1997, 78, 99–110. [Google Scholar] [CrossRef]
  22. Van Kammen, D.P.; Poltorak, M.; Kelley, M.E.; Yao, J.K.; Gurklis, J.A.; Peters, J.L.; Hemperly, J.J.; Wright, R.D.; Freed, W.J. Further studies of elevated cerebrospinal fluid neuronal cell adhesion molecule in schizophrenia. Biol. Psychiatry 1998, 43, 680–686. [Google Scholar] [CrossRef]
  23. Vawter, M.P.; Howard, A.L.; Hyde, T.M.; Kleinman, J.E.; Freed, W.J. Alterations of hippocampal secreted N-CAM in bipolar disorder and synaptophysin in schizophrenia. Mol. Psychiatry 1999, 4, 467–475. [Google Scholar] [CrossRef] [PubMed]
  24. Vawter, M.P.; Frye, M.A.; Hemperly, J.J.; VanderPutten, D.M.; Usen, N.; Doherty, P.; Saffell, J.L.; Issa, F.; Post, R.M.; Wyatt, R.J.; et al. Elevated concentration of N-CAM VASE isoforms in schizophrenia. J. Psychiatr. Res. 2000, 34, 25–34. [Google Scholar] [CrossRef]
  25. Vawter, M.P.; Freed, W.J.; Kleinman, J.E. Neuropathology of bipolar disorder. Biol. Psychiatry 2000, 48, 486–504. [Google Scholar] [CrossRef]
  26. Panicker, A.K.; Buhusi, M.; Thelen, K.; Maness, P.F. Cellular signalling mechanisms of neural cell adhesion molecules. Front. Biosci. 2003, 8, d900–d911. [Google Scholar] [CrossRef] [PubMed]
  27. Pillai-Nair, N.; Panicker, A.K.; Rodriguiz, R.M.; Gilmore, K.L.; Demyanenko, G.P.; Huang, J.Z.; Wetsel, W.C.; Maness, P.F. Neural cell adhesion molecule-secreting transgenic mice display abnormalities in GABAergic interneurons and alterations in behavior. J. Neurosci. 2005, 25, 4659–4671. [Google Scholar] [CrossRef] [PubMed]
  28. Walmod, P.S.; Kolkova, K.; Berezin, V.; Bock, E. Zippers make signals: NCAM-mediated molecular interactions and signal transduction. Neurochem. Res. 2004, 29, 2015–2035. [Google Scholar] [CrossRef] [PubMed]
  29. Persohn, E.; Pollerberg, G.E.; Schachner, M. Immunoelectron-microscopic localization of the 180 kD component of the neural cell adhesion molecule N-CAM in postsynaptic membranes. J. Comp. Neurol. 1989, 288, 92–100. [Google Scholar] [CrossRef] [PubMed]
  30. Dityatev, A.; Dityateva, G.; Schachner, M. Synaptic strength as a function of post- versus presynaptic expression of the neural cell adhesion molecule NCAM. Neuron 2000, 26, 207–217. [Google Scholar] [CrossRef]
  31. Noble, M.; Albrechtsen, M.; Møller, C.; Lyles, J.; Bock, E.; Goridis, C.; Watanabe, M.; Rutishauser, U. Glial cells express N-CAM/D2-CAM-like polypeptides in vitro. Nature 1985, 316, 725–728. [Google Scholar] [CrossRef] [PubMed]
  32. Andersson, A.M.; Olsen, M.; Zhernosekov, D.; Gaardsvoll, H.; Krog, L.; Linnemann, D.; Bock, E. Age-related changes in expression of the neural cell adhesion molecule in skeletal muscle: A comparative study of newborn, adult and aged rats. Biochem. J. 1993, 290, 641–648. [Google Scholar] [CrossRef] [PubMed]
  33. Gaardsvoll, H.; Krog, L.; Zhernosekov, D.; Andersson, A.M.; Edvardsen, K.; Olsen, M.; Bock, E.; Linnemann, D. Age-related changes in expression of neural cell adhesion molecule (NCAM) in heart: A comparative study of newborn, adult and aged rats. Eur. J. Cell Biol. 1993, 61, 100–107. [Google Scholar] [PubMed]
  34. Langley, O.K.; Aletsee-Ufrecht, M.C.; Grant, N.J.; Gratzl, M. Expression of the neural cell adhesion molecule NCAM in endocrine cells. J. Histochem. Cytochem. 1989, 37, 781–791. [Google Scholar] [CrossRef] [PubMed]
  35. Moller, C.J.; Christgau, S.; Williamson, M.R.; Madsen, O.D.; Niu, Z.P.; Bock, E.; Baekkeskov, S. Differential expression of neural cell adhesion molecule and cadherins in pancreatic islets, glucagonomas, and insulinomas. Mol. Endocrinol. 1992, 6, 1332–1342. [Google Scholar] [PubMed]
  36. Thor, G.; Probstmeier, R.; Schachner, M. Characterization of the cell adhesion molecules L1, N-CAM and J1 in the mouse intestine. EMBO J. 1987, 6, 2581–2586. [Google Scholar] [PubMed]
  37. Lanier, L.L.; Le, A.M.; Civin, C.I.; Loken, M.R.; Phillips, J.H. The relationship of CD16 (Leu-11) and Leu-19 (NKH-1) antigen expression on human peripheral blood NK cells and cytotoxic T lymphocytes. J. Immunol. 1986, 136, 4480–4486. [Google Scholar] [PubMed]
  38. Schmidt, R.E.; Murray, C.; Daley, J.F.; Schlossman, S.F.; Ritz, J. A subset of natural killer cells in peripheral blood displays a mature T cell phenotype. J. Exp. Med. 1986, 164, 351–356. [Google Scholar] [CrossRef] [PubMed]
  39. Crossin, K.L.; Chuong, C.M.; Edelman, G.M. Expression sequences of cell adhesion molecules. Proc. Natl. Acad. Sci. USA 1985, 82, 6942–6946. [Google Scholar] [CrossRef] [PubMed]
  40. Sasaki, H.; Yoshida, K.; Ikeda, E.; Asou, H.; Inaba, M.; Otani, M.; Kawase, T. Expression of the neural cell adhesion molecule in astrocytic tumors: An inverse correlation with malignancy. Cancer 1998, 82, 1921–1931. [Google Scholar] [CrossRef]
  41. Krushel, L.A.; Tai, M.H.; Cunningham, B.A.; Edelman, G.M.; Crossin, K.L. Neural cell adhesion molecule (N-CAM) domains and intracellular signaling pathways involved in the inhibition of astrocyte proliferation. Proc. Natl. Acad. Sci. USA 1998, 95, 2592–2596. [Google Scholar] [CrossRef] [PubMed]
  42. Glüer, S.; Zense, M.; Radtke, E.; von Schweinitz, D. Polysialylated neural cell adhesion molecule in childhood ganglioneuroma and neuroblastoma of different histological grade and clinical stage. Langenbecks Arch. Surg. 1998, 383, 340–344. [Google Scholar] [PubMed]
  43. Komminoth, P.; Roth, J.; Saremaslani, P.; Matias-Guiu, X.; Wolfe, H.J.; Heitz, P.U. Polysialic acid of the neural cell adhesion molecule in the human thyroid: A marker for medullary thyroid carcinoma and primary C-cell hyperplasia. An immunohistochemical study on 79 thyroid lesions. Am. J. Surg. Pathol. 1994, 18, 399–411. [Google Scholar] [CrossRef] [PubMed]
  44. Lantuejoul, S.; Moro, D.; Michalides, R.J.; Brambilla, C.; Brambilla, E. Neural cell adhesion molecules (NCAM) and NCAM-PSA expression in neuroendocrine lung tumors. Am. J. Surg. Pathol. 1998, 22, 1267–1276. [Google Scholar] [CrossRef] [PubMed]
  45. Trouillas, J.; Daniel, L.; Guigard, M.P.; Tong, S.; Gouvernet, J.; Jouanneau, E.; Jan, M.; Perrin, G.; Fischer, G.; Tabarin, A.; et al. Polysialylated neural cell adhesion molecules expressed in human pituitary tumors and related to extrasellar invasion. J. Neurosurg. 2003, 98, 1084–1093. [Google Scholar] [CrossRef] [PubMed]
  46. Zecchini, S.; Cavallaro, U. Neural cell adhesion molecule in cancer: Expression and mechanisms. Adv. Exp. Med. Biol. 2010, 663, 319–333. [Google Scholar] [PubMed]
  47. Zecchini, S.; Bombardelli, L.; Decio, A.; Bianchi, M.; Mazzarol, G.; Sanguineti, F.; Aletti, G.; Maddaluno, L.; Berezin, V.; Bock, E.; et al. The adhesion molecule NCAM promotes ovarian cancer progression via FGFR signalling. EMBO Mol. Med. 2011, 3, 480–494. [Google Scholar] [CrossRef] [PubMed]
  48. Ćirović, S.; Vještica, J.; Mueller, C.A.; Tatić, S.; Vasiljević, J.; Milenković, S.; Mueller, G.A.; Marković-Lipkovski, J. NCAM and FGFR1 coexpression and colocalization in renal tumors. Int. J. Clin. Exp. Pathol. 2014, 7, 1402–1414. [Google Scholar] [PubMed]
  49. Johnson, J.P. Cell adhesion molecules of the immunoglobulin supergene family and their role in malignant transformation and progression to metastatic disease. Cancer Metastasis Rev. 1991, 10, 11–22. [Google Scholar] [CrossRef] [PubMed]
  50. Kaiser, U.; Auerbach, B.; Oldenburg, M. The neural cell adhesion molecule NCAM in multiple myeloma. Leuk. Lymphoma 1996, 20, 389–395. [Google Scholar] [CrossRef] [PubMed]
  51. Lipinski, M.; Hirsch, M.R.; Deagostini-Bazin, H.; Yamada, O.; Tursz, T.; Goridis, C. Characterization of neural cell adhesion molecules (NCAM) expressed by ewing and neuroblastoma cell lines. Int. J. Cancer 1987, 40, 81–86. [Google Scholar] [CrossRef] [PubMed]
  52. Moolenaar, C.E.; Pieneman, C.; Walsh, F.S.; Mooi, W.J.; Michalides, R.J. Alternative splicing of neural-cell-adhesion molecule mRNA in human small-cell lung-cancer cell line H69. Int. J. Cancer 1992, 51, 238–243. [Google Scholar] [CrossRef] [PubMed]
  53. Roth, J.; Zuber, C.; Wagner, P.; Taatjes, D.J.; Weisgerber, C.; Heitz, P.U.; Goridis, C.; Bitter-Suermann, D. Reexpression of poly(sialic acid) units of the neural cell adhesion molecule in Wilms tumor. Proc. Natl. Acad. Sci. USA 1988, 85, 2999–3003. [Google Scholar] [CrossRef] [PubMed]
  54. Jimbo, T.; Nakayama, J.; Akahane, K.; Fukuda, M. Effect of polysialic acid on the tumor xenografts implanted into nude mice. Int. J. Cancer 2001, 94, 192–199. [Google Scholar] [CrossRef] [PubMed]
  55. Seidenfaden, R.; Krauter, A.; Schertzinger, F.; Gerardy-Schahn, R.; Hildebrandt, H. Polysialic acid directs tumor cell growth by controlling heterophilic neural cell adhesion molecule interactions. Mol. Cell. Biol. 2003, 23, 5908–5918. [Google Scholar] [CrossRef] [PubMed]
  56. Kiselyov, V.V.; Soroka, V.; Berezin, V.; Bock, E. Structural biology of NCAM homophilic binding and activation of FGFR. J. Neurochem. 2005, 94, 1169–1179. [Google Scholar] [CrossRef] [PubMed]
  57. Soroka, V.; Kiryushko, D.; Novitskaya, V.; Ronn, L.C.; Poulsen, F.M.; Holm, A.; Bock, E.; Berezin, V. Induction of neuronal differentiation by a peptide corresponding to the homophilic binding site of the second Ig module of the neural cell adhesion molecule. J. Biol. Chem. 2002, 277, 24676–24683. [Google Scholar] [CrossRef] [PubMed]
  58. Horstkorte, R.; Schachner, M.; Magyar, J.P.; Vorherr, T.; Schmitz, B. The fourth immunoglobulin-like domain of NCAM contains a carbohydrate recognition domain for oligomannosidic glycans implicated in association with L1 and neurite outgrowth. J. Cell Biol. 1993, 121, 1409–1421. [Google Scholar] [CrossRef] [PubMed]
  59. Milev, P.; Maurel, P.; Häring, M.; Margolis, R.K.; Margolis, R.U. TAG-1/axonin-1 is a high-affinity ligand of neurocan, phosphacan/protein-tyrosine phosphatase-zeta/beta, and N-CAM. J. Biol. Chem. 1996, 271, 15716–15723. [Google Scholar] [PubMed]
  60. Heiland, P.C.; Griffith, L.S.; Lange, R.; Schachner, M.; Hertlein, B.; Traub, O.; Schmitz, B. Tyrosine and serine phosphorylation of the neural cell adhesion molecule L1 is implicated in its oligomannosidic glycan dependent association with NCAM and neurite outgrowth. Eur. J. Cell Biol. 1998, 75, 97–106. [Google Scholar] [CrossRef]
  61. Cavallaro, U.; Niedermeyer, J.; Fuxa, M.; Christofori, G. N-CAM modulates tumour-cell adhesion to matrix by inducing FGF-receptor signalling. Nat. Cell Biol. 2001, 3, 650–657. [Google Scholar] [CrossRef] [PubMed]
  62. Francavilla, C.; Loeffler, S.; Piccini, D.; Kren, A.; Christofori, G.; Cavallaro, U. Neural cell adhesion molecule regulates the cellular response to fibroblast growth factor. J. Cell Sci. 2007, 120, 4388–4394. [Google Scholar] [CrossRef] [PubMed]
  63. Kiselyov, V.V.; Skladchikova, G.; Hinsby, A.M.; Jensen, P.H.; Kulahin, N.; Soroka, V.; Pedersen, N.; Tsetlin, V.; Poulsen, F.M.; Berezin, V.; et al. Structural basis for a direct interaction between FGFR1 and NCAM and evidence for a regulatory role of ATP. Structure 2003, 11, 691–701. [Google Scholar] [CrossRef]
  64. Francavilla, C.; Cattaneo, P.; Berezin, V.; Bock, E.; Ami, D.; de Marco, A.; Christofori, G.; Cavallaro, U. The binding of NCAM to FGFR1 induces a specific cellular response mediated by receptor trafficking. J. Cell Biol. 2009, 187, 1101–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Brennaman, L.H.; Zhang, X.; Guan, H.; Triplett, J.W.; Brown, A.; Demyanenko, G.P.; Manis, P.B.; Landmesser, L.; Maness, P.F. Polysialylated NCAM and EphrinA/EphA regulate synaptic development of GABAergic interneurons in prefrontal cortex. Cereb. Cortex 2013, 23, 162–177. [Google Scholar] [CrossRef] [PubMed]
  66. Brennaman, L.H.; Moss, M.L.; Maness, P.F. EphrinA/EphA-induced ectodomain shedding of neural cell adhesion molecule regulates growth cone repulsion through ADAM10 metalloprotease. J. Neurochem. 2014, 128, 267–279. [Google Scholar] [CrossRef] [PubMed]
  67. Paratcha, G.; Ledda, F.; Ibáñez, C.F. The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands. Cell 2003, 113, 867–879. [Google Scholar] [CrossRef]
  68. Cole, G.J.; Schubert, D.; Glaser, L. Cell-substratum adhesion in chick neural retina depends upon protein-heparan sulfate interactions. J. Cell Biol. 1985, 100, 1192–1199. [Google Scholar] [CrossRef] [PubMed]
  69. Burg, M.A.; Halfter, W.; Cole, G.J. Analysis of proteoglycan expression in developing chicken brain: Characterization of a heparan sulfate proteoglycan that interacts with the neural cell adhesion molecule. J. Neurosci. Res. 1995, 41, 49–64. [Google Scholar] [CrossRef] [PubMed]
  70. Herndon, M.E.; Stipp, C.S.; Lander, A.D. Interactions of neural glycosaminoglycans and proteoglycans with protein ligands: Assessment of selectivity, heterogeneity and the participation of core proteins in binding. Glycobiology 1999, 9, 143–155. [Google Scholar] [CrossRef] [PubMed]
  71. Santuccione, A.; Sytnyk, V.; Leshchyns’ka, I.; Schachner, M. Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J. Cell Biol. 2005, 169, 341–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Prodromidou, K.; Papastefanaki, F.; Sklaviadis, T.; Matsas, R. Functional cross-talk between the cellular prion protein and the neural cell adhesion molecule NCAM is critical for neuronal differentiation of neural stem/precursor cells. Stem Cells 2014, 32, 1674–1687. [Google Scholar] [CrossRef] [PubMed]
  73. Dzhandzhugazyan, K.; Bock, E. Demonstration of an extracellular ATP-binding site in NCAM: Functional implications of nucleotide binding. Biochemistry 1997, 36, 15381–15395. [Google Scholar] [CrossRef] [PubMed]
  74. Pollerberg, G.E.; Schachner, M.; Davoust, J. Differentiation state-dependent surface mobilities of two forms of the neural cell adhesion molecule. Nature 1986, 324, 462–465. [Google Scholar] [CrossRef] [PubMed]
  75. Leshchyns’ka, I.; Sytnyk, V.; Morrow, J.S.; Schachner, M. Neural cell adhesion molecule (NCAM) association with PKCbeta2 via betaI spectrin is implicated in NCAM-mediated neurite outgrowth. J. Cell Biol. 2003, 161, 625–639. [Google Scholar] [CrossRef] [PubMed]
  76. Rodriguez, M.M.; Ron, D.; Touhara, K.; Chen, C.H.; Mochly-Rosen, D. RACK1, a protein kinase C anchoring protein, coordinates the binding of activated protein kinase C and select pleckstrin homology domains in vitro. Biochemistry 1999, 38, 13787–13794. [Google Scholar] [CrossRef] [PubMed]
  77. Kolkova, K.; Novitskaya, V.; Pedersen, N.; Berezin, V.; Bock, E. Neural cell adhesion molecule-stimulated neurite outgrowth depends on activation of protein kinase C and the Ras-mitogen-activated protein kinase pathway. J. Neurosci. 2000, 20, 2238–2246. [Google Scholar] [PubMed]
  78. He, Q.; Meiri, K.F. Isolation and characterization of detergent-resistant microdomains responsive to NCAM-mediated signaling from growth cones. Mol. Cell. Neurosci. 2002, 19, 18–31. [Google Scholar] [CrossRef] [PubMed]
  79. Büttner, B.; Kannicht, C.; Reutter, W.; Horstkorte, R. Novel cytosolic binding partners of the neural cell adhesion molecule: Mapping the binding domains of PLCγ, LANP, TOAD-64, Syndapin, PP1, and PP2A. Biochemistry 2005, 44, 6938–6947. [Google Scholar] [CrossRef] [PubMed]
  80. Büttner, B.; Kannicht, C.; Reutter, W.; Horstkorte, R. The neural cell adhesion molecule is associated with major components of the cytoskeleton. Biochem. Biophys. Res. Commun. 2003, 310, 967–971. [Google Scholar] [CrossRef] [PubMed]
  81. Wobst, H.; Schmitz, B.; Schachner, M.; Diestel, S.; Leshchyns’ka, I.; Sytnyk, V. Kinesin-1 promotes post-Golgi trafficking of NCAM140 and NCAM180 to the cell surface. J. Cell Sci. 2015, 128, 2816–2829. [Google Scholar] [CrossRef] [PubMed]
  82. Schuch, U.; Lohse, M.J.; Schachner, M. Neural cell adhesion molecules influence second messenger systems. Neuron 1989, 3, 13–20. [Google Scholar] [CrossRef]
  83. Doherty, P.; Ashton, S.V.; Moore, S.E.; Walsh, F.S. Morphoregulatory activities of NCAM and N-cadherin can be accounted for by G protein-dependent activation of L- and N-type neuronal Ca2+ channels. Cell 1991, 67, 21–33. [Google Scholar] [CrossRef]
  84. Frei, T.; von Bohlen und Halbach, F.; Wille, W.; Schachner, M. Different extracellular domains of the neural cell adhesion molecule (N-CAM) are involved in different functions. J. Cell Biol. 1992, 118, 177–194. [Google Scholar] [CrossRef] [PubMed]
  85. Jessen, U.; Novitskaya, V.; Pedersen, N.; Serup, P.; Berezin, V.; Bock, E. The transcription factors CREB and c-Fos play key roles in NCAM-mediated neuritogenesis in PC12-E2 cells. J. Neurochem. 2001, 79, 1149–1160. [Google Scholar] [CrossRef] [PubMed]
  86. Beggs, H.E.; Baragona, S.C.; Hemperly, J.J.; Maness, P.F. NCAM140 interacts with the focal adhesion kinase p125(fak) and the SRC-related tyrosine kinase p59(fyn). J. Biol. Chem. 1997, 272, 8310–8319. [Google Scholar] [CrossRef] [PubMed]
  87. Bodrikov, V.; Leshchyns’ka, I.; Sytnyk, V.; Overvoorde, J.; den Hertog, J.; Schachner, M. RPTPalpha is essential for NCAM-mediated p59fyn activation and neurite elongation. J. Cell Biol. 2005, 168, 127–139. [Google Scholar] [CrossRef] [PubMed]
  88. Schmid, R.S.; Graff, R.D.; Schaller, M.D.; Chen, S.; Schachner, M.; Hemperly, J.J.; Maness, P.F. NCAM stimulates the Ras-MAPK pathway and CREB phosphorylation in neuronal cells. J. Neurobiol. 1999, 38, 542–558. [Google Scholar] [CrossRef]
  89. Niethammer, P.; Delling, M.; Sytnyk, V.; Dityatev, A.; Fukami, K.; Schachner, M. Cosignaling of NCAM via lipid rafts and the FGF receptor is required for neuritogenesis. J. Cell Biol. 2002, 157, 521–532. [Google Scholar] [CrossRef] [PubMed]
  90. Little, E.B.; Edelman, G.M.; Cunningham, B.A. Palmitoylation of the cytoplasmic domain of the neural cell adhesion molecule N-CAM serves as an anchor to cellular membranes. Cell Adhes. Commun. 1998, 6, 415–430. [Google Scholar] [CrossRef] [PubMed]
  91. Sorkin, B.C.; Hoffman, S.; Edelman, G.M.; Cunningham, B.A. Sulfation and phosphorylation of the neural cell adhesion molecule, N-CAM. Science 1984, 225, 1476–1478. [Google Scholar] [CrossRef] [PubMed]
  92. Pollscheit, J.; Glaubitz, N.; Haller, H.; Horstkorte, R.; Bork, K. Phosphorylation of serine 774 of the neural cell adhesion molecule is necessary for cyclic adenosine monophosphate response element binding protein activation and neurite outgrowth. J. Neurosci. Res. 2012, 90, 1577–1582. [Google Scholar] [CrossRef] [PubMed]
  93. Mackie, K.; Sorkin, B.C.; Nairn, A.C.; Greengard, P.; Edelman, G.M.; Cunningham, B.A. Identification of two protein kinases that phosphorylate the neural cell-adhesion molecule, N-CAM. J. Neurosci. 1989, 9, 1883–1896. [Google Scholar] [PubMed]
  94. Diestel, S.; Laurini, C.; Traub, O.; Schmitz, B. Tyrosine 734 of NCAM180 interferes with FGF receptor-dependent signaling implicated in neurite growth. Biochem. Biophys. Res. Commun. 2004, 322, 186–196. [Google Scholar]
  95. Cassens, C.; Kleene, R.; Xiao, M.F.; Friedrich, C.; Dityateva, G.; Schafer-Nielsen, C.; Schachner, M. Binding of the receptor tyrosine kinase TrkB to the neural cell adhesion molecule (NCAM) regulates phosphorylation of NCAM and NCAM-dependent neurite outgrowth. J. Biol. Chem. 2010, 285, 28959–28967. [Google Scholar] [CrossRef] [PubMed]
  96. Homrich, M.; Wobst, H.; Laurini, C.; Sabrowski, J.; Schmitz, B.; Diestel, S. Cytoplasmic domain of NCAM140 interacts with ubiquitin-fold modifier-conjugating enzyme-1 (Ufc1). Exp. Cell Res. 2014, 324, 192–199. [Google Scholar] [CrossRef] [PubMed]
  97. Albach, C.; Damoc, E.; Denzinger, T.; Schachner, M.; Przybylski, M.; Schmitz, B. Identification of N-glycosylation sites of the murine neural cell adhesion molecule NCAM by MALDI-TOF and MALDI-FTICR mass spectrometry. Anal. Bioanal. Chem. 2004, 378, 1129–1135. [Google Scholar] [CrossRef] [PubMed]
  98. Rutishauser, U. Polysialic acid in the plasticity of the developing and adult vertebrate nervous system. Nat. Rev. Neurosci. 2008, 9, 26–35. [Google Scholar] [CrossRef] [PubMed]
  99. Bonfanti, L.; Theodosis, D.T. Polysialic acid and activity-dependent synapse remodeling. Cell Adhes. Migr. 2009, 3, 43–50. [Google Scholar] [CrossRef]
  100. Rutishauser, U.; Landmesser, L. Polysialic acid in the vertebrate nervous system: A promoter of plasticity in cell-cell interactions. Trends Neurosci. 1996, 19, 422–427. [Google Scholar] [CrossRef]
  101. Storms, S.D.; Rutishauser, U. A role for polysialic acid in neural cell adhesion molecule heterophilic binding to proteoglycans. J. Biol. Chem. 1998, 273, 27124–27129. [Google Scholar] [CrossRef] [PubMed]
  102. Hübschmann, M.V.; Skladchikova, G.; Bock, E.; Berezin, V. Neural cell adhesion molecule function is regulated by metalloproteinase-mediated ectodomain release. J. Neurosci. Res. 2005, 80, 826–837. [Google Scholar] [CrossRef] [PubMed]
  103. Kalus, I.; Bormann, U.; Mzoughi, M.; Schachner, M.; Kleene, R. Proteolytic cleavage of the neural cell adhesion molecule by ADAM17/TACE is involved in neurite outgrowth. J. Neurochem. 2006, 98, 78–88. [Google Scholar] [CrossRef] [PubMed]
  104. Kleene, R.; Mzoughi, M.; Joshi, G.; Kalus, I.; Bormann, U.; Schulze, C.; Xiao, M.F.; Dityatev, A.; Schachner, M. NCAM-induced neurite outgrowth depends on binding of calmodulin to NCAM and on nuclear import of NCAM and fak fragments. J. Neurosci. 2010, 30, 10784–10798. [Google Scholar] [CrossRef] [PubMed]
  105. Rathjen, F.G.; Schachner, M. Immunocytological and biochemical characterization of a new neuronal cell surface component (L1 antigen) which is involved in cell adhesion. EMBO J. 1984, 3, 1–10. [Google Scholar] [PubMed]
  106. Kamiguchi, H.; Long, K.E.; Pendergast, M.; Schaefer, A.W.; Rapoport, I.; Kirchhausen, T.; Lemmon, V. The neural cell adhesion molecule L1 interacts with the AP-2 adaptor and is endocytosed via the clathrin-mediated pathway. J. Neurosci. 1998, 18, 5311–5321. [Google Scholar] [PubMed]
  107. Jacob, J.; Haspel, J.; Kane-Goldsmith, N.; Grumet, M. L1 mediated homophilic binding and neurite outgrowth are modulated by alternative splicing of exon 2. J. Neurobiol. 2002, 51, 177–189. [Google Scholar] [CrossRef] [PubMed]
  108. De Angelis, E.; Watkins, A.; Schäfer, M.; Brümmendorf, T.; Kenwrick, S. Disease-associated mutations in L1 CAM interfere with ligand interactions and cell-surface expression. Hum. Mol. Genet. 2002, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  109. Nolte, C.; Moos, M.; Schachner, M. Immunolocalization of the neural cell adhesion molecule L1 in epithelia of rodents. Cell Tissue Res. 1999, 298, 261–273. [Google Scholar] [CrossRef] [PubMed]
  110. Balaian, L.B.; Moehler, T.; Montgomery, A.M. The human neural cell adhesion molecule L1 functions as a costimulatory molecule in T cell activation. Eur. J. Immunol. 2000, 30, 938–943. [Google Scholar] [CrossRef]
  111. Fields, R.D.; Itoh, K. Neural cell adhesion molecules in activity-dependent development and synaptic plasticity. Trends Neurosci. 1996, 19, 473–480. [Google Scholar] [CrossRef]
  112. Dihné, M.; Bernreuther, C.; Sibbe, M.; Paulus, W.; Schachner, M. A new role for the cell adhesion molecule L1 in neural precursor cell proliferation, differentiation, and transmitter-specific subtype generation. J. Neurosci. 2003, 23, 6638–6650. [Google Scholar] [PubMed]
  113. Roonprapunt, C.; Huang, W.; Grill, R.; Friedlander, D.; Grumet, M.; Chen, S.; Schachner, M.; Young, W. Soluble cell adhesion molecule L1-Fc promotes locomotor recovery in rats after spinal cord injury. J. Neurotrauma 2003, 20, 871–882. [Google Scholar] [CrossRef] [PubMed]
  114. Lutz, D.; Kataria, H.; Kleene, R.; Loers, G.; Chaudhary, H.; Guseva, D.; Wu, B.; Jakovcevski, I.; Schachner, M. Myelin basic protein cleaves cell adhesion molecule L1 and improves regeneration after injury. Mol. Neurobiol. 2015. [Google Scholar] [CrossRef] [PubMed]
  115. Jouet, M.; Moncla, A.; Paterson, J.; McKeown, C.; Fryer, A.; Carpenter, N.; Holmberg, E.; Wadelius, C.; Kenwrick, S. New domains of neural cell-adhesion molecule L1 implicated in X-linked hydrocephalus and MASA syndrome. Am. J. Hum. Genet. 1995, 56, 1304–1314. [Google Scholar] [PubMed]
  116. Fransen, E.; van Camp, G.; Vits, L.; Willems, P.J. L1-associated diseases: Clinical geneticists divide, molecular geneticists unite. Hum. Mol. Genet. 1997, 6, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  117. Weller, S.; Gärtner, J. Genetic and clinical aspects of X-linked hydrocephalus (L1 disease): Mutations in theL1CAM gene. Hum. Mutat. 2001, 18, 1–12. [Google Scholar] [CrossRef] [PubMed]
  118. Kanemura, Y.; Okamoto, N.; Sakamoto, H.; Shofuda, T.; Kamiguchi, H.; Yamasaki, M. Molecular mechanisms and neuroimaging criteria for severe L1 syndrome with X-linked hydrocephalus. J. Neurosurg. 2006, 105, 403–412. [Google Scholar] [CrossRef] [PubMed]
  119. Schäfer, M.K.; Altevogt, P. L1CAM malfunction in the nervous system and human carcinomas. Cell. Mol. Life Sci. 2010, 67, 2425–2437. [Google Scholar] [CrossRef] [PubMed]
  120. Fogel, M.; Gutwein, P.; Mechtersheimer, S.; Riedle, S.; Stoeck, A.; Smirnov, A.; Edler, L.; Ben-Arie, A.; Huszar, M.; Altevogt, P. L1 expression as a predictor of progression and survival in patients with uterine and ovarian carcinomas. Lancet 2003, 362, 869–875. [Google Scholar] [CrossRef]
  121. Altevogt, P.; Doberstein, K.; Fogel, M. L1CAM in human cancer. Int. J. Cancer 2015. [Google Scholar] [CrossRef] [PubMed]
  122. Kenwrick, S.; Watkins, A.; de Angelis, E. Neural cell recognition molecule L1: Relating biological complexity to human disease mutations. Hum. Mol. Genet. 2000, 9, 879–886. [Google Scholar] [CrossRef] [PubMed]
  123. Lemmon, V.; Farr, K.L.; Lagenaur, C. L1-mediated axon outgrowth occurs via a homophilic binding mechanism. Neuron 1989, 2, 1597–1603. [Google Scholar] [CrossRef]
  124. Brümmendorf, T.; Hubert, M.; Treubert, U.; Leuschner, R.; Tárnok, A.; Rathjen, F.G. The axonal recognition molecule F11 is a multifunctional protein: Specific domains mediate interactions with Ng-CAM and restrictin. Neuron 1993, 10, 711–727. [Google Scholar] [CrossRef]
  125. Grumet, M.; Flaccus, A.; Margolis, R.U. Functional characterization of chondroitin sulfate proteoglycans of brain: Interactions with neurons and neural cell adhesion molecules. J. Cell Biol. 1993, 120, 815–824. [Google Scholar] [CrossRef] [PubMed]
  126. Friedlander, D.R.; Milev, P.; Karthikeyan, L.; Margolis, R.K.; Margolis, R.U.; Grumet, M. The neuronal chondroitin sulfate proteoglycan neurocan binds to the neural cell adhesion molecules Ng-CAM/L1/NILE and N-CAM, and inhibits neuronal adhesion and neurite outgrowth. J. Cell Biol. 1994, 125, 669–680. [Google Scholar] [CrossRef] [PubMed]
  127. Milev, P.; Friedlander, D.R.; Sakurai, T.; Karthikeyan, L.; Flad, M.; Margolis, R.K.; Grumet, M.; Margolis, R.U. Interactions of the chondroitin sulfate proteoglycan phosphacan, the extracellular domain of a receptor-type protein tyrosine phosphatase, with neurons, glia, and neural cell adhesion molecules. J. Cell Biol. 1994, 127, 1703–1715. [Google Scholar] [CrossRef] [PubMed]
  128. Kadmon, G.; Imhof, B.A.; Altevogt, P.; Schachner, M. Adhesive hierarchy involving the cell adhesion molecules L1, CD24, and alpha 6 integrin in murine neuroblastoma N2A cells. Biochem. Biophys. Res. Commun. 1995, 214, 94–101. [Google Scholar] [CrossRef] [PubMed]
  129. DeBernardo, A.P.; Chang, S. Heterophilic interactions of DM-GRASP: GRASP-NgCAM interactions involved in neurite extension. J. Cell Biol. 1996, 133, 657–666. [Google Scholar] [CrossRef] [PubMed]
  130. Buchstaller, A.; Kunz, S.; Berger, P.; Kunz, B.; Ziegler, U.; Rader, C.; Sonderegger, P. Cell adhesion molecules NgCAM and axonin-1 form heterodimers in the neuronal membrane and cooperate in neurite outgrowth promotion. J. Cell Biol. 1996, 135, 1593–1607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Rader, C.; Kunz, B.; Lierheimer, R.; Giger, R.J.; Berger, P.; Tittmann, P.; Gross, H.; Sonderegger, P. Implications for the domain arrangement of axonin-1 derived from the mapping of its NgCAM binding site. EMBO J. 1996, 15, 2056–2068. [Google Scholar] [PubMed]
  132. Oleszewski, M.; Beer, S.; Katich, S.; Geiger, C.; Zeller, Y.; Rauch, U.; Altevogt, P. Integrin and neurocan binding to L1 involves distinct Ig domains. J. Biol. Chem. 1999, 274, 24602–24610. [Google Scholar] [CrossRef] [PubMed]
  133. Oleszewski, M.; Gutwein, P.; von der Lieth, W.; Rauch, U.; Altevogt, P. Characterization of the L1-neurocan-binding site. Implications for L1-L1 homophilic binding. J. Biol. Chem. 2000, 275, 34478–34485. [Google Scholar] [CrossRef] [PubMed]
  134. Mechtersheimer, S.; Gutwein, P.; Agmon-Levin, N.; Stoeck, A.; Oleszewski, M.; Riedle, S.; Postina, R.; Fahrenholz, F.; Fogel, M.; Lemmon, V.; et al. Ectodomain shedding of L1 adhesion molecule promotes cell migration by autocrine binding to integrins. J. Cell Biol. 2001, 155, 661–673. [Google Scholar] [CrossRef] [PubMed]
  135. Castellani, V.; Chédotal, A.; Schachner, M.; Faivre-Sarrailh, C.; Rougon, G. Analysis of the L1-deficient mouse phenotype reveals cross-talk between Sema3A and L1 signaling pathways in axonal guidance. Neuron 2000, 27, 237–249. [Google Scholar] [CrossRef]
  136. Castellani, V.; de Angelis, E.; Kenwrick, S.; Rougon, G. Cis and trans interactions of L1 with neuropilin-1 control axonal responses to semaphorin 3A. EMBO J. 2002, 21, 6348–6357. [Google Scholar] [CrossRef] [PubMed]
  137. Thelen, K.; Kedar, V.; Panicker, A.K.; Schmid, R.S.; Midkiff, B.R.; Maness, P.F. The neural cell adhesion molecule L1 potentiates integrin-dependent cell migration to extracellular matrix proteins. J. Neurosci. 2002, 22, 4918–4931. [Google Scholar] [PubMed]
  138. Kulahin, N.; Li, S.; Hinsby, A.; Kiselyov, V.; Berezin, V.; Bock, E. Fibronectin type III (FN3) modules of the neuronal cell adhesion molecule L1 interact directly with the fibroblast growth factor (FGF) receptor. Mol. Cell. Neurosci. 2008, 37, 528–536. [Google Scholar] [CrossRef] [PubMed]
  139. Kadmon, G.; Kowitz, A.; Altevogt, P.; Schachner, M. The neural cell adhesion molecule N-CAM enhances L1-dependent cell-cell interactions. J. Cell Biol. 1990, 110, 193–208. [Google Scholar] [CrossRef] [PubMed]
  140. Simon, H.; Klinz, S.; Fahrig, T.; Schachner, M. Molecular association of the neural adhesion molecules L1 and N-CAM in the surface membrane of neuroblastoma cells is shown by chemical cross-linking. Eur. J. Neurosci. 1991, 3, 634–640. [Google Scholar] [CrossRef] [PubMed]
  141. Dickson, T.C.; Mintz, C.D.; Benson, D.L.; Salton, S.R. Functional binding interaction identified between the axonal CAM L1 and members of the ERM family. J. Cell Biol. 2002, 157, 1105–1112. [Google Scholar] [CrossRef] [PubMed]
  142. Dahlin-Huppe, K.; Berglund, E.O.; Ranscht, B.; Stallcup, W.B. Mutational analysis of the L1 neuronal cell adhesion molecule identifies membrane-proximal amino acids of the cytoplasmic domain that are required for cytoskeletal anchorage. Mol. Cell. Neurosci. 1997, 9, 144–156. [Google Scholar] [CrossRef] [PubMed]
  143. Cheng, L.; Itoh, K.; Lemmon, V. L1-mediated branching is regulated by two ezrin-radixin-moesin (ERM)-binding sites, the RSLE region and a novel juxtamembrane ERM-binding region. J. Neurosci. 2005, 25, 395–403. [Google Scholar] [CrossRef] [PubMed]
  144. Sakurai, T.; Gil, O.D.; Whittard, J.D.; Gazdoiu, M.; Joseph, T.; Wu, J.; Waksman, A.; Benson, D.L.; Salton, S.R.; Felsenfeld, D.P. Interactions between the L1 cell adhesion molecule and ezrin support traction-force generation and can be regulated by tyrosine phosphorylation. J. Neurosci. Res. 2008, 86, 2602–2614. [Google Scholar] [CrossRef] [PubMed]
  145. Wong, E.V.; Schaefer, A.W.; Landreth, G.; Lemmon, V. Involvement of p90rsk in neurite outgrowth mediated by the cell adhesion molecule L1. J. Biol. Chem. 1996, 271, 18217–18223. [Google Scholar] [CrossRef] [PubMed]
  146. Schaefer, A.W.; Kamiguchi, H.; Wong, E.V.; Beach, C.M.; Landreth, G.; Lemmon, V. Activation of the MAPK signal cascade by the neural cell adhesion molecule L1 requires L1 internalization. J. Biol. Chem. 1999, 274, 37965–37973. [Google Scholar] [CrossRef] [PubMed]
  147. Schaefer, A.W.; Kamei, Y.; Kamiguchi, H.; Wong, E.V.; Rapoport, I.; Kirchhausen, T.; Beach, C.M.; Landreth, G.; Lemmon, S.K.; Lemmon, V. L1 endocytosis is controlled by a phosphorylation-dephosphorylation cycle stimulated by outside-in signaling by L1. J. Cell Biol. 2002, 157, 1223–1232. [Google Scholar] [CrossRef] [PubMed]
  148. Schultheis, M.; Diestel, S.; Schmitz, B. The role of cytoplasmic serine residues of the cell adhesion molecule L1 in neurite outgrowth, endocytosis, and cell migration. Cell. Mol. Neurobiol. 2007, 27, 11–31. [Google Scholar] [CrossRef] [PubMed]
  149. Kamiguchi, H.; Lemmon, V. Recycling of the cell adhesion molecule L1 in axonal growth cones. J. Neurosci. 2000, 20, 3676–3686. [Google Scholar] [PubMed]
  150. Long, K.E.; Asou, H.; Snider, M.D.; Lemmon, V. The role of endocytosis in regulating L1-mediated adhesion. J. Biol. Chem. 2001, 276, 1285–1290. [Google Scholar] [CrossRef] [PubMed]
  151. Kamiguchi, H.; Yoshihara, F. The role of endocytic l1 trafficking in polarized adhesion and migration of nerve growth cones. J. Neurosci. 2001, 21, 9194–9203. [Google Scholar] [PubMed]
  152. Yap, C.C.; Wisco, D.; Kujala, P.; Lasiecka, Z.M.; Cannon, J.T.; Chang, M.C.; Hirling, H.; Klumperman, J.; Winckler, B. The somatodendritic endosomal regulator NEEP21 facilitates axonal targeting of L1/NgCAM. J. Cell Biol. 2008, 180, 827–842. [Google Scholar] [CrossRef] [PubMed]
  153. Horiuchi, H.; Lippé, R.; McBride, H.M.; Rubino, M.; Woodman, P.; Stenmark, H.; Rybin, V.; Wilm, M.; Ashman, K.; Mann, M.; et al. A novel Rab5 GDP/GTP exchange factor complexed to Rabaptin-5 links nucleotide exchange to effector recruitment and function. Cell 1997, 90, 1149–1159. [Google Scholar] [CrossRef]
  154. Aikawa, Y. Rabex-5 Protein Regulates the Endocytic Trafficking Pathway of Ubiquitinated Neural Cell Adhesion Molecule L1. J. Biol. Chem. 2012, 287, 32312–32323. [Google Scholar] [CrossRef] [PubMed]
  155. Bennett, V.; Chen, L. Ankyrins and cellular targeting of diverse membrane proteins to physiological sites. Curr. Opin. Cell Biol. 2001, 13, 61–67. [Google Scholar] [CrossRef]
  156. Davis, J.Q.; Bennett, V. Ankyrin binding activity shared by the neurofascin/L1/NrCAM family of nervous system cell adhesion molecules. J. Biol. Chem. 1994, 269, 27163–27166. [Google Scholar] [PubMed]
  157. Bennett, V.; Baines, A.J. Spectrin and ankyrin-based pathways: Metazoan inventions for integrating cells into tissues. Physiol. Rev. 2001, 81, 1353–1392. [Google Scholar] [PubMed]
  158. Hortsch, M.; Nagaraj, K.; Godenschwege, T.A. The interaction between L1-type proteins and ankyrins—A master switch for L1-type CAM function. Cell. Mol. Biol. Lett. 2009, 14, 57–69. [Google Scholar] [CrossRef] [PubMed]
  159. Nishimura, K.; Yoshihara, F.; Tojima, T.; Ooashi, N.; Yoon, W.; Mikoshiba, K.; Bennett, V.; Kamiguchi, H. L1-dependent neuritogenesis involves ankyrinB that mediates L1-CAM coupling with retrograde actin flow. J. Cell Biol. 2003, 163, 1077–1088. [Google Scholar] [CrossRef] [PubMed]
  160. Gil, O.D.; Sakurai, T.; Bradley, A.E.; Fink, M.Y.; Cassella, M.R.; Kuo, J.A.; Felsenfeld, D.P. Ankyrin binding mediates L1CAM interactions with static components of the cytoskeleton and inhibits retrograde movement of L1CAM on the cell surface. J. Cell Biol. 2003, 162, 719–730. [Google Scholar] [CrossRef] [PubMed]
  161. Garver, T.D.; Ren, Q.; Tuvia, S.; Bennett, V. Tyrosine phosphorylation at a site highly conserved in the L1 family of cell adhesion molecules abolishes ankyrin binding and increases lateral mobility of neurofascin. J. Cell Biol. 1997, 137, 703–714. [Google Scholar] [CrossRef] [PubMed]
  162. Tuvia, S.; Garver, T.D.; Bennett, V. The phosphorylation state of the FIGQY tyrosine of neurofascin determines ankyrin-binding activity and patterns of cell segregation. Proc. Natl. Acad. Sci. USA 1997, 94, 12957–12962. [Google Scholar] [CrossRef] [PubMed]
  163. Whittard, J.D.; Sakurai, T.; Cassella, M.R.; Gazdoiu, M.; Felsenfeld, D.P. MAP kinase pathway-dependent phosphorylation of the L1-CAM ankyrin binding site regulates neuronal growth. Mol. Biol. Cell 2006, 17, 2696–2706. [Google Scholar] [CrossRef] [PubMed]
  164. Guan, H.; Maness, P.F. Perisomatic GABAergic Innervation in Prefrontal Cortex Is Regulated by Ankyrin Interaction with the L1 Cell Adhesion Molecule. Cereb. Cortex 2010, 20, 2684–2693. [Google Scholar] [CrossRef] [PubMed]
  165. Kizhatil, K.; Wu, Y.X.; Sen, A.; Bennett, V. A new activity of doublecortin in recognition of the phospho-FIGQY tyrosine in the cytoplasmic domain of neurofascin. J. Neurosci. 2002, 22, 7948–7958. [Google Scholar] [PubMed]
  166. Silletti, S.; Yebra, M.; Perez, B.; Cirulli, V.; McMahon, M.; Montgomery, A.M. Extracellular signal-regulated kinase (ERK)-dependent gene expression contributes to L1 cell adhesion molecule-dependent motility and invasion. J. Biol. Chem. 2004, 279, 28880–28888. [Google Scholar] [CrossRef] [PubMed]
  167. Schmid, R.S.; Midkiff, B.R.; Kedar, V.P.; Maness, P.F. Adhesion molecule L1 stimulates neuronal migration through Vav2-Pak1 signaling. Neuroreport 2004, 15, 2791–2794. [Google Scholar] [PubMed]
  168. Lutz, D.; Wolters-Eisfeld, G.; Joshi, G.; Djogo, N.; Jakovcevski, I.; Schachner, M.; Kleene, R. Generation and nuclear translocation of sumoylated transmembrane fragment of cell adhesion molecule L1. J. Biol. Chem. 2012, 287, 17161–17175. [Google Scholar] [CrossRef] [PubMed]
  169. Hall, H.; Williams, E.J.; Moore, S.E.; Walsh, F.S.; Prochiantz, A.; Doherty, P. Inhibition of FGF-stimulated phosphatidylinositol hydrolysis and neurite outgrowth by a cell-membrane permeable phosphopeptide. Curr. Biol. 1996, 6, 580–587. [Google Scholar] [CrossRef]
  170. Doherty, P.; Walsh, F.S. CAM-FGF Receptor Interactions: A Model for Axonal Growth. Mol. Cell. Neurosci. 1996, 8, 99–111. [Google Scholar] [CrossRef] [PubMed]
  171. Brittis, P.A.; Silver, J.; Walsh, F.S.; Doherty, P. Fibroblast growth factor receptor function is required for the orderly projection of ganglion cell axons in the developing mammalian retina. Mol. Cell. Neurosci. 1996, 8, 120–128. [Google Scholar] [CrossRef] [PubMed]
  172. Saffell, J.L.; Williams, E.J.; Mason, I.J.; Walsh, F.S.; Doherty, P. Expression of a dominant negative FGF receptor inhibits axonal growth and FGF receptor phosphorylation stimulated by CAMs. Neuron 1997, 18, 231–242. [Google Scholar] [CrossRef]
  173. Walsh, F.S.; Doherty, P. Neural cell adhesion molecules of the immunoglobulin superfamily: Role in axon growth and guidance. Annu. Rev. Cell Dev. Biol. 1997, 13, 425–456. [Google Scholar] [CrossRef] [PubMed]
  174. Nakamura, M.; Masuda, H.; Horii, J.; Kuma, K.; Yokoyama, N.; Ohba, T.; Nishitani, H.; Miyata, T.; Tanaka, M.; Nishimoto, T. When overexpressed, a novel centrosomal protein, RanBPM, causes ectopic microtubule nucleation similar to gamma-tubulin. J. Cell Biol. 1998, 143, 1041–1052. [Google Scholar] [CrossRef] [PubMed]
  175. Cheng, L.; Lemmon, S.; Lemmon, V. RanBPM is an L1-interacting protein that regulates L1-mediated mitogen-activated protein kinase activation. J. Neurochem. 2005, 94, 1102–1110. [Google Scholar] [CrossRef] [PubMed]
  176. Bechara, A.; Nawabi, H.; Moret, F.; Yaron, A.; Weaver, E.; Bozon, M.; Abouzid, K.; Guan, J.L.; Tessier-Lavigne, M.; Lemmon, V.; et al. FAK-MAPK-dependent adhesion disassembly downstream of L1 contributes to semaphorin3A-induced collapse. EMBO J. 2008, 27, 1549–1562. [Google Scholar] [CrossRef] [PubMed]
  177. Wong, E.V.; Schaefer, A.W.; Landreth, G.; Lemmon, V. Casein kinase II phosphorylates the neural cell adhesion molecule L1. J. Neurochem. 1996, 66, 779–786. [Google Scholar] [CrossRef] [PubMed]
  178. Kamiguchi, H.; Lemmon, V. A neuronal form of the cell adhesion molecule L1 contains a tyrosine-based signal required for sorting to the axonal growth cone. J. Neurosci. 1998, 18, 3749–3756. [Google Scholar] [PubMed]
  179. Beer, S.; Oleszewski, M.; Gutwein, P.; Geiger, C.; Altevogt, P. Metalloproteinase-mediated release of the ectodomain of L1 adhesion molecule. J. Cell Sci. 1999, 112, 2667–2675. [Google Scholar] [PubMed]
  180. Gutwein, P.; Oleszewski, M.; Mechtersheimer, S.; Agmon-Levin, N.; Krauss, K.; Altevogt, P. Role of Src kinases in the ADAM-mediated release of L1 adhesion molecule from human tumor cells. J. Biol. Chem. 2000, 275, 15490–15497. [Google Scholar] [CrossRef] [PubMed]
  181. Gutwein, P.; Mechtersheimer, S.; Riedle, S.; Stoeck, A.; Gast, D.; Joumaa, S.; Zentgraf, H.; Fogel, M.; Altevogt, D.P. ADAM10-mediated cleavage of L1 adhesion molecule at the cell surface and in released membrane vesicles. FASEB J. 2003, 17, 292–294. [Google Scholar] [CrossRef] [PubMed]
  182. Kalus, I.; Schnegelsberg, B.; Seidah, N.G.; Kleene, R.; Schachner, M. The proprotein convertase PC5A and a metalloprotease are involved in the proteolytic processing of the neural adhesion molecule L1. J. Biol. Chem. 2003, 278, 10381–10388. [Google Scholar] [CrossRef] [PubMed]
  183. Maretzky, T.; Schulte, M.; Ludwig, A.; Rose-John, S.; Blobel, C.; Hartmann, D.; Altevogt, P.; Saftig, P.; Reiss, K. L1 Is Sequentially processed by two differently activated metalloproteases and presenilin/-secretase and regulates neural cell adhesion, cell migration, and neurite outgrowth. Mol. Cell. Biol. 2005, 25, 9040–9053. [Google Scholar] [CrossRef] [PubMed]
  184. Riedle, S.; Kiefel, H.; Gast, D.; Bondong, S.; Wolterink, S.; Gutwein, P.; Altevogt, P. Nuclear translocation and signalling of L1-CAM in human carcinoma cells requires ADAM10 and presenilin/γ-secretase activity. Biochem. J. 2009, 420, 391–402. [Google Scholar] [CrossRef] [PubMed]
  185. Lutz, D.; Loers, G.; Kleene, R.; Oezen, I.; Kataria, H.; Katagihallimath, N.; Braren, I.; Harauz, G.; Schachner, M. Myelin basic protein cleaves cell adhesion molecule L1 and promotes neuritogenesis and cell survival. J. Biol. Chem. 2014, 289, 13503–13518. [Google Scholar] [CrossRef] [PubMed]
  186. Lehmann, J.M.; Holzmann, B.; Breitbart, E.W.; Schmiegelow, P.; Riethmüller, G.; Johnson, J.P. Discrimination between benign and malignant cells of melanocytic lineage by two novel antigens, a glycoprotein with a molecular weight of 113,000 and a protein with a molecular weight of 76,000. Cancer Res. 1987, 47, 841–845. [Google Scholar] [PubMed]
  187. Johnson, J.P.; Rummel, M.M.; Rothbächer, U.; Sers, C. MUC18: A cell adhesion molecule with a potential role in tumor growth and tumor cell dissemination. Curr. Top. Microbiol. Immunol. 1996, 213, 95–105. [Google Scholar] [PubMed]
  188. Shih, I.M. The role of CD146 (Mel-CAM) in biology and pathology. J. Pathol. 1999, 189, 4–11. [Google Scholar] [CrossRef]
  189. Shih, I.M.; Speicher, D.; Hsu, M.Y.; Levine, E.; Herlyn, M. Melanoma cell-cell interactions are mediated through heterophilic Mel-CAM/ligand adhesion. Cancer Res. 1997, 57, 3835–3840. [Google Scholar] [PubMed]
  190. Sorrentino, A.; Ferracin, M.; Castelli, G.; Biffoni, M.; Tomaselli, G.; Baiocchi, M.; Fatica, A.; Negrini, M.; Peschle, C.; Valtieri, M. Isolation and characterization of CD146+ multipotent mesenchymal stromal cells. Exp. Hematol. 2008, 36, 1035–1046. [Google Scholar] [CrossRef] [PubMed]
  191. Xu, J.; Wang, W.; Kapila, Y.; Lotz, J.; Kapila, S. Multiple differentiation capacity of STRO-1+/CD146+ PDL mesenchymal progenitor cells. Stem Cells Dev. 2009, 18, 487–496. [Google Scholar] [CrossRef] [PubMed]
  192. Lv, F.J.; Tuan, R.S.; Cheung, K.M.; Leung, V.Y. Concise review: The surface markers and identity of human mesenchymal stem cells. Stem Cells 2014, 32, 1408–1419. [Google Scholar] [CrossRef] [PubMed]
  193. Gruenloh, W.; Kambal, A.; Sondergaard, C.; McGee, J.; Nacey, C.; Kalomoiris, S.; Pepper, K.; Olson, S.; Fierro, F.; Nolta, J.A. Characterization and in vivo testing of mesenchymal stem cells derived from human embryonic stem cells. Tissue Eng. A 2011, 17, 1517–1525. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, Z.; Yan, X. CD146, a multi-functional molecule beyond adhesion. Cancer Lett. 2013, 330, 150–162. [Google Scholar] [CrossRef] [PubMed]
  195. Wu, G.J.; Wu, M.W.; Wang, S.W.; Liu, Z.; Qu, P.; Peng, Q.; Yang, H.; Varma, V.A.; Sun, Q.C.; Petros, J.A.; et al. Isolation and characterization of the major form of human MUC18 cDNA gene and correlation of MUC18 over-expression in prostate cancer cell lines and tissues with malignant progression. Gene 2001, 279, 17–31. [Google Scholar] [CrossRef]
  196. Shih, I.; Wang, T.; Wu, T.; Kurman, R.J.; Gearhart, J.D. Expression of Mel-CAM in implantation site intermediate trophoblastic cell line, IST-1, limits its migration on uterine smooth muscle cells. J. Cell Sci. 1998, 111, 2655–2664. [Google Scholar] [PubMed]
  197. Lehmann, J.M.; Riethmüller, G.; Johnson, J.P. MUC18, a marker of tumor progression in human melanoma, shows sequence similarity to the neural cell adhesion molecules of the immunoglobulin superfamily. Proc. Natl. Acad. Sci. USA 1989, 86, 9891–9895. [Google Scholar] [CrossRef] [PubMed]
  198. Vainio, O.; Dunon, D.; Aïssi, F.; Dangy, J.P.; McNagny, K.M.; Imhof, B.A. HEMCAM, an adhesion molecule expressed by c-kit+ hemopoietic progenitors. J. Cell Biol. 1996, 135, 1655–1668. [Google Scholar] [CrossRef] [PubMed]
  199. Kohama, K.; Tsukamoto, Y.; Furuya, M.; Okamura, K.; Tanaka, H.; Miki, N.; Taira, E. Molecular cloning and analysis of the mouse gicerin gene. Neurochem. Int. 2005, 46, 465–470. [Google Scholar] [CrossRef] [PubMed]
  200. Guezguez, B.; Vigneron, P.; Lamerant, N.; Kieda, C.; Jaffredo, T.; Dunon, D. Dual role of melanoma cell adhesion molecule (MCAM)/CD146 in lymphocyte endothelium interaction: MCAM/CD146 promotes rolling via microvilli induction in lymphocyte and is an endothelial adhesion receptor. J. Immunol. 2007, 179, 6673–6685. [Google Scholar] [CrossRef] [PubMed]
  201. Taira, E.; Nagino, T.; Taniura, H.; Takaha, N.; Kim, C.H.; Kuo, C.H.; Li, B.S.; Higuchi, H.; Miki, N. Expression and functional analysis of a novel isoform of gicerin, an immunoglobulin superfamily cell adhesion molecule. J. Biol. Chem. 1995, 270, 28681–28687. [Google Scholar] [CrossRef] [PubMed]
  202. Bardin, N.; Reumaux, D.; Geboes, K.; Colombel, J.F.; Blot-Chabaud, M.; Sampol, J.; Duthilleul, P.; Dignat-George, F. Increased expression of CD146, a new marker of the endothelial junction in active inflammatory bowel disease. Inflamm. Bowel Dis. 2006, 12, 16–21. [Google Scholar] [CrossRef] [PubMed]
  203. Bardin, N.; Blot-Chabaud, M.; Despoix, N.; Kebir, A.; Harhouri, K.; Arsanto, J.P.; Espinosa, L.; Perrin, P.; Robert, S.; Vely, F.; et al. CD146 and its soluble form regulate monocyte transendothelial migration. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 746–753. [Google Scholar] [CrossRef] [PubMed]
  204. Schwarz, M.J.; Müller, N.; Körschenhausen, D.; Kirsch, K.H.; Penning, R.; Ackenheil, M.; Johnson, J.P.; Hampel, H. Melanoma-associated adhesion molecule MUC18/MCAM (CD146) and transcriptional regulator mader in normal human CNS. Neuroimmunomodulation 1998, 5, 270–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Kato, S.; Taniura, H.; Taira, E.; Miki, N. Involvement of a receptor for neurite outgrowth factor (NOFR) in cerebellar neurogenesis. Neurosci. Lett. 1992, 140, 78–80. [Google Scholar] [CrossRef]
  206. Hiroi, S.; Taira, E.; Ogawa, K.; Tsukamoto, Y. Neurite extension of DRG neurons by gicerin expression is enhanced by nerve growth factor. Int. J. Mol. Med. 2005, 16, 1009–1014. [Google Scholar] [CrossRef] [PubMed]
  207. Hiroi, S.; Tsukamoto, Y.; Sasaki, F.; Miki, N.; Taira, E. Involvement of gicerin, a cell adhesion molecule, in development and regeneration of chick sciatic nerve. FEBS Lett. 2003, 554, 311–314. [Google Scholar] [CrossRef]
  208. Bardin, N.; Moal, V.; Anfosso, F.; Daniel, L.; Brunet, P.; Sampol, J.; Dignat George, F. Soluble CD146, a novel endothelial marker, is increased in physiopathological settings linked to endothelial junctional alteration. Thromb. Haemost. 2003, 90, 915–920. [Google Scholar] [CrossRef] [PubMed]
  209. Daniel, L.; Bardin, N.; Moal, V.; Dignat-George, F.; Berland, Y.; Figarella-Branger, D. Tubular CD146 expression in nephropathies is related to chronic renal failure. Nephron Exp. Nephrol. 2005, 99, e105–e111. [Google Scholar] [CrossRef] [PubMed]
  210. Małyszko, J.; Małyszko, J.S.; Brzosko, S.; Wołczynski, S.; Myśliwiec, M. Markers of endothelial cell activation/injury: CD146 and thrombomodulin are related to adiponectin in kidney allograft recipients. Am. J. Nephrol. 2005, 25, 203–210. [Google Scholar] [PubMed]
  211. Małyszko, J.; Małyszko, J.S.; Brzosko, S.; Wołczynski, S.; Myśliwiec, M. Adiponectin is related to CD146, a novel marker of endothelial cell activation/injury in chronic renal failure and peritoneally dialyzed patients. J. Clin. Endocrinol. Metab. 2004, 89, 4620–4627. [Google Scholar] [CrossRef] [PubMed]
  212. Şeftalioğlu, A.; Karakoç, L. Expression of CD146 adhesion molecules (MUC18 or MCAM) in the thymic microenvironment. Acta Histochem. 2000, 102, 69–83. [Google Scholar] [CrossRef] [PubMed]
  213. Tsukamoto, Y.; Matsumoto, T.; Kotani, T.; Taira, E.; Takaha, N.; Miki, N.; Yamate, J.; Sakuma, S. The expression of gicerin, a cell adhesion molecule, in regenerating process of collecting ducts and ureters of the chicken kidney following infection with a nephrotropic strain of infectious bronchitis virus. Avian Pathol. 1997, 26, 245–255. [Google Scholar] [CrossRef] [PubMed]
  214. Tsukamoto, Y.; Taira, E.; Nakane, Y.; Tsudzuki, M.; Kohama, K.; Amin, H.; Miki, N.; Sasaki, F. Expression of gicerin, a cell adhesion molecule, in the abnormal retina in silver plumage color mutation of Japanese quail (Coturnix japonica). Neurosci. Lett. 1999, 266, 53–56. [Google Scholar] [CrossRef]
  215. Holzmann, B.; Bröcker, E.B.; Lehmann, J.M.; Ruiter, D.J.; Sorg, C.; Riethmüller, G.; Johnson, J.P. Tumor progression in human malignant melanoma: Five stages defined by their antigenic phenotypes. Int. J. Cancer 1987, 39, 466–471. [Google Scholar] [CrossRef] [PubMed]
  216. Sers, C.; Kirsch, K.; Rothbächer, U.; Riethmüller, G.; Johnson, J.P. Genomic organization of the melanoma-associated glycoprotein MUC18: Implications for the evolution of the immunoglobulin domains. Proc. Natl. Acad. Sci. USA 1993, 90, 8514–8518. [Google Scholar] [CrossRef] [PubMed]
  217. Melnikova, V.O.; Bar-Eli, M. Bioimmunotherapy for melanoma using fully human antibodies targeting MCAM/MUC18 and IL-8. Pigment Cell Res. 2006, 19, 395–405. [Google Scholar] [CrossRef] [PubMed]
  218. Kraus, A.; Masat, L.; Johnson, J.P. Analysis of the expression of intercellular adhesion molecule-1 and MUC18 on benign and malignant melanocytic lesions using monoclonal antibodies directed against distinct epitopes and recognizing denatured, non-glycosylated antigen. Melanoma Res. 1997, 7, S75–S81. [Google Scholar] [CrossRef] [PubMed]
  219. Tang, X.; Chen, X.; Xu, Y.; Qiao, Y.; Zhang, X.; Wang, Y.; Guan, Y.; Sun, F.; Wang, J. CD166 positively regulates MCAM via inhibition to ubiquitin E3 ligases Smurf1 and βTrCP through PI3K/AKT and c-Raf/MEK/ERK signaling in Bel-7402 hepatocellular carcinoma cells. Cell. Signal. 2015, 27, 1694–1702. [Google Scholar] [CrossRef] [PubMed]
  220. Wu, G.J.; Wu, M.W.; Wang, C.; Liu, Y. Enforced expression of METCAM/MUC18 increases tumorigenesis of human prostate cancer LNCaP cells in nude mice. J. Urol. 2011, 185, 1504–1512. [Google Scholar] [CrossRef] [PubMed]
  221. Satyamoorthy, K.; Muyrers, J.; Meier, F.; Patel, D.; Herlyn, M. Mel-CAM-specific genetic suppressor elements inhibit melanoma growth and invasion through loss of gap junctional communication. Oncogene 2001, 20, 4676–4684. [Google Scholar] [CrossRef] [PubMed]
  222. Mills, L.; Tellez, C.; Huang, S.; Baker, C.; McCarty, M.; Green, L.; Gudas, J.M.; Feng, X.; Bar-Eli, M. Fully human antibodies to MCAM/MUC18 inhibit tumor growth and metastasis of human melanoma. Cancer Res. 2002, 62, 5106–5114. [Google Scholar] [PubMed]
  223. Dagur, P.K.; McCoy, J.P. Endothelial-binding, proinflammatory T cells identified by MCAM (CD146) expression: Characterization and role in human autoimmune diseases. Autoimmun. Rev. 2015, 14, 415–422. [Google Scholar] [CrossRef] [PubMed]
  224. Taira, E.; Takaha, N.; Taniura, H.; Kim, C.H.; Miki, N. Molecular Cloning and Functional Expression of Gicerin, a Novel Cell Adhesion Molecule That Binds to Neurite Outgrowth Factor. Neuron 1994, 12, 861–872. [Google Scholar] [CrossRef]
  225. Taira, E.; Nagino, T.; Tsukamoto, Y.; Okumura, S.; Muraoka, O.; Sakuma, F.; Miki, N. Cytoplasmic domain is not essential for the cell adhesion activities of gicerin, an Ig-superfamily molecule. Exp. Cell Res. 1999, 253, 697–703. [Google Scholar] [CrossRef] [PubMed]
  226. Taira, E.; Kohama, K.; Tsukamoto, Y.; Okumura, S.; Miki, N. Gicerin/CD146 is involved in neurite extension of NGF-treated PC12 cells. J. Cell. Physiol. 2005, 204, 632–637. [Google Scholar] [CrossRef] [PubMed]
  227. Taira, E.; Kohama, K.; Tsukamoto, Y.; Okumura, S.; Miki, N. Characterization of Gicerin/MUC18/CD146 in the rat nervous system. J. Cell. Physiol. 2004, 198, 377–387. [Google Scholar] [CrossRef] [PubMed]
  228. Taira, E.; Nagino, T.; Tsukamoto, Y.; Ding, Y.; Sakuma, S.; Miki, N. Neurite promotion from ciliary ganglion neurons by gicerin. Neurochem. Int. 1998, 32, 23–29. [Google Scholar] [CrossRef]
  229. Bardin, N.; Francès, V.; Lesaule, G.; Horschowski, N.; George, F.; Sampol, J. Identification of the S-Endo 1 endothelial-associated antigen. Biochem. Biophys. Res. Commun. 1996, 218, 210–216. [Google Scholar] [CrossRef] [PubMed]
  230. Johnson, J.P.; Bar-Eli, M.; Jansen, B.; Markhof, E. Melanoma progression-associated glycoprotein MUC18/MCAM mediates homotypic cell adhesion through interaction with a heterophilic ligand. Int. J. Cancer 1997, 73, 769–774. [Google Scholar] [CrossRef]
  231. Bardin, N.; Anfosso, F.; Massé, J.M.; Cramer, E.; Sabatier, F.; Le Bivic, A.; Sampol, J.; Dignat-George, F. Identification of CD146 as a component of the endothelial junction involved in the control of cell-cell cohesion. Blood 2001, 98, 3677–3684. [Google Scholar] [CrossRef] [PubMed]
  232. Luo, Y.; Zheng, C.; Zhang, J.; Lu, D.; Zhuang, J.; Xing, S.; Feng, J.; Yang, D.; Yan, X. Recognition of CD146 as an ERM-binding protein offers novel mechanisms for melanoma cell migration. Oncogene 2012, 31, 306–321. [Google Scholar] [CrossRef] [PubMed]
  233. Li, G.; Kalabis, J.; Xu, X.; Meier, F.; Oka, M.; Bogenrieder, T.; Herlyn, M. Reciprocal regulation of MelCAM and AKT in human melanoma. Oncogene 2003, 22, 6891–6899. [Google Scholar] [CrossRef] [PubMed]
  234. Xu, Q.; Simpson, S.E.; Scialla, T.J.; Bagg, A.; Carroll, M. Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood 2003, 102, 972–980. [Google Scholar] [CrossRef] [PubMed]
  235. Anfosso, F.; Bardin, N.; Frances, V.; Vivier, E.; Camoin-Jau, L.; Sampol, J.; Dignat-George, F. Activation of human endothelial cells via S-Endo-1 antigen (CD146) stimulates the tyrosine phosphorylation of focal adhesion kinase p125FAK. J. Biol. Chem. 1998, 273, 26852–26856. [Google Scholar] [CrossRef] [PubMed]
  236. Anfosso, F.; Bardin, N.; Vivier, E.; Sabatier, F.; Sampol, J.; Dignat-George, F. Outside-in signaling pathway linked to CD146 engagement in human endothelial cells. J. Biol. Chem. 2001, 276, 1564–1569. [Google Scholar] [CrossRef] [PubMed]
  237. Bu, P.; Zhuang, J.; Feng, J.; Yang, D.; Shen, X.; Yan, X. Visualization of CD146 dimerization and its regulation in living cells. Biochim. Biophys. Acta Mol. Cell Res. 2007, 1773, 513–520. [Google Scholar] [CrossRef] [PubMed]
  238. Bu, P.; Gao, L.; Zhuang, J.; Feng, J.; Yang, D.; Yan, X. Anti-CD146 monoclonal antibody AA98 inhibits angiogenesis via suppression of nuclear factor-kappaB activation. Mol. Cancer Ther. 2006, 5, 2872–2878. [Google Scholar] [CrossRef] [PubMed]
  239. Wang, J.; Tang, X.; Weng, W.; Qiao, Y.; Lin, J.; Liu, W.; Liu, R.; Ma, L.; Yu, W.; Yu, Y.; et al. The membrane protein melanoma cell adhesion molecule (MCAM) is a novel tumor marker that stimulates tumorigenesis in hepatocellular carcinoma. Oncogene 2015, 34, 5781–5795. [Google Scholar] [CrossRef] [PubMed]
  240. Burns, F.R.; von Kannen, S.; Guy, L.; Raper, J.A.; Kamholz, J.; Chang, S. DM-GRASP, a novel immunoglobulin superfamily axonal surface protein that supports neurite extension. Neuron 1991, 7, 209–220. [Google Scholar] [CrossRef]
  241. Tanaka, H.; Obata, K. Developmental changes in unique cell surface antigens of chick embryo spinal motoneurons and ganglion cells. Dev. Biol. 1984, 106, 26–37. [Google Scholar] [CrossRef]
  242. Pourquié, O.; Coltey, M.; Thomas, J.L.; le Douarin, N.M. A widely distributed antigen developmentally regulated in the nervous system. Development 1990, 109, 743–752. [Google Scholar] [PubMed]
  243. Bowen, M.A.; Patel, D.D.; Li, X.; Modrell, B.; Malacko, A.R.; Wang, W.C.; Marquardt, H.; Neubauer, M.; Pesando, J.M.; Francke, U.; et al. Cloning, mapping, and characterization of activated leukocyte-cell adhesion molecule (ALCAM), a CD6 ligand. J. Exp. Med. 1995, 181, 2213–2220. [Google Scholar] [CrossRef] [PubMed]
  244. Denzinger, T.; Diekmann, H.; Bruns, K.; Laessing, U.; Stuermer, C.A.; Przybylski, M. Isolation, primary structure characterization and identification of the glycosylation pattern of recombinant goldfish neurolin, a neuronal cell adhesion protein. J. Mass Spectrom. 1999, 34, 435–446. [Google Scholar] [CrossRef]
  245. Swart, G.W. Activated leukocyte cell adhesion molecule (CD166/ALCAM): Developmental and mechanistic aspects of cell clustering and cell migration. Eur. J. Cell Biol. 2002, 81, 313–321. [Google Scholar] [CrossRef] [PubMed]
  246. Ikeda, K.; Quertermous, T. Molecular isolation and characterization of a soluble isoform of activated leukocyte cell adhesion molecule that modulates endothelial cell function. J. Biol. Chem. 2004, 279, 55315–55323. [Google Scholar] [CrossRef] [PubMed]
  247. Bech-Serra, J.J.; Santiago-Josefat, B.; Esselens, C.; Saftig, P.; Baselga, J.; Arribas, J.; Canals, F. Proteomic identification of desmoglein 2 and activated leukocyte cell adhesion molecule as substrates of ADAM17 and ADAM10 by difference gel electrophoresis. Mol. Cell. Biol. 2006, 26, 5086–5095. [Google Scholar] [CrossRef] [PubMed]
  248. Rosso, O.; Piazza, T.; Bongarzone, I.; Rossello, A.; Mezzanzanica, D.; Canevari, S.; Orengo, A.M.; Puppo, A.; Ferrini, S.; Fabbi, M. The ALCAM Shedding by the Metalloprotease ADAM17/TACE Is Involved in Motility of Ovarian Carcinoma Cells. Mol. Cancer Res. 2007, 5, 1246–1253. [Google Scholar] [CrossRef] [PubMed]
  249. Pourquié, O.; Corbel, C.; le Caer, J.P.; Rossier, J.; le Douarin, N.M. BEN, a surface glycoprotein of the immunoglobulin superfamily, is expressed in a variety of developing systems. Proc. Natl. Acad. Sci. USA 1992, 89, 5261–5265. [Google Scholar] [CrossRef] [PubMed]
  250. Diekmann, H.; Stuermer, C.A. Zebrafish neurolin-a and -b, orthologs of ALCAM, are involved in retinal ganglion cell differentiation and retinal axon pathfinding. J. Comp. Neurol. 2009, 513, 38–50. [Google Scholar] [CrossRef] [PubMed]
  251. Fraboulet, S.; Schmidt-Petri, T.; Dhouailly, D.; Pourquié, O. Expression of DM-GRASP/BEN in the developing mouse spinal cord and various epithelia. Mech. Dev. 2000, 95, 221–224. [Google Scholar] [CrossRef]
  252. Hirata, H.; Murakami, Y.; Miyamoto, Y.; Tosaka, M.; Inoue, K.; Nagahashi, A.; Jakt, L.M.; Asahara, T.; Iwata, H.; Sawa, Y.; et al. ALCAM (CD166) is a surface marker for early murine cardiomyocytes. Cells Tissues Organs 2006, 184, 172–180. [Google Scholar] [CrossRef] [PubMed]
  253. Swart, G.W.; Lunter, P.C.; Kilsdonk, J.W.; Kempen, L.C. Activated leukocyte cell adhesion molecule (ALCAM/CD166): Signaling at the divide of melanoma cell clustering and cell migration? Cancer Metastasis Rev. 2005, 24, 223–236. [Google Scholar] [CrossRef] [PubMed]
  254. Hansen, A.G.; Swart, G.W.; Zijlstra, A. ALCAM: Basis Sequence: Mouse. AFCS Nat. Mol. Pages 2011. [Google Scholar] [CrossRef] [PubMed]
  255. Prat-Vidal, C.; Roura, S.; Farré, J.; Gálvez, C.; Llach, A.; Molina, C.E.; Hove-Madsen, L.; Garcia, J.; Cinca, J.; Bayes-Genis, A. Umbilical cord blood-derived stem cells spontaneously express cardiomyogenic traits. Transplant. Proc. 2007, 39, 2434–2437. [Google Scholar] [CrossRef] [PubMed]
  256. Liu, F.; Akiyama, Y.; Tai, S.; Maruyama, K.; Kawaguchi, Y.; Muramatsu, K.; Yamaguchi, K. Changes in the expression of CD106, osteogenic genes, and transcription factors involved in the osteogenic differentiation of human bone marrow mesenchymal stem cells. J. Bone Miner. Metab. 2008, 26, 312–320. [Google Scholar] [CrossRef] [PubMed]
  257. Gonzalez, R.; Griparic, L.; Vargas, V.; Burgee, K.; Santacruz, P.; Anderson, R.; Schiewe, M.; Silva, F.; Patel, A. A putative mesenchymal stem cells population isolated from adult human testes. Biochem. Biophys. Res. Commun. 2009, 385, 570–575. [Google Scholar] [CrossRef] [PubMed]
  258. Hua, J.; Yu, H.; Dong, W.; Yang, C.; Gao, Z.; Lei, A.; Sun, Y.; Pan, S.; Wu, Y.; Dou, Z. Characterization of mesenchymal stem cells (MSCs) from human fetal lung: Potential differentiation of germ cells. Tissue Cell 2009, 41, 448–455. [Google Scholar] [CrossRef] [PubMed]
  259. Risbud, M.V.; Guttapalli, A.; Tsai, T.T.; Lee, J.Y.; Danielson, K.G.; Vaccaro, A.R.; Albert, T.J.; Gazit, Z.; Gazit, D.; Shapiro, I.M. Evidence for skeletal progenitor cells in the degenerate human intervertebral disc. Spine 2007, 32, 2537–2544. [Google Scholar] [CrossRef] [PubMed]
  260. Karaöz, E.; Doğan, B.N.; Aksoy, A.; Gacar, G.; Akyüz, S.; Ayhan, S.; Genç, Z.S.; Yürüker, S.; Duruksu, G.; Demircan, P.C.; et al. Isolation and in vitro characterisation of dental pulp stem cells from natal teeth. Histochem. Cell Biol. 2010, 133, 95–112. [Google Scholar] [CrossRef] [PubMed]
  261. Choi, S.; Kobayashi, M.; Wang, J.; Habelhah, H.; Okada, F.; Hamada, J.; Moriuchi, T.; Totsuka, Y.; Hosokawa, M. Activated leukocyte cell adhesion molecule (ALCAM) and annexin II are involved in the metastatic progression of tumor cells after chemotherapy with Adriamycin. Clin. Exp. Metastasis 2000, 18, 45–50. [Google Scholar] [CrossRef] [PubMed]
  262. Lunter, P.C.; van Kilsdonk, J.W.; van Beek, H.; Cornelissen, I.M.; Bergers, M.; Willems, P.H.; van Muijen, G.N.; Swart, G.W. Activated leukocyte cell adhesion molecule (ALCAM/CD166/MEMD), a novel actor in invasive growth, controls matrix metalloproteinase activity. Cancer Res. 2005, 65, 8801–8808. [Google Scholar] [CrossRef] [PubMed]
  263. Van Kempen, L.C.; Meier, F.; Egeblad, M.; Kersten-Niessen, M.J.; Garbe, C.; Weidle, U.H.; van Muijen, G.N.; Herlyn, M.; Bloemers, H.P.; Swart, G.W. Truncation of activated leukocyte cell adhesion molecule: A gateway to melanoma metastasis. J. Investig. Dermatol. 2004, 122, 1293–1301. [Google Scholar] [CrossRef] [PubMed]
  264. Ofori-Acquah, S.F.; King, J.A. Activated leukocyte cell adhesion molecule: A new paradox in cancer. Transl. Res. 2008, 151, 122–128. [Google Scholar] [CrossRef] [PubMed]
  265. Kristiansen, G.; Pilarsky, C.; Wissmann, C.; Kaiser, S.; Bruemmendorf, T.; Roepcke, S.; Dahl, E.; Hinzmann, B.; Specht, T.; Pervan, J.; et al. Expression profiling of microdissected matched prostate cancer samples reveals CD166/MEMD and CD24 as new prognostic markers for patient survival. J. Pathol. 2005, 205, 359–376. [Google Scholar] [CrossRef] [PubMed]
  266. Weichert, W.; Knösel, T.; Bellach, J.; Dietel, M.; Kristiansen, G. ALCAM/CD166 is overexpressed in colorectal carcinoma and correlates with shortened patient survival. J. Clin. Pathol. 2004, 57, 1160–1164. [Google Scholar] [CrossRef] [PubMed]
  267. Davies, S.; Jiang, W.G. ALCAM, activated leukocyte cell adhesion molecule, influences the aggressive nature of breast cancer cells, a potential connection to bone metastasis. Anticancer Res. 2010, 30, 1163–1168. [Google Scholar] [PubMed]
  268. Davies, S.R.; Dent, C.; Watkins, G.; King, J.A.; Mokbel, K.; Jiang, W.G. Expression of the cell to cell adhesion molecule, ALCAM, in breast cancer patients and the potential link with skeletal metastasis. Oncol. Rep. 2008, 19, 555–561. [Google Scholar] [CrossRef] [PubMed]
  269. Ihnen, M.; Köhler, N.; Kersten, J.F.; Milde-Langosch, K.; Beck, K.; Höller, S.; Müller, V.; Witzel, I.; Jänicke, F.; Kilic, E. Expression levels of Activated Leukocyte Cell Adhesion Molecule (ALCAM/CD166) in primary breast carcinoma and distant breast cancer metastases. Dis. Markers 2010, 28, 71–78. [Google Scholar] [CrossRef] [PubMed]
  270. Sawhney, M.; Matta, A.; Macha, M.A.; Kaur, J.; DattaGupta, S.; Shukla, N.K.; Ralhan, R. Cytoplasmic accumulation of activated leukocyte cell adhesion molecule is a predictor of disease progression and reduced survival in oral cancer patients. Int. J. Cancer 2009, 124, 2098–2105. [Google Scholar] [CrossRef] [PubMed]
  271. Van den Brand, M.; Takes, R.P.; Blokpoel-deRuyter, M.; Slootweg, P.J.; van Kempen, L.C. Activated leukocyte cell adhesion molecule expression predicts lymph node metastasis in oral squamous cell carcinoma. Oral Oncol. 2010, 46, 393–398. [Google Scholar] [CrossRef] [PubMed]
  272. Kahlert, C.; Weber, H.; Mogler, C.; Bergmann, F.; Schirmacher, P.; Kenngott, H.G.; Matterne, U.; Mollberg, N.; Rahbari, N.N.; Hinz, U.; et al. Increased expression of ALCAM/CD166 in pancreatic cancer is an independent prognostic marker for poor survival and early tumour relapse. Br. J. Cancer 2009, 101, 457–464. [Google Scholar] [CrossRef] [PubMed]
  273. Corrias, M.V.; Gambini, C.; Gregorio, A.; Croce, M.; Barisione, G.; Cossu, C.; Rossello, A.; Ferrini, S.; Fabbi, M. Different subcellular localization of ALCAM molecules in neuroblastoma: Association with relapse. Cell. Oncol. 2010, 32, 77–86. [Google Scholar] [PubMed]
  274. Mezzanzanica, D.; Fabbi, M.; Bagnoli, M.; Staurengo, S.; Losa, M.; Balladore, E.; Alberti, P.; Lusa, L.; Ditto, A.; Ferrini, S.; et al. Subcellular localization of activated leukocyte cell adhesion molecule is a molecular predictor of survival in ovarian carcinoma patients. Clin. Cancer Res. 2008, 14, 1726–1733. [Google Scholar] [CrossRef] [PubMed]
  275. Van Kempen, L.C.; van den Oord, J.J.; van Muijen, G.N.; Weidle, U.H.; Bloemers, H.P.; Swart, G.W. Activated Leukocyte Cell Adhesion Molecule/CD166, a Marker of Tumor Progression in Primary Malignant Melanoma of the Skin. Am. J. Pathol. 2000, 156, 769–774. [Google Scholar]
  276. Tanaka, H.; Matsui, T.; Agata, A.; Tomura, M.; Kubota, I.; McFarland, K.C.; Kohr, B.; Lee, A.; Phillips, H.S.; Shelton, D.L. Molecular cloning and expression of a novel adhesion molecule, SC1. Neuron 1991, 7, 535–545. [Google Scholar]
  277. DeBernardo, A.P.; Chang, S. Native and recombinant DM-GRASP selectively support neurite extension from neurons that express GRASP. Dev. Biol. 1995, 169, 65–75. [Google Scholar] [PubMed]
  278. Pollerberg, G.E.; Mack, T.G. Cell adhesion molecule SC1/DMGRASP is expressed on growing axons of retina ganglion cells and is involved in mediating their extension on axons. Dev. Biol. 1994, 165, 670–687. [Google Scholar] [CrossRef] [PubMed]
  279. Avci, H.X.; Zelina, P.; Thelen, K.; Pollerberg, G.E. Role of cell adhesion molecule DM-GRASP in growth and orientation of retinal ganglion cell axons. Dev. Biol. 2004, 271, 291–305. [Google Scholar] [CrossRef] [PubMed]
  280. Weiner, J.A.; Koo, S.J.; Nicolas, S.; Fraboulet, S.; Pfaff, S.L.; Pourquié, O.; Sanes, J.R. Axon fasciculation defects and retinal dysplasias in mice lacking the immunoglobulin superfamily adhesion molecule BEN/ALCAM/SC1. Mol. Cell. Neurosci. 2004, 27, 59–69. [Google Scholar] [CrossRef] [PubMed]
  281. Heffron, D.S.; Golden, J.A. DM-GRASP is necessary for nonradial cell migration during chick diencephalic development. J. Neurosci. 2000, 20, 2287–2294. [Google Scholar] [PubMed]
  282. Stephan, J.P.; Bald, L.; Roberts, P.E.; Lee, J.; Gu, Q.; Mather, J.P. Distribution and function of the adhesion molecule BEN during rat development. Dev. Biol. 1999, 212, 264–277. [Google Scholar] [CrossRef] [PubMed]
  283. Chédotal, A.; Pourquié, O.; Ezan, F.; San Clemente, H.; Sotelo, C. BEN as a presumptive target recognition molecule during the development of the olivocerebellar system. J. Neurosci. 1996, 16, 3296–3310. [Google Scholar] [PubMed]
  284. Ott, H.; Diekmann, H.; Stuermer, C.A.; Bastmeyer, M. Function of Neurolin (DM-GRASP/SC-1) in Guidance of Motor Axons during Zebrafish Development. Dev. Biol. 2001, 235, 86–97. [Google Scholar] [CrossRef] [PubMed]
  285. Buhusi, M.; Demyanenko, G.P.; Jannie, K.M.; Dalal, J.; Darnell, E.P.; Weiner, J.A.; Maness, P.F. ALCAM regulates mediolateral retinotopic mapping in the superior colliculus. J. Neurosci. 2009, 29, 15630–15641. [Google Scholar] [CrossRef] [PubMed]
  286. Thelen, K.; Maier, B.; Faber, M.; Albrecht, C.; Fischer, P.; Pollerberg, G.E. Translation of the cell adhesion molecule ALCAM in axonal growth cones—Regulation and functional importance. J. Cell Sci. 2012, 125, 1003–1014. [Google Scholar] [CrossRef] [PubMed]
  287. Thelen, K.; Jaehrling, S.; Spatz, J.P.; Pollerberg, G.E. Depending on Its Nano-Spacing, ALCAM Promotes Cell Attachment and Axon Growth. PLoS ONE 2012, 7, e40493. [Google Scholar] [CrossRef] [PubMed]
  288. Wade, A.; Thomas, C.; Kalmar, B.; Terenzio, M.; Garin, J.; Greensmith, L.; Schiavo, G. Activated leukocyte cell adhesion molecule modulates neurotrophin signaling. J. Neurochem. 2012, 121, 575–586. [Google Scholar] [CrossRef] [PubMed]
  289. Wagner, M.; Bilinska, M.; Pokryszko-Dragan, A.; Sobczynski, M.; Cyrul, M.; Kusnierczyk, P.; Jasek, M. ALCAM and CD6—Multiple sclerosis risk factors. J. Neuroimmunol. 2014, 276, 98–103. [Google Scholar] [CrossRef] [PubMed]
  290. Zhou, P.; Du, L.F.; Lv, G.Q.; Yu, X.M.; Gu, Y.L.; Li, J.P.; Zhang, C. Functional polymorphisms in CD166/ALCAM gene associated with increased risk for breast cancer in a Chinese population. Breast Cancer Res. Treat. 2011, 128, 527–534. [Google Scholar] [CrossRef] [PubMed]
  291. Van Kempen, L.C.; Nelissen, J.M.; Degen, W.G.; Torensma, R.; Weidle, U.H.; Bloemers, H.P.; Figdor, C.G.; Swart, G.W. Molecular Basis for the Homophilic Activated Leukocyte Cell Adhesion Molecule (ALCAM)-ALCAM Interaction. J. Biol. Chem. 2001, 276, 25783–25790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Bowen, M.A.; Bajorath, J.; Siadak, A.W.; Modrell, B.; Malacko, A.R.; Marquardt, H.; Nadler, S.G.; Aruffo, A. The amino-terminal immunoglobulin-like domain of activated leukocyte cell adhesion molecule binds specifically to the membrane-proximal scavenger receptor cysteine-rich domain of CD6 with a 1:1 stoichiometry. J. Biol. Chem. 1996, 271, 17390–17396. [Google Scholar] [CrossRef] [PubMed]
  293. Hassan, N.J.; Barclay, A.N.; Brown, M.H. Frontline: Optimal T cell activation requires the engagement of CD6 and CD166. Eur. J. Immunol. 2004, 34, 930–940. [Google Scholar] [CrossRef] [PubMed]
  294. Te Riet, J.; Zimmerman, A.W.; Cambi, A.; Joosten, B.; Speller, S.; Torensma, R.; van Leeuwen, F.N.; Figdor, C.G.; de Lange, F. Distinct kinetic and mechanical properties govern ALCAM-mediated interactions as shown by single-molecule force spectroscopy. J. Cell Sci. 2007, 120, 3965–3976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Gimferrer, I.; Calvo, M.; Mittelbrunn, M.; Farnós, M.; Sarrias, M.R.; Enrich, C.; Vives, J.; Sánchez-Madrid, F.; Lozano, F. Relevance of CD6-mediated interactions in T cell activation and proliferation. J. Immunol. 2004, 173, 2262–2270. [Google Scholar] [CrossRef] [PubMed]
  296. Hassan, N.J.; Simmonds, S.J.; Clarkson, N.G.; Hanrahan, S.; Puklavec, M.J.; Bomb, M.; Barclay, A.N.; Brown, M.H. CD6 regulates T-cell responses through activation-dependent recruitment of the positive regulator SLP-76. Mol. Cell. Biol. 2006, 26, 6727–6738. [Google Scholar] [CrossRef] [PubMed]
  297. Singer, N.G.; Richardson, B.C.; Powers, D.; Hooper, F.; Lialios, F.; Endres, J.; Bott, C.M.; Fox, D.A. Role of the CD6 glycoprotein in antigen-specific and autoreactive responses of cloned human T lymphocytes. Immunology 1996, 88, 537–543. [Google Scholar] [PubMed]
  298. Zimmerman, A.W.; Joosten, B.; Torensma, R.; Parnes, J.R.; van Leeuwen, F.N.; Figdor, C.G. Long-term engagement of CD6 and ALCAM is essential for T-cell proliferation induced by dendritic cells. Blood 2006, 107, 3212–3220. [Google Scholar] [CrossRef] [PubMed]
  299. Cayrol, R.; Wosik, K.; Berard, J.L.; Dodelet-Devillers, A.; Ifergan, I.; Kebir, H.; Haqqani, A.S.; Kreymborg, K.; Krug, S.; Moumdjian, R.; et al. Activated leukocyte cell adhesion molecule promotes leukocyte trafficking into the central nervous system. Nat. Immunol. 2008, 9, 137–145. [Google Scholar] [CrossRef] [PubMed]
  300. Bowen, M.A.; Aruffo, A.A.; Bajorath, J. Cell surface receptors and their ligands: In vitro analysis of CD6-CD166 interactions. Proteins 2000, 40, 420–428. [Google Scholar] [CrossRef]
  301. Zimmerman, A.W.; Nelissen, J.M.; van Emst-de Vries, S.E.; Willems, P.H.; de Lange, F.; Collard, J.G.; van Leeuwen, F.N.; Figdor, C.G. Cytoskeletal restraints regulate homotypic ALCAM-mediated adhesion through PKC independently of Rho-like GTPases. J. Cell Sci. 2004, 117, 2841–2852. [Google Scholar] [CrossRef] [PubMed]
  302. Tomita, K.; van Bokhoven, A.; Jansen, C.F.; Bussemakers, M.J.; Schalken, J.A. Coordinate recruitment of E-cadherin and ALCAM to cell-cell contacts by alpha-catenin. Biochem. Biophys. Res. Commun. 2000, 267, 870–874. [Google Scholar] [CrossRef] [PubMed]
  303. Mertens, A.E.; Roovers, R.C.; Collard, J.G. Regulation of Tiam1-Rac signalling. FEBS Lett. 2003, 546, 11–16. [Google Scholar] [CrossRef]
  304. Uhlenbrock, K.; Eberth, A.; Herbrand, U.; Daryab, N.; Stege, P.; Meier, F.; Friedl, P.; Collard, J.G.; Ahmadian, M.R. The RacGEF Tiam1 inhibits migration and invasion of metastatic melanoma via a novel adhesive mechanism. J. Cell Sci. 2004, 117, 4863–4871. [Google Scholar] [CrossRef] [PubMed]
  305. Piazza, T.; Cha, E.; Bongarzone, I.; Canevari, S.; Bolognesi, A.; Polito, L.; Bargellesi, A.; Sassi, F.; Ferrini, S.; Fabbi, M. Internalization and recycling of ALCAM/CD166 detected by a fully human single-chain recombinant antibody. J. Cell Sci. 2005, 118, 1515–1525. [Google Scholar] [CrossRef] [PubMed]
  306. Finne, J.; Finne, U.; Deagostini-Bazin, H.; Goridis, C. Occurrence of alpha 2–8 linked polysialosyl units in a neural cell adhesion molecule. Biochem. Biophys. Res. Commun. 1983, 112, 482–487. [Google Scholar] [CrossRef]
  307. Hoffman, S.; Sorkin, B.C.; White, P.C.; Brackenbury, R.; Mailhammer, R.; Rutishauser, U.; Cunningham, B.A.; Edelman, G.M. Chemical characterization of a neural cell adhesion molecule purified from embryonic brain membranes. J. Biol. Chem. 1982, 257, 7720–7729. [Google Scholar] [PubMed]
  308. Faissner, A.; Teplow, D.B.; Kübler, D.; Keilhauer, G.; Kinzel, V.; Schachner, M. Biosynthesis and membrane topography of the neural cell adhesion molecule L1. EMBO J. 1985, 4, 3105–3313. [Google Scholar] [PubMed]
  309. Wolff, J.M.; Rathjen, F.G.; Frank, R.; Roth, S. Biochemical characterization of polypeptide components involved in neurite fasciculation and elongation. Eur. J. Biochem. 1987, 168, 551–561. [Google Scholar] [CrossRef] [PubMed]
  310. Schmitz, B.; Peter-Katalinic, J.; Egge, H.; Schachner, M. Monoclonal antibodies raised against membrane glycoproteins from mouse brain recognize N-linked oligomannosidic glycans. Glycobiology 1993, 3, 609–617. [Google Scholar] [CrossRef] [PubMed]
  311. Fahrig, T.; Schmitz, B.; Weber, D.; Kücherer-Ehret, A.; Faissner, A.; Schachner, M. Two monoclonal antibodies recognizing carbohydrate epitopes on neural adhesion molecules interfere with cell interactions. Eur. J. Neurosci. 1990, 2, 153–161. [Google Scholar] [CrossRef] [PubMed]
  312. Linnemann, D.; Edvardsen, K.; Bock, E. Developmental study of the cell adhesion molecule L1. Dev. Neurosci. 1988, 10, 34–42. [Google Scholar] [CrossRef] [PubMed]
  313. Rutishauser, U.; Hoffman, S.; Edelman, G.M. Binding properties of a cell adhesion molecule from neuraltissue. Proc. Natl. Acad. Sci. USA 1982, 9, 685–689. [Google Scholar] [CrossRef]
  314. Angata, K.; Suzuki, M.; Fukuda, M. Differential and cooperative polysialylation of the neural cell adhesion molecule by two polysialyltransferases, PST and STX. J. Biol. Chem. 1998, 273, 28524–28532. [Google Scholar] [CrossRef] [PubMed]
  315. Ebeling, O.; Duczmal, A.; Aigner, S.; Geiger, C.; Schollhammer, S.; Kemshead, J.T.; Moller, P.; Schwartz-Albiez, R.; Altevogt, P. L1 adhesion molecule on human lymphocytes and monocytes: Expression and involvement in binding to alpha v beta 3 integrin. Eur. J. Immunol. 1996, 26, 2508–2516. [Google Scholar] [CrossRef] [PubMed]
  316. Jouve, N.; Despoix, N.; Espeli, M.; Gauthier, L.; Cypowyj, S.; Fallague, K.; Schiff, C.; Dignat-George, F.; Vély, F.; Leroyer, A.S. The involvement of CD146 and its novel ligand galectin-1 in apoptotic regulation of endothelial cells. J. Biol. Chem. 2013, 288, 2571–2579. [Google Scholar] [CrossRef] [PubMed]
  317. Flanagan, K.; Fitzgerald, K.; Baker, J.; Regnstrom, K.; Gardai, S.; Bard, F.; Mocci, S.; Seto, P.; You, M.; Larochelle, C.; et al. Laminin-411 Is a vascular ligand for MCAM and facilitates TH17 cell entry into the CNS. PLoS ONE 2012, 7, e4044. [Google Scholar] [CrossRef] [PubMed]
  318. Taniura, H.; Kuo, C.H.; Hayashi, Y.; Miki, N. Purification and Characterization of an 82-kD Membrane Protein as a neurite outgrowth factor binding protein: Possible involvement of NOF binding protein in axonal outgrowth in developing retina. J. Cell Biol. 1991, 112, 313–322. [Google Scholar] [CrossRef] [PubMed]
  319. Jiang, T.; Zhuang, J.; Duan, H.; Luo, Y.; Zeng, Q.; Fan, K.; Yan, H.; Lu, D.; Ye, Z.; Hao, J.; et al. CD146 is a coreceptor for VEGFR-2 in tumor angiogenesis. Blood 2012, 120, 2330–2339. [Google Scholar] [CrossRef] [PubMed]
  320. Ye, Z.; Zhang, C.; Tu, T.; Sun, M.; Liu, D.; Lu, D.; Feng, J.; Yang, D.; Liu, F.; Yan, X. Wnt5a uses CD146 as a receptor to regulate cell motility and convergent extension. Nat. Commun. 2013. [Google Scholar] [CrossRef] [PubMed]
  321. El-Deeb, S.; Thompson, S.C.; Covault, J. Characterization of a cell surface adhesion molecule expressed by a subset of developing chick neurons. Dev. Biol. 1992, 149, 213–227. [Google Scholar] [CrossRef]
  322. Hershko, A.; Ciechanover, A. The ubiquitin system. Annu. Rev. Biochem. 1998, 67, 425–479. [Google Scholar] [CrossRef] [PubMed]
  323. Pickart, C.M.; Eddins, M.J. Ubiquitin: Structures, functions, mechanisms. Biochim. Biophys. Acta 2004, 1695, 55–72. [Google Scholar] [CrossRef] [PubMed]
  324. Varshavsky, A. The ubiquitin system. Trends Biochem. Sci. 1997, 22, 383–387. [Google Scholar] [CrossRef]
  325. Ciechanover, A.; Heller, H.; Elias, S.; Haas, A.L.; Hershko, A. ATP-dependent conjugation of reticulocyte proteins with the polypeptide required for protein degradation. Proc. Natl. Acad. Sci. USA 1980, 77, 1365–1368. [Google Scholar] [CrossRef] [PubMed]
  326. Pickart, C.M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 2001, 70, 503–533. [Google Scholar] [CrossRef] [PubMed]
  327. Hoeller, D.; Dikic, I. Targeting the ubiquitin system in cancer therapy. Nature 2009, 458, 438–444. [Google Scholar] [CrossRef] [PubMed]
  328. Hershko, A. Ubiquitin: Roles in protein modification and breakdown. Cell 1983, 34, 11–12. [Google Scholar] [CrossRef]
  329. Nakayama, K.I.; Nakayama, K. Ubiquitin ligases: Cell-cycle control and cancer. Nat. Rev. Cancer 2006, 6, 369–381. [Google Scholar] [CrossRef] [PubMed]
  330. Levkowitz, G.; Waterman, H.; Ettenberg, S.A.; Katz, M.; Tsygankov, A.Y.; Alroy, I.; Lavi, S.; Iwai, K.; Reiss, Y.; Ciechanover, A.; et al. Ubiquitin ligase activity and tyrosine phosphorylation underlie suppression of growth factor signaling by c-Cbl/Sli-1. Mol. Cell 1999, 4, 1029–1040. [Google Scholar] [CrossRef]
  331. Marchese, A.; Raiborg, C.; Santini, F.; Keen, J.H.; Stenmark, H.; Benovic, J.L. The E3 ubiquitin ligase AIP4 mediates ubiquitination and sorting of the G protein-coupled receptor CXCR4. Dev. Cell 2003, 5, 709–722. [Google Scholar] [CrossRef]
  332. Piper, R.C.; Dikic, I.; Lukacs, G.L. Ubiquitin-dependent sorting in endocytosis. Cold Spring Harb. Perspect. Biol. 2014. [Google Scholar] [CrossRef] [PubMed]
  333. Ryu, K.S.; Choi, Y.S.; Ko, J.; Kim, S.O.; Kim, H.J.; Cheong, H.K.; Jeon, Y.H.; Choi, B.S.; Cheong, C. Direct characterization of E2-dependent target specificity and processivity using an artificial p27-linker-E2 ubiquitination system. BMB Rep. 2008, 41, 852–857. [Google Scholar] [CrossRef] [PubMed]
  334. Ikeda, F.; Dikic, I. Atypical ubiquitin chains: New molecular signals. “Protein Modifications: Beyond the Usual Suspects” review series. EMBO Rep. 2008, 9, 536–542. [Google Scholar] [CrossRef] [PubMed]
  335. Haglund, K.; Dikic, I. The role of ubiquitylation in receptor endocytosis and endosomal sorting. J. Cell Sci. 2012, 125, 265–275. [Google Scholar] [CrossRef] [PubMed]
  336. Clague, M.J.; Urbé, S. Ubiquitin: Same molecule, different degradation pathways. Cell 2010, 143, 682–685. [Google Scholar] [CrossRef] [PubMed]
  337. Paul, S. Dysfunction of the ubiquitin-proteasome system in multiple disease conditions: Therapeutic approaches. Bioessays 2008, 30, 1172–1184. [Google Scholar] [CrossRef] [PubMed]
  338. Peng, J.; Schwartz, D.; Elias, J.E.; Thoreen, C.C.; Cheng, D.; Marsischky, G.; Roelofs, J.; Finley, D.; Gygi, S.P. A proteomics approach to understanding protein ubiquitination. Nat. Biotechnol. 2003, 21, 921–926. [Google Scholar] [CrossRef] [PubMed]
  339. Pickart, C.M.; Fushman, D. Polyubiquitin chains: Polymeric protein signals. Curr. Opin. Chem. Biol. 2004, 8, 610–616. [Google Scholar] [CrossRef] [PubMed]
  340. Clague, M.J.; Urbé, S. Endocytosis: The DUB version. Trends Cell Biol. 2006, 16, 551–559. [Google Scholar] [CrossRef] [PubMed]
  341. Huen, M.S.; Sy, S.M.; Chen, J. BRCA1 and its toolbox for the maintenance of genome integrity. Nat. Rev. Mol. Cell Biol. 2010, 11, 138–148. [Google Scholar] [CrossRef] [PubMed]
  342. Zimmermann, M.; de Lange, T. 53BP1: Pro choice in DNA repair. Trends Cell Biol. 2014, 24, 108–117. [Google Scholar] [CrossRef] [PubMed]
  343. Chen, J.; Chen, Z.J. Regulation of NFκB by ubiquitination. Curr. Opin. Immunol. 2013, 25, 4–12. [Google Scholar] [CrossRef] [PubMed]
  344. Spence, J.; Gali, R.R.; Dittmar, G.; Sherman, F.; Karin, M.; Finley, D. Cell cycle-regulated modification of the ribosome by a variant multiubiquitin chain. Cell 2000, 102, 67–76. [Google Scholar] [CrossRef]
  345. Xu, P.; Duong, D.M.; Seyfried, N.T.; Cheng, D.; Xie, Y.; Robert, J.; Rush, J.; Hochstrasser, M.; Finley, D.; Peng, J. Quantitative proteomics reveals the function of unconventional ubiquitin chains in proteasomal degradation. Cell 2009, 137, 133–145. [Google Scholar] [CrossRef] [PubMed]
  346. Dynek, J.N.; Goncharov, T.; Dueber, E.C.; Fedorova, A.V.; Izrael-Tomasevic, A.; Phu, L.; Helgason, E.; Fairbrother, W.J.; Deshayes, K.; Kirkpatrick, D.S.; et al. c-IAP1 and UbcH5 promote K11-linked polyubiquitination of RIP1 in TNF signalling. EMBO J. 2010, 29, 4198–4209. [Google Scholar] [CrossRef] [PubMed]
  347. Jin, L.; Williamson, A.; Banerjee, S.; Philipp, I.; Rape, M. Mechanism of ubiquitin-chain formation by the human anaphase-promoting complex. Cell 2008, 133, 653–665. [Google Scholar] [CrossRef] [PubMed]
  348. Hatakeyama, S.; Yadam, M.; Matsumoto, M.; Ishida, N.; Nakayama, K.I. U box proteins as a new family of ubiquitin-protein ligases. J. Biol. Chem. 2001, 276, 33111–33120. [Google Scholar] [CrossRef] [PubMed]
  349. Johnson, E.S.; Ma, P.C.; Ota, I.M.; Varshavsky, A. A proteolytic pathway that recognizes ubiquitin as a degradation signal. J. Biol. Chem. 1995, 270, 17442–17456. [Google Scholar] [CrossRef] [PubMed]
  350. Chastagner, P.; Israël, A.; Brou, C. Itch/AIP4 mediates Deltex degradation through the formation of K29-linked polyubiquitin chains. EMBO Rep. 2006, 7, 1147–1153. [Google Scholar] [CrossRef] [PubMed]
  351. Kölling, R.; Hollenberg, C.P. The ABC-transporter Ste6 accumulates in the plasma membrane in a ubiquitinated form in endocytosis mutants. EMBO J. 1994, 13, 3261–3271. [Google Scholar] [PubMed]
  352. Hicke, L. A new ticket for entry into budding vesicles-ubiquitin. Cell 2001, 106, 527–530. [Google Scholar] [CrossRef]
  353. Geoffroy, M.C.; Hay, R.T. An additional role for SUMO in ubiquitin-mediated proteolysis. Nat. Rev. Mol. Cell Biol. 2009, 10, 564–568. [Google Scholar] [CrossRef] [PubMed]
  354. Henne, W.M.; Buchkovich, N.J.; Emr, S.D. The ESCRT pathway. Dev. Cell 2011, 21, 77–91. [Google Scholar] [CrossRef] [PubMed]
  355. Rahighi, S.; Ikeda, F.; Kawasaki, M.; Akutsu, M.; Suzuki, N.; Kato, R.; Kensche, T.; Uejima, T.; Bloor, S.; Komander, D.; et al. Specific Recognition of Linear Ubiquitin Chains by NEMO Is Important for NF-κB Activation. Cell 2009, 136, 1098–1109. [Google Scholar] [CrossRef] [PubMed]
  356. Husnjak, K.; Dikic, I. Ubiquitin-binding proteins: Decoders of ubiquitin-mediated cellular functions. Annu. Rev. Biochem. 2012, 81, 291–322. [Google Scholar] [CrossRef] [PubMed]
  357. Sen, A.; Madhivanan, K.; Mukherjee, D.; Aguilar, R.C. The epsin protein family: Coordinators of endocytosis and signaling. Biomol. Concepts 2012, 3, 117–126. [Google Scholar] [CrossRef] [PubMed]
  358. De Melker, A.A.; van der Horst, G.; Borst, J. Ubiquitin ligase activity of c-Cbl guides the epidermal growth factor receptor into clathrin-coated pits by two distinct modes of Eps15 recruitment. J. Biol. Chem. 2004, 279, 55465–55473. [Google Scholar] [CrossRef] [PubMed]
  359. Stang, E. Cbl-dependent ubiquitination is required for progression of EGF receptors into clathrin-coated pits. Mol. Biol. Cell 2004, 15, 3591–3604. [Google Scholar] [CrossRef] [PubMed]
  360. Sigismund, S.; Woelk, T.; Puri, C.; Maspero, E.; Tacchetti, C.; Transidico, P.; di Fiore, P.P.; Polo, S. Clathrin-independent endocytosis of ubiquitinated cargos. Proc. Natl. Acad. Sci. USA 2005, 102, 2760–2765. [Google Scholar] [CrossRef] [PubMed]
  361. Gucwa, A.L.; Brown, D.A. UIM domain-dependent recruitment of the endocytic adaptor protein Eps15 to ubiquitin-enriched endosomes. BMC Cell Biol. 2014. [Google Scholar] [CrossRef] [PubMed]
  362. Clague, M.J.; Liu, H.; Urbé, S. Governance of endocytic trafficking and signaling by reversible ubiquitylation. Dev. Cell 2012, 23, 457–467. [Google Scholar] [CrossRef] [PubMed]
  363. Lobert, V.H.; Brech, A.; Pedersen, N.M.; Wesche, J.; Oppelt, A.; Malerød, L.; Stenmark, H. Ubiquitination of α5β1 Integrin Controls Fibroblast Migration through Lysosomal Degradation of Fibronectin-Integrin Complexes. Dev. Cell 2010, 19, 148–159. [Google Scholar] [CrossRef] [PubMed]
  364. Umebayashi, K. The roles of ubiquitin and lipids in protein sorting along the endocytic pathway. Cell Struct. Funct. 2003, 28, 443–453. [Google Scholar] [PubMed]
  365. Reyes-Turcu, F.E.; Ventii, K.H.; Wilkinson, K.D. Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu. Rev. Biochem. 2009, 78, 363–397. [Google Scholar] [PubMed]
  366. Schwarz, L.A.; Patrick, G.N. Ubiquitin-dependent endocytosis, trafficking and turnover of neuronal membrane proteins. Mol. Cell. Neurosci. 2012, 49, 387–393. [Google Scholar] [PubMed]
  367. Bomberger, J.M.; Barnaby, R.L.; Stanton, B.A. The deubiquitinating enzyme USP10 regulates the post-endocytic sorting of cystic fibrosis transmembrane conductance regulator in airway epithelial cells. J. Biol. Chem. 2009, 284, 18778–18789. [Google Scholar] [PubMed]
  368. Berthouze, M.; Venkataramanan, V.; Li, Y.; Shenoy, S.K. The deubiquitinases USP33 and USP20 coordinate beta2 adrenergic receptor recycling and resensitization. EMBO J. 2009, 28, 1684–1696. [Google Scholar] [PubMed]
  369. Butterworth, M.B.; Edinger, R.S.; Ovaa, H.; Burg, D.; Johnson, J.P.; Frizzell, R.A. The deubiquitinating enzyme UCH-L3 regulates the apical membrane recycling of the epithelial sodium channel. J. Biol. Chem. 2007, 282, 37885–37893. [Google Scholar] [CrossRef] [PubMed]
  370. Diestel, S.; Schaefer, D.; Cremer, H.; Schmitz, B. NCAM is ubiquitylated, endocytosed and recycled in neurons. J. Cell Sci. 2007, 120, 4035–4049. [Google Scholar] [CrossRef] [PubMed]
  371. Bailey, C.H.; Chen, M.; Keller, F.; Kandel, E.R. Serotonin-mediated endocytosis of apCAM: An early step of learning-related synaptic growth in Aplysia. Science 1992, 256, 645–649. [Google Scholar] [CrossRef] [PubMed]
  372. Montarolo, P.G.; Goelet, P.; Castellucci, V.F.; Morgan, J.; Kandel, E.R.; Schacher, S. A critical period for macromolecular synthesis in long-term heterosynaptic facilitation in Aplysia. Science 1986, 234, 1249–1254. [Google Scholar] [CrossRef] [PubMed]
  373. Glanzman, D.L.; Mackey, S.L.; Hawkins, R.D.; Dyke, A.M.; Lloyd, P.E.; Kandel, E.R. Depletion of serotonin in the nervous system of Aplysia reduces the behavioral enhancement of gill withdrawal as well as the heterosynaptic facilitation produced by tail shock. J. Neurosci. 1989, 9, 4200–4213. [Google Scholar] [PubMed]
  374. Hu, Y.; Barzilai, A.; Chen, M.; Bailey, C.H.; Kandel, E.R. 5-HT and cAMP induce the formation of coated pits and vesicles and increase the expression of clathrin light chain in sensory neurons of aplysia. Neuron 1993, 10, 921–929. [Google Scholar] [CrossRef]
  375. Miñana, R.; Duran, J.M.; Tomas, M.; Renau-Piqueras, J.; Guerri, C. Neural cell adhesion molecule is endocytosed via a clathrin-dependent pathway. Eur. J. Neurosci. 2001, 13, 749–756. [Google Scholar] [CrossRef] [PubMed]
  376. Bailey, C.H.; Kaang, B.K.; Chen, M.; Martin, K.C.; Lim, C.S.; Casadio, A.; Kandel, E.R. Mutation in the phosphorylation sites of MAP kinase blocks learning-related internalization of apCAM in Aplysia sensory neurons. Neuron 1997, 18, 913–924. [Google Scholar] [CrossRef]
  377. Mayford, M.; Barzilai, A.; Keller, F.; Schacher, S.; Kandel, E.R. Modulation of an NCAM-related adhesion molecule with long-term synaptic plasticity in Aplysia. Science 1992, 256, 638–644. [Google Scholar] [CrossRef] [PubMed]
  378. Bailey, C.H.; Kandel, E.R.; Si, K. The persistence of long-term memory: A molecular approach to self-sustaining changes in learning-induced synaptic growth. Neuron 2004, 44, 49–57. [Google Scholar] [CrossRef] [PubMed]
  379. Foley, A.G.; Hartz, B.P.; Gallagher, H.C.; Rønn, L.C.; Berezin, V.; Bock, E.; Regan, C.M. A synthetic peptide ligand of neural cell adhesion molecule (NCAM) IgI domain prevents NCAM internalization and disrupts passive avoidance learning. J. Neurochem. 2000, 74, 2607–2613. [Google Scholar] [CrossRef] [PubMed]
  380. Monzo, H.J.; Park, T.I.; Dieriks, B.V.; Jansson, D.; Faull, R.L.; Dragunow, M.; Curtis, M.A. Insulin and IGF1 modulate turnover of polysialylated neural cell adhesion molecule (PSA-NCAM) in a process involving specific extracellular matrix components. J. Neurochem. 2013, 6, 758–770. [Google Scholar] [CrossRef] [PubMed]
  381. Baska, K.M.; Manandhar, G.; Feng, D.; Agca, Y.; Tengowski, M.W.; Sutovsky, M.; Yi, Y.J.; Sutovsky, P. Mechanism of extracellular ubiquitination in the mammalian epididymis. J. Cell. Physiol. 2008, 215, 684–696. [Google Scholar] [CrossRef] [PubMed]
  382. Terrell, J.; Shih, S.; Dunn, R.; Hicke, L. A function for monoubiquitination in the internalization of a G protein-coupled receptor. Mol. Cell 1998, 1, 193–202. [Google Scholar] [CrossRef]
  383. Lucero, P.; Peñalver, E.; Vela, L.; Lagunas, R. Monoubiquitination is sufficient to signal internalization of the maltose transporter in Saccharomyces cerevisiae. J. Bacteriol. 2000, 182, 241–243. [Google Scholar] [CrossRef] [PubMed]
  384. Nakatsu, F.; Sakuma, M.; Matsuo, Y.; Arase, H.; Yamasaki, S.; Nakamura, N.; Saito, T.; Ohno, H. A Di-leucine signal in the ubiquitin moiety. Possible involvement in ubiquitination-mediated endocytosis. J. Biol. Chem. 2000, 275, 26213–26219. [Google Scholar] [CrossRef] [PubMed]
  385. Roth, A.F.; Davis, N.G. Ubiquitination of the PEST-like endocytosis signal of the yeast a-factor receptor. J. Biol. Chem. 2000, 275, 8143–8153. [Google Scholar] [CrossRef] [PubMed]
  386. Barriere, H.; Nemes, C.; Lechardeur, D.; Khan-Mohammad, M.; Fruh, K.; Lukacs, G.L. Molecular basis of oligoubiquitin-dependent internalization of membrane proteins in Mammalian cells. Traffic 2006, 7, 282–297. [Google Scholar] [CrossRef] [PubMed]
  387. Hawryluk, M.J.; Keyel, P.A.; Mishra, S.K.; Watkins, S.C.; Heuser, J.E.; Traub, L.M. Epsin 1 is a polyubiquitin-selective clathrin-associated sorting protein. Traffic 2006, 7, 262–281. [Google Scholar] [CrossRef] [PubMed]
  388. Wobst, H.; Förster, S.; Laurini, C.; Sekulla, A.; Dreiseidler, M.; Höhfeld, J.; Schmitz, B.; Diestel, S. UCHL1 regulates ubiquitination and recycling of the neural cell adhesion molecule NCAM. FEBS J. 2012, 279, 4398–4409. [Google Scholar] [CrossRef] [PubMed]
  389. Liu, Y.; Fallon, L.; Lashuel, H.A.; Liu, Z.; Lansbury, P.T., Jr. The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson’s disease susceptibility. Cell 2002, 111, 209–218. [Google Scholar] [CrossRef]
  390. Larsen, C.N.; Krantz, B.A.; Wilkinson, K.D. Substrate Specificity of Deubiquitinating Enzymes: Ubiquitin C-Terminal Hydrolases. Biochemistry 1998, 37, 3358–3368. [Google Scholar] [CrossRef] [PubMed]
  391. Osaka, H.; Wang, Y.L.; Takada, K.; Takizawa, S.; Setsuie, R.; Li, H.; Sato, Y.; Nishikawa, K.; Sun, Y.J.; Sakurai, M.; et al. Ubiquitin carboxy-terminal hydrolase L1 binds to and stabilizes monoubiquitin in neuron. Hum. Mol. Genet. 2003, 12, 1945–1958. [Google Scholar] [CrossRef] [PubMed]
  392. Schäfer, M.K.; Schmitz, B.; Diestel, S. L1CAM ubiquitination facilitates its lysosomal degradation. FEBS Lett. 2010, 584, 4475–4480. [Google Scholar] [CrossRef] [PubMed]
  393. Itoh, K.; Fujisaki, K.; Watanabe, M. Human L1CAM carrying the missense mutations of the fibronectin-like type III domains is localized in the endoplasmic reticulum and degraded by polyubiquitylation. J. Neurosci. Res. 2011, 89, 1637–1645. [Google Scholar] [CrossRef] [PubMed]
  394. Castellani, V.; Falk, J.; Rougon, G. Semaphorin3A-induced receptor endocytosis during axon guidance responses is mediated by L1 CAM. Mol. Cell. Neurosci. 2004, 26, 89–100. [Google Scholar] [CrossRef] [PubMed]
  395. Ewan, L.C.; Jopling, H.M.; Jia, H.; Mittar, S.; Bagherzadeh, A.; Howell, G.J.; Walker, J.H.; Zachary, I.C.; Ponnambalam, S. Intrinsic tyrosine kinase activity is required for vascular endothelial growth factor receptor 2 ubiquitination, sorting and degradation in endothelial Cells. Traffic 2006, 7, 1270–1282. [Google Scholar] [CrossRef] [PubMed]
  396. Hicke, L.; Zanolari, B.; Riezman, H. Cytoplasmic tail phosphorylation of the alpha-factor receptor is required for its ubiquitination and internalization. J. Cell Biol. 1998, 141, 349–358. [Google Scholar] [CrossRef] [PubMed]
  397. Kumar, K.G.; Krolewski, J.J.; Fuchs, S.Y. Phosphorylation and specific ubiquitin acceptor sites are required for ubiquitination and degradation of the IFNAR1 subunit of type I interferon receptor. J. Biol. Chem. 2004, 279, 46614–46620. [Google Scholar] [CrossRef] [PubMed]
  398. Aikawa, Y. Ubiquitination within the membrane-proximal ezrin-radixin-moesin (ERM)-binding region of the L1 cell adhesion molecule. Commun. Integr. Biol. 2013. [Google Scholar] [CrossRef] [PubMed]
  399. Thelen, K.; Georg, T.; Bertuch, S.; Zelina, P.; Pollerberg, G.E. Ubiquitination and endocytosis of cell adhesion molecule DM-GRASP regulate its cell surface presence and affect its role for axon navigation. J. Biol. Chem. 2008, 283, 32792–32801. [Google Scholar] [CrossRef] [PubMed]
  400. Dalerba, P.; Dylla, S.J.; Park, I.K.; Liu, R.; Wang, X.; Cho, R.W.; Hoey, T.; Gurney, A.; Huang, E.H.; Simeone, D.M.; et al. Phenotypic characterization of human colorectal cancer stem cells. Proc. Natl. Acad. Sci. USA 2007, 104, 10158–10163. [Google Scholar] [CrossRef] [PubMed]
  401. Levin, T.G.; Powell, A.E.; Davies, P.S.; Silk, A.D.; Dismuke, A.D.; Anderson, E.C.; Swain, J.R.; Wong, M.H. Characterization of the intestinal cancer stem cell marker CD166 in the human and mouse gastrointestinal tract. Gastroenterology 2010, 139, 2072–2082.e5. [Google Scholar] [CrossRef] [PubMed]
  402. Jiao, J.; Hindoyan, A.; Wang, S.; Tran, L.M.; Goldstein, A.S.; Lawson, D.; Chen, D.; Li, Y.; Guo, C.; Zhang, B.; et al. Identification of CD166 as a surface marker for enriching prostate stem/progenitor and cancer initiating cells. PLoS ONE 2012, 7, e42564. [Google Scholar] [CrossRef] [PubMed]
  403. Jezierska, A.; Matysiak, W.; Motyl, T. ALCAM/CD166 protects breast cancer cells against apoptosis and autophagy. Med. Sci. Monit. 2006, 12, BR263–BR273. [Google Scholar] [PubMed]
  404. Ma, L.; Wang, J.; Lin, J.; Pan, Q.; Yu, Y.; Sun, F. Cluster of differentiation 166 CD166) regulated by phosphatidylinositide 3-Kinase (PI3K)/AKT signaling to exert its anti-apoptotic role via yes-associated protein (YAP) in liver cancer. J. Biol. Chem. 2014, 289, 6921–6933. [Google Scholar] [CrossRef] [PubMed]
  405. Ma, L.; Pan, Q.; Sun, F.; Yu, Y.; Wang, J. Cluster of differentiation 166 (CD166) regulates cluster of differentiation (CD44) via NF-κB in liver cancer cell line Bel-7402. Biochem. Biophys. Res. Commun. 2014, 451, 334–338. [Google Scholar] [CrossRef] [PubMed]
  406. Witze, E.S.; Litman, E.S.; Argast, G.M.; Moon, R.T.; Ahn, N.G. Wnt5a control of cell polarity and directional movement by polarized redistribution of adhesion receptors. Science 2008, 320, 365–369. [Google Scholar] [CrossRef] [PubMed]
  407. Nichols, B.J. GM1-containing lipid rafts are depleted within clathrin-coated pits. Curr. Biol. 2003, 13, 686–690. [Google Scholar] [CrossRef]
  408. Subtil, A.; Gaidarov, I.; Kobylarz, K.; Lampson, M.A.; Keen, J.H.; McGraw, T.E. Acute cholesterol depletion inhibits clathrin-coated pit budding. Proc. Natl. Acad. Sci. USA 1999, 96, 6775–6780. [Google Scholar] [CrossRef] [PubMed]
  409. Zuhorn, I.S.; Kalicharan, R.; Hoekstra, D. Lipoplex-mediated transfection of mammalian cells occurs through the cholesterol-dependent clathrin-mediated pathway of endocytosis. J. Biol. Chem. 2002, 277, 18021–18028. [Google Scholar] [CrossRef] [PubMed]
  410. Gu, F.; Gruenberg, J. Biogenesis of transport intermediates in the endocytic pathway. FEBS Lett. 1999, 452, 61–66. [Google Scholar] [CrossRef]
  411. Chen, H.; de Camilli, P. The association of epsin with ubiquitinated cargo along the endocytic pathway is negatively regulated by its interaction with clathrin. Proc. Natl. Acad. Sci. USA 2005, 102, 2766–2771. [Google Scholar] [CrossRef] [PubMed]
  412. Belouzard, S.; Rouillé, Y. Ubiquitylation of leptin receptor OB-Ra regulates its clathrin-mediated endocytosis. EMBO J. 2006, 25, 932–942. [Google Scholar] [CrossRef] [PubMed]
  413. Raiborg, C.; Bache, K.G.; Gillooly, D.J.; Madshus, I.H.; Stang, E.; Stenmark, H. Hrs sorts ubiquitinated proteins into clathrin-coated microdomains of early endosomes. Nat. Cell Biol. 2002, 4, 394–398. [Google Scholar] [CrossRef] [PubMed]
  414. Piper, R.C.; Luzio, J.P. Ubiquitin-dependent sorting of integral membrane proteins for degradation in lysosomes. Curr. Opin. Cell Biol. 2007, 19, 459–465. [Google Scholar] [CrossRef] [PubMed]
  415. Biederer, T.; Sara, Y.; Mozhayeva, M.; Atasoy, D.; Liu, X.; Kavalali, E.T.; Südhof, T.C. SynCAM, a synaptic adhesion molecule that drives synapse assembly. Science 2002, 297, 1525–1531. [Google Scholar] [CrossRef] [PubMed]
  416. Stagi, M.; Fogel, A.I.; Biederer, T. SynCAM 1 participates in axo-dendritic contact assembly and shapes neuronal growth cones. Proc. Natl. Acad. Sci. USA 2010, 107, 7568–7573. [Google Scholar] [CrossRef] [PubMed]
  417. Fogel, A.I.; Stagi, M.; Perez de Arce, K.; Biederer, T. Lateral assembly of the immunoglobulin protein SynCAM 1 controls its adhesive function and instructs synapse formation. EMBO J. 2011, 30, 4728–4738. [Google Scholar] [CrossRef] [PubMed]
  418. Kidd, T.; Brose, K.; Mitchell, K.J.; Fetter, R.D.; Tessier-Lavigne, M.; Goodman, C.S.; Tear, G. Roundabout controls axon crossing of the CNS midline and defines a novel subfamily of evolutionarily conserved guidance receptors. Cell 1998, 92, 205–215. [Google Scholar] [CrossRef]
  419. Seeger, M.; Tear, G.; Ferres-Marco, D.; Goodman, C.S. Mutations affecting growth cone guidance in Drosophila: Genes necessary for guidance toward or away from the midline. Neuron 1993, 10, 409–426. [Google Scholar] [CrossRef]
  420. Ypsilanti, A.R.; Zagar, Y.; Chédotal, A. Moving away from the midline: New developments for Slit and Robo. Development 2010, 137, 1939–1952. [Google Scholar] [CrossRef] [PubMed]
  421. Chance, R.K.; Bashaw, G.J. Slit-dependent endocytic trafficking of the robo receptor is required for son of sevenless recruitment and midline axon repulsion. PLoS Genet. 2015, 1, e1005402. [Google Scholar] [CrossRef] [PubMed]
  422. Yuasa-Kawada, J.; Kinoshita-Kawada, M.; Rao, Y.; Wu, J.Y. Deubiquitinating enzyme USP33/VDU1 is required for Slit signaling in inhibiting breast cancer cell migration. Proc. Natl. Acad. Sci. USA 2009, 106, 14530–14535. [Google Scholar] [CrossRef] [PubMed]
  423. Kim, T.H.; Lee, H.K.; Seo, I.A.; Bae, H.R.; Suh, D.J.; Wu, J.; Rao, Y.; Hwang, K.G.; Park, H.T. Netrin induces down-regulation of its receptor, deleted in colorectal cancer, through the ubiquitin-proteasome pathway in the embryonic cortical neuron. J. Neurochem. 2005, 95, 1–8. [Google Scholar] [CrossRef] [PubMed]
  424. Tian, N.; Leshchyns’ka, I.; Welch, J.H.; Diakowski, W.; Yang, H.; Schachner, M.; Sytnyk, V. Lipid raft-dependent endocytosis of close homolog of adhesion molecule L1 (CHL1) promotes neuritogenesis. J. Biol. Chem. 2012, 287, 44447–44463. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Homrich, M.; Gotthard, I.; Wobst, H.; Diestel, S. Cell Adhesion Molecules and Ubiquitination—Functions and Significance. Biology 2016, 5, 1. https://doi.org/10.3390/biology5010001

AMA Style

Homrich M, Gotthard I, Wobst H, Diestel S. Cell Adhesion Molecules and Ubiquitination—Functions and Significance. Biology. 2016; 5(1):1. https://doi.org/10.3390/biology5010001

Chicago/Turabian Style

Homrich, Mirka, Ingo Gotthard, Hilke Wobst, and Simone Diestel. 2016. "Cell Adhesion Molecules and Ubiquitination—Functions and Significance" Biology 5, no. 1: 1. https://doi.org/10.3390/biology5010001

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop