Oncogene (2010) 29, 1865–1882 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc REVIEW Dependence receptors: a new paradigm in cell signaling and cancer therapy

D Goldschneider and P Mehlen

Apoptosis, Cancer and Development Laboratory- Equipe labellise´e ‘La Ligue’, CNRS UMR5238, Centre Le´on Be´rard, Universite´ de Lyon, Lyon, France

Dependence receptors (DRs) now form a family of more (Stupack et al., 2001). All of them are involved in both than a dozen membrane receptors that are not linked by nervous system development and cancer progression. their structure, but by common functional traits. The most notable is their ability to trigger two opposite signaling pathways: in the presence of ligand, these receptors Dependence receptors: a short history activate classic signaling pathways implicated in cell survival, migration and differentiation. In the absence of p75NTR was the first DR to be ligand, they do not stay inactive, rather they elicit an described. P75NTR was discovered as one of two apoptotic signal. Thus, cells expressing this kind of receptors able to bind nerve (Chao receptor are dependent on the presence of ligand in the et al., 1986), the other being TrkA (Kaplan et al., extracellular environment to survive. This review will 1991). TrkA was rapidly shown to mediate the known recapitulate the increasing data regarding the molecular responses to NGF, such as neurite outgrowth and mechanisms associated with DRs, their potential implica- neuronal survival (Lee et al., 2001a; Lykissas et al., tion during development, as well as their deregulation 2007), whereas the precise biological role of p75NTR during tumorigenesis and, finally, their emergence as new remained misunderstood. p75NTR was shown to collabo- possible therapeutic targets for cancer treatment. rate with TrkA to form high-affinity sites for NGF Oncogene (2010) 29, 1865–1882; doi:10.1038/onc.2010.13; binding (Hempstead et al., 1991). In addition, p75NTR published online 22 February 2010 was shown to alter the ligand specificity of other Trk receptors. For example, brain-derived neurotrophic Keywords: dependence receptors; ; ; factor, NT3 and NT4/5 can all bind TrkB in the absence tumor progression; cancer therapy of p75NTR, whereas only brain-derived neurotrophic factor does so in the presence of p75NTR. In contrast, coexpression of p75NTR with TrkC results in a relaxation Membrane receptors are classically considered as inactive in its absolute specificity for NT3 (Hempstead, 2002). At unless bound to their ligand. However, increasing observa- the time of its discovery, p75NTR was considered as a tions demonstrate that some receptors, in addition to their unique type of but, subsequently, a large ‘positive’ signaling when their ligand is present, transduce a superfamily of tumor necrosis factor (TNF) receptors ‘negative’ signal that induces apoptosis in the absence of were found to share the overall structure of p75NTR ligand (Figure 1). Cells expressing these receptors are thus (Liepinsh et al., 1997). Identification of this superfamily dependent on the presence of ligand to survive. These helped elucidate some of the biological functions of receptors are named ‘dependence receptors.’ To date, the p75NTR, including its link to cell death regulation. The dependence receptor (DR) family is composed of more relationship between these TNF death receptors, which than a dozen members including DCC (deleted in color- induce cell death on binding of proapoptotic ligand such ectal carcinoma) (Mehlen et al., 1998), UNC5Hs (un- as TNF or FasL, and p75NTR, which binds NGF, a coordinated 5 homologs), neogenin (Matsunaga et al., trophic factor known to induce cell survival, led DE NTR 2004), p75 (p75 neurotrophin receptor) (Rabizadeh Bredesen and colleagues to propose that p75NTR induces et al., 1993), RET (rearranged during transfection) cell death when unoccupied by NGF, whereas binding (Bordeaux et al., 2000), TrkC (tyrosine receptor C) of NGF blocks apoptosis (Rabizadeh et al., 1993) (Tauszig-Delamasure et al., 2007), Ptc (patched) (Thibert (Figure 2 and Table 1). This finding suggested that et al., 2003), EphA4 ( type A receptor 4) (Furne p75NTR expression creates a state of cellular dependence et al., 2009), ALK (anaplastic lymphoma kinase) (Mourali on NGF. Further studies with knockout mice confirmed et al.,2006),MET(Tulasneet al., 2004) and some this notion. First, p75NTR-deficient mice have an increased number of cholinergic neurons, somal hyper- Correspondence: Dr P Mehlen, Apoptosis, Cancer and Development trophy and hyperinnervation in some areas of the Laboratory- Equipe labellise´e ‘La Ligue’, CNRS UMR5238, Centre hippocampus (Yeo et al., 1997; Naumann et al., 2002). Le´on Be´rard, Universite´de Lyon, 28 rue Laennec, Lyon, Rhone 69008, In addition, crossing NGF hemizygous mice, which France. E-mail: [email protected] display a reduction in cholinergic cell numbers, with NTR NTR Received 1 October 2009; revised 2 January 2010; accepted 6 January p75 null mice showed that loss of p75 partially 2010; published online 22 February 2010 restores cholinergic cell numbers (Naumann et al., A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1866 2002). However, the overall picture of p75NTR function binding rather than ligand withdrawal (Casaccia-Bon- became more complicated when some studies showed nefil et al., 1996; Frade et al., 1996). Particularly, in that p75NTR induced apoptosis in response to ligand addition to its ability to bind mature ,

Table 1 DRs and their known ligands Receptors Ligands

p75NTR NGF, proNGF, BDNF, NT-3, NT-4/5, b-amyloid, prion DCC netrin-1, netrin-4 Neogenin netrin-1, RGMa, RGMb, RGMc, netrin-3, netrin-4 Unc50s netrin-1, netrin-4 RET GDNF, , , Ptc Shh TrkC NT-3 EphA4 ephrinB3, ephrinA1, ephrinA4 ALK , midkin, jeb Survival, MET HGF Migration, APOPTOSIS AR androgens Differentiation avb3 extracellular matrix and a5b1 Figure 1 The DR model. DRs have two faces: in the presence of their respective ligand, they transduce a positive signal of The above listed are the DRs and their known ligands. In case of differentiation, migration or survival, whereas in the absence of multiple ligands, those which were shown to block apoptotic function their ligand, they do not stay inactive but, rather, induce apoptosis. are underlined. ALK is a particular case: its putative ligands have not Thus, cells expressing such receptors are dependent on ligand yet been tested for blocking apoptosis, only agonist antibodies have availability for survival. been used.

Figure 2 Representation of the DR family. The functional domains present in extra and intracellular domains are represented. DRs are not related to each other according to their structure, but according to their ability to induce apoptosis in the absence of ligand. All of them are caspase substrates, except p75NTR and integrins. The position of caspase cleavage sites is indicated. Localization of ADD, which has been more or less precisely determined depending on receptor, is indicated by double arrows.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1867 other cell-death-inducing ligands have been proposed embryogenesis. In vertebrates, besides netrin-1, four for p75NTR, such as pro-NGF (Lee et al., 2001b), other netrins have been described: netrin-2/3/2like, b-amyloid (Yaar et al., 2002) and prion (Della-Bianca netrin-4/b, netrin-G1 and netrin-G2 (Puschel, 1999; et al., 2001) . The decision between ligand- Mehlen and Mazelin, 2003). Netrins are structurally induced apoptosis and ligand-inhibited apoptosis related to the short arm of laminin (g for netrin 1–3 and mediated by p75NTR likely depends on cell type and b for netrin-4, G1 and G2). Netrins 1–4 are secreted, development stage (Barrett and Bartlett, 1994). Because whereas netrin-G1 and G2 are membrane anchored by the idea of a receptor triggering apoptosis when means of a glycophosphatidylinol tail. Secreted netrins unbound to its ligand contradicted the dogma regarding exert their biological functions by binding to receptors receptor signaling and the trophic theory, considering such as DCC, UNC5, neogenin and DSCAM, whereas p75NTR as a classic death receptor had more success than netrin Gs do not interact with these receptors (Rajase- considering it as a DR. Consequently, DR p75NTR has kharan and Kennedy, 2009). Interestingly, netrin-5 has been rather forgotten, although it was the first to be recently appeared in databases, which seems to be described and no definitive evidence has demonstrated related to the netrin 1–3 group according to its sequence. that it is not a DR. Netrin-1 is the most studied member of the netrin family The concept reemerged with DCC. DCC was dis- and to date it seems to be the main ligand for DCC, as covered in 1990 as a putative cell-surface receptor well as for UNC5 receptors (see below), although a encoded by a frequently deleted through allelic recent report mentioned that netrin-4 could interact with loss in colorectal carcinoma (Fearon et al., 1990). DCC and UNC5H1 (Qin et al., 2007). Observation that DCC expression is reduced or lost in The UNC5 receptor was initially identified in colorectal cancer led to the proposal that DCC Caenorhabditis elegans as an axonal guidance trans- expression represented a constraint for disease progres- membrane receptor (Leung-Hagesteijn et al., 1992) and, sion and is therefore a tumor suppressor. This hypo- on the basis of a genetic screen, was predicted to interact thesis was supported by the fact that DCC expression is with UNC6 (the C. elegans netrin-1 ortholog) (Hedge- lost or reduced in various cancers (Mehlen and Fearon, cock et al., 1990). Four homologs of UNC5 have been 2004) and that its loss of expression is associated with described in mammals (UNC5H1, 2, 3 and 4 in rodents poor prognosis (Shibata et al., 1996; Sun et al., 1999). In and UNC5A, B, C and D in humans) (Leonardo et al., addition, restoration of DCC expression can suppress 1997). Although DCC alone is implicated in the tumorigenic property in vitro and in nude mice chemoattractive effect of netrin-1, it has been proposed (Klingelhutz et al., 1993; Velcich et al., 1999). The that UNC5, associated with DCCs through their DCC extracellular domain shares structural features intracellular domains, is responsible for the repulsive with certain types of cell-adhesion molecules, such as effect of netrin-1 (Hong et al., 1999). Besides this role, NCAM (Cho et al., 1994) (Figure 2 and Table 1), but its UNC5 receptors are now known to have critical roles in intracellular domain shows little similarity with known other cellular processes, such as neuronal migration ; hence in spite of the above-mentioned studies (Mehlen and Furne, 2005) and embryonic angiogenesis on cancers, little was known about the precise biological (Lu et al., 2004). Interestingly, UNC5 proteins contain a role of DCC. It was rediscovered as the receptor for death domain related to the death domain of the TNF netrin-1 (Keino-Masu et al., 1996), a diffusible molecule receptor superfamily in their cytoplasmic part. As originally identified as a chemoattractant for commis- UNC5 receptors were netrin-1 receptors and contained sural axons in the vertebral spinal cord (Serafini et al., a death domain, it was suggested that they were possible 1994). The classic view for netrin-1 is that a gradient of DRs (Figure 2 and Table 1). Along this line, UNC5 this cue diffuses from a ventral spinal cord structure, the receptors are able to induce apoptosis in the absence of floor plate, and orients the growth of commissural axons netrin-1, whereas addition of ligand efficiently blocked as they extend circumferentially toward the ventral this effect (Llambi et al., 2001; Wang et al., 2008). midline of the embryonic nervous system. The key role A DCC homolog was discovered and called neogenin of DCC and netrin-1 in mediating axon outgrowth and (Vielmetter et al., 1994; Meyerhardt et al., 1997). pathfinding is supported by a large number of studies, Although there is only one member of the DCC receptor particularly the analysis of DCC and netrin-1 knockout family in C. elegans and Drosophila (UNC40 and mice, which display similar defects in the central nervous frazzled, respectively), vertebrates have evolved two system (Serafini et al., 1996; Fazeli et al., 1997). Such a closely related orthologs, DCC and neogenin. Owing to dual role for a receptor, implicated during development its identity of sequence with DCCs, especially in their and functioning as a putative tumor suppressor, seems ectodomain, neogenin was initially considered to be a to be a common trait for DRs. DCC was thus proposed netrin-1 receptor as well. However, more recently, the as a DR when it was shown that its expression in various propensity of netrin-1 to function as a ligand for cancer cell lines that lacked endogenous DCC expres- neogenin was challenged when neogenin seemed to have sion induced cell death and that addition of netrin-1 much higher affinity for RGM (repulsive guidance blocked apoptosis (Mehlen et al., 1998; Chen et al., molecule), another guidance molecule, and to mediate 1999). its repulsive effect (Rajagopalan et al., 2004). RGM was Netrin-1 is in fact the founding member of a family of first identified as a repulsive, membrane-bound cue extracellular proteins found throughout the animal responsible for the mapping of temporal retinal axons to kingdom and that direct cell and axon migration during the posterior region of the chick tectum (Monnier et al.,

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1868 2002). In mammals, three RGMs exist: RGMa, the These ‘non-liganded integrins,’ which are either un- closest ortholog of chick RGM, RGMb/DRAGON and ligated or occupied with a soluble antagonist, not only RGMc/hemojuvelin. RGMa and b are both expressed in disrupt survival signaling but also actively induce the central nervous system and follow a complementary apoptosis, hence supporting the view of integrins as expression pattern (Niederkofler et al., 2004), whereas DRs (Stupack et al., 2001) (Figure 2 and Table 1). RGMc is mainly expressed in striated muscles and liver Beside these receptors, some classical (Schmidtmer and Engelkamp, 2004). Neogenin is a DR receptors have also emerged as DRs. RET was the first in that it can induce apoptosis when overexpressed in one (Figure 2 and Table 1). RET is the signaling chick neural tube, and its ligand, RGM, but not netrin- component of a multisubunit complex that functions as 1, counteracts this process (Matsunaga et al., 2004) a receptor for glial cell line-derived neurotrophic factor (Figure 2 and Table 1). It should be noted that the (Jing et al., 1996), neurturin, artemin and persephin question of neogenin’s ligand remains tricky: despite its (Kotzbauer et al., 1996), four homologous neurotrophic higher affinity for RGM, neogenin seems to be able to factors related to the transforming growth factor-b mediate netrin-1 signaling in axon attraction (Wilson family. The receptor complex also includes (GPI)- and Key, 2006) or cell adhesion (Srinivasan et al., 2003). anchored proteins GFRa1, 2, 3 and 4 that are required Moreover, neogenin has also been proposed to mediate for RET dimerization and dictate ligand selectivity netrin-3 signaling during myotube formation (Kang (Baloh et al., 2000; Scott and Ibanez, 2001). After et al., 2004), as well as netrin-4 signaling in angiogenesis interaction with its ligands, RET undergoes autopho- (Lejmi et al., 2008). sphorylation and then interacts with multiple effectors Patched (Ptc) is a 12-transmembrane receptor that is such as phospholipase C, Shc, enigma, Grb2, Grb7/ part of the complex responsible for sonic hedgehog Grb10, Src kinase and Ras-GAP (Santoro et al., 1994; (Shh) morphogen signaling (Figure 2 and Table 1). Shh Arighi et al., 1997; Lorenzo et al., 1997). Gain- binds Ptc and thus abolishes the Ptc-repressing effect on of-function of the RET gene have been smoothened (Smo). Smo is a seven-transmembrane associated with multiple endocrine neoplasia type 2 receptor that activates downstream Gli transcription (MEN 2), an autosomal dominant inherited cancer factors (Murone et al., 1999). Shh is a glycoprotein syndrome (Mulligan et al., 1993), whereas loss-of- secreted by the notochord and floor plate during function mutations of RET have been associated with development, after a ventro-dorsal concentration gra- Hirschsprung disease (aganglionosis, HSCR), a frequent dient in the ventral neural tube. This gradient deter- congenital intestinal malformation (1 in 5000 live births) mines the induction and specification of ventral neurons characterized by the absence of neural crest-derived in the vertebrate neural tube (Jessell, 2000). In addition parasympathetic neurons of the hindgut (Edery et al., to its morphogen activity, Shh was also shown to be a 1994; Romeo et al., 1994). In vitro, MEN 2-associated survival factor: indeed, Le Douarin and colleagues mutations lead to ligand-independent constitutive acti- discovered that experimental withdrawal of Shh in vation of RET kinase activity either through covalent chick embryos by partial destruction of the notochord dimerization of the receptor (MEN 2A) (Santoro et al., leads to massive cell death in the developing neural tube 1995) or through direct structural changes in its kinase (Charrier et al., 1999, 2001). It was subsequently demon- domains (MEN 2B) (Songyang et al., 1995). In contrast, strated that Shh functions as a survival factor by the mechanisms leading to the absence of intramural inhibiting the apoptotic function of Ptc (Thibert et al., ganglion cells of the hindgut observed in HSCR remain 2003). Thus, Ptc is a DR and can signal independently incompletely understood. The observation that RET is of Smo. involved in both cancer progression and nervous system Integrins are the main receptors that mediate cellular development, similar to previously identified DRs, led to interactions with extracellular matrix ligands such as the question as to whether it could also be one of them. laminins, collagens and fibronectins (Hood and Cher- It was then shown that, in different settings, expression esh, 2002). They are heterodimeric (ab) type I trans- of RET induced apoptosis in the absence, but not in the membrane receptors, and provide a connection between presence, of glial cell line-derived neurotrophic factor the matrix and the cytoskeleton. Integrins have tradi- (Bordeaux et al., 2000; Canibano et al., 2007). tionally been considered as prosurvival receptors, on the Trk receptors (TrkA, B and C) are the main basis of the concept of ‘anchorage dependence’ (Stupack neurotrophin receptors. TrkA is the receptor for NGF, and Cheresh, 2002). Integrin-mediated adhesion sup- TrkB is the receptor for brain-derived neurotrophic ports the formation of cytoskeletal and contractile factor and NT4/5, whereas TrkC is the receptor for NT3 elements, promotes cellular resistance to exogenous (Kaplan and Miller, 2000). The classic neurotrophic apoptotic stimuli and facilitates signaling by trophic theory proposes that neuronal survival depends on the factor receptors. Most cells require integrin-mediated presence of trophic factors such as neurotrophins (Levi- adhesion to respond to trophic factors. This has led to Montalcini and Angeletti, 1963; Huang and Reichardt, the proposal that controlling cell adhesion and geome- 2001), and that cell death, which occurs when these try, thereby permitting responsiveness to survival factors become limited, is strictly due to loss of survival factors, may be the critical function of integrins in signals. On neurotrophin binding, Trk receptors activate maintaining cell viability. However, expression of PI3K/Akt and Ras/MAP kinase pathways that are certain b3orb1 integrins can also induce apoptosis, if thought to inhibit the naturally occurring death pro- immobilized substrates are not available as ligands. gram in neurons (Kaplan and Miller, 2000). However, in

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1869 light of what was known about DRs, it was tempting to To close this list, it should be mentioned that a hypothesize that neurotrophin binding served also to nontransmembrane receptor, (AR), block an active apoptotic signal from Trks. Interest- has also been proposed to be a DR (Ellerby et al., 1999). ingly, when Trk receptors were evaluated as possible AR is a member of the nuclear receptor superfamily of DRs, TrkC was the only one the enforced expression of ligand-activated transcription factors. Binding of andro- which induced cell death in the absence of ligand gens such as by the AR leads to nuclear (Tauszig-Delamasure et al., 2007). Thus, TrkC is a DR, translocation and transcriptional activity. Gene regula- whereas TrkA and B are not, suggesting that even a tion by the AR affects widespread processes such as closely related receptor can acquire a different activity male gonadal development, cell survival and muscular with regard to cell survival and apoptosis (Figure 2 and development, among many others (Lee and Chang, Table 1). 2003). AR displays a profile similar to that of membrane Anaplastic lymphoma kinase (ALK) tyrosine kinase is DRs; it is a caspase substrate and its expression induces a member of the receptor superfamily (Figure 2 apoptosis in the absence of ligand, whereas the addition and Table 1). It was initially identified as part of the of ligand inhibits receptor-induced cell death (Ellerby oncogenic nucleophosmin–ALK fusion protein resulting et al., 1999). Mutations in the AR are associated with from the t(2;5) translocation that is frequently asso- both prostate cancer and neurodegeneration. Neurode- ciated with anaplastic large-cell lymphoma (Morris generation-associated mutants give rise to Kennedy’s et al., 1994). Nucleophosmin allows dimerization of disease, a syndrome associated with the degeneration of the fusion protein, causing constitutive activation of motor neurons in the brainstem and spinal cord, ALK kinase and downstream activation of phospholi- resulting in weakness and muscular atrophy. Interest- pase C-g, PI3K, STATs and pp60c-src (Allouche, 2007). ingly, mutations associated with neurodegeneration The native full-length ALK receptor is mainly expressed consist in expansion (430) of polyglutamine tracts in discrete regions of the developing central and present in the N-terminal part of the receptor, whereas peripheral nervous system (Iwahara et al., 1997). shortened tracts (p22) are associated with a greater Mourali et al. (2006) forced ALK expression in cells of risk of developing prostate cancer (Nelson and Witte, lymphoid and neuronal origin to investigate wild-type 2002; Clark et al., 2003). AR, similar to p75NTR, has been ALK functions. They observed that ALK enhanced or quite forgotten as a DR. Paradoxically, the word triggered apoptosis in these cells and that treatment with ‘dependence’ is frequently associated with AR in the agonist antibodies mimicking ALK ligand prevented cell field of prostate cancer. Indeed, studies have led to the death induction (Mourali et al., 2006). concept that prostate secretory epithelial cells require Eph receptors constitute the largest family of tyrosine testosterone for survival, and the withdrawal of kinase receptors and bind ligands called testosterone leads to apoptosis in these cells (Craft and (Figure 2 and Table 1). Eph receptors regulate a diverse Sawyers, 1998). Thus, neoplastic prostate epithelial cells array of cellular processes during development, such as are often treated by hormone deprivation because it axon guidance, angiogenesis, or cell migration and leads to apoptosis as a result of their dependence on positioning (Pasquale, 2005). More recently, some testosterone. It is quite regrettable that no link has ever ephrin and Eph receptors have been found to affect cell been made between the dependency phenomenon of death in neurogenic regions. Activation of EphA7 by prostate cancer cells and the fact that AR could behave overexpressed ephrinA5 in the embryonic cortex re- as a DR. sulted in neural progenitor apoptosis (Depaepe et al., 2005). On the other hand, lack of ephrinB3 is associated with apoptosis in the subventricular zone of adult mice DRs are caspase substrates (Ricard et al., 2006), suggesting that its receptor could function as a DR. EphA4 has been shown to function as The molecular hallmark of a DR, the apoptotic activity of which is impaired by its (apoptosis) is the activation of (Thornberry ligand ephrinB3 (Furne et al., 2009). and Lazebnik, 1998). During apoptosis, caspases, which Another tyrosine kinase receptor should probably be form a family of cysteine-dependent aspartate-directed added to the list: MET, the receptor for hepatocyte proteases, can cleave a wide range of substrates, thereby growth factor (Figure 2 and Table 1). MET is well inactivating survival and activating proapoptotic me- known for its essential role in normal development and chanisms. Except for the fact that they induce apoptosis cell survival. Interestingly, it was reported that MET in the absence of their ligand, the other most common was cleaved under stress conditions by caspase, thereby characteristic of DR is that they are cleaved by caspases. generating an apoptotic fragment (Tulasne et al., 2004). Receptor cleavage is important for apoptotic function, As we will see below, caspase cleavage is one of the most as of the cleavage site abolishes cell death common traits for apoptotic signaling of DRs. More- induction. DCC, neogenin, Ptc, ALK, EphA4 are over, inhibits this caspase cleaved once, roughly in the middle of their intracellular cleavage and concomitantly apoptosis. Therefore, domain (Mehlen et al., 1998; Thibert et al., 2003; although it is not clear whether MET is able to initiate Matsunaga et al., 2004; Mourali et al., 2006; Furne apoptosis by itself in the absence of ligand, as other et al., 2009). The cleavage site of UNC5 receptors is very DRs do, this receptor presents some striking similarities close to the plasma membrane (Llambi et al., 2001), with DRs. whereas RET, Trkc and MET have two cleavage sites

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1870 Table 2 Conservation of caspase cleavage sites of DR among species Homo Pan Bos Canis Mus Rattus Gallus Xenopus Xenopus Danio sapiens troglodytes taurus familiaris musculus norvegicus gallus tropicalis laevis rerio

DCC LSVD LSVD NA LSVD LSVD LSVD NA NA LTVD No UNC5B DITD DITD DITD DITD DITD DITD DITD NA DITD EITD Neogenin CCTD CCTD CCTD CCTD CCTD CCTD PCAD? GPED? NA CTTD Ptc PETD PETD PETD PETD PETD PETD NEDD? HEND? HEND? No RET VSVD VSVD VSVD VSVD VPVD VSVD VSVD NA MSVD VAID DYLD DYLD DYLD DYLD DYLD DYLD DYLD DYLD DYLD TrkC SSLD SSLD SSLD SSLD SSLD SSLD SSLD NA NA NA ILVD ILVD ILVD ILVD ILVD ILVD ILVD EphA4 LEDD LEDD LEDD LEDD LEDD LEDD LEDD LEDD LEDD LEED ALK DELD DELD DELD DELD DELD DELD DELD DEMD NA DELD MET ESVD ESVD ESVD ESVD ESVD ESVD ESVD ESVD ESVD ESVD DNADDNAD No DNID DNID DNID DNTDNoNo SNLD?

Caspase cleavage sites of DR have been mapped by experiment on human or rodent proteins. This table lists these sites for various DRs and the amino acids found at the same position in sequence from other species. Caspase cleavage sites are well conserved in mammals and variably conserved in other vertebrates: for example, cleavage site of EphA4 is better conserved than those of neogenin or patched. In some cases, sequence is not perfectly conserved but only a slight variation is observed between species (amino acid is shown in bold). In other cases an aspartate residue is found at the correct position, but the surrounding residues are not conserved (shown in italic). No: means that no asparte residue has been found by sequence alignment. NA: means not analyzed or not found in databases. Alignments have been performed using sequence from Ensembl or NCBI databases.

(Bordeaux et al., 2000; Foveau et al., 2007; Tauszig- cleavage inhibition. In addition, ligand binding has been Delamasure et al., 2007). The cases of p75NTR and proposed to have other structural effects, such as integrins are less clear, as there is no solid evidence that receptor multimerization. In fact, with the exception of caspase cleavage is needed for their proapoptotic effect. Ptc and integrins, most DRs display homo-multimeriza- Interestingly, caspase cleavage sites of DRs seem to be tion properties in the presence of their respective ligand conserved in mammals, variably in other vertebrates but (Wang et al., 2000; Manie et al., 2001; Stein et al., 2001; never in orthologs in lower organisms, such as nematode Mille et al., 2009a). Initially described as being or Drosophila (Table 2) (Mehlen and Thibert, 2004). important for positive signaling, receptor multimeriza- These findings may suggest that the appearance as a tion also seems to have a role in blocking apoptosis caspase substrate, and therefore the mediation of the induction. P75NTR exerts its proapoptotic effect as a dependence state, is a relatively late event in the monomer, whereas multimerization abrogates this effect evolution of these proteins. This may make sense, given (Rabizadeh et al., 2000). In the same way, results the greater plasticity of the mammalian nervous system obtained with DCC and UNC5H2 showed that these compared with those of invertebrates and the necessity receptors trigger cell death when their ligand-induced for more complex and higher lifespan mammals to multimerization is hindered (Mille et al., 2009a). Netrin- develop antitumor mechanisms. 1 has also been proposed to suppress the apoptotic function of UNC5H2 by inducing interaction of the receptor with the GTPase PIKE-L (Tang et al., 2008). This interaction triggers the activation of PI3 kinase Role of ligand binding signaling and consequently the inhibition of UNC5H2 proapoptotic function. Furthermore, recent data from Another common feature of DRs is the inhibition of cristallography evidenced that the UNC5H2 intra- apoptosis induction on ligand binding. The main cellular domain adopts an autoinhibited closed con- hypothesis with regard to the role of ligand binding is formation. In this conformation, ZU5 and death inhibition of the caspase cleavage. This hypothesis has domains bind to each other and are thus unable to been partly confirmed for some DRs, UNC5H2 induce cell death. Netrin-1 is unable to prevent (Tanikawa et al., 2003), TrkC (Tauszig-Delamasure apoptosis induced by the UNC5H2 mutant that has a et al., 2007), EphA4 (Furne et al., 2009) and MET constitutive open conformation, which leads the authors (Tulasne et al., 2004; Foveau et al., 2007). For other to suggest that netrin-1 somehow stabilizes the auto- receptors, the effect of ligand on caspase cleavage inhibited conformation of UNC5H2 (Wang et al., 2009). cannot be assessed because cleavage products have It must be noted that, for receptors accepting more relatively short half-lives and are thus hardly detectable than one ligand, the antiapoptotic effect has not been in vivo. On the other hand, ectopic expression of a demonstrated for all ligands. For example, only glial cell truncated receptor, mimicking the caspase cleaved line-derived neurotrophic factor was shown to block receptor, leads to apoptosis induction even in the RET-induced apoptosis (Bordeaux et al., 2000), no data presence of ligand (Mehlen et al., 1998; Thibert et al., are available for neurturin, artemin and persephin, 2003; Matsunaga et al., 2004). This is an indirect although neurturin, similar to glial cell line-derived argument in favor of a role for the ligand in caspase neurotrophic factor, was recently proposed to be a

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1871 survival factor for parasympathetic neurons (Peterziel netrin-1, DCC recruits and activates caspase 9, thereby et al., 2007). In the case of neogenin, the ligand allowing caspase 3 activation, but this process does not antiapoptotic effect was reported only for chick RGM/ require cytochrome c release and subsequent formation RGMa (Matsunaga et al., 2004) but not for other of an apoptosome (cytochrome c/apaf-1/caspase 9) members of the RGM family or for netrin-1/3/4. EphA4- complex, as is the case in the classic mitochondrial associated cell death induction is hindered by ephrinB3, pathway (Forcet et al., 2001). DCC does not interact but not by its other known ligands, ephrinA1 and directly with caspase 9, hence it may recruit one or more ephrinA4 (Furne et al., 2009). With regard to ALK, only adaptor proteins (Figure 3). One of them could be ligand-mimicking antibodies have been used because the DIP13a (DCC-interacting protein 13-a), a protein identity of the receptor’s physiological ligand is still a identified as an interactor of DCC ADD, and shown matter of debate. Pleiotrophin and , two heparin- to be important for DCC-induced cell death (Liu et al., binding growth factors, have been proposed to function 2002). However, the precise role of DIP13a in DCC- as ALK ligand but these proteins fail to make a general triggered apoptosis remains quite obscure, as it does not consensus. This controversy is further supported by the seem to mediate interaction of DCC with caspase 9 and genetic identification in Drosophila melanogaster of an further studies performed on DIP13a, also known as ALK ligand, jelly belly (jeb), which has no similarities APPL1 (adaptor protein containing PH domain, PTB with pleiotrophin or midkine (Englund et al., 2003; Lee domain and leucine zipper motif 1), have not provided et al.,2003;Lorenet al., 2003). clear evidence for a role of this protein in apoptosis Finally, in the particular case of integrins, the ligand induction. In addition, palmitoylation and lipid raft must be an immobilized substrate ligand to block localization were reported to be a prerequisite for DCC apoptosis as, contrary to other DRs, soluble ligand proapoptotic activity, both in vitro and in primary is not sufficient to counteract cell death induction commissural neurons (Furne et al., 2006). Lipid rafts are (Stupack, 2005). ordered membrane microdomains enriched in sphingo- lipids and cholesterol, and are proposed to have an important role in cell signaling, in particular through the organization of surface receptors, signaling and Proapoptotic signaling by DRs adaptor molecules into complexes at specific sites in the membrane (Simons and Toomre, 2000; Hueber, 2003). It DRs share the property of being caspase amplifiers; was then shown that DCC presence in lipid rafts is indeed most of them fail to induce apoptosis in the required to allow caspase-9/DCC interaction, suggesting presence of general caspase inhibitors such as zVADfmk that this caspase-activating complex occurs in and takes (Mehlen and Thibert, 2004). The way that leads to advantage of lipid rafts. caspase activation and amplification has begun to be The discovery of a caspase-activating complex re- decrypted specifically for some of them. First, all DRs cruited to a DR was recently made for Patched. Ptc was contain, in their intracellular part, a domain required for indeed found to interact through its ADD, and only in apoptosis induction. This domain, the ADD (addiction/ the absence of its ligand Shh, with DRAL/FHL2 (Mille dependence domain) (Bredesen et al., 1998), is required et al., 2009b). DRAL was already known to promote and often sufficient for cell death induction. Caspase apoptosis through an unknown mechanism in a wide cleavage, except for p75NTR and integrins, is thought to be variety of cells when overexpressed (Scholl et al., 2000) responsible for unmasking the ADD. In most cases, and to interact with TUCAN, a CARD-containing ADD is borne by the remaining membrane-anchored adaptor protein for caspases 1 and 9 (Stilo et al., 2002). fragment. In the case of UNC5H, RET, TrkC and MET Mille and colleagues showed that the Ptc–DRAL receptors, however, it is the cytosolic generated fragment association serves as a platform for recruiting TUCAN that is proapoptotic. ADDs are usually unique regions (and/or NALP1, a protein closely related to TUCAN) that are not structurally related to known protein and caspase 9 (Figure 4). This then allows caspase 9 functional domains. Two notable exceptions are p75NTR recruitment to Ptc and caspase 9 activation. The and UNC5H receptors: in those two cases, two regions complex involving DRAL, TUCAN and caspase 9 was corresponding to known functional domains are respon- named dependosome, by analogy to other known sible for apoptosis induction. The first region is their caspase-activating complexes such as the apoptosome death domain, which is structurally related to the death (comprising Apaf-1, cytochrome c and caspase 9), DISC domain of receptors of the TNF receptor superfamily (comprising Fas, FADD and caspase 8), the PIDDo- (Hofmann and Tschopp, 1995; Bredesen et al.,1998; some (comprising PIDD, RAID and caspase 2) and the Llambi et al., 2001). The second region is the chopper inflammosome (comprising NALPs, ASC, caspases 1 domain for p75NTR (Coulson et al., 2000) and the ZU5 and 5). Additional studies are required to determine domain for UNC5H (Williams et al.,2003). whether this dependosome is a common platform for After ADD release/exposition, caspase amplification other DRs. seems to be more or less direct, depending on receptors. In any case, if such a dependosome existed and were In some cases, ADD recruits caspase-activating com- common to DRs, the initiator recruited caspase could plexes that are different from those implicated in death not always be caspase 9. Indeed, Stupack et al. (2001) receptors and in intrinsic mitochondrial classical elegantly showed that integrins, as DRs, trigger apoptotic pathways. For example, in the absence of apoptosis through recruitment of caspase 8.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1872

Netrin-1

DIP13α X

C ASP Cleavage ASE 9 Caspase 3

Caspase 3 activation Amplification of receptor cleavage

Apoptosis Figure 3 Model of cell death induction by DCC. In the presence of netrin-1, DCC is dimerized and interacts with procaspase 3. Following ligand withdrawal, DCC becomes a monomer and is cleaved, possibly by bound caspase 3 or another activated protease. Cleavage leads to ADD exposure and to its direct interaction with apoptotic partners such as DIP13a, the role of which remains unclear, or to indirect interaction with caspase 9. Consequently, these interactions lead to caspase 9 activation, which in turn activates caspase 3.

Shh

Shh

Cleavage

Caspase 3 activation Amplification of receptor cleavage

Apoptosis Figure 4 Model of cell death induction by Ptc. Following ligand withdrawal, Ptc is cleaved by caspase (or by another activated protease), thus allowing exposure of its ADD. Ptc recruits DRAL, which in turns recruits TUCAN (or NALP1) and caspase 9. Formation of this complex leads to caspase 9 activation and consequently to caspase 3 activation.

The formation of a caspase-activating complex does fact, apoptotic pathways downstream of UNC5H3 and not seem to be the only mechanism used by DRs to 4 have not yet been documented, and functional data are trigger apoptosis. As an example, the mechanism of available only for UNC5H1 and 2. UNC5H1 induces UNC5H-induced apoptosis has been documented. apoptosis by interacting with NRAGE (neurotrophin Despite their structural homology, members of the receptor-interacting MAGE homolog) through its ZU5 UNC5H family seem to mediate their apoptotic signal domain (Williams et al., 2003). NRAGE possibly by interacting preferentially with distinct partners. In transduces UNC5H1-induced apoptosis through at least

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1873

Unc5H2

Netrin-1

Cleavage ZU5 ZU5 ZU5 5 DD DD P ZU DAPK Amplification of DD Closed receptor cleavage DA PK conformation P DAPK

Caspase 3 activation

Apoptosis Figure 5 Model of cell death induction by UNC5H2/UNC5B. In the presence of netrin-1, UNC5H2 and UNC5B are dimerized, and their intracellular domain adopts a close conformation in which ZU5 and death domains interact with each other. DAPK interacts with the closed intracellular domain, but is in an inactive autophosphorylated state. Following ligand withdrawal, UNC5H2 receptor becomes a monomer, whereas the intracellular domain undergoes both caspase (or another protease) cleavage and opening/ dissociation of ZU5 and DD in parallel. Relation and chronology between cleavage and opening remain unclear, but these modifications should allow interaction between the DD of UNC5H and DAPK, thus leading to DAPK activation and initiation of an apoptotic program. Precisely how activated DAPK induces apoptosis remains to be determined. two pathways: one implicating the degradation of the UNC5H2 through their non-‘death domain’-interacting caspase inhibitor XIAP, and the other implicating regions, whereas, in the absence of netrin-1, DAPK activation of the proapoptotic JNK signaling pathway. interacts with the death domain of UNC5H2, NRAGE can also interact with UNC5H2 and 3, but which allows DAPK activation (Figure 5). This hypoth- with a much weaker binding affinity. On the other hand, esis strongly fits with the more recent study by UNC5H2 triggers apoptosis mainly by recruiting the Wang et al. (2009), arguing that UNC5H2 can adopt a serine/threonine death-associated closed conformation, preventing association of the (DAPK) (Llambi et al., 2005). UNC5H2 and DAPK death domain with other proteins. Downstream bind each other in part through their respective death effectors of DAPK in UNC5H2-mediated cell death domain, but not only so, as deletion of these domains is remain to be identified, but DAPK is already known to not sufficient to abrogate their association. Surprisingly, trigger cell death through p53-dependent and -indepen- although UNC5H2-mediated induction of DAPK ac- dent mechanisms. Moreover, phosphorylation of the tivity is observed only in the absence of netrin-1 and myosin light chain by DAPK leads to membrane requires UNC5H2 caspase cleavage, DAPK seems to blebbing, a hallmark of programmed cell death interact constitutively with UNC5H2, that is, not only (Gozuacik and Kimchi, 2006). Interestingly, it was in the absence of netrin-1. Indeed, DAPK is known to recently suggested that UNC5H2 was not the only DR be capable of autophosphorylation, which inhibits to trigger apoptosis through DAPK, as neogenin was its activity by inducing a conformational change also shown to interact with DAPK and to require (Shohat et al., 2001). On this basis, and according to DAPK for apoptosis induction (Fujita et al., 2008), in their observations, Llambi and colleagues proposed the same way that DCC, UNC5H1, 2 and 3 also require that, in the presence of netrin-1, DAPK is in an inactive lipid raft association to induce cell death (Maisse et al., autophosphorylated state, and it interacts with 2008).

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1874 The fact that DRs are at the same time caspase TrkC-induced apoptosis in the classic neurotrophin substrates and caspase activators/amplifiers points out a theory. The classic dogma suggests that each neuron is paradox. How can a receptor that requires caspase moving toward death unless a survival/neurotrophin cleavage to be a proapoptotic molecule participate in signal is provided. The integration of the DR notion apoptosis induction? One possibility could be the within this neurotrophin theory would then be that each initiation of DR cleavage by a noncaspase protease, neuron is actively pushed toward apoptosis by a DR, which would be sufficient to generate a caspase such as TrkC, in the context of ligand limitation, amplification loop. Another view could be that caspases whereas ligand presence not only activates survival are never completely inactive, even in nonapoptotic signals but also blocks the active process of cell death. cells, and that this residual caspase activity is sufficient The idea of TrkC being a DR is particularly attractive to detect receptors disengaged from their ligand. while analyzing data from knockout mice for neuro- Interestingly, caspase 3 was observed to interact with trophins and their respective receptors. Indeed, inactiva- DCC, downstream of its cleavage site, only in non- tion of TrkA or NGF in mice results in the same amount apoptotic conditions, that is, in the presence of netrin-1, of sensory neuron loss at birth (that is, nociceptive or when the DCC caspase cleavage site is mutated neurons) (Crowley et al., 1994). Similarly, inactivation (Forcet et al., 2001). It is tempting to speculate that, of either TrkB or brain-derived neurotrophic factor when dimerized in the presence of its ligand, DCC results in an equivalent loss of mechanoceptive neurons adopts a conformation that prevents its cleavage by (Ernfors et al., 1994; Minichiello et al., 1995). On the caspases, whereas it can be efficiently cleaved as a other hand, neonates invalidated for TrkC present a loss monomer in the absence of ligand. This event results in of 30% DRG neurons, whereas NT-3À/À neonates have unmasking its ADD, thus allowing capase 9 activation lost 70% of them (Tessarollo et al., 1994, 1997). This is and caspase 3 amplification. in agreement with the view that TrkC, contrary to TrkA and B, can trigger, when deprived of ligand, an active apoptotic signaling. A confirmation that TrkC apopto- tic signaling controls the fate of sensory neurons is Role of DRs during embryonic development provided by an experience in which microinjection in sensory neurons of a mutated intracellular domain of Before being identified as DRs, all members of this TrkC, known to interfere with TrkC apoptotic function, functional family were already known to be implicated dramatically enhanced survival of NT-3-deprived neu- in nervous system development. Implications of their rons (Tauszig-Delamasure et al., 2007). positive signaling in the presence of their ligand are The ligand/DR pair ephrinB3/EphA4 was also largely documented, whereas a role for negative recently demonstrated, by studying knockout mice, to proapoptotic signaling long remained essentially spec- have an important role in regulating cell number in an ulative. However, several lines of evidence now accu- adult mouse subventricular zone through apoptosis mulate to further support the view that DRs participate modulation. Indeed, extinction of eprhinB3 results in in nervous system development through their proapop- increased apoptosis in subventricular zone, whereas the totic function as well. Because of this ability to trigger absence of EphA4 results in an excessive number of apoptosis in settings of ligand absence or limitation, neuroblasts in this zone (Furne et al., 2009). In addition, they were hypothesized to control cell numbers in some infusion of soluble ephrinB3 into the lateral ventricle specific areas of the developing brain and to dictate reduced cell death. Thus, it is tempting to speculate that adequate territories of neuron migration and axon EphA4, as a DR, is important in regulating the fate of projection by eliminating those localized out of regions neuronal stem cells during brain development. of ligand availability. For example, netrin-1 receptors do An elegant study from Palmer and colleagues not only mediate the chemotropic effect of netrin-1 in evidenced a role for the ALK/jeb pair in the Drosophi- the developing nervous system but also seem to regulate la-developing visual system. ALK is expressed and the survival of olivary neurons, as these cells, known to required in target neurons in the optic lobe, whereas express DCC and UNC5H receptors, display increased jeb is primarily generated by photoreceptor axons and apoptosis in netrin-1À/À mice (Llambi et al., 2001). functions in the eye to control target selection of specific Moreover, netrin-1 functions as a survival factor for photoreceptor cell axons. Interestingly, the level of spinal cord commissural neurons, which was shown in neuronal cell death (measured by active caspase 3 level) both primary neuron cultures and animal models (Furne in the ALK expressing optic lobe medulla increases in et al., 2008). mutants lacking an expression of jeb. Moreover, Similarly, it was shown that Shh, the ligand of DR Ptc, caspase-dependent neuronal apoptosis dramatically is not only a morphogen but also a survival factor decreases in mutants overexpressing jeb (Bazigou (Charrier et al., 1999, 2001). Interestingly, it was et al., 2007). These results suggest that ALK could have demonstrated that Shh functions as a survival factor by a role in the physiological negative selection of neurons inhibiting the apoptotic function of Ptc and that this shaping the optic lobe in the Drosophila nervous system proapoptotic function is crucial for adequate neural tube by favoring apoptosis in the absence of the ALK ligand. development (Thibert et al., 2003; Mille et al.,2009b). Interestingly, even though the data reported so far on The recent observation that neurotrophin receptor the role of DRs during development seem to be linked to TrkC is a DR also led to questioning the implication of nervous system development, it has recently been

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1875 unbound receptor apoptotic cancer cell

bound receptor cancer cell

normal cell

Figure 6 DRs and tumor suppression. Normal cells located in their normal environment express DRs that are bound to ligands supplied by local source. In contrast, in this limited and constant ligand concentration environment, highly proliferative tumor cells display some unbound DRs and thus undergo apoptosis. In the case of metastatic tumor cells escaping from the primary site, migration away from locally supplied ligands leads to apoptosis because of unbound receptors. DRs could thus counteract tumor growth and/or invasiveness. Loss of expression of these receptors or overexpression of ligands would represent a selective advantage for cancer cells. proposed that these DRs may also be involved outside on 18q, DCC is submitted to loss of the developing nervous system. Along this line, netrin-1 heterozygosity in over 70% of colorectal cancers was recently shown to control the survival of endothelial (Fearon et al., 1990). DCC is also submitted to loss of cells and to promote angiogenesis, at least in part, by heterozygosity and/or to decreased expression in various blocking apoptosis triggered by its unbound UNC5H2 other cancers including gastric, prostate, endometrial, receptor (Castets et al., 2009). The DR activity of ovarian, esophageal, breast and testicular cancer, as well UNC5H2 can indeed conciliate conflicting results as neuroblastoma and hematological malignancies regarding the implication of netrin-1 in angiogenesis (Mehlen and Fearon, 2004). Loss of DCC expression (Lu et al., 2004; Wilson et al., 2006). is often associated with poor prognosis and advanced cancer or (Shibata et al., 1996; Saito et al., 1999), suggesting a role of DCC loss in cancer progression rather than in cancer initiation. Moreover, DRs are altered during tumor progression restoration of DCC expression can suppress tumorigenic growth properties in vitro or in nude mice (Klingelhutz In addition to a role during embryonic development, the et al., 1993; Velcich et al., 1999; Kato et al., 2000; DR model also predicts a role for such receptors as Rodrigues et al., 2007). On the other hand, the fact that tumor suppressors, because of their ability to promote only 10–15% of colon cancers carry mutations in DCC cell death when disengaged from their ligand. A tumor and the lack of a tumor predisposing effect of DCC cell submitted to an abnormal environment (highly inactivation in mouse models (Fazeli et al., 1997) led proliferative cells in an environment with limited and some investigators to conclude that DCC had little or no constant ligand concentration or metastatic cells migrat- biological role in colon cancer, and that its inactivation ing to sites in which ligand is absent) would display was an epiphenomenon. However, such a categoric unbound DRs and thus undergo apoptosis (Figure 6). judgment was partly due to the lack of understanding of This mechanism would represent an alternative safe- the biological roles of DCC, which has since been guard mechanism to limit tumor progression. It is then compensated by its characterization as a DR (Grady, expected that in aggressive tumors, tumor cells have to 2007). turndown this DR pathway. Consistent with this view, a In the same way, the UNC5H receptor family is loss of receptor expression would then represent a downregulated in human cancers, including colorectal, selective advantage for tumor cells and seem to be a breast, ovary, uterus, stomach, lung or kidney cancers primary method to overcome this safeguard mechanism. (Thiebault et al., 2003). Recently, two studies have Since its discovery in the 1990s, DCC has been focused on UNC5H3/UNC5C alteration in colorectal suspected to be a tumor suppressor gene, even though carcinoma. UNC5C is indeed the most downregulated no definitive evidence has been proposed so far. Located member of the UNC5H family (74–77% of cases,

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1876 whereas UNC5H1/UNC5A and H2/B show a reduced expression by small interfering RNA or netrin-1 expression in 48 and 27% of the cases, respectively). titration by decoy soluble receptor ectodomain causes This loss of expression observed in human primary apoptosis and prevents metastasis formation both in a tumors, as well as in cell lines, is essentially due to syngenic mouse model and in a xenograft model promoter methylation (Bernet et al., 2007; Shin et al., (Fitamant et al., 2008). In the same way, high levels of 2007). Furthermore, Bernet and colleagues took advan- netrin-1 were detected in almost 50% of non-small-cell tage of a natural UNC5H3 loss-of-function occurring in lung cancer and in a large fraction of aggressive mice (rcm, rostral cerebellar malformation) to demon- neuroblastoma. As in the breast cancer study, strategies strate that UNC5H3 loss of function is associated with disrupting the netrin-1 autocrine loop led to apoptosis tumor progression: mice that carry the APC1638N germ- induction and tumor growth inhibition in xenografted line mutation, known to predispose mice to the models (Delloye-Bourgeois et al., 2009a, b). In these development of low-grade adenoma (Sieber et al., three cases, apoptosis resulting from netrin-1 inhibition 2000), and are heterozygous or homozygous for mutant seems to be mediated by UNC5H receptors, rather than UNC5H3, develop adenomas that progress to adeno- by DCC. Interestingly, in the case of aggressive carcinoma at a higher frequency than what is seen in neuroblastomas, netrin-1 expression levels were found APC1638N mice. Interestingly, loss of UNC5H3 function to have prognosis significance. Aggressive stage 4 in mice also correlates with apoptosis reduction in mice metastatic neuroblastoma is divided into three groups: tumors. Another clue in favor of a role of the UNC5H stage 4 in children aged more than 1 year has the worse family in cancer is that UNC5B and D are p53 target prognosis, whereas stage 4S and stage 4 in children less , able to mediate part of p53 proapoptotic activity than 1-year old generally have a more favorable (Tanikawa et al., 2003; Wang et al., 2008). prognosis, even though many infants succumb to Patched is also a known tumor suppressor (Stone disease. High levels of netrin-1 were shown to correlate et al., 1996). Inactive mutations of Ptc, as well as loss of with adverse outcome of stage 4S and stage 4 (o1 year). expression, are found in basal cell carcinoma and A study by Link et al. (2007), reporting that netrin-1 medulloblastoma (Wicking and McGlinn, 2001). Ptc expression has significant impact on the overall survival expression inhibits the hallmarks of cell transformation of patients with poorly differentiated pancreas tumors, in vitro (Koike et al., 2002) and, interestingly, Ptc also completes this description of netrin-1 upregulation in inhibits growth in soft agar of transformed cells. This is human neoplasias. Although the mechanism for auto- linked to its proapoptotic function, as growth inhibition crine production of netrin-1 remains to be determined, it does not occur in the presence of Shh, or of a general could be at least in part a result of nuclear factor-kB caspase inhibitor, or when Ptc is mutated on its caspase activation, as netrin-1 is a direct target gene of this cleavage site (Thibert et al., 2003). However, there is no transcription factor (Paradisi et al., 2008). Moreover, strong in vivo evidence so far that Ptc functions as a according to the well-admitted link between inflamma- tumor suppressor because it triggers apoptosis. tion and colorectal cancer predisposition, it has been There is a wide spectrum of data supporting the role suggested that nuclear factor-kB activation resulting of most DRs as tumor suppressors. As an example, from inflammatory stimulus could lead to local netrin-1 EphA4 is downregulated in invasive forms of breast production, and thus to tumor promotion by apoptosis cancers (Fox and Kandpal, 2004), in liver and kidney inhibition. Along this line, colorectal tumor formation cancers (Hafner et al., 2004) and in metastatic melano- in an animal model for chronic inflammation was mas (Easty et al., 1997), whereas a progressive decrease inhibited by treatment with netrin-1 titrating agents in p75NTR expression is described in prostate cancers (Paradisi et al., 2009). (Pflug and Djakiew, 1998). TrkC is associated with good Finally, a third possible way for tumor cells to escape prognosis in several cancers (Yamashiro et al., 1997). the proapoptotic activity of DRs would be to inactivate However, the tumor suppressive functions of these their downstream signaling pathways. Notably, three receptors are yet to be demonstrated per se. effectors of DRs display functional inactivation in Whereas DR loss during tumorigenesis occurs in a human cancers: DAPK, DRAL and caspase 8, which wide fraction of cancers, another selective advantage for transduce UNC5H2-, Ptc- and integrin-mediated apop- tumor cells would be to constitutively overexpress tosis, respectively. DAPK loss of expression, essentially ligand. There is now accumulating evidence with regard through promoter methylation, has now been described to netrin-1 to support this idea. Indeed, forced expres- in a wide variety of cancers, including lymphoma, sion of netrin-1 in the digestive tract of transgenic mice leukemia, brain tumors, bladder, breast, renal, cervix, has been associated with decreased apoptosis in the prostate and colorectal carcinomas (Kissil et al., 1997; intestine, development of advanced adenomas and Raveh and Kimchi, 2001; Gozuacik and Kimchi, 2006). tumor progression to adenomacarcinoma in a setting Moreover, DAPK loss of expression is associated with a of adenoma predisposition (Mazelin et al., 2004). More more malignant tumor phenotype and increased meta- recently, high levels of netrin-1 were detected in a large static capacity. DAPK is absent in highly metastatic panel of human cancers from distinct organs, and variants of mouse lung cancer cell lines, and is present in netrin-1 overexpression was correlated with a blocking the low metastatic variants of those same cell lines of the proapoptotic functions of netrin-1 receptors. (Inbal et al., 1997). In lung and head and neck cancers, First, in breast cancer, netrin-1 was shown to be a DAPK promoter methylation was associated with marker of metastatic disease: decrease in netrin-1 aggressive disease and poor survival (Sanchez-Cespedes

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1877 et al., 2000; Tang et al., 2000; Kim et al., 2001; Kong window. However, it remains to be determined whether et al., 2005). DRAL was initially characterized as a interfering with DR/ligand pairs will stand the test of downregulated gene in rhabdomyosarcoma (Genini clinical tests. The situation in human patients is often et al., 1997). Low or no transcript levels were observed more complex than that in murine experimental systems, in lymphoblastic leukemia, promyelocytic leukemia and and potential secondary effects due to perturbation of Burkitt’s lymphoma cells (Johannessen et al., 2006; netrin-1 in normal tissues need to be evaluated. Finally, Desmond et al., 2007). Finally, caspase 8 expression is this therapeutic approach could be extended to other selectively lost during establishment of neuroblastoma DR ligands besides netrin-1, if further investigations metastases, rendering these cells refractory to unligated show abnormal expression levels in cancers. integrin-mediated cell death (Stupack et al., 2006).

Future members of the family? DRs as new therapeutic targets The list of DRs is of course not closed, and future Reactivating proapoptotic pathways in human cancer studies will probably reveal the unexpected ability of cells is one of the main strategies that is being developed some receptors to induce apoptosis in the absence of in oncology nowadays. Therefore, DRs can join the list ligand. Some receptors already share some intriguing of promising targets for cancer treatment. Whereas characteristics with DRs. For example, ErbB2, a restoring the expression of an extinguished receptor- related to the EGF receptor coding gene does not seem conceivable, interfering with family, can be cleaved by caspase, and the generated ligand binding seems to be a more pertinent approach, cytosolic fragment triggers apoptosis (Tikhomirov et al., further supported by recent studies reporting an over- 2005; Strohecker et al., 2008). Interestingly, this frag- expression of netrin-1 in various cancers (Link et al., ment contains a BH3-like domain related to the Bcl 2007; Fitamant et al., 2008; Delloye-Bourgeois et al., involved in mitochondrial apoptosis. An 2009a, b). These studies provide an idea of what the important issue would be to check whether ErbB2 future therapeutic molecules used in clinical studies cleavage is affected by the presence of ligand. Unfortu- could be; for example, a decoy receptor corresponding nately, ErbB2 remains, to date, an orphan receptor. The to the entire ectodomain of DCC receptor that would epidermal and Notch1 receptors titrate the ligand, thus leading to unbound membrane are also caspase substrates, but no effect of ligand receptors (Fitamant et al., 2008). However, the large size binding has been reported, and caspase cleavage has (1100 aa) of the complete extracellular domain of DCC only been proposed as a mechanism for turning down may complicate its use in vivo. An alternative could be to signaling by these receptors (He et al., 2003, 2006; use shorter polypeptides, corresponding to the fourth or Cohen et al., 2005). An even more promising candidate fifth fibronectin-like domain of DCC (Fitamant et al., is the b-amyloid precursor protein (APP), a transmem- 2008; Delloye-Bourgeois et al., 2009a, b), which are both brane receptor, the biological function of which remains known to interact with netrin-1 (Geisbrecht et al., 2003). largely unknown. APP abnormal processing by secretase Unlike the complete ectodomain, the fifth and fourth proteases led to accumulation and extracellular deposit fibronectin-like domains do not counteract interaction of the small Ab associated with the development between netrin-1 and its receptors, but rather block of Alzheimer’s disease, a common neurodegenerative receptor multimerization (Mille et al., 2009a). Alterna- disorder (Koo, 2002). Until recently, APP was an tively, other interfering molecules, such as blocking orphan receptor, yet it was recently proposed as a antibodies or peptidomimetic compounds, could block netrin-1 receptor (Lourenco et al., 2009). In addition, the interaction between netrin-1 and its receptors. APP is known to be cleaved in its intracellular domain Interestingly, cancers that show high levels of netrin-1, by caspase, thus yielding a toxic fragment (Nguyen that is, metastatic breast cancers, neuroblastoma, non- et al., 2008). Whether APP is a netrin-1 DR remains an small-cell lung cancer and pancreatic cancers, are all intriguing question. In addition, if APP is a DR, one tumors in which there is a need for effective new would expect it to function as a tumor suppressor. So therapies. Moreover, and importantly, therapy resis- far, no convincing data link APP to cancer, but this tance in cancer is frequently associated with deregula- remains to be carefully assessed. tion in the mechanisms that control apoptosis, and cancer cells are often reliant on these molecular aberrations for survival. Inducing a novel apoptosis Conflict of interest pathway may add one new component to the combined treatment of cancer, thereby increasing the therapeutic The authors declare no conflict of interest.

References

Allouche M. (2007). ALK is a novel dependence receptor: Arighi E, Alberti L, Torriti F, Ghizzoni S, Rizzetti MG, Pelicci G et al. potential implications in development and cancer. Cell Cycle 6: (1997). Identification of Shc docking site on Ret tyrosine kinase. 1533–1538. Oncogene 14: 773–782.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1878 Baloh RH, Enomoto H, Johnson Jr EM, Milbrandt J. (2000). The Delloye-Bourgeois C, Fitamant J, Paradisi A, Cappellen D, Douc- GDNF family ligands and receptors—implications for neural Rasy S, Raquin MA et al. (2009b). Netrin-1 acts as a survival factor development. Curr Opin Neurobiol 10: 103–110. for aggressive neuroblastoma. J Exp Med 206: 833–847. Barrett GL, Bartlett PF. (1994). The p75 receptor Depaepe V, Suarez-Gonzalez N, Dufour A, Passante L, Gorski JA, mediates survival or death depending on the stage of sensory neuron Jones KR et al. (2005). Ephrin signalling controls brain development. Proc Natl Acad Sci USA 91: 6501–6505. size by regulating apoptosis of neural progenitors. Nature 435: Bazigou E, Apitz H, Johansson J, Loren CE, Hirst EM, Chen PL et al. 1244–1250. (2007). Anterograde Jelly belly and Alk receptor tyrosine kinase Desmond JC, Raynaud S, Tung E, Hofmann WK, Haferlach T, signaling mediates retinal axon targeting in Drosophila. Cell 128: Koeffler HP. (2007). Discovery of epigenetically silenced genes in 961–975. acute myeloid leukemias. Leukemia 21: 1026–1034. Bernet A, Mazelin L, Coissieux MM, Gadot N, Ackerman SL, Scoazec Easty DJ, Mitchell PJ, Patel K, Florenes VA, Spritz RA, Bennett DC. JY et al. (2007). Inactivation of the UNC5C Netrin-1 receptor is (1997). Loss of expression of receptor tyrosine kinase family associated with tumor progression in colorectal malignancies. genes PTK7 and SEK in metastatic melanoma. Int J Cancer 71: Gastroenterology 133: 1840–1848. 1061–1065. Bordeaux MC, Forcet C, Granger L, Corset V, Bidaud C, Billaud M Edery P, Lyonnet S, Mulligan LM, Pelet A, Dow E, Abel L et al. et al. (2000). The RET proto-oncogene induces apoptosis: a novel (1994). Mutations of the RET proto-oncogene in Hirschsprung’s mechanism for Hirschsprung disease. EMBO J 19: 4056–4063. disease. Nature 367: 378–380. Bredesen DE, Ye X, Tasinato A, Sperandio S, Wang JJ, Assa-Munt N Ellerby LM, Hackam AS, Propp SS, Ellerby HM, Rabizadeh S, et al. (1998). p75NTR and the concept of cellular dependence: seeing Cashman NR et al. (1999). Kennedy’s disease: caspase cleavage of how the other half die. Cell Death Differ 5: 365–371. the androgen receptor is a crucial event in cytotoxicity. J Neurochem Canibano C, Rodriguez NL, Saez C, Tovar S, Garcia-Lavandeira M, 72: 185–195. Borrello MG et al. (2007). The dependence receptor Ret induces Englund C, Loren CE, Grabbe C, Varshney GK, Deleuil F, Hallberg B apoptosis in somatotrophs through a Pit-1/p53 pathway, preventing et al. (2003). Jeb signals through the Alk receptor tyrosine kinase to tumor growth. EMBO J 26: 2015–2028. drive visceral muscle fusion. Nature 425: 512–516. Casaccia-Bonnefil P, Carter BD, Dobrowsky RT, Chao MV. (1996). Ernfors P, Lee KF, Jaenisch R. (1994). Mice lacking brain-derived Death of oligodendrocytes mediated by the interaction of nerve neurotrophic factor develop with sensory deficits. Nature 368: growth factor with its receptor p75. Nature 383: 716–719. 147–150. Castets M, Coissieux MM, Delloye-Bourgeois C, Bernard L, Delcros Fazeli A, Dickinson SL, Hermiston ML, Tighe RV, Steen RG, Small JG, Bernet A et al. (2009). Inhibition of endothelial cell apoptosis by CG et al. (1997). Phenotype of mice lacking functional Deleted in netrin-1 during angiogenesis. Dev Cell 16: 614–620. colorectal cancer (Dcc) gene. Nature 386: 796–804. Chao MV, Bothwell MA, Ross AH, Koprowski H, Lanahan AA, Fearon ER, Cho KR, Nigro JM, Kern SE, Simons JW, Ruppert JM Buck CR et al. (1986). Gene transfer and molecular cloning of the et al. (1990). Identification of a chromosome 18q gene that is altered human NGF receptor. Science 232: 518–521. in colorectal cancers. Science 247: 49–56. Charrier JB, Lapointe F, Le Douarin NM, Teillet MA. (2001). Anti- Fitamant J, Guenebeaud C, Coissieux MM, Guix C, Treilleux I, apoptotic role of Sonic hedgehog protein at the early stages of Scoazec JY et al. (2008). Netrin-1 expression confers a selective nervous system organogenesis. Development 128: 4011–4020. advantage for tumor cell survival in metastatic breast cancer. Proc Charrier JB, Teillet MA, Lapointe F, Le Douarin NM. (1999). Natl Acad Sci USA 105: 4850–4855. Defining subregions of Hensen’s node essential for caudalward Forcet C, Ye X, Granger L, Corset V, Shin H, Bredesen DE et al. movement, midline development and cell survival. Development 126: (2001). The dependence receptor DCC (deleted in colorectal cancer) 4771–4783. defines an alternative mechanism for caspase activation. Proc Natl Chen YQ, Hsieh JT, Yao F, Fang B, Pong RC, Cipriano SC et al. Acad Sci USA 98: 3416–3421. (1999). Induction of apoptosis and G2/M cell cycle arrest by DCC. Foveau B, Leroy C, Ancot F, Deheuninck J, Ji Z, Fafeur V et al. Oncogene 18: 2747–2754. (2007). Amplification of apoptosis through sequential caspase Cho KR, Oliner JD, Simons JW, Hedrick L, Fearon ER, Preisinger cleavage of the MET tyrosine kinase receptor. Cell Death Differ AC et al. (1994). The DCC gene: structural analysis and mutations 14: 752–764. in colorectal carcinomas. Genomics 19: 525–531. Fox BP, Kandpal RP. (2004). Invasiveness of breast carcinoma cells Clark PE, Irvine RA, Coetzee GA. (2003). The androgen receptor and transcript profile: Eph receptors and ephrin ligands as CAG repeat and prostate cancer risk. Methods Mol Med 81: molecular markers of potential diagnostic and prognostic applica- 255–266. tion. Biochem Biophys Res Commun 318: 882–892. Cohen LY, Bourbonniere M, Sabbagh L, Bouchard A, Chew T, Frade JM, Rodriguez-Tebar A, Barde YA. (1996). Induction of cell Jeannequin P et al. (2005). Notch1 antiapoptotic activity is death by endogenous nerve growth factor through its p75 receptor. abrogated by caspase cleavage in dying T lymphocytes. Cell Death Nature 383: 166–168. Differ 12: 243–254. Fujita Y, Taniguchi J, Uchikawa M, Endo M, Hata K, Kubo T et al. Coulson EJ, Reid K, Baca M, Shipham KA, Hulett SM, Kilpatrick TJ (2008). Neogenin regulates neuronal survival through DAP kinase. et al. (2000). Chopper, a new death domain of the p75 neurotrophin Cell Death Differ 15: 1593–1608. receptor that mediates rapid neuronal cell death. J Biol Chem 275: Furne C, Corset V, Herincs Z, Cahuzac N, Hueber AO, Mehlen P. 30537–30545. (2006). The dependence receptor DCC requires lipid raft Craft N, Sawyers CL. (1998). Mechanistic concepts in androgen- localization for cell death signaling. Proc Natl Acad Sci USA 103: dependence of prostate cancer. Cancer Metastasis Rev 17: 421–427. 4128–4133. Crowley C, Spencer SD, Nishimura MC, Chen KS, Pitts-Meek S, Furne C, Rama N, Corset V, Chedotal A, Mehlen P. (2008). Netrin-1 Armanini MP et al. (1994). Mice lacking nerve growth factor display is a survival factor during commissural neuron navigation. Proc perinatal loss of sensory and sympathetic neurons yet develop basal Natl Acad Sci USA 105: 14465–14470. forebrain cholinergic neurons. Cell 76: 1001–1011. Furne C, Ricard J, Cabrera JR, Pays L, Bethea JR, Mehlen P et al. Della-Bianca V, Rossi F, Armato U, Dal-Pra I, Costantini C, Perini G (2009). EphrinB3 is an anti-apoptotic ligand that inhibits the et al. (2001). Neurotrophin p75 receptor is involved in neuronal dependence receptor functions of EphA4 receptors during adult damage by prion peptide-(106-126). J Biol Chem 276: 38929–38933. neurogenesis. Biochim Biophys Acta 1793: 231–238. Delloye-Bourgeois C, Brambilla E, Coissieux MM, Guenebeaud C, Geisbrecht BV, Dowd KA, Barfield RW, Longo PA, Leahy DJ. Pedeux R, Firlej V et al. (2009a). Interference with netrin-1 and (2003). Netrin binds discrete subdomains of DCC and UNC5 and tumor cell death in non-small cell lung cancer. J Natl Cancer Inst mediates interactions between DCC and heparin. J Biol Chem 278: 101: 237–247. 32561–32568.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1879 Genini M, Schwalbe P, Scholl FA, Remppis A, Mattei MG, Schafer Kim DH, Nelson HH, Wiencke JK, Christiani DC, Wain JC, Mark EJ BW. (1997). Subtractive cloning and characterization of DRAL, a et al. (2001). Promoter methylation of DAP-kinase: association novel LIM-domain protein down-regulated in rhabdomyosarcoma. with advanced stage in non-small cell lung cancer. Oncogene 20: DNA Cell Biol 16: 433–442. 1765–1770. Gozuacik D, Kimchi A. (2006). DAPk protein family and cancer. Kissil JL, Feinstein E, Cohen O, Jones PA, Tsai YC, Knowles MA Autophagy 2: 74–79. et al. (1997). DAP-kinase loss of expression in various carcinoma Grady WM. (2007). Making the case for DCC and UNC5C as tumor- and B-cell lymphoma cell lines: possible implications for role as suppressor genes in the colon. Gastroenterology 133: 2045–2049. tumor suppressor gene. Oncogene 15: 403–407. Hafner C, Schmitz G, Meyer S, Bataille F, Hau P, Langmann T et al. Klingelhutz AJ, Smith PP, Garrett LR, McDougall JK. (1993). (2004). Differential of Eph receptors and ephrins in Alteration of the DCC tumor-suppressor gene in tumorigenic benign human tissues and cancers. Clin Chem 50: 490–499. HPV-18 immortalized human keratinocytes transformed by nitro- He YY, Huang JL, Chignell CF. (2006). Cleavage of epidermal growth somethylurea. Oncogene 8: 95–99. factor receptor by caspase during apoptosis is independent of its Koike C, Mizutani T, Ito T, Shimizu Y, Yamamichi N, Kameda T internalization. Oncogene 25: 1521–1531. et al. (2002). Introduction of wild-type patched gene suppresses the He YY, Huang JL, Gentry JB, Chignell CF. (2003). Epidermal growth oncogenic potential of human squamous cell carcinoma cell lines factor receptor down-regulation induced by UVA in human including A431. Oncogene 21: 2670–2678. keratinocytes does not require the receptor kinase activity. J Biol Kong WJ, Zhang S, Guo C, Zhang S, Wang Y, Zhang D. (2005). Chem 278: 42457–42465. Methylation-associated silencing of death-associated protein Hedgecock EM, Culotti JG, Hall DH. (1990). The unc-5, unc-6, and kinase gene in laryngeal squamous cell cancer. Laryngoscope 115: unc-40 genes guide circumferential migrations of pioneer axons and 1395–1401. mesodermal cells on the epidermis in C elegans. Neuron 4: 61–85. Koo EH. (2002). The beta-amyloid precursor protein (APP) and Hempstead BL. (2002). The many faces of p75NTR. Curr Opin Alzheimer’s disease: does the tail wag the dog? Traffic 3: 763–770. Neurobiol 12: 260–267. Kotzbauer PT, Lampe PA, Heuckeroth RO, Golden JP, Creedon DJ, Hempstead BL, Martin-Zanca D, Kaplan DR, Parada LF, Chao MV. Johnson Jr EM et al. (1996). Neurturin, a relative of glial-cell-line- (1991). High-affinity NGF binding requires coexpression of the trk derived neurotrophic factor. Nature 384: 467–470. proto-oncogene and the low-affinity NGF receptor. Nature 350: Lee DK, Chang C. (2003). Molecular communication between 678–683. androgen receptor and general transcription machinery. J Steroid Hofmann K, Tschopp J. (1995). The death domain motif found in Fas Biochem Mol Biol 84: 41–49. (Apo-1) and TNF receptor is present in proteins involved in Lee FS, Kim AH, Khursigara G, Chao MV. (2001a). The uniqueness apoptosis and axonal guidance. FEBS Lett 371: 321–323. of being a neurotrophin receptor. Curr Opin Neurobiol 11: 281–286. Hong K, Hinck L, Nishiyama M, Poo MM, Tessier-Lavigne M, Stein Lee HH, Norris A, Weiss JB, Frasch M. (2003). Jelly belly protein E. (1999). A ligand-gated association between cytoplasmic domains activates the receptor tyrosine kinase Alk to specify visceral muscle of UNC5 and DCC family receptors converts netrin-induced growth pioneers. Nature 425: 507–512. cone attraction to repulsion. Cell 97: 927–941. Lee R, Kermani P, Teng KK, Hempstead BL. (2001b). Regulation of Hood JD, Cheresh DA. (2002). Role of integrins in cell invasion and cell survival by secreted proneurotrophins. Science 294: 1945–1948. migration. Nat Rev Cancer 2: 91–100. Lejmi E, Leconte L, Pedron-Mazoyer S, Ropert S, Raoul W, Lavalette Huang EJ, Reichardt LF. (2001). Neurotrophins: roles in neuronal S et al. (2008). Netrin-4 inhibits angiogenesis via binding to development and function. Annu Rev Neurosci 24: 677–736. neogenin and recruitment of Unc5B. Proc Natl Acad Sci USA Hueber AO. (2003). Role of membrane microdomain rafts in TNFR- 105: 12491–12496. mediated . Cell Death Differ 10: 7–9. Leonardo ED, Hinck L, Masu M, Keino-Masu K, Ackerman SL, Inbal B, Cohen O, Polak-Charcon S, Kopolovic J, Vadai E, Eisenbach Tessier-Lavigne M. (1997). Vertebrate homologues of C. elegans L et al. (1997). DAP kinase links the control of apoptosis to UNC-5 are candidate netrin receptors. Nature 386: 833–838. metastasis. Nature 390: 180–184. Leung-Hagesteijn C, Spence AM, Stern BD, Zhou Y, Su MW, Iwahara T, Fujimoto J, Wen D, Cupples R, Bucay N, Arakawa T et al. Hedgecock EM et al. (1992). UNC-5, a transmembrane protein with (1997). Molecular characterization of ALK, a receptor tyrosine immunoglobulin and thrombospondin type 1 domains, guides cell kinase expressed specifically in the nervous system. Oncogene 14: and pioneer axon migrations in C elegans. Cell 71: 289–299. 439–449. Levi-Montalcini R, Angeletti PU. (1963). Essential role of the nerve Jessell TM. (2000). Neuronal specification in the spinal cord: inductive growth factor in the survival and maintenance of dissociated signals and transcriptional codes. Nat Rev Genet 1: 20–29. sensory and sympathetic embryonic nerve cells in vitro. Dev Biol 7: Jing S, Wen D, Yu Y, Holst PL, Luo Y, Fang M et al. (1996). GDNF- 653–659. induced activation of the ret protein tyrosine kinase is mediated by Liepinsh E, Ilag LL, Otting G, Ibanez CF. (1997). NMR structure of GDNFR-alpha, a novel receptor for GDNF. Cell 85: 1113–1124. the death domain of the p75 neurotrophin receptor. EMBO J 16: Johannessen M, Moller S, Hansen T, Moens U, Van Ghelue M. 4999–5005. (2006). The multifunctional roles of the four-and-a-half-LIM only Link BC, Reichelt U, Schreiber M, Kaifi JT, Wachowiak R, Bogoevski protein FHL2. Cell Mol Life Sci 63: 268–284. D et al. (2007). Prognostic implications of netrin-1 expression and Kang JS, Yi MJ, Zhang W, Feinleib JL, Cole F, Krauss RS. (2004). its receptors in patients with adenocarcinoma of the pancreas. Ann Netrins and neogenin promote myotube formation. J Cell Biol 167: Surg Oncol 14: 2591–2599. 493–504. Liu J, Yao F, Wu R, Morgan M, Thorburn A, Finley Jr RL et al. Kaplan DR, Martin-Zanca D, Parada LF. (1991). Tyrosine phosphor- (2002). Mediation of the DCC apoptotic signal by DIP13 alpha. J ylation and tyrosine kinase activity of the trk proto-oncogene Biol Chem 277: 26281–26285. product induced by NGF. Nature 350: 158–160. Llambi F, Causeret F, Bloch-Gallego E, Mehlen P. (2001). Netrin-1 Kaplan DR, Miller FD. (2000). Neurotrophin signal transduction in acts as a survival factor via its receptors UNC5H and DCC. EMBO the nervous system. Curr Opin Neurobiol 10: 381–391. J 20: 2715–2722. Kato H, Zhou Y, Asanoma K, Kondo H, Yoshikawa Y, Watanabe K Llambi F, Lourenco FC, Gozuacik D, Guix C, Pays L, Del Rio G et al. (2000). Suppressed tumorigenicity of human endometrial et al. (2005). The dependence receptor UNC5H2 mediates apoptosis cancer cells by the restored expression of the DCC gene. Br J Cancer through DAP-kinase. EMBO J 24: 1192–1201. 82: 459–466. Loren CE, Englund C, Grabbe C, Hallberg B, Hunter T, Palmer RH. Keino-Masu K, Masu M, Hinck L, Leonardo ED, Chan SS, Culotti (2003). A crucial role for the anaplastic lymphoma kinase receptor JG et al. (1996). Deleted in Colorectal Cancer (DCC) encodes a tyrosine kinase in gut development in Drosophila melanogaster. netrin receptor. Cell 87: 175–185. EMBO Rep 4: 781–786.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1880 Lorenzo MJ, Gish GD, Houghton C, Stonehouse TJ, Pawson T, Naumann T, Casademunt E, Hollerbach E, Hofmann J, Dechant G, Ponder BA et al. (1997). Ret alternate splicing influences the Frotscher M et al. (2002). Complete deletion of the neurotrophin interaction of activated RET with the SH2 and PTB domains of receptor p75NTR leads to long-lasting increases in the number of Shc, and the SH2 domain of Grb2. Oncogene 14: 763–771. basal forebrain cholinergic neurons. J Neurosci 22: 2409–2418. Lourenco FC, Galvan V, Fombonne J, Corset V, Llambi F, Muller U Nelson KA, Witte JS. (2002). Androgen receptor CAG repeats and et al. (2009). Netrin-1 interacts with amyloid precursor protein and prostate cancer. Am J Epidemiol 155: 883–890. regulates amyloid-beta production. Cell Death Differ 16: 655–663. Nguyen TV, Galvan V, Huang W, Banwait S, Tang H, Zhang J et al. Lu X, Le Noble F, Yuan L, Jiang Q, De Lafarge B, Sugiyama D et al. (2008). Signal transduction in Alzheimer disease: p21-activated (2004). The netrin receptor UNC5B mediates guidance events con- kinase signaling requires C-terminal cleavage of APP at Asp664. J trolling morphogenesis of the vascular system. Nature 432: 179–186. Neurochem 104: 1065–1080. Lykissas MG, Batistatou AK, Charalabopoulos KA, Beris AE. (2007). Niederkofler V, Salie R, Sigrist M, Arber S. (2004). Repulsive guidance The role of neurotrophins in axonal growth, guidance, and molecule (RGM) gene function is required for neural tube closure regeneration. Curr Neurovasc Res 4: 143–151. but not retinal topography in the mouse visual system. J Neurosci Maisse C, Rossin A, Cahuzac N, Paradisi A, Klein C, Haillot ML 24: 808–818. et al. (2008). Lipid raft localization and palmitoylation: identifica- Paradisi A, Maisse C, Bernet A, Coissieux MM, Maccarrone M, tion of two requirements for cell death induction by the tumor Scoazec JY et al. (2008). NF-kappaB regulates netrin-1 expression suppressors UNC5H. Exp Cell Res 314: 2544–2552. and affects the conditional tumor suppressive activity of the netrin-1 Manie S, Santoro M, Fusco A, Billaud M. (2001). The RET receptor: receptors. Gastroenterology 135: 1248–1257. function in development and dysfunction in congenital malforma- Paradisi A, Maisse C, Coissieux MM, Gadot N, Lepinasse F, Delloye- tion. Trends Genet 17: 580–589. Bourgeois C et al. (2009). Netrin-1 up-regulation in inflammatory Matsunaga E, Tauszig-Delamasure S, Monnier PP, Mueller BK, bowel diseases is required for colorectal cancer progression. Proc Strittmatter SM, Mehlen P et al. (2004). RGM and its receptor Natl Acad Sci USA 106: 17146–17151. neogenin regulate neuronal survival. Nat Cell Biol 6: 749–755. Pasquale EB. (2005). Eph receptor signalling casts a wide net on cell Mazelin L, Bernet A, Bonod-Bidaud C, Pays L, Arnaud S, Gespach C behaviour. Nat Rev Mol Cell Biol 6: 462–475. et al. (2004). Netrin-1 controls colorectal tumorigenesis by regulat- Peterziel H, Paech T, Strelau J, Unsicker K, Krieglstein K. (2007). ing apoptosis. Nature 431: 80–84. Specificity in the crosstalk of TGFbeta/GDNF family members is Mehlen P, Fearon ER. (2004). Role of the dependence receptor DCC determined by distinct GFR alpha receptors. J Neurochem 103: in colorectal cancer pathogenesis. J Clin Oncol 22: 3420–3428. 2491–2504. Mehlen P, Furne C. (2005). Netrin-1: when a neuronal guidance cue Pflug B, Djakiew D. (1998). Expression of p75NTR in a human turns out to be a regulator of tumorigenesis. Cell Mol Life Sci 62: prostate epithelial tumor cell line reduces nerve growth factor- 2599–2616. induced cell growth by activation of programmed cell death. Mol Mehlen P, Mazelin L. (2003). The dependence receptors DCC and Carcinog 23: 106–114. UNC5H as a link between neuronal guidance and survival. Biol Cell PuschelPuschel AW. (1999). Divergent properties of mouse netrins. 95: 425–436. Mech Dev 83: 65–75. Mehlen P, Rabizadeh S, Snipas SJ, Assa-Munt N, Salvesen GS, Qin S, Yu L, Gao Y, Zhou R, Zhang C. (2007). Characterization of the Bredesen DE. (1998). The DCC gene product induces apoptosis by a receptors for axon guidance factor netrin-4 and identification of the mechanism requiring receptor proteolysis. Nature 395: 801–804. binding domains. Mol Cell Neurosci 34: 243–250. Mehlen P, Thibert C. (2004). Dependence receptors: between life and Rabizadeh S, Oh J, Zhong LT, Yang J, Bitler CM, Butcher LL et al. death. Cell Mol Life Sci 61: 1854–1866. (1993). Induction of apoptosis by the low-affinity NGF receptor. Meyerhardt JA, Look AT, Bigner SH, Fearon ER. (1997). Identifica- Science 261: 345–348. tion and characterization of neogenin, a DCC-related gene. Rabizadeh S, Ye X, Sperandio S, Wang JJ, Ellerby HM, Ellerby LM Oncogene 14: 1129–1136. et al. (2000). Neurotrophin dependence domain: a domain required Mille F, Llambi F, Guix C, Delloye-Bourgeois C, Guenebeaud C, for the mediation of apoptosis by the p75 neurotrophin receptor. Castro-Obregon S et al. (2009a). Interfering with multimerization of J Mol Neurosci 15: 215–229. netrin-1 receptors triggers tumor cell death. Cell Death Differ 16: Rajagopalan S, Deitinghoff L, Davis D, Conrad S, Skutella T, 1344–1351. Chedotal A et al. (2004). Neogenin mediates the action of repulsive Mille F, Thibert C, Fombonne J, Rama N, Guix C, Hayashi H et al. guidance molecule. Nat Cell Biol 6: 756–762. (2009b). The Patched dependence receptor triggers apoptosis Rajasekharan S, Kennedy TE. (2009). The netrin protein family. through a DRAL-caspase-9 complex. Nat Cell Biol 11: 739–746. Genome Biol 10: 239. Minichiello L, Piehl F, Vazquez E, Schimmang T, Hokfelt T, Represa Raveh T, Kimchi A. (2001). DAP kinase-a proapoptotic gene that J et al. (1995). Differential effects of combined functions as a tumor suppressor. Exp Cell Res 264: 185–192. mutations on dorsal root ganglion and inner ear sensory neurons. Ricard J, Salinas J, Garcia L, Liebl DJ. (2006). EphrinB3 regulates cell Development 121: 4067–4075. proliferation and survival in adult neurogenesis. Mol Cell Neurosci Monnier PP, Sierra A, Macchi P, Deitinghoff L, Andersen JS, Mann 31: 713–722. M et al. (2002). RGM is a repulsive guidance molecule for retinal Rodrigues S, De Wever O, Bruyneel E, Rooney RJ, Gespach C. (2007). axons. Nature 419: 392–395. Opposing roles of netrin-1 and the dependence receptor DCC in Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, cancer cell invasion, tumor growth and metastasis. Oncogene 26: Saltman DL et al. (1994). Fusion of a kinase gene, ALK, to a 5615–5625. nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science Romeo G, Ronchetto P, Luo Y, Barone V, Seri M, Ceccherini I et al. 263: 1281–1284. (1994). Point mutations affecting the tyrosine kinase domain of the Mourali J, Benard A, Lourenco FC, Monnet C, Greenland C, Moog- RET proto-oncogene in Hirschsprung’s disease. Nature 367: Lutz C et al. (2006). Anaplastic lymphoma kinase is a dependence 377–378. receptor whose proapoptotic functions are activated by caspase Saito M, Yamaguchi A, Goi T, Tsuchiyama T, Nakagawara G, Urano cleavage. Mol Cell Biol 26: 6209–6222. T et al. (1999). Expression of DCC protein in colorectal tumors and Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C, Gardner E its relationship to tumor progression and metastasis. Oncology 56: et al. (1993). Germ-line mutations of the RET proto-oncogene in 134–141. multiple endocrine neoplasia type 2A. Nature 363: 458–460. Sanchez-Cespedes M, Esteller M, Wu L, Nawroz-Danish H, Yoo GH, Murone M, Rosenthal A, de Sauvage FJ. (1999). Sonic hedgehog Koch WM et al. (2000). Gene promoter hypermethylation in tumors signaling by the patched-smoothened receptor complex. Curr Biol 9: and serum of head and neck cancer patients. Cancer Res 60: 76–84. 892–895.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1881 Santoro M, Carlomagno F, Romano A, Bottaro DP, Dathan NA, Sun XF, Rutten S, Zhang H, Nordenskjold B. (1999). Expression of Grieco M et al. (1995). Activation of RET as a dominant the deleted in colorectal cancer gene is related to prognosis in DNA transforming gene by germline mutations of MEN2A and MEN2B. diploid and low proliferative colorectal adenocarcinoma. J Clin Science 267: 381–383. Oncol 17: 1745–1750. Santoro M, Dathan NA, Berlingieri MT, Bongarzone I, Paulin C, Tang X, Jang SW, Okada M, Chan CB, Feng Y, Liu Y et al. (2008). Grieco M et al. (1994). Molecular characterization of RET/PTC3; a Netrin-1 mediates neuronal survival through PIKE-L interaction novel rearranged version of the RETproto-oncogene in a human with the dependence receptor UNC5B. Nat Cell Biol 10: 698–706. thyroid papillary carcinoma. Oncogene 9: 509–516. Tang X, Khuri FR, Lee JJ, Kemp BL, Liu D, Hong WK et al. (2000). Schmidtmer J, Engelkamp D. (2004). Isolation and expression pattern Hypermethylation of the death-associated protein (DAP) kinase of three mouse homologues of chick Rgm. Gene Expr Patterns 4: promoter and aggressiveness in stage I non-small-cell lung cancer. 105–110. J Natl Cancer Inst 92: 1511–1516. Scholl FA, McLoughlin P, Ehler E, de Giovanni C, Schafer BW. Tanikawa C, Matsuda K, Fukuda S, Nakamura Y, Arakawa H. (2000). DRAL is a p53-responsive gene whose four and a half LIM (2003). p53RDL1 regulates p53-dependent apoptosis. Nat Cell Biol domain protein product induces apoptosis. J Cell Biol 151: 495–506. 5: 216–223. Scott RP, Ibanez CF. (2001). Determinants of ligand binding Tauszig-Delamasure S, Yu LY, Cabrera JR, Bouzas-Rodriguez J, specificity in the glial cell line-derived neurotrophic factor family Mermet-Bouvier C, Guix C et al. (2007). The TrkC receptor induces receptor alpha S. J Biol Chem 276: 1450–1458. apoptosis when the dependence receptor notion meets the neuro- Serafini T, Colamarino SA, Leonardo ED, Wang H, Beddington R, trophin paradigm. Proc Natl Acad Sci USA 104: 13361–13366. Skarnes WC et al. (1996). Netrin-1 is required for commissural Tessarollo L, Tsoulfas P, Donovan MJ, Palko ME, Blair-Flynn J, axon guidance in the developing vertebrate nervous system. Cell 87: Hempstead BL et al. (1997). Targeted deletion of all isoforms of the 1001–1014. trkC gene suggests the use of alternate receptors by its ligand Serafini T, Kennedy TE, Galko MJ, Mirzayan C, Jessell TM, Tessier- neurotrophin-3 in neuronal development and implicates trkC in Lavigne M. (1994). The netrins define a family of axon outgrowth- normal cardiogenesis. Proc Natl Acad Sci USA 94: 14776–14781. promoting proteins homologous to C. elegans UNC-. Cell 78: 409–424. Tessarollo L, Vogel KS, Palko ME, Reid SW, Parada LF. (1994). Shibata D, Reale MA, Lavin P, Silverman M, Fearon ER, Steele Jr G Targeted mutation in the neurotrophin-3 gene results in loss of et al. (1996). The DCC protein and prognosis in colorectal cancer. N muscle sensory neurons. Proc Natl Acad Sci USA 91: 11844–11848. Engl J Med 335: 1727–1732. Thibert C, Teillet MA, Lapointe F, Mazelin L, Le Douarin NM, Shin SK, Nagasaka T, Jung BH, Matsubara N, Kim WH, Carethers Mehlen P. (2003). Inhibition of neuroepithelial patched-induced JM et al. (2007). Epigenetic and genetic alterations in Netrin-1 apoptosis by sonic hedgehog. Science 301: 843–846. receptors UNC5C and DCC in human colon cancer. Gastroenter- Thiebault K, Mazelin L, Pays L, Llambi F, Joly MO, Scoazec JY et al. ology 133: 1849–1857. (2003). The netrin-1 receptors UNC5H are putative tumor Shohat G, Spivak-Kroizman T, Cohen O, Bialik S, Shani G, Berrisi H suppressors controlling cell death commitment. Proc Natl Acad et al. (2001). The pro-apoptotic function of death-associated protein Sci USA 100: 4173–4178. kinase is controlled by a unique inhibitory autophosphorylation- Thornberry NA, Lazebnik Y. (1998). Caspases: enemies within. based mechanism. J Biol Chem 276: 47460–47467. Science 281: 1312–1316. Sieber OM, Tomlinson IP, Lamlum H. (2000). The adenomatous Tikhomirov O, Dikov M, Carpenter G. (2005). Identification of polyposis coli (APC) tumour suppressor—genetics, function and proteolytic fragments from ErbB-2 that induce apoptosis. Oncogene disease. Mol Med Today 6: 462–469. 24: 3906–3913. Simons K, Toomre D. (2000). Lipid rafts and signal transduction. Nat Tulasne D, Deheuninck J, Lourenco FC, Lamballe F, Ji Z, Leroy C Rev Mol Cell Biol 1: 31–39. et al. (2004). Proapoptotic function of the MET tyrosine kinase Songyang Z, Carraway IIIrd KL, Eck MJ, Harrison SC, Feldman RA, receptor through caspase cleavage. Mol Cell Biol 24: 10328–10339. Mohammadi M et al. (1995). Catalytic specificity of protein-tyrosine Velcich A, Corner G, Palumbo L, Augenlicht L. (1999). Altered is critical for selective signaling. Nature 373: 536–539. phenotype of HT29 colonic adenocarcinoma cells following expres- Srinivasan K, Strickland P, Valdes A, Shin GC, Hinck L. (2003). sion of the DCC gene. Oncogene 18: 2599–2606. Netrin-1/neogenin interaction stabilizes multipotent progenitor cap Vielmetter J, Kayyem JF, Roman JM, Dreyer WJ. (1994). Neogenin, cells during mammary gland morphogenesis. Dev Cell 4: 371–382. an avian cell surface protein expressed during terminal neuronal Stein E, Zou Y, Poo M, Tessier-Lavigne M. (2001). Binding of DCC differentiation, is closely related to the human tumor suppressor by netrin-1 to mediate axon guidance independent of adenosine molecule deleted in colorectal cancer. J Cell Biol 127: 2009–2020. A2B receptor activation. Science 291: 1976–1982. Wang H, Ozaki T, Shamim Hossain M, Nakamura Y, Kamijo T, Xue Stilo R, Leonardi A, Formisano L, Di Jeso B, Vito P, Liguoro D. X et al. (2008). A newly identified dependence receptor UNC5H4 is (2002). TUCAN/CARDINAL and DRAL participate in a common induced during DNA damage-mediated apoptosis and transcrip- pathway for modulation of NF-kappaB activation. FEBS Lett 521: tional target of tumor suppressor p53. Biochem Biophys Res 165–169. Commun 370: 594–598. Stone DM, Hynes M, Armanini M, Swanson TA, Gu Q, Johnson RL Wang JJ, Rabizadeh S, Tasinato A, Sperandio S, Ye X, Green M et al. et al. (1996). The tumour-suppressor gene patched encodes a (2000). Dimerization-dependent block of the proapoptotic effect of candidate receptor for Sonic hedgehog. Nature 384: 129–134. p75(NTR). J Neurosci Res 60: 587–593. Strohecker AM, Yehiely F, Chen F, Cryns VL. (2008). Caspase Wang R, Wei Z, Jin H, Wu H, Yu C, Wen W et al. (2009). cleavage of HER-2 releases a Bad-like cell death effector. J Biol Autoinhibition of UNC5b revealed by the cytoplasmic domain Chem 283: 18269–18282. structure of the receptor. Mol Cell 33: 692–703. Stupack DG. (2005). Integrins as a distinct subtype of dependence Wicking C, McGlinn E. (2001). The role of hedgehog signalling in receptors. Cell Death Differ 12: 1021–1030. tumorigenesis. Cancer Lett 173: 1–7. Stupack DG, Cheresh DA. (2002). ECM remodeling regulates Williams ME, Strickland P, Watanabe K, Hinck L. (2003). UNC5H1 angiogenesis: endothelial integrins look for new ligands. Sci STKE induces apoptosis via its juxtamembrane region through an 2002: PE7. interaction with NRAGE. J Biol Chem 278: 17483–17490. Stupack DG, Puente XS, Boutsaboualoy S, Storgard CM, Cheresh Wilson BD, Ii M, Park KW, Suli A, Sorensen LK, Larrieu-Lahargue F DA. (2001). Apoptosis of adherent cells by recruitment of caspase-8 et al. (2006). Netrins promote developmental and therapeutic to unligated integrins. J Cell Biol 155: 459–470. angiogenesis. Science 313: 640–644. Stupack DG, Teitz T, Potter MD, Mikolon D, Houghton PJ, Kidd VJ Wilson NH, Key B. (2006). Neogenin interacts with RGMa and et al. (2006). Potentiation of neuroblastoma metastasis by loss of netrin-1 to guide axons within the embryonic vertebrate forebrain. caspase-8. Nature 439: 95–99. Dev Biol 296: 485–498.

Oncogene A new paradigm in cell signaling and cancer therapy D Goldschneider and P Mehlen 1882 Yaar M, Zhai S, Fine RE, Eisenhauer PB, Arble BL, Stewart KB et al. Trk-C in favourable human neuroblastomas. Eur J Cancer 33: (2002). binds trimers as well as monomers of the 75- 2054–2057. kDa neurotrophin receptor and activates receptor signaling. J Biol Yeo TT, Chua-Couzens J, Butcher LL, Bredesen DE, Cooper JD, Chem 277: 7720–7725. Valletta JS et al. (1997). Absence of p75NTR causes increased basal Yamashiro DJ, Liu XG, Lee CP, Nakagawara A, Ikegaki N, forebrain cholinergic neuron size, choline acetyltransferase activity, McGregor LM et al. (1997). Expression and function of and target innervation. J Neurosci 17: 7594–7605.

Oncogene