<<

Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Structure and Physiology of the RET

Carlos F. Iba´n˜ ez

Department of Neuroscience, Karolinska Institute, S-17177 Stockholm, Sweden; Life Sciences Institute, Department of Physiology, National University of Singapore 117456, Singapore Correspondence: [email protected]

The identification of the ret by Masahide Takahashi and Geoffrey Cooper in 1985 was both serendipitous and paradigmatic (Takahashi et al. 1985). By transfecting total DNA from a human lymphoma into mouse NIH3T3 cells, they obtained one clone, which in secondary transformants yielded more than 100-fold improvement in transformation effi- ciency. Subsequent investigations revealed that the ret oncogene was not present as such in the primary lymphoma, but was derived by DNA rearrangement during transfection from normal human sequences of the ret locus. At the time, activation by DNA rearrangement had not been previously described for a transforming with the NIH3T3 transfection assay. The discovery of ret opened a field of study that has had a profound impact in research, developmental biology, and neuroscience, and that continues to yield surprises and impor- tant insights to this day.

AN UNUSUAL RECEPTOR 2000). A molecular modeling study of the ex- WITH CADHERIN REPEATS tracellular domain of RET later revealed four cadherin repeats—termed cadherin-like do- solation of ret cDNA clones revealed a car- mains or CLDs 1–4—each of about 110 resi- Iboxy-terminal domain with high homology dues, and one Ca2þ- between CLD2 with members of the tyrosine kinase gene fam- and CLD3 (Anders et al. 2001). Ca2þ binding is ily preceded by a hydrophobic sequence charac- required for the functional integrity of the RET teristic of a transmembrane domain, suggesting and for its ability to interact with . that the ret oncogene encoded a cell-surface re- Following the four CLDs, the extracellular do- ceptor (Takahashi and Cooper 1987). The char- main of RET contains a Cys-rich region of 120 acterization of the human (Takahashi et al. residues connected to the transmembrane do- 1988) and mouse (Iwamoto et al. 1993; Pachnis main. The intracellular region of RET begins et al. 1993) ret proto- revealed the full with a juxtamembrane portion of 50 residues, primary structure of the RET protein and the a tyrosine kinase domain split by a 14-residue unusual presence in its extracellular region of a linker, and a 100-residue-long carboxy-termi- sequence with similarity to cadherins, trans- nal tail, which comes in two flavors as a result membrane that mediate Ca2þ-depen- of . After position 1063, dent homophilic cell adhesion (Nollet et al. the “short” RET isoform contains nine unique

Editors: and Mark A. Lemmon Additional Perspectives on Receptor Tyrosine available at www.cshperspectives.org Copyright # 2013 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a009134 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134

1 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

C.F. Iba´n˜ ez

carboxy-terminal residues (RET9), whereas the highly related in sequence to GDNF, known re- “long” contains 51 (RET51). spectively as (Kotzbauer et al. 1996), (Milbrandt et al. 1998), and (Baloh et al. 1998b), were subsequently also ONE GENE, MANY DISEASES identified as ligands of RET. However, this li- Mutations in the RET gene have been found in gand–receptor relationship proved to be a little a number of human diseases, including sever- more unusual than initially expected. Neither al different of neuroendocrine origin ligand is able to bind RETon its own, but require and a gut syndrome characterized by intestinal a ligand-binding subunit acting as coreceptor, obstruction known as Hirschsprung’s disease. known as the GDNF family receptor a (GFRa) Four different human cancers carry mutations component. Four different GFRas have been in the RET gene, including papillary thyroid described (GFRa1–4), each with selectivity— carcinoma (PTC) (Grieco et al. 1990), medul- although not absolute specificity—for each of lary thyroid carcinoma (familial and sporadic) the four distinct members of the GDNF ligand (Donis-Keller et al. 1993; Hofstra et al. 1994), family (Jing et al. 1996; Baloh et al. 1997, 1998a; and the multiple endocrine neoplasias type 2A Buj-Bello et al. 1997; Klein et al. 1997; Sanicola (MEN2A) (Donis-Keller et al. 1993; Mulligan et al. 1997; Naveilhan et al. 1998; Trupp et al. et al. 1993) and 2B (MEN2B) (Hofstra et al. 1998; Worby et al. 1998; Masure et al. 2000). 1994). Dozens of different substitutions and re- GDNF can be chemically cross-linked to RET arrangements in the RET gene underlie these (Trupp et al. 1996), indicating that it does syndromes, a complexity that has profound im- make direct contact with the receptor, although plications for our understating of genotype/ its binding affinity is too low to stabilize a com- phenotype relationships and the molecular plex. On the other hand, GDNF has high affinity mechanisms of (for an in- for GFRa1 independently of RET. A model ini- depth review of the cancer biology of RET, see tially proposed had GDNF forming first a com- the article by Santoro and Carlomagno 2013). plex with GFRa1 and subsequently recruiting Although RETmutations that lead to tumor for- RET to the complex (Massague´ 1996). An alter- mation have in most cases been described as gain native model, in which GFRa1 and RETare pre- of function, mutations that result in Hirsch- associated to some extent before ligand binding, sprung’s disease—of which more than 50 are was later proposed based on binding and site- known so far—invariably result in loss of RET directed mutagenesis studies (Eketja¨ll et al. function by targeting its kinase activity (Iwa- 1999; Cik et al. 2000). It should be noted that shita et al. 1996; Pelet et al. 1998), docking sites two additional receptors for GDNF have also for intracellular signaling effectors (Geneste been described, namely, the neural cell adhesion et al. 1999), or residues in the RETextracellular molecule NCAM (Paratcha et al. 2003) and syn- domain that affect RET processing in the endo- decan-3 (Bespalov et al. 2011), which are able to plasmic reticulum and prevent RET expression transmit GDNF signals independently of RET. at the cell surface (Iwashita et al. 1996; Cosma et al. 1998; Kjaer and Ibanez 2003b). EVOLUTIONARY RELATIONSHIPS Only one ret gene is known to exist in higher A WEALTH OF RET LIGANDS AND organisms. RET, GDNF family ligands, and CORECEPTORS GFRa proteins have been found in all vertebrate The RET ligand has been identified as glial cell species investigated so far. A ret orthologue has line-derived neurotrophic factor (GDNF) (Dur- also been found in the genome of the cephalo- bec et al. 1996; Trupp et al. 1996), a dimeric chordate Amphioxus, along with sequences cor- -like protein distantly related to responding to one GDNF-like and one GFRa- members of the transforming growth factor-b like encoded protein product. RET is also found (TGF-b) superfamily. Three additional proteins in Drosophila melanogaster. Intriguingly, its ex-

2 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Structure and Physiology of the RET RTK

pression pattern in the fly is to some extent sic susceptibility to misfolding of mammalian reminiscent of the one found in vertebrates RETECD may be the result of a trade-off that (Hahn and Bishop 2001). However, no GDNF helps to avoid an increased incidence of tu- or GFRa proteins appear to be encoded in the fly mors, at the expense of a greater vulnerability genome. Drosophila RET is unable to interact to Hirschsprung’s disease. with GDNF or GFRa1 of mammalian origin, Sequence and functional divergences be- nor is it capable of mediating cell adhesion tween the ectodomains of mammalian and (Abrescia et al. 2005). A chimeric approach amphibian RET molecules have been exploited was used to show that Drosophila RET contains to map binding determinants in the human an active tyrosine kinase that is competent to RETECD responsible for its interaction with induce neuronal differentiation on activation the GDNF-GFRa1 complex through homolog- in PC12 cells (Abrescia et al. 2005). The physio- scanning mutagenesis. It was found that Xe- logical function of RET in Drosophila remains nopus RETECD was unable to bind to GDNF- unknown. GFRa1 complexes of mammalian origin. How- ever, a chimeric molecule containing CLD1, 2, and 3 from human RETECD, but neither do- STRUCTURE–FUNCTION STUDIES OF RET main alone, had similar binding activity than EXTRACELLULAR AND KINASE DOMAINS full-length human RETECD (Kjaer and Ibanez Loss-of-function mutations in RET cause ab- 2003a). This suggested the existence of an ex- normal development of the enteric nervous sys- tended ligand-binding surface within the three tem, leading to Hirschsprung’s disease. Hirsch- amino-terminal cadherin-like domains of hu- sprung mutations in the extracellular domain of man RETECD.Subsequently,astudy usingchem- RET (RETECD) affect processing in the endo- ical cross-linking of a reconstituted GDNF/ plasmic reticulum (ER) and prevent RETexpres- GFRa1/RET complex followed by matrix-assis- sion at the cell surface. Most Hirschsprung mu- ted laser desorption/ionization (MALDI) mass tations examined prevent the maturation of spectrometry analysis, indicated that CLD4 RETECD in the ER, indicating defects in protein and the carboxy-terminal cysteine-rich domain folding (Kjaerand Ibanez 2003b). Maturation of (CRD) of the RETECD are in direct contact with RETECD mutants can be rescued by allowing GFRa1 in complex with GDNF (Amoresano protein expression to proceed at 308C, a condi- et al. 2005). This study failed to identify any tion known to facilitate protein folding, regain- direct contacts between RET and GDNF, al- ing their ability to bind to the GDNF/GFRa1. though, as mentioned earlier, those were known Analysis of autonomous folding subunits in the to exist from previous cross-linking experiments RETECD has indicated an intrinsic propensity to (Trupp et al. 1996). These discrepant sets of re- misfolding in the amino-terminal CLDs 1–3 sults could be reconciled if the role of the amino- (Kjaer and Ibanez 2003b), which also concen- terminal CLD1–3 in ligand binding was indi- trate the majority of Hirschsprung mutations rect, rather than in establishing physical contact affecting the RETECD. A recent crystal structure with the GDNF/GFRa1 complex. In this sce- of the two amino-terminal CLD1–2 domains of nario, CLD1–3 would be necessary for RET to the RETECD has revealed these two CLDs folded adopt a conformation that is competent for onto each other in a compact clam-shell ar- binding and complex assembly but not directly rangement distinct from that of classical cadher- involved in contacting the RET ligands (Amor- ins (Kjaer et al. 2010). CLD1 structural elements esano et al. 2005). At the time of this writing, and disulfide composition are unique to mam- efforts to solve the three-dimensional structure mals, indicating an unexpected structural diver- of the tripartite GDNF/GFRa1/RET complex sity within higher and lower vertebrate RET are still ongoing, but preliminary results would CLD regions. The same study identified two un- seem to offer support for this latter model. paired cysteines that predispose human RET to The crystal structures of the nonphosphory- maturation impediments in the ER. The intrin- lated (inactive) and phosphorylated (active) RET

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 3 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

C.F. Iba´n˜ ez

kinase have been determined and shown to mology 2 (SH2) or phosphotyrosine-binding adopt the same active kinase conformation (PTB) domains. Previous studies have indicat- competent to bind ATPand (Knowles ed that at least 14 of the 18 tyrosine residues et al. 2006). Both structures show a preorgan- present in the intracellular region of RET can ized activation loop conformation that is inde- become phosphorylated (Liu et al. 1996; Kawa- pendent of status. In agree- moto et al. 2004; Knowles et al. 2006). Among ment with the structural data, kinetic those, Tyr900 and Tyr905 are present in the ki- data showed that autophosphorylation pro- nase activation loop and are known to contrib- duces only a modest increase in activity (Know- ute to full kinase activation (Knowles et al. les et al. 2006). Longer forms of the RET intra- 2006). Autophosphorylation of the key residue cellular domain containing the juxtamembrane Tyr 1062 is required for activation of Ras/MAP domain and carboxy-terminal tail showed sim- kinase and PI3 kinase/AKT (Besset et al. 2000; ilar kinetic behavior as the isolated kinase, indi- Hayashi et al. 2000; Segouffin-Cariou and Bil- cating the absence of a cis-inhibitory mech- laud 2000; Coulpier et al. 2002). This residue anism within the RET intracellular domain appears to be critical for RET function, and (Knowles et al. 2006). Unlike the situation of mice with a point mutation in Tyr1062 show a most other receptor tyrosine kinases, these re- severe loss-of-function phenotype (Jijiwa et al. sults suggest the existence of alternative inhibi- 2004; Wong et al. 2005; Jain et al. 2006a). Phos- tory mechanisms, possibly in trans, for the au- phorylation of Tyr1096, present only in the long toregulation of RET kinase activity. RET51 isoform, also contributes to these path- ways. On ligand stimulation, at least two distinct protein complexes assemble on phosphorylated RET SIGNALING MECHANISMS Tyr 1062 of RET via Shc, one leading to activa- Dimerization of receptor tyrosine kinases is tion of the Ras/MAP kinase pathway through known to be required, although most likely not recruitment of Grb2 and Sos, and another to the sufficient, for ligand-induced kinase transphos- PI3K/AKT pathway through recruitment of phorylation and activation. Several receptor adaptors Grb2 and Gab2 followed by p85PI3K tyrosine kinases are found as preformed homo- and the SHP2 tyrosine (Besset et dimers at the plasma membrane independently al. 2000). of ligand binding. Using atransmembrane (TM) The adaptor protein FRS2 can also bind to domain self-association assay, Kjaer et al. ob- phosphorylated Tyr1062 in the activated RET served strong self-association of the RET-TM receptor (Kurokawa et al. 2001; Melillo et al. in a biological membrane (Kjaer et al. 2006). 2001). FRS2 competes with Shc for binding to These investigators found that mutagenesis of Tyr 1062, and it has been shown that Shc but not specific residues in the RET-TM domain re- FRS2 is responsible for cell survival effects of duced receptor homodimerization and abol- RET in neuroblastoma cells (Lundgren et al. ished the transforming activity of MEN2A 2006). This differential signaling may be medi- RET, one of the strongest oncogenic variants of ated from different compartments in the plasma the RET protein, suggesting that self-association membrane, as RET has been shown to interact of RET TM domains contributes to the mecha- with FRS2 in lipid rafts, but with Shc outside nism of activation of RET (Kjaer et al. 2006). lipid rafts (Paratcha et al. 2001). A series of Like the majority of receptor tyrosine ki- adaptor molecules from the p62dok family nases studied, signaling pathways initiated by have also been shown to interact with the acti- the RET receptor include the Ras/MAP kinase, vated RETreceptor. Dok-1, -2, -4, -5, and -6 all PI3 kinase/AKT, and -g interact with phosphorylated Tyr1062 via their (PLCg)pathways.Onactivation, RETundergoes PTB domains (Grimm et al. 2001; Crowder autophosphorylation of intracellular tyrosine et al. 2004; Kurotsuchi et al. 2010) and are residues, which then serve as docking sites for thought to contribute to neuronal differentia- downstream signaling effectors carrying Src ho- tion (Grimm et al. 2001; Crowder et al. 2004).

4 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Structure and Physiology of the RET RTK

The Grb2/Gab2 complex can also assem- residue Tyr687 (Fukuda et al. 2002). In line with ble directly onto phosphorylated Tyr1096, offer- this, activation of PKA by forskolin was found to ing an alternative route to PI3K activation by impair the recruitment of SHP2 to RET and GDNF.Regarding the remaining autophosphor- negativelyaffected ligand-mediated neurite out- ylation sites, it has been found that phosphory- growth (Perrinjaquet et al. 2010). Moreover, lation of Tyr1015 leads to activation of PLCg mutation of Ser696 enhanced SHP2 binding to (Borrello et al. 1996), and phospho-Tyr981 binds the receptor and eliminated the effect of forsko- the Src cytoplasmic tyrosine kinase (Encinas lin on ligand-induced neurite outgrowth. To- et al. 2004). Recently, a yeast-two-hybrid screen gether, these findings established Tyr687 as a crit- led to the identification of the GTPase-activat- ical platform for integration of RET and PKA ing protein (GAP) for Rap1, Rap1GAP,as a nov- signals. el RET-binding protein (Jiao et al. 2011). Like Among the interactions not mediated by Src, Rap1GAP was also found to require phos- phosphorylation, the PDZ domain-containing phorylation of Tyr981 for RET binding and sup- Shank3 protein was found to interact with a pressed GDNF-induced activation of ERK and PDZ-binding motif present in the RET9 but neurite outgrowth. A recent study has estab- not in the RET51 isoform (Schuetz et al. lished a biochemical function and a physiolog- 2004). Shank3 was shown to mediate sustained ical role for the phosphorylation of Tyr687 in the MAP kinase and PI3 kinase signaling, and the juxtamembrane region of the RET intracellular formation of branched tubular structures in domain (Perrinjaquet et al. 2010). Using a phage three-dimensional cultures of epithelial cells. display strategy, Perrinjaquet et al. found that the phosphotyrosine phosphatase SHP2 binds RET FUNCTION IN KIDNEY DEVELOPMENT to phospho-Tyr687 on ligand-induced RET ac- tivation. SHP2 is recruited to activated RET in Knockout studies have shown that RET inacti- a cooperative fashion, such that both interaction vation results in renal agenesis or severe hypo- with Tyr687 and association with components of dysplasia, owing to failure of the ureteric bud to the Tyr1062 signaling complex are required for evaginate from the Wolffian duct and branch stable recruitment of SHP2 to the receptor. normally (Schuchardt et al. 1994, 1996). RET SHP2 recruitment was found to contribute to expression defines a population of ureteric the ability of RET to activate the PI3K/AKT bud tip cells that proliferate under GDNF stim- pathway and promote survival and neurite ulation from the metanephric mesenchyme. In outgrowth in primary neurons (Perrinjaquet the absence of RET, tip cells change fate and et al. 2010). instead contribute to the ureteric bud trunk In addition to tyrosine autophosphoryla- (Shakya et al. 2005). Studies in knock-in mice tion, RET has been found to undergo serine have provided evidence for differential and iso- phosphorylation at Ser696 by A form-specific roles of RET phospho-Tyr dock- (PKA) (Fukuda et al. 2002). Mutation of Ser696 ing sites in kidney development. One earlier affected the ability of RET to activate the small study initially reported that mice expressing GTPase Rac1 and stimulate formation of cell only RET9 were normal, whereas those express- lamellipodia (Fukuda et al. 2002). Homozygous ing only RET51 showed kidney hypodysplasia knock-in mice carrying this mutation lacked (de Graaff et al. 2001). In contrast, a later study neuronal elements of the enteric nervous system reported that mice monoisomorphic for either in the distal colon, resulting from a migration RET9 or RET51 were viable and showed normal defect of enteric cells (Asai et al. kidneys, indicating redundant roles of RET iso- 2006), indicating a physiological role for PKA- forms in kidney development (Jain et al. 2006a). dependent modification of RET function. In- As discussed elsewhere, a possible reason for this terestingly, the signaling deficits of the Ser696 discrepancy may lie in the use of chimeric RET mutant could be alleviated—at least in vi- mouse–human knock-in cDNAs in the first tro—by simultaneous mutation of the nearby study. It has also been reported that wild-type

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 5 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

C.F. Iba´n˜ ez

human RET51 and RET9 are both able to (Pachnis et al. 1993; Trupp et al. 1997; Garce`s promote branching morphogenesis to a simi- et al. 2000). GDNF has potent survival activities lar extent, but mutation of Tyr1062 abrogates in spinal motoneurons (Henderson et al. 1994) this activity only in RET9 and not in RET51, and is required for their in vivo survival during presumably because of redundancy through late embryogenesis (Oppenheim et al. 2000). At Tyr 1096 (Jain et al. 2006a). In contrast, mutation early stages of development, RET is required for of Tyr1015 produced clear defects in ureteric bud the topographic projection of hind limb-inner- outgrowth in the context of either isoform, pro- vating axons, functioning as an instructive viding evidence for the importance of PLCg guidance signal for motor axons (Kramer et al. signaling downstream from RET in renal de- 2006). In this case, RETwas shown to cooperate velopment. Despite the prominent role of RET with the EphA4 to en- signaling in kidney development, no human force the precision of this binary choice in mo- RETmutations have yet been uncovered in chil- tor axon guidance. More recent studies using dren suffering from renal tract malformations. conditional alleles have shown that the effect of RET on motoneuron survival during pro- grammed cell death is restricted to the early RET FUNCTIONS IN NERVOUS SYSTEM neonatal development of the subpopulation of DEVELOPMENT g-motoneurons that innervates muscle spindles Enteric Nervous System (Gould et al. 2008). RET signaling would thus appear to have multiple effects on motoneuron Hirschsprung’s disease is a of survival and connectivity. neural crest development characterized by the absence of enteric parasympathetic neurons in the lower regions of the gut. In agreement with a Ventral Midbrain Dopaminergic Neurons role in neural crest development, RET is ex- GDNF was identified on the basis of its survival- pressed in several neuronal subpopulations de- promoting effects on ventral midbrain dopami- rived from this structure, including cells in the nergic neurons, which are important in the enteric, sensory, and sympathetic nervous sys- pathogenesis of Parkinson’s disease (Lin et al. tems (Pachnis et al. 1993). Mice that are homo- 1993). RET is expressed at high levels in adult zygotes for a targeted mutation in the RET gene ventral midbrain dopaminergic neurons of the lack enteric neurons throughout the digestive substantia nigra, and exogenous application of tract (Schuchardt et al. 1994). A subpopulation GDNF was shown to protect RET-expressing of enteric neural crest was found to undergo neurons in this structure from cell death induced apoptotic cell death specifically in the foregut by 6-hydroxydopamine, an animal model of of embryos lacking the RET receptor (Taraviras Parkinson’s disease (Trupp et al. 1996). The ro- et al. 1999). Together with defects in kidney bust effects of GDNF/RET signaling on do- organogenesis, this leads to the death of RET paminergic neuron survival in several lesion null animals at birth. It has more recently paradigms naturally raised expectations of an been found that conditional ablation of RET, important physiological function for RET in or the GFRa1 coreceptor, in postmigratory en- dopaminergic neurons, where it is expressed teric neurons causes widespread neuronal death from very early stages of development. In agree- in the colon, leading to colonic aganglionosis ment with this, knock-in of a constitutive allele that resembles Hirschsprung’s disease (Uesaka of RET resulted in increased numbers of do- et al. 2007, 2008). paminergic neurons and profound elevation of dopamine concentration, suggesting that RET signaling can have a direct biological effect Motoneurons in the brain dopaminergic system (Mijatovic RET is expressed in all spinal cord motoneurons et al. 2007). Despite the successes of gain-of- from the earliest stages of their development function approaches, the results from loss-of-

6 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Structure and Physiology of the RET RTK

function studies have been less clear-cut with the bulk of other receptor tyrosine kinases. Its regard to the importance of RET activity in ligand system, with the GFRa coreceptors, is dopaminergic neurons. Conditional ablation of also rather unique, and upcoming crystal struc- RET in dopaminergic neurons has failed to re- tures of the full ternary complex promise to re- veal a prominent role for RET signaling in dop- veal the mechanism of complex formation, and aminergic neuron development or mainte- perhaps explain how a distant TGF-b superfam- nance, at least during the average life span of ily member ended up binding a receptor tyro- the mouse (Jain et al. 2006b; Kramer et al. sine kinase. These insights may also find appli- 2007). One of these two studies, however, exam- cations in drug discovery. Indeed, although its ined aging mutant mice and found that RET physiological significance in dopaminergic neu- ablation caused progressive, but moderate, rons remains unclear, the robust effects of RET adult-onset loss of dopaminergic neurons in gain-of-function on the survival and function of the substantia nigra and reduced dopaminergic these neurons has encouraged efforts to identify nerve terminals in striatum, reaching 38% re- agonists for treatment of Parkinson’s disease duction by 2 years of age (Kramer et al. 2007). (Aron and Klein 2011). Such compounds may In contrast to those mild effects, another study circumvent the intrinsic problems of protein induced adult deletion of a conditional allele of delivery of current GDNF-based approaches. Gdnf and found widespread deficits in dopami- nergic and noradrenergic neurons (Pascual et al. 2008). As discussed elsewhere (Ibanez 2008), the ACKNOWLEDGMENTS striking discrepancy between these studies could The author apologizes to all of the colleagues be explained by the presence of alternative re- whose work could not be cited owing to space ceptors for GDNF in dopaminergic neurons, constraints. Work at the author’s laboratory is compensatory effects in developing RET-defi- funded by research grants from the Swedish Re- cient neurons, or the possibility that RET func- search Council, Swedish Cancer Society, Swed- tions as a “dependence receptor” in dopaminer- ish Foundation for Strategic Research, European gic neurons. Dependence receptors make cells Research Council, and the National Institutes of that express them dependent on their ligands, Health. and evidence from transformed cell lines has been provided suggesting that RETmight indeed function in this way (Bordeaux et al. 2000). At REFERENCES the time of this writing, independent efforts are Reference is also in this collection. underway to replicate several of the above-men- tioned studies and so it is likely that the contro- Abrescia C, Sjo¨strand D, Kjaer S, Ibanez CF.2005. Drosophila versyover the physiological role of RETsignaling RET contains an active tyrosine kinase and elicits neuro- trophic activities in mammalian cells. FEBS Lett 579: in dopaminergic neuron survival and mainte- 3789–3796. nance will become clarified before long. Amoresano A, Incoronato M, Monti G, Pucci P, de Fran- ciscis V, Cerchia L. 2005. Direct interactions among Ret, GDNF and GFRa1 molecules reveal new insights into CONCLUDING REMARKS the assembly of a functional three-. Cell Signal 17: 717–727. Nearly three decades after its discovery, unique Anders J, Kjar S, Ibanez CF.2001. Molecular modeling of the aspects of RET function and physiology contin- extracellular domain of the RETreceptor tyrosine kinase ue to fascinate biologists and biochemists. Un- reveals multiple cadherin-like domains and a calcium- binding site. J Biol Chem 276: 35808–35817. like other receptor tyrosine kinases, autophos- Aron L, Klein R. 2011. Repairing the parkinsonian brain phorylation has only a modest effect on kinase with . Trends Neurosci 34: 88–100. activity, and so the mechanism of activation Asai N, Fukuda T, Wu Z, Enomoto A, Pachnis V, Taka- of the RET kinase remains elusive. The possi- hashi M, Costantini F. 2006. Targeted mutation of ser- ine 697 in the Ret tyrosine kinase causes migration defect bility of autoinhibition in trans is tantalizing of enteric neural crest cells. Development 133: 4507– and would mechanistically set RET apart from 4516.

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 7 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

C.F. Iba´n˜ ez

Baloh RH, Tansey MG, Golden JP, Creedon DJ, Heuck- Donis-Keller H, Dou S, Chi D, Carlson KM, Toshima K, eroth RO, Keck CL, Zimonjic DB, Popescu NC, Lairmore TC, Howe JR, Moley JF, Goodfellow P, Johnson EM, Milbrandt J. 1997. TrnR2, a novel receptor Wells SA. 1993. Mutations in the RET proto-oncogene that mediates neurturin and GDNF signaling through are associated with MEN 2A and FMTC. Hum Mol Genet Ret. Neuron 18: 793–802. 2: 851–856. Baloh RH, Gorodinsky A, Golden JP, Tansey MG, Keck CL, Durbec P, Marcos-Gutierrez CV, Kilkenny C, Grigoriou M, Popescu NC, Johnson EM, Milbrandt J. 1998a. GFRa3is Wartiowaara K, Suvanto P, Smith D, Ponder B, Cos- an orphan member of the GDNF/neurturin/persephin tantini FD, Saarma M, et al. 1996. GDNF signalling receptor family. Proc Natl Acad Sci 95: 5801–5806. through the Ret receptor tyrosine kinase. Nature 381: Baloh RH, Tansey MG, Lampe PA, Fahrner TJ, Enomoto H, 789–793. Simburger KS, Leitner ML, Araki T, Johnson EM, Eketja¨ll S, Fainzilber M, Murray-Rust J, Ibanez CF. 1999. Milbrandt J. 1998b. Artemin, a novel member of the Distinct structural elements in GDNF mediate binding GDNF ligand family, supports peripheral and central to GFRa1 and activation of the GFRa1-c-Ret receptor neurons and signals through the GFRa3-RET receptor complex. EMBO J 18: 5901–5910. complex. Neuron 21: 1291–1302. Encinas M, Crowder RJ, Milbrandt J, Johnson EM. 2004. Bespalov MM, Sidorova YA, Tumova S, Ahonen-Bishopp A, Tyrosine 981, a novel ret autophosphorylation site, binds Magalha˜es AC, Kulesskiy E, Paveliev M, Rivera C, c-Src to mediate neuronal survival. J Biol Chem 279: Rauvala H, Saarma M. 2011. Heparan sulfate proteogly- 18262–18269. can syndecan-3 is a novel receptor for GDNF, neurturin, Fukuda T, Kiuchi K, Takahashi M. 2002. Novel mechanism and artemin. J Cell Biol 192: 153–169. of regulation of Rac activity and lamellipodia formation Besset V,Scott RP,Ibanez CF.2000. Signaling complexes and by RET tyrosine kinase. J Biol Chem 277: 19114–19121. protein–protein interactions involved in the activation of Garce`s A, Haase G, Airaksinen MS, Livet J, Filippi P, Dela- the Ras and phosphatidylinositol 3-kinase pathways by peyrie`re O. 2000. GFRa 1 is required for development of the c-Ret receptor tyrosine kinase. J Biol Chem 275: distinct subpopulations of motoneuron. J Neurosci 20: 39159–39166. 4992–5000. Bordeaux MC, Forcet C, Granger L, Corset V, Bidaud C, Geneste O, Bidaud C, De Vita G, Hofstra RM, Tartare- Billaud M, Bredesen DE, Edery P, Mehlen P. 2000. The Deckert S, Buys CH, Lenoir GM, Santoro M, Billaud M. RET proto-oncogene induces : A novel mech- 1999. Twodistinct mutations of the RETreceptor causing anism for Hirschsprung disease. EMBO J 19: 4056–4063. Hirschsprung’s disease impair the binding of signalling Borrello MG, Alberti L, Arighi E, Bongarzone I, Battistini C, effectors to a multifunctional docking site. Hum Mol Bardelli A, Pasini B, Piutti C, Rizzetti MG, Mondellini P, Genet 8: 1989–1999. et al. 1996. The full oncogenic activity of Ret/ptc2 de- Gould TW, Yonemura S, Oppenheim RW, Ohmori S, Eno- pends on tyrosine 539, a docking site for phospholipase moto H. 2008. The neurotrophic effects of glial cell line- Cg. Mol Cell Biol 16: 2151–2163. derived neurotrophic factor on spinal motoneurons are Buj-Bello A, Adu J, Pin˜o´n LG, Horton A, Thompson JF, restricted to fusimotor subtypes. J Neurosci 28: 2131– Rosenthal A, Chinchetru M, Buchman VL, Davies AM. 2146. 1997. Neurturin responsiveness requires a GPI-linked re- Grieco M, Santoro M, Berlingieri MT, Melillo RM, Don- ceptor and the Ret receptor tyrosine kinase. Nature 387: ghi R, Bongarzone I, Pierotti MA, Porta Della G, Fus- 721–724. co A, Vecchio G. 1990. PTC is a novel rearranged form Cik M, Masure S, Lesage AS, Van der Linden I, Van of the ret proto-oncogene and is frequently detected in Gompel P, Pangalos MN, Gordon RD, Leysen JE. 2000. vivo in human thyroid papillary carcinomas. Cell 60: Binding of GDNF and neurturin to human GDNF family 557–563. receptor a 1 and 2. Influence of cRET and cooperative Grimm J, Sachs M, Britsch S, Di Cesare S, Schwarz- interactions. J Biol Chem 275: 27505–27512. Romond T, Alitalo K, Birchmeier W. 2001. Novel Cosma MP,Cardone M, Carlomagno F,Colantuoni V.1998. p62dok family members, dok-4 and dok-5, are substrates Mutations in the extracellular domain cause RET loss of of the c-Ret receptor tyrosine kinase and mediate neu- function by a dominant negative mechanism. Mol Cell ronal differentiation. J Cell Biol 154: 345–354. Biol 18: 3321–3329. Hahn M, Bishop J. 2001. Expression pattern of Drosophila Coulpier M, Anders J, Ibanez CF. 2002. Coordinated activa- ret suggests a common ancestral origin between the meta- tion of autophosphorylation sites in the RET receptor morphosis precursors in insect endoderm and the verte- tyrosine kinase: Importance of tyrosine 1062 for GDNF brate enteric neurons. Proc Natl Acad Sci 98: 1053–1058. mediated neuronal differentiation and survival. J Biol Hayashi H, Ichihara M, Iwashita T,Murakami H, Shimono Y, Chem 277: 1991–1999. Kawai K, Kurokawa K, Murakumo Y,Imai T,Funahashi H, Crowder R, Enomoto H, Yang M, Johnson E, Milbrandt J. et al. 2000. Characterization of intracellular signals via 2004. Dok-6, a novel p62 Dok family member, promotes tyrosine 1062 in RET activated by glial cell line-derived Ret-mediated neurite outgrowth. J Biol Chem 279: 42072– neurotrophic factor. Oncogene 19: 4469–4475. 42081. Henderson C, Phillips H, Pollock R, Davies A, Lemeulle C, de Graaff E, Srinivas S, Kilkenny C, D’Agati V, Mankoo BS, Armanini M, Simpson L, Moffet B, Vandlen R, Koli- Costantini FD, Pachnis V. 2001. Differential activities of atsos V, et al. 1994. Gdnf: A potent survival factor for the RET tyrosine kinase receptor isoforms during mam- motoneurons present in peripheral-nerve and muscle. malian embryogenesis. Dev 15: 2433–2444. Science 266: 1062–1064.

8 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Structure and Physiology of the RET RTK

Hofstra RM, Landsvater RM, Ceccherini I, Stulp RP, Stel- Knowles PP, Murray-Rust J, Kjaer S, Scott RP, Heilshorn S, wagen T, Luo Y, Pasini B, Ho¨ppener JW, van Amstel HK, Santoro M, Ibanez CF, McDonald NQ. 2006. Structure Romeo G. 1994. A mutation in the RET proto-oncogene and chemical inhibition of the RET tyrosine kinase do- associated with multiple endocrine neoplasia type 2B and main. J Biol Chem 281: 33577–33587. sporadic medullary thyroid carcinoma. Nature 367: 375– Kotzbauer PT, Lampe PA, Heuckeroth RO, Golden JP, 376. Creedon DJ, Johnson EM, Milbrandt J. 1996. Neurturin, Ibanez CF. 2008. Catecholaminergic neuron survival: Get- a relative of glial-cell-line-derived neurotrophic factor. ting hooked on GDNF. Nat Neurosci 11: 735–736. Nature 384: 467–470. Iwamoto T, Taniguchi M, Asai N, Ohkusu K, Nakashima I, Kramer ER, Knott L, Su F, Dessaud E, Krull CE, Helm- Takahashi M. 1993. cDNA cloning of mouse ret proto- bacher F, Klein R. 2006. Cooperation between GDNF/ oncogene and its sequence similarity to the cadherin Ret and ephrinA/EphA4 signals for motor-axon pathway superfamily. Oncogene 8: 1087–1091. selection in the limb. Neuron 50: 35–47. Iwashita T,Murakami H, Asai N, Takahashi M. 1996. Mech- Kramer, Aron L, Ramakers, Seitz, Zhuang, Beyer, Smidt, anism of ret dysfunction by Hirschsprung mutations af- Klein. 2007. Absence of Ret signaling in mice causes pro- fecting its extracellular domain. Hum Mol Genet 5: gressive and late degeneration of the nigrostriatal system. 1577–1580. PLoS Biol 5: e39. Jain S, Encinas M, Johnson EM, Milbrandt J. 2006a. Critical Kurokawa K, Iwashita T, Murakami H, Hayashi H, Kawai K, and distinct roles for key RET tyrosine docking sites in Takahashi M. 2001. Identification of SNT/FRS2 docking renal development. Genes Dev 20: 321–333. site on RETreceptor tyrosine kinase and its role for signal transduction. Oncogene 20: 1929–1938. Jain S, Golden JP, Wozniak D, Pehek E, Johnson EM, Milbrandt J. 2006b. RET is dispensable for maintenance Kurotsuchi A, Murakumo Y, Jijiwa M, Kurokawa K, Itoh Y, of midbrain dopaminergic neurons in adult mice. J Neu- Kodama Y, Kato T, Enomoto A, Asai N, Terasaki H, et al. rosci 26: 11230–11238. 2010. Analysis of DOK-6 function in downstream signal- ing of RET in human neuroblastoma cells. Cancer Sci Jiao L, Zhang Y, Hu C, Wang Y-G, Huang A, He C. 2011. 101: 1147–1155. Rap1GAP interacts with RET and suppresses GDNF-in- duced neurite outgrowth. Cell Res 21: 327–337. Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F. 1993. GDNF: A glial cell line-derived neurotrophic factor for Jijiwa M, Fukuda T, Kawai K, Nakamura A, Kurokawa K, midbrain dopaminergic neurons. Science 260: 1130– Murakumo Y,Ichihara M, TakahashiM. 2004. A targeting 1132. mutation of tyrosine 1062 in Ret causes a marked de- crease of enteric neurons and renal hypoplasia. Mol Cell Liu X, VegaQC, Decker RA, Pandey A, Worby CA, Dixon JE. Biol 24: 8026–8036. 1996. Oncogenic RET receptors display different auto- phosphorylation sites and substrate binding specificities. Jing S, Wen D, Yu Y, Holst PL, Luo Y, Fang M, Tamir R, J Biol Chem 271: 5309–5312. Antonio L, Hu Z, Cupples R, et al. 1996. GDNF-induced activation of the ret protein tyrosine kinase is mediated Lundgren TK, Scott RP,Smith M, Pawson T,Ernfors P.2006. Engineering the recruitment of phosphotyrosine binding by GDNFR-a, a novel receptor for GDNF. Cell 85: domain-containing adaptor proteins reveals distinct 1113–1124. roles for RET receptor-mediated cell survival. J Biol Kawamoto Y, Takeda K, Okuno Y, Yamakawa Y, Ito Y, Chem 281: 29886–29896. TaguchiR, Kato M, Suzuki H, Takahashi M, Nakashima I. Massague´ J. 1996. Neurotrophic factors. Crossing receptor 2004. Identification of RETautophosphorylation sites by boundaries. Nature 382: 29–30. mass spectrometry. J Biol Chem 279: 14213–14224. Masure S, Cik M, Hoefnagel E, Nosrat CA, Vander Linden I, Kjaer S, Ibanez CF. 2003a. Identification of a surface for Scott RP, Van Gompel P, Lesage AS, Verhasselt P, binding to the GDNF-GFR a 1 complex in the first cad- Ibanez CF, et al. 2000. Mammalian GFRa-4, a divergent herin-like domain of RET. J Biol Chem 278: 47898– member of the GFRa family of coreceptors for glial cell 47904. line-derived neurotrophic factor family ligands, is a re- Kjaer S, Ibanez CF. 2003b. Intrinsic susceptibility to mis- ceptor for the neurotrophic factor persephin. J Biol Chem folding of a hot-spot for Hirschsprung disease mutations 275: 39427–39434. in the ectodomain of RET. Hum Mol Genet 12: 2133– Melillo RM, Santoro M, Ong SH, Billaud M, Fusco A, 2144. Hadari YR, Schlessinger J, Lax I. 2001. Docking protein Kjaer S, Kurokawa K, Perrinjaquet M, Abrescia C, Ibanez CF. FRS2 links the protein tyrosine kinase RETand its onco- 2006. Self-association of the transmembrane domain of genic forms with the mitogen-activated protein kinase RET underlies oncogenic activation by MEN2A muta- signaling cascade. Mol Cell Biol 21: 4177–4187. tions. Oncogene 25: 7086–7095. Mijatovic J, Airavaara M, Planken A, Auvinen P,Raasmaja A, Kjaer S, Heilshorn S, Totty N, McDonald NQ. 2010. Mam- Piepponen TP,Costantini FD, Ahtee L, Saarma M. 2007. mal-restricted elements predispose human RET to fold- Constitutive Ret activity in knock-in multiple endocrine ing impairment by HSCR mutations. Nat Struct Biol 17: neoplasia type B mice induces profound elevation of 726–731. brain dopamine concentration via enhanced synthesis Klein RD, Sherman D, Ho WH, Stone D, Bennett GL, and increases the number of TH-positive cells in the sub- Moffat B, Vandlen R, Simmons L, Gu Q, Hongo JA, et stantia nigra. J Neurosci 27: 4799–4809. al. 1997. A GPI-linked protein that interacts with Ret to Milbrandt J, de Sauvage FJ, Fahrner TJ, Baloh RH, Leit- form a candidate neurturin receptor. Nature 387: ner ML, Tansey MG, Lampe PA, Heuckeroth RO, Kotz- 717–721. bauer PT, Simburger KS, et al. 1998. Persephin, a novel

Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 9 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

C.F. Iba´n˜ ez

neurotrophic factor related to GDNF and neurturin. Schuetz G, Rosa´rio M, Grimm J, Boeckers TM, Gundel- Neuron 20: 245–253. finger ED, Birchmeier W. 2004. The neuronal scaffold Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C, protein Shank3 mediates signaling and biological func- Gardner E, Love DR, Mole SE, Moore JK, Papi L. 1993. tion of the receptor tyrosine kinase Ret in epithelial cells. J Germ-line mutations of the RET proto-oncogene in mul- Cell Biol 167: 945–952. tiple endocrine neoplasia type 2A. Nature 363: 458–460. Segouffin-Cariou C, Billaud M. 2000. Transforming ability Naveilhan P, Baudet C, Mikaels A, Shen L, Westphal H, of MEN2A-RET requires activation of the phosphatidy- Ernfors P. 1998. Expression and regulation of GFRa3, a linositol 3-kinase/AKT signaling pathway. J Biol Chem glial cell line-derived neurotrophic factor family receptor. 275: 3568–3576. Proc Natl Acad Sci 95: 1295–1300. Shakya R, Watanabe T, Costantini FD. 2005. The role of Nollet F, Kools P, van Roy F. 2000. Phylogenetic analysis of GDNF/Ret signaling in ureteric bud cell fate and branch- the cadherin superfamily allows identification of six ma- ing morphogenesis. Dev Cell 8: 65–74. jor subfamilies besides several solitary members. J Mol Takahashi M, Cooper GM. 1987. ret transforming gene en- Biol 299: 551–572. codes a fusion protein homologous to tyrosine kinases. Oppenheim RW, Houenou LJ, Parsadanian AS, Prevette D, Mol Cell Biol 7: 1378–1385. Snider WD, Shen L. 2000. Glial cell line-derived neuro- TakahashiM, Ritz J, Cooper GM. 1985. Activation of a novel trophic factor and developing mammalian motoneurons: human transforming gene, ret, by DNA rearrangement. Regulation of programmed cell death among motoneu- Cell 42: 581–588. ron subtypes. J Neurosci 20: 5001–5011. Takahashi M, Buma Y, Iwamoto T, Inaguma Y, Ikeda H, Pachnis V, Mankoo B, Costantini FD. 1993. Expression of Hiai H. 1988. Cloning and expression of the ret proto- the c-ret proto-oncogene during mouse embryogenesis. oncogene encoding a tyrosine kinase with two potential Development 119: 1005–1017. transmembrane domains. Oncogene 3: 571–578. Paratcha G, Ledda F,Baars L, Coulpier M, Besset V,Anders J, Taraviras S, Marcos-Gutierrez CV, Durbec P, Jani H, Gri- Scott RP, Ibanez CF. 2001. Released GFRa1 potentiates goriou M, Sukumaran M, Wang LC, Hynes MA, Rais- downstream signaling, neuronal survival, and differenti- man G, Pachnis V. 1999. Signalling by the RET receptor ation via a novel mechanism of recruitment of c-Ret to tyrosine kinase and its role in the development of the lipid rafts. Neuron 29: 171–184. mammalian enteric nervous system. Development 126: 2785–2797. Paratcha G, Ledda F, Ibanez CF. 2003. The neural cell adhe- sion molecule NCAM is an alternative signaling receptor Trupp M, Arenas E, Fainzilber M, Nilsson AS, Sieber B-A, for GDNF family ligands. Cell 113: 867–879. Grigoriou M, Kilkenny C, Salazar-Grueso E, Pachnis V, Arumae U, et al. 1996. Functional receptor for GDNF Pascual A, Hidalgo-Figueroa M, Piruat JI, Pintado CO, Go´- encoded by the c-ret proto-oncogene. Nature 381: 785– mez-Dı´az R, Lo´pez-Barneo J. 2008. Absolute require- 789. ment of GDNF for adult catecholaminergic neuron sur- vival. Nat Neurosci 11: 755–761. TruppM, Belluardo N, Funakoshi H, Ibanez CF.1997. Com- plementary and overlapping expression of glial cell line- Pelet A, Geneste O, Edery P, Pasini A, Chappuis S, Atti T, derived neurotrophic factor (GDNF), c-ret proto-onco- Munnich A, Lenoir G, Lyonnet S, Billaud M. 1998. Var- gene, and GDNF receptor-a indicates multiple mecha- ious mechanisms cause RET-mediated signaling defects nisms of trophic actions in the adult rat CNS. J Neurosci in Hirschsprung’s disease. J Clin Invest 101: 1415–1423. 17: 3554–3567. Perrinjaquet M, Vilar M, Ibanez CF. 2010. Protein-tyrosine Trupp M, Raynoschek C, Belluardo N, Ibanez CF. 1998. phosphatase SHP2 contributes to GDNF neurotrophic Multiple GPI-anchored receptors control GDNF-depen- activity through direct binding to phospho-Tyr687 in dent and independent activation of the c-Ret receptor the RET receptor tyrosine kinase. J Biol Chem 285: tyrosine kinase. Mol Cell Neurosci 11: 47–63. 31867–31875. Uesaka T, Jain S, Yonemura S, Uchiyama Y, Milbrandt J, Sanicola M, Hession CA, Worley D, Carmillo P,Ehrenfels C, Enomoto H. 2007. Conditional ablation of GFRa1in Walus L, Robinson S, Jaworski G, Wei H, Tizard R, et al. postmigratory enteric neurons triggers unconventional 1997. Glial cell line-derived neurotrophic factor-depen- neuronal death in the colon and causes a Hirschsprung’s dent RETactivation can be mediated by two different cell- disease phenotype. Development 134: 2171–2181. surface accessory proteins. Proc Natl Acad Sci 94: Uesaka T, Nagashimada M, Yonemura S, Enomoto H. 2008. 6238–6243. Diminished Ret expression compromises neuronal sur- Santoro M, Carlomagno F. 2013. Central role of RET in vival in the colon and causes intestinal aganglionosis in thyroid cancer. Cold Spring Harb Perspect Biol doi: mice. J Clin Invest 118: 1890–1898. 10.1101/cshperspect.a009233. Wong A, Bogni S, Kotka P, de Graaff E, D’Agati V, Schuchardt A, D’Agati V, Larsson-Blomberg L, Costan- Costantini FD, Pachnis V. 2005. Phosphotyrosine 1062 tini FD, Pachnis V.1994. Defects in the kidney and enteric is critical for the in vivo activity of the Ret9 receptor nervous system of mice lacking the tyrosine kinase recep- tyrosine kinase isoform. Mol Cell Biol 25: 9661–9673. tor Ret. Nature 367: 380–383. Worby CA, Vega QC, Chao HH, Seasholtz AF, Thomp- Schuchardt A, D’Agati V, Pachnis V, Costantini F. 1996. Re- son RC, Dixon JE. 1998. Identification and characteriza- nal agenesis and hypodysplasia in ret-k- mutant mice tion of GFRa-3, a novel co-receptor belonging to the glial result from defects in ureteric bud development. Devel- cell line-derived neurotrophic receptor family. J Biol opment 122: 1919–1929. Chem 273: 3502–3508.

10 Cite this article as Cold Spring Harb Perspect Biol 2013;5:a009134 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Structure and Physiology of the RET Receptor Tyrosine Kinase

Carlos F. Ibáñez

Cold Spring Harb Perspect Biol 2013; doi: 10.1101/cshperspect.a009134

Subject Collection Signaling by Receptor Tyrosine Kinases

CSF-1 Receptor Signaling in Myeloid Cells The Genesis of E. Richard Stanley and Violeta Chitu Tony Hunter The EGFR Family: Not So Prototypical Receptor Structure-Function Relationships of ErbB RTKs in Tyrosine Kinases the Plasma Membrane of Living Cells Mark A. Lemmon, Joseph Schlessinger and Donna J. Arndt-Jovin, Michelle G. Botelho and Kathryn M. Ferguson Thomas M. Jovin Tie2 and Eph Receptor Tyrosine Kinase Activation Receptor Tyrosine Kinases: Legacy of the First and Signaling Two Decades William A. Barton, Annamarie C. Dalton, Tom C.M. Joseph Schlessinger Seegar, et al. The Spatiotemporal Organization of ErbB The Role of Ryk and Ror Receptor Tyrosine Receptors: Insights from Microscopy Kinases in Wnt Signal Transduction Christopher C. Valley, Keith A. Lidke and Diane S. Jennifer Green, Roel Nusse and Renée van Lidke Amerongen Receptor Signaling in Normal and Regulation of Receptor Tyrosine Kinase Ligand Insulin-Resistant States Processing Jérémie Boucher, André Kleinridders and C. Colin Adrain and Matthew Freeman Ronald Kahn Central Role of RET in Thyroid Cancer Molecular Mechanisms of SH2- and Massimo Santoro and Francesca Carlomagno PTB-Domain-Containing Proteins in Receptor Tyrosine Kinase Signaling Melany J. Wagner, Melissa M. Stacey, Bernard A. Liu, et al. Receptor Tyrosine Kinase-Mediated Angiogenesis Eph Receptor Signaling and Michael Jeltsch, Veli-Matti Leppänen, Pipsa Erika M. Lisabeth, Giulia Falivelli and Elena B. Saharinen, et al. Pasquale Biology of the TAM Receptors Effects of Membrane Trafficking on Signaling by Greg Lemke Receptor Tyrosine Kinases Marta Miaczynska For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2013 Cold Spring Harbor Laboratory Press; all rights reserved Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2013 Cold Spring Harbor Laboratory Press; all rights reserved