You are on page 1of 8

Opinion

GDNF and GFRa: a versatile molecular


complex for developing neurons
Gustavo Paratcha and Fernanda Ledda
Laboratory of Molecular and Cellular Neuroscience, Department of Neuroscience, Karolinska Institute, S-17177 Stockholm, Sweden

The GDNF family ligands (GFLs) signal through the of the GFRa co-receptor. The other component of the
canonical signaling receptor Ret and a glycosyl-phos- GDNF receptor system, GFRa1, was identified in 1996
phatidylinositol-anchored co-receptor, GFRa. In recent by expression cloning and screening of GDNF binding
years, signaling by GFLs has been shown to be more protein [11,12]. The family of GFRa co-receptors includes
complex than originally assumed. The discrepant four members (GFRa 1–4) which determine ligand speci-
expression between GFRas and Ret has suggested the ficity. Thus, GFRa 1–4 are the preferential co-receptors for
existence of additional signal-transducing GDNF recep- GDNF, NRTN, ARTN and PSPN, respectively, but alterna-
tors, such as NCAM. Here we summarize novel functions tive ligand–co-receptor interactions can also occur [13].
and Ret-independent signaling mechanisms for GDNF Following homodimeric GFL binding to the cognate GFRa
and GFRa, focusing on developing neurons. Emerging co-receptor, Ret becomes dimerized and tyrosine phos-
evidence indicates a prominent role of GDNF and GFRa phorylated, and triggers different pathways, including
in the control of neuroblast migration and chemoattrac- the Ras-MAPK (mitogen-activated protein kinase), the
tion and in the formation of neuronal synapses by a new phosphatidylinositol-3 kinase (PI3K)-Akt, the PLC-g and
mechanism of ligand-induced cell adhesion. Therefore, the Src signaling pathways (Figure 1) [13,14].
these data highlight the importance of this versatile Several observations have indicated that GDNF
molecular complex for nervous system development, signaling is more complex than originally supposed. In
function and regeneration. several regions of the nervous system, GFRas are highly
expressed where Ret is undetectable [2,13]. One possibility
Introduction to explain this discordance is that GFRas can activate Ret
Glial cell line-derived neurotrophic factor (GDNF) family signaling in a non-cell-autonomous fashion, presenting
ligands (GFLs) play a crucial role in the development and GFLs in trans to Ret-expressing cells (see Figure 1)
function of the nervous system [1,2]. GDNF was originally [15–17]. Furthermore, the discrepant expression of Ret
characterized in 1993 as a growth factor promoting the and GFRas prompted many groups to intensively explore
survival of ventral midbrain dopaminergic neurons, those for the existence of additional GFL receptors that signal
that degenerate in Parkinson’s disease [3]. Later, it was cooperatively with GFRas but independently of Ret
found to have a potent survival effect on motor neurons and (Table 1) [18–24]. Thus, in 2003, NCAM was identified
other neuronal subpopulations in the central and periph- as an alternative signaling receptor for GFLs [21]. This
eral nervous systems (CNS and PNS, respectively) [1,2,4]. large body of evidence points out the relevance of GFRa co-
In addition to its role as a survival factor, GDNF is also receptors for the versatility of GFL signaling (see Figure 1
essential for proliferation, migration and differentiation of and Box 1).
neuronal cells [2,5–8]. The biological effects summarized in This review summarizes recent advances made in our
Table 1 highlight the pleotropic roles played by GDNF understanding of the Ret-dependent and -independent
during neuronal development. signaling mechanisms used by the GFL-GFRa complex
The GFLs consist of four members, GDNF, Neurturin and their functional implications for nervous system de-
(NRTN), Artemin (ARTN) and Persephin (PSPN), which velopment. Based on the potential therapeutic implica-
are involved in the survival, proliferation and differen- tions of GDNF and Ret in multiple neurodegenerative
tiation of several neuronal populations in the CNS and diseases, we also draw attention to some new mechanisms
PNS [2]. A common feature of the receptor complex for the through which Ret receptor signaling and biology are
GFLs is the requirement of two receptor subunits, one restricted in neuronal cells.
specialized in transmembrane signaling, the receptor tyro-
sine kinase Ret, and the other involved in ligand binding, Novel physiological functions of GFRa-Ret signaling
the glycosyl-phosphatidylinositol (GPI)-anchored co-recep- Mice lacking Ret die a few hours after birth owing to
tor GFRa (GDNF family receptor a) [1]. In 1996, the kidney agenesis and show absence of enteric and many
transmembrane receptor tyrosine kinase Ret was ident- parasympathetic neurons [2]. A similar phenotype was
ified as a GDNF receptor [9,10]. Ret is shared by all of the described for Gdnf- and Gfra1-null mutant mice
GFLs as their common signaling receptor. However, Ret is [2,25,26]. By contrast, Gfra2-, Gfra3- and Gfra4-mutant
unable to bind GDNF or any of the other GFLs in absence mice are viable and fertile [2]. In light of the known in vitro
Corresponding authors: Paratcha, G. (gustavo.paratcha@ki.se);
survival effects of GDNF/Ret signaling on dopaminergic
Ledda, F. (fernanda.ledda@ki.se). and motor neurons, it was surprising that mice lacking
384 0166-2236/$ – see front matter ß 2008 Elsevier Ltd. All rights reserved. doi:10.1016/j.tins.2008.05.003 Available online 1 July 2008
Opinion Trends in Neurosciences Vol.31 No.8

Table 1. Summary of GDNF effects on neuroblasts and mature neurons


Biological functions Cell type Receptor system Refs
Neuroblast
Proliferation Enteric progenitors Ret/GFRa [6,8]

Migration/chemoattraction Enteric progenitors Ret/GFRa [5,7]


MGE-derived progenitors ?/GFRa [20]
RMS-derived progenitors NCAM/GFRa [18]

Differentiation Enteric progenitors Ret/GFRa [8]


MGE-derived progenitors ?/GFRa [20]

Neuron
Axonal growth Sensory and sympathetic neurons Ret/GFRa* [16]
Motor neurons Ret/GFRa [52]
Hippocampal neurons NCAM/GFRa [21]
Cortical GABAergic neurons ?/GFRa [20]
Axon guidance Sensory and sympathetic neurons Ret/GFRa* [16]
Spinal chord motor neurons Ret/GFRa [29]

Survival VM dopaminergic neurons Ret/GFRa [3]


Spinal chord motor neurons [2,4]
Sympathetic, parasympathetic and sensory neurons [2]
Enteric neurons [8,27]

Synapse formation Midbrain dopaminergic neurons Ret/GFRa [50]


Motor neurons [32,52]
Hippocampal and cortical neurons NCAM/GFRa [19]

The table describes the distinct biological functions of GDNF at different time points during neuronal development. The neuronal type and the receptor system through which
GDNF ligands induce these biological effects are also indicated. Asterisks indicate that the effect was obtained using soluble GFRa presented in trans and ‘‘?’’ indicates that the
signaling receptor is unknown. MGE = medial ganglionic eminence; RMS = rostral migratory stream; VM = ventral midbrain.

Ret, GDNF or GFRa1 show only minor phenotypes in the development [33]. Recently, two groups have reported the
CNS during embryonic development. However, the generation of different strains of mice with the specific
early lethality of these animals precluded postnatal deletion of Ret in dopaminergic neurons [30,31]. Surpris-
analyses of the nervous system. To overcome this obstacle, ingly, these animals showed normal development and
recently different groups have generated mice with selec- maturation of the nigrostriatal system. A detailed study,
tive ablation of Ret or GFRa1 in different neuronal popu- including dopaminergic neuron number in substantia
lations that are compatible with postnatal survival nigra pars compacta (SNpc) and ventral tegmental area
[27–32]. Here we attempt to summarize the current knowl- (VTA), fiber density in the striatum and nucleus accum-
edge regarding the in vivo function of the Ret receptors as bens, and dopamine levels, indicated that Ret is not
determined from the analysis of tissue-specific Ret-knock- required for the maintenance of midbrain dopaminergic
out and -inducible Gfra1-deficient mice that have been neurons in young adult mice. However, in one of these
generated during the last 2 years. studies, Kramer and coworkers found that, only in aged
animals, Ret ablation leads to a progressive and cell-type-
Dopaminergic neurons specific loss of SNpc dopaminergic neurons and their
Although GDNF was originally characterized as a potent afferents, thereby indicating that Ret is a critical regulator
survival factor for midbrain dopaminergic neurons, for the long-term maintenance of the nigrostriatal system
analysis of knockout mice has shown that GFRa1-Ret [30].
signaling is not essential for the embryonic development The significance of Ret signaling for the dopaminergic
of these neurons, illustrating the complex network of sig- system was additionally studied in knockin multiple endo-
naling pathways and compensatory mechanisms that crine neoplasia type B (MEN2B) mice by introducing the
regulate neural development. In agreement with this, it Ret-MEN2B mutation (Met919Thr), which renders the
was suggested that the neurotrophic factor CDNF (con- Ret receptor tyrosine kinase constitutively active. In this
served dopamine neurotrophic factor) might compensate work, the authors showed that the constitutive Ret
GDNF-GFRa1-Ret deficiency during embryonic midbrain activity in mice is sufficient to increase brain dopamine

385
Opinion Trends in Neurosciences Vol.31 No.8

Figure 1. Schematic representation of GFL signaling through Ret and p140NCAM receptors. The left side of the figure describes GFL-induced Ret receptor tyrosine kinase
activation and signaling by membrane-bound (cis-signaling) and soluble GFRa (sGFRa, trans-signaling) molecules released from the plasma membrane. During activation
in cis, GFL dimers bind to GPI-anchored GFRa receptors located in lipid rafts. The heterocomplex GFL-GFRa associates with Ret resulting in its dimerization, tyrosine
autophosphorylation and activation of downstream signaling pathways. During activation in trans, GPI-anchored GFRa is released from the membrane of neighboring cells
by the action of unknown phospholipases or proteases. Soluble GFRa molecules bind to and activate Ret inside and outside lipid rafts. For simplicity, the figure only shows
Ret activation by GFRa in trans in raft domains. Only the Ras/MAPK and PI3K/Akt signaling pathways are represented in the scheme. The right side of the figure summarizes
NCAM signaling. In the absence of GFLs, short-range signaling by homophilic interactions between p140NCAM molecules can be disrupted by the presence of GFRa. This is a
GFL-independent mechanism of extracellular signaling crosstalk between GFRa and p140NCAM molecules. In the presence of GFLs, the association of membrane-bound
GFRa and p140NCAM results in the activation of the Fyn-FAK-MAPK signaling pathway and in the ability of p140NCAM to mediate long-range intercellular communication.
Binding affinities of GDNF for Ret and NCAM in the presence of GFRa1 are as follows: DdRet + GFRa1 = 0.27  0.12 nM and DdNCAM + GFRa1 = 1.1  0.32 nM. For simplicity, the
molecules are represented in monomeric form.

concentration and the number of dopaminergic neurons in of Ret in premigratory neural crest cells, including pro-
the SNpc [34]. genitors of DRG neurons, was generated [28]. These
animals survive at birth; however, the majority of them
Motor neurons die by 3 weeks of age. Using these mice, the authors found
The overexpression of GDNF in skeletal muscle induces that Ret signaling is not required for the viability of non-
hyperinnervation of the neuromuscular junction (NMJ) peptidergic DRG neurons in vivo but is important for the
[35], indicating an important role for GDNF in motor acquisition of normal neuronal size, axonal innervation of
neuron target innervation. Using Ret-null mice and a the epidermis, postnatal extinction of the NGF receptor
mutant mouse strain with a conditional inactivation of TrkA and expression of nonpeptidergic neuron-specific
Ret in the embryonic spinal cord, Kramer and coworkers genes.
demonstrated that GDNF-Ret signaling functions as an
instructive guidance cue for certain motor axons toward Enteric neurons
dorsal limb muscles. Thus, in these Ret-mutant mice, Analysis of Ret-null mice embryos revealed that enteric
motor axons follow an aberrant ventral trajectory away nervous system (ENS) progenitors are reduced in number
from the dorsal territory enriched in GDNF. Interestingly, and fail to enter the midgut, precluding the analysis of the
this phenotype is enhanced in mutant mice lacking both physiological relevance of GDNF signaling in late ENS
Ret and Eph receptor tyrosine kinase A4 (EphA4), development. To study this, Enomoto and coworkers have
suggesting that Ret and EphA4 signals cooperate in the engineered mice in which the function of GFRa1 can be
motor neuron guidance decision [29]. Recently, Baudet and conditionally inactivated in late ENS development. Dis-
coworkers have generated another strain of mice with a ruption of GFRa1 at this late stage resulted in a rapid and
specific deletion of Ret in cranial motor neurons. In these widespread neuronal death in the colon, leading to agan-
animals, the absence of Ret results in a developmental glionosis reminiscent of Hirschsprung’s disease [27].
deficit in motor neuron maturation and specialization of
presynaptic neuromuscular terminals in specific motor New insights into the negative control of Ret signaling
neuron pools [32]. Despite the critical role played by GDNF and Ret in
orchestrating the development and maintenance of specific
Sensory neurons neuronal subpopulations, the molecular mechanisms that
In peripheral neurons of the dorsal root ganglia (DRG), Ret restrict Ret activation and downstream signaling are not
is expressed by a subpopulation of nonpeptidergic cells entirely understood. Undoubtedly, the understanding of
that mediates pain sensation. To analyze the influence of the mechanisms that regulate Ret signaling in neuronal
Ret signaling on postnatal development of sensory cells could open new therapeutic possibilities for the treat-
neurons, a new strain of mice with conditional deletion ment of both neurodegenerative disorders and nerve

386
Opinion Trends in Neurosciences Vol.31 No.8

Box 1. GFL signaling: advantages to having a GFRa Other negative controls include antagonists that
co-receptor undergo transcriptional upregulation upon GDNF-induced
Ret signaling. These inhibitors function through negative
Ret and NCAM signaling receptors require the presence of GFRa co- feedback loops to restrict Ret activation and signaling in a
receptors for both high-affinity GFL binding and receptor signaling
activation. Therefore, GFRas allow the GFLs to influence many
reversible manner. Examples of this type of Ret inhibitors
biological processes by signaling through different transmembrane include members of the Sprouty family of proteins [36,37]
receptors (Figure 1) [13]. and the leucine-rich repeat and immunoglobulin-like
By virtue of their GPI anchor domain, GFRas are localized in domain protein Lrig1 [38].
specialized microenvironments on the cell surface known as lipid rafts
A genetic study revealed that loss of the Sprouty2 gene
[17,38,40,59]. Rafts are dynamic assemblies of cholesterol and
sphingolipids scattered within the lipid bilayer that have emerged in mice resulted in enteric nerve hyperplasia due to GDNF-
as membrane platforms specialized in signal transduction events induced hyperactivation of MAPK and Akt in enteric nerve
[60,61]. An important function of GPI-anchored GFRas is to induce a cells [36]. In agreement with this, Ishida and coworkers
rapid recruitment of Ret to lipid rafts in response to GFLs (cis also demonstrated that Sprouty2 is able to regulate growth
mechanism) [17,59]. As in many other GPI-linked molecules, GPI-
and differentiation of human neuroblastoma cells through
anchored GFRas can function in both membrane-bound and -soluble
forms after release by proteinases and/or phospholipases. Notably, in the Ret-MAPK pathway [37].
cells lacking GFRa, Ret can also be recruited to raft compartments by In addition to Sprouty, Lrig1 has also been identified as
simultaneous treatment with a GFL and its cognate GFRa partner in an endogenous inhibitor of Ret activation and signaling. In
soluble form (trans mechanism) (Figure 1) [17]. In particular, particular, physical association between Lrig1 and Ret
recruitment of Ret in trans is delayed and sustained. In this case,
activated Ret can also be detected outside rafts, suggesting the
inhibits GDNF binding, recruitment of Ret to raft domains,
existence of a dynamic equilibrium in which Ret is partitioning receptor autophosphorylation and MAPK activation in
between raft and nonraft compartments. Indeed, trans stimulation response to GDNF [38].
has the capacity to activate different adaptor signaling molecules
depending on whether Ret is activated outside or inside rafts, NCAM: an alternative signaling receptor for GFLs
allowing this receptor to transmit different signals depending on its
membrane localization [17]. In addition, released GFRas have the
The identification of alternative signal-transducing GDNF
capacity to influence axonal guidance by creating positional informa- receptors has remained an unresolved issue for several
tion for Ret-positive axons even in the presence of a uniform years. Recently, NCAM has been identified as an alterna-
concentration of GFLs [16]. Clearly, all of this evidence indicates that tive signaling receptor for GFLs [21]. Interestingly, high-
the release of GFRa molecules could represent a general strategy to
affinity GDNF binding and signaling through NCAM
diversify and potentiate both signaling and biological functions of
GFLs. The fact that both Schwann cells and injured sciatic nerves requires the presence of GFRa co-receptors. Indeed, in
release GFRa1 molecules to the extracellular space [17] could increase cultured Schwann cells and cortical neurons, both of which
the therapeutic possibilities of GFLs for the treatment of nerve injuries express GFRa and p140NCAM isoform, but not Ret, GDNF
and neurodegeneration. treatment activates the cytoplasmic protein tyrosine
Interestingly, GFRa molecules interfere with NCAM function
kinases Fyn and FAK, two signaling mediators down-
silencing NCAM homophilic interactions and NCAM-mediated
cell adhesion (Figure 1) [21]. More recent evidence indicates that stream of p140NCAM [21]. Structure–function analysis
GDNF mediates cell adhesion between GFRa1-expressing cells identified the third immunoglobulin domain of NCAM as
through a novel mechanism of ligand-induced cell adhesion [19], the necessary and sufficient determinant for its interaction
revealing the ability of GFRa1 and GDNF to function as a remarkably with GDNF [42]. A distinctive characteristic of GFRa is its
versatile molecular complex with capacity to modulate intercellular
communication.
ability to act as a negative regulator of the NCAM homo-
philic interaction in the absence of GDNF [21]. Interest-
ingly, the role of GFRa in silencing NCAM–NCAM
injury. Research over the last 3 years has led to the interactions but promoting GDNF binding to NCAM
identification and characterization of the mechanism of represents a novel mechanism of extracellular crosstalk
action of several Ret signaling inhibitors [36–41]. The between soluble growth factor (long-range) and contact-
picture arising from these studies is that Ret signaling mediated (short-range) signaling with the capacity to influ-
is tightly regulated through the coordinated action of ence important biological processes during nervous system
different protein inhibitors that function at multiple levels development (Figure 1).
of the signaling cascade and at different time points after The most distinctive abnormality of the CNS of mice
receptor engagement. lacking NCAM is a dramatic reduction in the size of the
A prevalent mechanism to control Ret is its downregu- olfactory bulb (OB) [43], caused by defects in the migration
lation by ligand-induced receptor ubiquitination, which of neuronal precursors in the rostral migratory stream
sorts active Ret molecules for intracellular degradation (RMS). The prominent expression of GFRa1 and NCAM,
(irreversible inhibition). Indeed, Ret degradation is accom- but absence of Ret, in the developing and adult RMS
plished via the recruitment and polyubiquitination of the indicates that Ret-independent GDNF signaling via
receptor by Cbl ubiquitin ligases and subsequent degra- NCAM could play a role in the migration and guidance
dation by the proteasomal pathway [39,40]. Interestingly, of neuroblasts and in the development of the RMS-OB
Pierchala and colleagues uncovered an additional role for pathway [21,44]. Consistent with this, Gfra1-deficient
lipid rafts in the control of GDNF signaling. These mem- newborn mice have a moderate enlargement in the width
brane domains sequester phosphorylated Ret away from of the developing RMS, a phenotype that is absent in
proteasomal degradation, thereby increasing the half-life newborn mice lacking Ret [21]. However, these findings
of activated Ret and strengthening its downstream sig- have been considered inconclusive because the RMS and
naling [40]. OB are only partially developed by postnatal day 0 (P0),

387
Opinion Trends in Neurosciences Vol.31 No.8

when GFRa1-mutant mice die. To further address this and tangential migration of cortical GABAergic neurons
issue, Enomoto and colleagues generated mutant mice via GFRa1 receptors but independently of Ret or NCAM,
lacking GFRa1 expression in cells devoid of Ret, such as suggesting that GFRa1 molecules might associate with as
those in the RMS [44]. Although no differences were yet unidentified signaling receptors [20]. In addition, Pop-
observed in either the size or the gross histological organ- sueva and coworkers have shown that in cells expressing
ization of the adult RMS and OB cell layers, it is possible GFRa1, but not Ret, GDNF transactivates Met signaling
that some defects in specific neuronal populations might be through a Src-dependent mechanism [22]. Therefore, the
found in these structures following a more detailed expression of the hepatocyte growth factor receptor Met in
analysis. More recently, it has been shown that long-range migrating GABAergic neurons of the forebrain [47] might
NCAM signaling by GDNF is able to evoke chemotropic be mediating the Ret- and NCAM-independent migratory
effects on neuronal precursors derived from the RMS [18], activity reported for GDNF and GFRa1 on these cells.
indicating that a more detailed analysis of the different
neuronal cell types present in the OB of mice in which Ret- GFRa as a ligand-induced cell-adhesion molecule
independent GFRa1 expression is specifically eliminated Recently, a new role for GFRa proteins as ligand-induced
deserves further investigation. cell-adhesion molecules (LICAMs) has been described [19].
In addition to providing guidance of neuroblasts in vitro, Unlike other known cell-adhesion molecules (CAMs) which
GDNF-GFRa-NCAM signaling stimulates neurite out- involve the direct interaction of membrane-associated
growth of cultured hippocampal and cortical neurons adhesion partners in trans, GFRa1-mediated cell adhesion
[21], regulates Schwann cell migration and function is dependent on the presence of its ligand, GDNF.
[21,45] and promotes presynaptic maturation and synapse Whereas stable cell interactions are necessary to main-
formation of hippocampal and cortical neurons in vitro and tain the structural integrity of tissues, dynamic cell-
in vivo [19]. Interestingly, a recent study demonstrating adhesion mechanisms are involved in the morphogenesis
that NCAM signaling plays a critical role in the analgesic of developing and regenerating tissues. The majority of the
effect of GDNF in a rat model of neuropathic pain might CAMs known to date are regulated at different levels
support the physiological relevance of this new signaling including transcription, expression and clustering. There
pathway [46]. are also dynamic CAMs that allow for the regulation of
Recently, the possibility that GFRas might also have intercellular interactions by conformational changes that
cell-autonomous functions independent of Ret or NCAM modify their adhesive properties through inside-out sig-
has been suggested. Intriguingly, Pozas and Ibáñez naling in response to external cues (e.g. integrins, cadher-
demonstrated that GDNF regulates the differentiation ins) [48,49]. However, the mechanism of ligand-induced

Figure 2. Model describing the mechanism of ligand-induced cell adhesion by GDNF and GFRa1. (a) Scheme summarizing GDNF-induced trans-homophilic cell adhesion
between GFRa1-expressing cells. (b) Hypothetical model describing trans-synaptic adhesion and presynaptic differentiation induced by GDNF and GPI-anchored GFRa1
molecules in hippocampal neurons. In the presence of GDNF, GFRa1, located at pre- and postsynaptic endings, holds the terminals together triggering presynaptic
maturation. The potential role of GDNF and GFRa1 promoting postsynaptic organization remains to be studied (?). The recruitment of synaptic vesicles (SV) to the
presynaptic terminal is dependent on GFRa1 and partially dependent on p140NCAM, suggesting the involvement of additional transmembrane signaling molecules (X). The
figure only describes the possibility that GDNF binding triggers a conformational change in GFRa1 molecules required for trans-homophilic binding. The alternative option
that GDNF could act as a bridge between the GFRa1 partners remains open and is not included in the scheme. For simplicity, the molecules are represented in monomeric
form. NtR = neurotransmitter receptors; SP = scaffolding proteins.

388
Opinion Trends in Neurosciences Vol.31 No.8

cell adhesion represents a new way to regulate cell–cell between neurons and their targets. Trans-synaptic
interaction in which the ligand–receptor system is the adhesion molecules influence synapses at different levels
actual mediator of the contact, giving a new level of regu- from the establishment of the synaptic contact and differ-
lation to the adhesive process (Figure 2a). The role of entiation of pre- and postsynaptic specialization to the
GFRa1 as a LICAM predicts that other GPI-linked or modulation of the function of synaptic channels and recep-
transmembrane receptors and their soluble ligands might tors [53,54]. Recent evidence indicates that, in the presence
induce cell adhesion working in a similar fashion. of GDNF, a localized source of exogenous GFRa1 that
The molecular bases of GDNF-induced trans-homophi- mimics its postsynaptic localization was able to induce
lic interactions between GFRa1 molecules are not clear presynaptic differentiation of hippocampal neurons. This
yet. The domains of GFRa1 underlying its LICAM activity effect was visualized by the clustering of vesicular proteins
have been studied using GFRa1 deletion mutants. This and neurotransmitter transporters and by activity-de-
study indicated that GFRa1-mediated cell adhesion pendent vesicle recycling. Similar results were observed
requires the presence of an intact GDNF binding domain in excitatory and inhibitory hippocampal neurons [19].
in both interacting GFRa1 partners [19]. Because GDNF is However, whether this is a common mechanism used by
a dimeric molecule with twofold symmetry, it could act as a other neuronal populations remains to be established.
bridge between the GFRa1 partners. Another possibility is Interestingly, presynaptic differentiation induced by
that GDNF binding triggers a conformational change in GDNF was independent of Ret and partially dependent
GFRa1 molecules resulting in the reorientation and expo- on NCAM, suggesting that additional effector molecules
sition of the determinants involved in the trans-homophilic are involved in this process. These results are consistent
binding. with learning deficits reported in Gdnf-, Gfra2- and Ncam-
The ability of GFRa1 to be released from the plasma mutant mice [55–57]. In agreement with this evidence,
membrane of neuronal and glial cells indicates an Gdnf-heterozygous mice showed a transient reduction in
additional way to control cell adhesion mediated by GPI- the synaptic localization of presynaptic proteins, indicat-
GFRa1. In agreement with this, GDNF- and GPI-GFRa- ing a physiological role for GDNF in hippocampal presyn-
induced cell adhesion can be disrupted in the presence of aptic assembly [19]. This transient defect could be
soluble GFRa1 [19]. Thus, ligand-induced trans-homophi- explained by the existence of either compensatory mech-
lic interactions between GFRa1 molecules represent a new anisms or the partial reduction in the amount of GDNF
way to regulate intercellular interactions that could have achieved in heterozygous mutants. Recently, Baudet and
important implications during the development of the coworkers have shown a role of Ret in NMJ synapse
nervous system, expanding the biological possibilities of formation, suggesting that GFLs and GFRa1 promote
the GDNF–GFRa1 signaling system. synaptogenesis functioning through different signal-trans-
ducing receptors along the nervous system [32].
Role of GDNF and GFRa receptors in synapse formation Because of the asymmetric nature of the synapse, the
During the last decade, numerous studies have shown that trans-synaptic interaction between GFRa1 molecules
GFLs participate in synapse development and plasticity might activate different signaling responses in pre- and
[2]. Enhancement of transmitter release promoted by postsynaptic compartments. However, the contribution of
GDNF from midbrain dopaminergic neurons and neuro- GDNF and GFRa1 to the postsynaptic density organiz-
muscular synapses led to the proposition that GDNF ation remains to be explored (Figure 2b).
regulates the establishment and functional properties of The function of the nervous system relies on the precise
synaptic terminals. An increase in the amount of presyn- establishment of neuronal connections, and aberrant con-
aptic vesicle clustering observed in midbrain dopaminergic nectivity can result in the development of nervous system
neurons indicated a role of GDNF in presynaptic differen- disorders. In particular, trans-synaptic adhesion molecules
tiation [50]. Yang and coworkers also demonstrated that have been associated with cognitive disorders such as
acute GDNF treatment modulates A-type K+ channels and autism, mental retardation and schizophrenia [53,54].
the excitability of these neurons in culture [51]. Further- Thus, elucidating the molecular basis underlying synapse
more, using nerve-muscle cocultures, it has been shown formation and maturation is critical to the understanding
that long-term application of GFLs induces a significant of the development of nervous system diseases and to
increase in the number and size of presynaptic vesicle designing therapeutic strategies for them. The ability of
aggregates as well as acetylcholine receptor (AChR) clus- GDNF to induce trans-homophilic interactions between
ters, indicating that GFLs are able to coordinate the de- GFRa1 molecules represents the first example of a new
velopment of neuromuscular synapses through both pre- synaptogenic mechanism that combines soluble and mem-
and postsynaptic mechanisms [52]. The temporal expres- brane-bound molecules with possible implications in cog-
sion pattern of GDNF and GFRa1 in the developing hippo- nitive disorders.
campus and the localization of GFRa1 to pre- and
postsynaptic membranes, together with the ability of this Conclusions and perspectives
receptor to function as a LICAM, have suggested that Many questions still remain unanswered. The historic
GDNF-GFRa1 complexes could act as synaptogenic factors question of why GFRas are more widely distributed than
by inducing adhesion between pre- and postsynaptic Ret is perhaps a key issue that remains to be addressed.
specializations (Figure 2b) [19]. Indeed, the physiological contribution of trans-signaling to
Neuronal synapses can be defined as a specialized type nervous system development and axon regeneration still
of cell–cell interaction that mediates the communication deserves additional analysis because the genetic models

389
Opinion Trends in Neurosciences Vol.31 No.8

currently available cannot completely eliminate the poten- system progenitor cells during mammalian embryogenesis.
Development 129, 5151–5160
tial compensatory effects of other GFRa molecules. It
6 Gianino, S. et al. (2003) GDNF availability determines enteric neuron
would also be of therapeutic value to find out whether number by controlling precursor proliferation. Development 130, 2187–
exogenous administration of soluble GFRa1 in combi- 2198
nation with GDNF could promote axonal regeneration 7 Young, H.M. et al. (2001) GDNF is a chemoattractant for enteric neural
after nerve injury. Furthermore, the identification of the cells. Dev. Biol. 229, 503–516
8 Taraviras, S. et al. (1999) Signalling by the RET receptor tyrosine
mechanism by which GPI-anchored GFRas are released
kinase and its role in the development of the mammalian enteric
could reveal additional therapeutic possibilities for nerve nervous system. Development 126, 2785–2797
regeneration. Animals genetically modified to express 9 Durbec, P. et al. (1996) GDNF signalling through the Ret receptor
GFRa1 exclusively in Ret-positive cells in GFRa1-null tyrosine kinase. Nature 381, 789–793
mice have represented a useful strategy to evaluate only 10 Trupp, M. et al. (1996) Functional receptor for GDNF encoded by the c-
ret proto-oncogene. Nature 381, 785–789
certain aspects of Ret-independent GFRa1 signaling 11 Jing, S. et al. (1996) GDNF-induced activation of the ret protein
during postnatal development [44]. Although the analysis tyrosine kinase is mediated by GDNFR-a, a novel receptor for
performed on these mice failed to detect structural GDNF. Cell 85, 1113–1124
abnormalities in the hippocampus, RMS and OB, it is 12 Treanor, J.J. et al. (1996) Characterization of a multicomponent
possible that functional deficits or morphological changes receptor for GDNF. Nature 382, 80–83
13 Sariola, H. and Saarma, M. (2003) Novel functions and signalling
in specific neuronal populations could be detected in these pathways for GDNF. J. Cell Sci. 116, 3855–3862
brain areas following a more detailed analysis. Only tissue- 14 Santoro, M. et al. (2004) RET: normal and abnormal functions.
specific or -inducible GFRa1 knockout alone or in combi- Endocrinology 145, 5448–5451
nation with mice deficient in other GFRas will allow us to 15 Worley, D.S. et al. (2000) Developmental regulation of GDNF response
and receptor expression in the enteric nervous system. Development
unequivocally determine the in vivo relevance of Ret-inde-
127, 4383–4393
pendent signaling during postnatal nervous system de- 16 Ledda, F. et al. (2002) Target-derived GFRa1 as an attractive guidance
velopment. A detailed morphological and functional signal for developing sensory and sympathetic axons via activation of
analysis of these mice, especially in the forebrain, would Cdk5. Neuron 36, 387–401
be essential to identify and dissect the physiological con- 17 Paratcha, G. et al. (2001) Released GFRa1 potentiates downstream
signaling, neuronal survival, and differentiation via a
tributions of GFLs and GFRa signaling through either
novel mechanism of recruitment of c-Ret to lipid rafts. Neuron 29,
NCAM or still unidentified signaling receptors. 171–184
From a therapeutic point of view, mice with specific 18 Paratcha, G. et al. (2006) GDNF is a chemoattractant factor for
deletion of Ret in the nigrostriatal system might be useful neuronal precursor cells in the rostral migratory stream. Mol. Cell.
for studying the molecular mechanisms involved in age- Neurosci. 31, 505–514
19 Ledda, F. et al. (2007) GDNF and GFRa1 promote formation of
related neurodegeneration, such as Parkinson’s disease neuronal synapses by ligand-induced cell adhesion. Nat. Neurosci.
[30]. The physiological requirement of Ret for the mainten- 10, 293–300
ance [30] and regeneration [58] of the dopaminergic system 20 Pozas, E. and Ibáñez, C.F. (2005) GDNF and GFRa1 promote
supports additional investigation toward optimizing the differentiation and tangential migration of cortical GABAergic
ongoing clinical trials for Parkinson’s disease using activa- neurons. Neuron 45, 701–713
21 Paratcha, G. et al. (2003) The neural cell adhesion molecule NCAM is
tors or reducing the action of endogenous antagonists of the an alternative signaling receptor for GDNF family ligands. Cell 113,
Ret signaling pathway. The huge complexity of GDNF- 867–879
GFRa signaling also argues for further characterization 22 Popsueva, A. et al. (2003) GDNF promotes tubulogenesis of GFRa1-
and dissection of the molecular mechanisms involved in expressing MDCK cells by Src-mediated phosphorylation of Met
receptor tyrosine kinase. J. Cell Biol. 161, 119–129
protection and/or regeneration of the nervous system.
23 Trupp, M. et al. (1999) Ret-dependent and -independent mechanisms of
glial cell line-derived neurotrophic factor signaling in neuronal cells. J.
Acknowledgements Biol. Chem. 274, 20885–20894
We thank Kimberly Dougherty and Julianna Kele Olovsson for critical 24 Poteryaev, D. et al. (1999) GDNF triggers a novel ret-independent Src
reading of this manuscript, and Sevgi Cakir for secretarial help. Work in kinase family-coupled signaling via a GPI-linked GDNF receptor a1.
the authors’ laboratory is funded by grants from the Swedish Medical FEBS Lett. 463, 63–66
Research Council, Parkinsonfonden, the Royal Swedish Academy of 25 Enomoto, H. et al. (1998) GFR a1-deficient mice have deficits in the
Science and the Karolinska Institute. F.L. and G.P. are supported by enteric nervous system and kidneys. Neuron 21, 317–324
assistant researcher positions from the Swedish Medical Research 26 Cacalano, G. et al. (1998) GFRa1 is an essential receptor component for
Council and the Karolinska Institute, respectively. The authors declare GDNF in the developing nervous system and kidney. Neuron 21, 53–62
that they have no competing financial interests. 27 Uesaka, T. et al. (2007) Conditional ablation of GFRa1 in
postmigratory enteric neurons triggers unconventional neuronal
References death in the colon and causes a Hirschsprung’s disease phenotype.
1 Airaksinen, M.S. et al. (1999) GDNF family neurotrophic factor Development 134, 2171–2181
signaling: four masters, one servant? Mol. Cell. Neurosci. 13, 313–325 28 Luo, W. et al. (2007) A hierarchical NGF signaling cascade controls Ret-
2 Airaksinen, M.S. and Saarma, M. (2002) The GDNF family: signalling, dependent and Ret-independent events during development of
biological functions and therapeutic value. Nat. Rev. Neurosci. 3, 383– nonpeptidergic DRG neurons. Neuron 54, 739–754
394 29 Kramer, E.R. et al. (2006) Cooperation between GDNF/Ret and
3 Lin, L.F. et al. (1993) GDNF: a glial cell line-derived neurotrophic factor ephrinA/EphA4 signals for motor-axon pathway selection in the
for midbrain dopaminergic neurons. Science 260, 1130–1132 limb. Neuron 50, 35–47
4 Henderson, C.E. et al. (1994) GDNF: a potent survival factor for 30 Kramer, E.R. et al. (2007) Absence of Ret signaling in mice causes
motoneurons present in peripheral nerve and muscle. Science 266, progressive and late degeneration of the nigrostriatal system. PLoS
1062–1064 Biol. 5, e39
5 Natarajan, D. et al. (2002) Requirement of signalling by receptor 31 Jain, S. et al. (2006) RET is dispensable for maintenance of midbrain
tyrosine kinase RET for the directed migration of enteric nervous dopaminergic neurons in adult mice. J. Neurosci. 26, 11230–11238

390
Opinion Trends in Neurosciences Vol.31 No.8

32 Baudet, C. et al. (2008) Retrograde signaling onto Ret during motor 46 Sakai, A. et al. (2007) Involvement of neural cell adhesion molecule
nerve terminal maturation. J. Neurosci. 28, 963–975 signaling in glial cell line-derived neurotrophic factor-induced
33 Lindholm, P. et al. (2007) Novel neurotrophic factor CDNF protects and analgesia in a rat model of neuropathic pain. Pain, DOI: 10.1016/
rescues midbrain dopamine neurons in vivo. Nature 448, 73–77 j.pain.2007.09.020 (http://www.elsevier.com/locate/jpain)
34 Mijatovic, J. et al. (2007) Constitutive Ret activity in knock-in multiple 47 Powell, E.M. et al. (2001) Hepatocyte growth factor/scatter factor is a
endocrine neoplasia type B mice induces profound elevation of brain motogen for interneurons migrating from the ventral to dorsal
dopamine concentration via enhanced synthesis and increases the telencephalon. Neuron 30, 79–89
number of TH-positive cells in the substantia nigra. J. Neurosci. 27, 48 Ginsberg, M.H. et al. (2005) Integrin regulation. Curr. Opin. Cell Biol.
4799–4809 17, 509–516
35 Nguyen, Q.T. et al. (1998) Hyperinnervation of neuromuscular 49 Gumbiner, B.M. (2005) Regulation of cadherin-mediated adhesion in
junctions caused by GDNF overexpression in muscle. Science 279, morphogenesis. Nat. Rev. Mol. Cell Biol. 6, 622–634
1725–1729 50 Bourque, M.J. and Trudeau, L.E. (2000) GDNF enhances the synaptic
36 Taketomi, T. et al. (2005) Loss of mammalian Sprouty2 leads to enteric efficacy of dopaminergic neurons in culture. Eur. J. Neurosci. 12, 3172–
neuronal hyperplasia and esophageal achalasia. Nat. Neurosci. 8, 855– 3180
857 51 Yang, F. et al. (2001) GDNF acutely modulates excitability and A-type
37 Ishida, M. et al. (2007) Sprouty2 regulates growth and differentiation of K(+) channels in midbrain dopaminergic neurons. Nat. Neurosci. 4,
human neuroblastoma cells through RET tyrosine kinase. Cancer Sci. 1071–1078
98, 815–821 52 Wang, C.Y. et al. (2002) Regulation of neuromuscular synapse
38 Ledda, F. et al. (2008) Lrig1 is an endogenous inhibitor of Ret receptor development by glial cell line-derived neurotrophic factor and
tyrosine kinase activation, downstream signaling, and biological neurturin. J. Biol. Chem. 277, 10614–10625
responses to GDNF. J. Neurosci. 28, 39–49 53 Dalva, M.B. et al. (2007) Cell adhesion molecules: signalling functions
39 Scott, R.P. et al. (2005) Distinct turnover of alternatively spliced isoforms at the synapse. Nat. Rev. Neurosci. 8, 206–220
of the RET kinase receptor mediated by differential recruitment of the 54 Scheiffele, P. (2003) Cell-cell signaling during synapse formation in the
Cbl ubiquitin ligase. J. Biol. Chem. 280, 13442–13449 CNS. Annu. Rev. Neurosci. 26, 485–508
40 Pierchala, B.A. et al. (2006) Glial cell line-derived neurotrophic factor- 55 Rutishauser, U. (2008) Polysialic acid in the plasticity of the developing
dependent recruitment of Ret into lipid rafts enhances signaling by and adult vertebrate nervous system. Nat. Rev. Neurosci. 9, 26–35
partitioning Ret from proteasome-dependent degradation. J. Neurosci. 56 Voikar, V. et al. (2004) Impaired behavioural flexibility and memory in
26, 2777–2787 mice lacking GDNF family receptor a2. Eur. J. Neurosci. 20, 308–312
41 Ledda, F. and Paratcha, G. (2007) Negative regulation of receptor 57 Gerlai, R. et al. (2001) Impaired water maze learning performance
tyrosine kinase (RTK) signaling: a developing field. Biomark. without altered dopaminergic function in mice heterozygous for the
Insights 2, 45–58 GDNF mutation. Eur. J. Neurosci. 14, 1153–1163
42 Sjostrand, D. et al. (2007) Disruption of the GDNF binding site in 58 Kowsky, S. et al. (2007) RET signaling does not modulate MPTP
NCAM dissociates ligand binding and homophilic cell adhesion. J. Biol. toxicity but is required for regeneration of dopaminergic axon
Chem. 282, 12734–12740 terminals. Proc. Natl. Acad. Sci. U. S. A. 104, 20049–20054
43 Gascon, E. et al. (2007) Polysialic acid-neural cell adhesion molecule in 59 Tansey, M.G. et al. (2000) GFRa-mediated localization of RET to lipid
brain plasticity: from synapses to integration of new neurons. Brain rafts is required for effective downstream signaling, differentiation,
Res. Rev. 56, 101–118 and neuronal survival. Neuron 25, 611–623
44 Enomoto, H. et al. (2004) GFRa1 expression in cells lacking RET is 60 Paratcha, G. and Ibanez, C.F. (2002) Lipid rafts and the control of
dispensable for organogenesis and nerve regeneration. Neuron 44, neurotrophic factor signaling in the nervous system: variations on a
623–636 theme. Curr. Opin. Neurobiol. 12, 542–549
45 Iwase, T. et al. (2005) Glial cell line-derived neurotrophic factor- 61 Rajendran, L. and Simons, K. (2005) Lipid rafts and membrane
induced signaling in Schwann cells. J. Neurochem. 94, 1488–1499 dynamics. J. Cell Sci. 118, 1099–1102

391

You might also like