9
REVIEW Wnt signalling in neuronal differentiation and development Nibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014 /Accepted: 25 August 2014 /Published online: 19 September 2014 # Springer-Verlag Berlin Heidelberg 2014 Abstract Wnts are secreted glycoproteins that play multiple roles in early development, including the differentiation of precursor cells. During this period, gradients of Wnts and other morphogens are formed and regulate the differentiation and migration of neural progenitor cells. Afterwards, Wnt signalling cascades participate in the formation of neuronal circuits, playing roles in dendrite and axon development, dendritic spine formation and synaptogenesis. Finally, in the adult brain, Wnts control hippocampal plasticity, regulating synaptic transmission and neurogenesis. In this review, we summarize the reported roles of Wnt signalling cascades in these processes with a particular emphasis on the role of Wnts in neuronal differentiation and development. Keywords Wnt signalling pathway . Neural progenitor cells . Neuronal differentiation . Neuronal maturation . Adult neurogenesis Introduction Wnts are secreted signalling molecules that activate signalling cascades involved in different aspects of embryonic develop- ment including cell fate specification, polarity and migration (Clevers and Nusse 2012; Nusse and Varmus 2012). The Wnt signalling pathway is activated by Wnt ligands, which are members of a family of 19 secreted glycoproteins in mammals that bind to seven-pass transmembrane Frizzled (FZD) recep- tors to activate different signalling cascades: the canonical Wnt/β-catenin signalling pathway and the non-canonical β- catenin-independent signalling cascades (Gordon and Nusse 2006) (Fig. 1). Importantly, the same ligand may activate different Wnt signalling cascades depending on the receptor context, and activation of a specific pathway may antagonize the activation of other pathways (Mikels and Nusse 2006). Canonical and non-canonical Wnt ligands can compete for binding to FZD receptors, thereby causing reciprocal pathway inhibition of both signalling pathways (Grumolato et al. 2010). In addition to FZDs, other membrane proteins have been identified as receptors or co-receptors of Wnts, including the low-density lipoprotein receptor-related protein 5 (LRP5), LRP6, receptor tyrosine kinase-like orphan receptor 1 (ROR1), ROR2 (Cadigan and Liu 2006; Gordon and Nusse 2006; Green et al. 2008), and the receptor-like tyrosine kinase Ryk that has been implicated in axon guidance in the develop- ing nervous system (Bovolenta et al. 2006; Fradkin et al. 2009). Soluble proteins are also important modulators of Wnts actions, where some of them, such as secreted FZD-related proteins (sFRPs), Dickkopf (DKK) proteins and Wnt inhibi- tory factor 1 (WIF1), act as inhibitors of the Wnt pathway (Cruciat and Niehrs 2013; Kawano and Kypta 2003; Niehrs 2006). There are also other physiological and pharmacologi- cal mechanisms that can activate the intracellular component of the canonical Wnt/β-catenin signalling cascade. For exam- ple, extracellular R-spondin (Rspo) proteins have been shown N. C. Inestrosa (*) Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 340, P. O. Box 114-D, Santiago, Chile e-mail: [email protected] N. C. Inestrosa Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia N. C. Inestrosa Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile L. Varela-Nallar (*) Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Republica 239, 8370146 Santiago, Chile e-mail: [email protected] Cell Tissue Res (2015) 359:215223 DOI 10.1007/s00441-014-1996-4

Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

REVIEW

Wnt signalling in neuronal differentiation and development

Nibaldo C. Inestrosa & Lorena Varela-Nallar

Received: 2 May 2014 /Accepted: 25 August 2014 /Published online: 19 September 2014# Springer-Verlag Berlin Heidelberg 2014

Abstract Wnts are secreted glycoproteins that play multipleroles in early development, including the differentiation ofprecursor cells. During this period, gradients of Wnts andother morphogens are formed and regulate the differentiationand migration of neural progenitor cells. Afterwards, Wntsignalling cascades participate in the formation of neuronalcircuits, playing roles in dendrite and axon development,dendritic spine formation and synaptogenesis. Finally, in theadult brain, Wnts control hippocampal plasticity, regulatingsynaptic transmission and neurogenesis. In this review, wesummarize the reported roles of Wnt signalling cascades inthese processes with a particular emphasis on the role of Wntsin neuronal differentiation and development.

Keywords Wnt signalling pathway . Neural progenitor cells .

Neuronal differentiation . Neuronal maturation . Adultneurogenesis

Introduction

Wnts are secreted signalling molecules that activate signallingcascades involved in different aspects of embryonic develop-ment including cell fate specification, polarity and migration(Clevers and Nusse 2012; Nusse and Varmus 2012). The Wntsignalling pathway is activated by Wnt ligands, which aremembers of a family of 19 secreted glycoproteins in mammalsthat bind to seven-pass transmembrane Frizzled (FZD) recep-tors to activate different signalling cascades: the canonicalWnt/β-catenin signalling pathway and the non-canonical β-catenin-independent signalling cascades (Gordon and Nusse2006) (Fig. 1). Importantly, the same ligand may activatedifferent Wnt signalling cascades depending on the receptorcontext, and activation of a specific pathway may antagonizethe activation of other pathways (Mikels and Nusse 2006).Canonical and non-canonical Wnt ligands can compete forbinding to FZD receptors, thereby causing reciprocal pathwayinhibition of both signalling pathways (Grumolato et al.2010). In addition to FZDs, other membrane proteins havebeen identified as receptors or co-receptors ofWnts, includingthe low-density lipoprotein receptor-related protein 5 (LRP5),LRP6, receptor tyrosine kinase-like orphan receptor 1(ROR1), ROR2 (Cadigan and Liu 2006; Gordon and Nusse2006; Green et al. 2008), and the receptor-like tyrosine kinaseRyk that has been implicated in axon guidance in the develop-ing nervous system (Bovolenta et al. 2006; Fradkin et al. 2009).

Soluble proteins are also important modulators of Wnt’sactions, where some of them, such as secreted FZD-relatedproteins (sFRPs), Dickkopf (DKK) proteins and Wnt inhibi-tory factor 1 (WIF1), act as inhibitors of the Wnt pathway(Cruciat and Niehrs 2013; Kawano and Kypta 2003; Niehrs2006). There are also other physiological and pharmacologi-cal mechanisms that can activate the intracellular componentof the canonical Wnt/β-catenin signalling cascade. For exam-ple, extracellular R-spondin (Rspo) proteins have been shown

N. C. Inestrosa (*)Centro de Envejecimiento y Regeneración (CARE), Departamentode Biología Celular y Molecular, Facultad de Ciencias Biológicas,Pontificia Universidad Católica de Chile, 340, P. O. Box 114-D,Santiago, Chilee-mail: [email protected]

N. C. InestrosaCenter for Healthy Brain Ageing, School of Psychiatry, Faculty ofMedicine, University of New South Wales, Sydney, Australia

N. C. InestrosaCentro de Excelencia en Biomedicina de Magallanes (CEBIMA),Universidad de Magallanes, Punta Arenas, Chile

L. Varela-Nallar (*)Center for Biomedical Research, Faculty of Biological Sciences andFaculty of Medicine, Universidad Andres Bello, Republica 239,8370146 Santiago, Chilee-mail: [email protected]

Cell Tissue Res (2015) 359:215–223DOI 10.1007/s00441-014-1996-4

Page 2: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

to bind LGR4, LGR5 and LGR6 G-protein coupled receptorsto physiologically activate this pathway (Carmon et al. 2011;Glinka et al. 2011). In addition, the long-standing pharmaco-logical agent lithium inhibits glycogen synthase kinase-3β(GSK-3β) intracellularly, thereby stabilizing β-catenin to ac-tivate the pathway independently of ligand–receptor interac-tions (Toledo and Inestrosa 2010; Valvezan and Klein 2012).

The activation of the canonical Wnt/β-catenin signallingcascade involves the formation of a Wnt–LRP–FZD complexthat activates the scaffold protein Dishevelled (Dvl) andcauses the dissociation of a multiprotein destruction complex,consisting of enzymes that phosphorylate and target β-catenin

for proteasomal degradation when the pathway has not beenactivated (Hart et al. 1999; Liu et al. 2002). Consequently, β-catenin accumulates in the cytoplasm and translocates to thenucleus where it interacts with members of the TCF/LEF1family of transcription factors to regulate the expression ofWnt target genes (Arrazola et al. 2009; Clevers and Nusse2012; Nusse and Varmus 2012). In the multiprotein destruc-tion complex, β-catenin is phosphorylated by casein kinase1α (CK1α) and GSK-3β which are associated with the scaf-fold protein Axin and adenomatous polyposis coli (APC)(Hart et al. 1998; Ikeda et al. 1998; Kishida et al. 1998;Sakanaka et al. 1998).

Fig. 1 The Wnt signalling pathway. Canonical and non-canonical sig-nalling cascades. Binding of a Wnt ligand to a Frizzled (FZD) receptoractivates different signalling cascades. In the canonical Wnt/β-cateninsignalling pathway (left) theWnt ligand also interacts with the co-receptorlow density lipoprotein-related protein 5 or 6 (LRP5/6) to activate theprotein Dishevelled (Dvl) and induce the stabilization of β-catenin bypreventing its phosphorylation by glycogen synthase kinase-3β (GSK-3β) in a multiprotein complex composed also of the scaffold protein Axinand adenomatous polyposis coli (APC). Consequently, β-catenin accu-mulates in the cytoplasm and enters the nucleus where it interacts with thetranscription factors TCF/LEF to induce the transcription of Wnt target

genes. In the non-canonical Wnt/Ca2+ signalling cascade (middle), acti-vation of the pathway triggers the activation of trimeric G proteins andsubsequently phospholipase C (PLC), increasing inositol triphosphate(IP3). IP3 induces the release of Ca2+ from the endoplasmic reticulum(ER), which then induces the activation of calcium/calmodulin-dependentprotein kinase II (CamKII) and protein kinase C (PKC). In the non-canonical Wnt/PCP signalling cascade (right), activation of the pathwayleads to the activation of the small GTPases Rho and Rac leading to theactivation of Rho-associated protein kinase (ROCK) and Jun N-terminalkinase (JNK) to regulate cytoskeleton dynamics

216 Cell Tissue Res (2015) 359:215–223

Page 3: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

The Wnt/FZD interaction can also activate the non-canonical Wnt/PCP or Wnt/Ca2+ pathways (Fig. 1). In theWnt/PCP pathway, the phosphorylation of Dvl causes theactivation of the small GTPases Rho and Rac and, conse-quently, the downstream Jun N-terminal kinase (JNK), whichregulates cytoskeleton dynamics (Gordon and Nusse 2006;Rosso and Inestrosa 2013; Rosso et al. 2005). In contrast, theWnt/Ca2+ pathway is almost exclusively a G-protein-dependent signalling pathway (Kohn and Moon 2005). Theactivation of the Wnt/Ca2+ pathway requires binding of Wntto FZD on the cell surface membrane to trigger stimulation ofheterotrimeric G-proteins (Slusarski et al. 1997a; Slusarskiet al. 1997b), which activate phospholipase-C (PLC). PLCcauses an increment in intracellular Ca2+ release that decreasescyclic guanosine monophosphate (cGMP) and activates theprotein kinases Ca2+/Calmodulin-dependent protein kinase II(CaMKII) and protein kinase-C (PKC) (Kohn and Moon2005; Montcouquiol et al. 2006; Veeman et al. 2003). Inaddition, it activates the transcription factor NFAT to inducethe transcription of target genes.

In the nervous system, Wnt signalling cascades are impor-tant for the formation of neuronal circuits. During develop-ment, Wnt signalling regulates self-renewal, maintenance anddifferentiation of neural progenitor cells (Hirabayashi et al.2004; Machon et al. 2007; Munji et al. 2011) and regulates thedevelopment of the cortex and hippocampus (Li and Pleasure2005; Machon et al. 2007). Wnts not only regulate earlyembryonic development but are also key regulators of lateembryonic and postnatal development of the central nervoussystem (CNS) (Inestrosa and Arenas 2010; Oliva et al. 2013).Various Wnt ligands regulate synaptic development and func-tion, including the formation and maturation of pre- andpostsynaptic sites and neurotransmission. Members of theWnt family of secreted signalling proteins are implicated inevery step of neural development. During vertebrate develop-ment, a Wnt signalling gradient that is high in the posteriorand low in the anterior is critical for the proper specification ofthe anterior–posterior axis of the neural plate (Kiecker andNiehrs 2001). In fact, several studies have demonstrated thatinhibition of Wnt signalling in the anterior, as well as theactivation of Wnt signalling in the posterior, is required forproper anterior-posterior patterning of the early CNS (Esteveet al. 2000; Glinka et al. 1998; Houart et al. 2002; Kazanskayaet al. 2000). During early development, after the neural plate isspecified, it invaginates to form the neural tube, a process thatis complete once the paired neural folds adhere at the dorsalmidline, and which is required for the development of thespinal cord and brain. Neural tube defects cause conditionslike spina bifida and anencephaly. It has been determined thatthe Wnt/PCP signalling pathway is relevant for neural tubeclosure and is involved in neural tube defects (Curtin et al.2003; Wen et al. 2010). Also, a role for the Wnt/β-cateninpathway in neural tube closure is indicated by the presence of

neural tube defects in mice with mutations in LRP6 (Carteret al. 2005) and Axin 1 (Perry et al. 1995), and in micecarrying ablated TCF–β-catenin interactions (Wu et al. 2012).

Neural progenitor cells that make up the neural tube pro-liferate, differentiate and migrate to form the many neuronalganglia, nuclei, and layers of the CNS. The Wnt/β-cateninsignalling pathway has been associated with both proliferationand specification of neural progenitor cells. It has been deter-mined that Wnt-1 and Wnt-3a ligands are expressed at thedorsal midline of the developing neural tube and are requiredfor the formation of brain structures; mutant analyses in miceshow that the midbrain is absent whenWnt-1 is not expressed(McMahon and Bradley 1990; Thomas and Capecchi 1990),and the hippocampus is absent when Wnt-3a is not expressed(Lee et al. 2000). The Wnt-3a mutant shows decreased celldivision within the hippocampal neuroepithelium; however,there remain small hippocampal subfields. This implies thatthe Wnt-3a ligand primarily regulates proliferation of hippo-campal neural precursor cells (Lee et al. 2000). Duringcorticogenesis, the levels of Wnt activity have been shownto be important in controlling the switch between proliferationand differentiation of progenitor cells (Bielen and Houart2014), which depends on β-catenin. Increased stabilizationof β-catenin decreases cell cycle exit in neural precursors,while decreased Wnt/β-catenin signalling is associated withdifferentiation (Chenn and Walsh 2002; Mutch et al. 2010).Later in development, Wnt/β-catenin signalling induces neu-ral differentiation. In cortical neural precursor cells, Wnt/β-catenin signalling inhibits the self-renewal capacity of progen-itors and promotes neuronal differentiation (Hirabayashi et al.2004). This effect on differentiation is stage-specific, since itis not observed in neural precursor cells at earlier develop-mental stages (Hirabayashi et al. 2004; Munji et al. 2011).Thus, Wnt signalling regulates both self-renewal and subse-quent cell fate specification of neuronal progeny, and theseeffects depend on transcriptional regulation mediated by theWnt/β-catenin signalling pathway. The effect on proliferationinvolves the transcriptional control of genes that regulate thecell cycle (reviewed in Niehrs and Acebron 2012) and neuro-nal differentiation involves the regulation of the expression ofproneural genes such as the neurogenin 1 gene (Hirabayashiet al. 2004).

The relevance of the Wnt signalling pathway in normalbrain function is supported by the fact that its deregulation hasbeen linked to different pathologies that affect the CNS,including mental disorders, mood disorders and neurodegen-erative diseases (De Ferrari and Moon 2006; Inestrosa et al.2012; Inestrosa and Varela-Nallar 2014; Lovestone et al.2007; Oliva et al. 2013). Interestingly, the Wnt signallingpathway shows neuroprotective properties in Alzheimer’sdisease (AD), a neurodegenerative illness characterized by aprogressive loss of cognitive abilities including learning andmemory (Ballard et al. 2011) that has been associated with an

Cell Tissue Res (2015) 359:215–223 217

Page 4: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

impairment in Wnt signalling (De Ferrari and Inestrosa 2000;Inestrosa and Arenas 2010). In cultured hippocampal neuronsand in hippocampal slices, Wnts show neuroprotective prop-erties against the toxicity of the amyloid-β (Aβ) peptide(Alvarez et al. 2004; Cerpa et al. 2010; Chacon et al. 2008;De Ferrari et al. 2003), which is associated with AD patho-genesis. This was supported by in vivo experiments showingthat the activation of the Wnt signalling pathway in a doubletransgenic mouse model of AD reduces spatial memory im-pairment and the histopathological hallmarks of the disease(Toledo and Inestrosa 2010), indicating that in vivo modula-tion of the Wnt signalling pathway may have therapeuticvalue in some pathological conditions.

Wnt signalling in neuronal maturation

Compelling evidence indicates that Wnt signalling cascadesare important for the regulation of several aspects of neuronaldevelopment. Neuronal polarization, axonal and dendriticdevelopment and synaptogenesis are crucial for the formationof neuronal circuits. Studies have shown that the Wnt signal-ling pathway is involved in all these steps of neuronal devel-opment. In cerebellar neurons, it has been shown that Wnt-7ainduced axon and growth cone remodelling (Hall et al. 2000;Lucas and Salinas 1997). Wnt-mediated changes in growthcone remodelling involve changes in microtubule organiza-tion through the regulation of APC (Purro et al. 2008).Wnt-5aalso regulates axonal behavior in sympathetic and corticalneurons (Bodmer et al. 2009; Li et al. 2009). Interestingly, incortical neurons, Wnt-5a stimulates axonal outgrowth andrepulsive axon guidance through different receptors. Axonaloutgrowth is mediated by the Ryk receptor, whereas axonalrepulsion requires both Ryk and FZD receptors (Li et al.2009). Recently, we determined that FZD5 regulates axondevelopment (Slater et al. 2013). Moreover, by gain- andloss-of-function experiments, we determined that FZD5 reg-ulates a very early event in neuronal development: the estab-lishment of neuronal polarity (Fig. 2). In cultured neurons,FZD5 shows a very polarized distribution being mainly pres-ent in the peripheral zone of growth cones. Overexpression ofFZD5 induces a loss of polarized distribution of the receptorand induces a mislocalization of axonal proteins, while FZD5knockdown induces a loss of axonal proteins (Slater et al.2013). These data suggest that FZD5 is important for neuronalpolarity. In addition, when the receptor is overexpressed afterthe acquisition of neuronal polarity, it does not revert polari-zation but alters neuronal morphogenesis by decreasing axo-nal length and increasing dendritic length and arborisation viaa JNK-dependent mechanism (Slater et al. 2013). Severalstudies have shown the involvement of Wnt signalling com-ponents in dendrite morphogenesis. In hippocampal neurons,β-catenin is a critical mediator of dendritic morphogenesis.

This effect of β-catenin is independent of gene transcriptionand is required for dendritic growth induced by depolarization(Yu and Malenka 2003). Another study demonstrated that, inhippocampal neurons, activation of NMDA receptors in-creases the expression of Wnt-2, which then stimulates den-dritic arborization (Wayman et al. 2006), indicating the in-volvement of Wnt in activity-dependent dendritic develop-ment. Wnt-7b also regulates dendritic arborization, and thiseffect is mediated by a non-canonical Wnt signalling cascadethat involves JNK activation (Rosso et al. 2005).

In addition to the participation in neuronal polarization andmorphogenesis, it is known that Wnt signalling regulates syn-apse formation and neurotransmission. Electrophysiologicalrecordings in rat hippocampal slices showed that a blockadeof Wnt signalling impairs long-term potentiation (LTP) (Chenet al. 2006), indicating the importance of this signalling path-way in synaptic plasticity (Vargas et al. 2014). Importantly, theexpression and release of Wnts are regulated by neuronalactivity (Chen et al. 2006; Wayman et al. 2006), supportingthe notion that these ligands may play a role during neuronaltransmission. Regarding the synaptic effects of Wnts, severalyears ago it was shown in cerebellar neurons that Wnt-7aregulates the clustering of the synaptic vesicle protein synapsinI (Hall et al. 2000; Lucas and Salinas 1997). The effect of Wntsignalling on presynaptic assembly has also been observed inhippocampal neurons, in which Wnt-7a, Wnt-3a and Wnt-7bincrease the presynaptic puncta and the synaptic vesicle cycle(Ahmad-Annuar et al. 2006; Cerpa et al. 2008; Davis et al.2008). Moreover, electrophysiological recordings on adult rathippocampal slices demonstrated that Wnt-7a increases neuro-transmitter release in CA3-CA1 synapses (Cerpa et al. 2008).The presynaptic effects of Wnts may be mediated by Wntreceptors located at the synaptic region. In hippocampal neu-rons, we determined that the Wnt receptor FZD1 is present atthe presynaptic site where it regulates the presynaptic assembly(Varela-Nallar et al. 2009). Also, FZD5 was shown to mediatesynaptogenesis induced by Wnt-7a (Sahores et al. 2010).These findings have demonstrated a relevant role for the Wntsignalling pathway in presynaptic differentiation and function.

In addition, non-canonical Wnt signalling cascades havebeen associated with the postsynaptic apparatus.Electrophysiological recordings showed that Wnt-5a in-creases the amplitude of field excitatory postsynaptic poten-tials (fEPSP) and upregulates synaptic NMDAR currents,facilitating the induction of LTP (Cerpa et al. 2010, 2011), atwo-step effect that is independently mediated by proteinkinase C (PKC) and JNK (Cerpa et al. 2011). At the structurallevel, Wnt-5a modulates postsynaptic assembly, increasingclustering of the postsynaptic density protein-95 (PSD-95)(Farias et al. 2009), which is a key scaffold protein of thepostsynaptic density.

TheWnt signalling pathway also modulates dendritic spinemorphogenesis in cultured hippocampal neurons, an early

218 Cell Tissue Res (2015) 359:215–223

Page 5: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

event in synapse formation that is crucial for synaptic plastic-ity. We observed that Wnt-5a treatment induces an increase indendritic protrusions that mature into dendritic spines (Varela-Nallar et al. 2010a). Interestingly, the effect on dendriticprotrusions was not observed by treatment with Wnt-3a, indi-cating that there is some level of specificity for this effect(Varela-Nallar et al. 2010a). Time-lapse imaging revealed thatWnt-5a induces the formation of new dendritic spines andincreases the size of preexisting ones (Varela-Nallar et al.2010a). Another ligand that regulates dendritic spines isWnt-7a, which was shown to increase the density andmaturityof dendritic spines through a mechanism involving CamKII(Ciani et al. 2011), suggesting that the non-canonical Wnt/Ca2+

signalling cascade is involved in this effect. Both Wnt-5a andWnt-7a increase intracellular Ca2+ concentration in neurons(Ciani et al. 2011; Varela-Nallar et al. 2010a), suggesting thata common Ca2+-dependent mechanism may be involved inthe Wnt-mediated regulation of dendritic spines. A potentialreceptor for the effect of Wnts on dendritic spines is ROR2,which is located in dendritic spines (Alfaro et al., unpublishedresults). We have determined that ROR2 is important for thematuration and maintenance of dendritic spines in hippocam-pal neurons (Fig. 3), and, interestingly, this receptor mediatedan inhibitory effect of Wnt-5a in a voltage-gated K+ current

(Parodi et al., unpublished results), indicating that this may bethe receptor or co-receptor mediating the described postsyn-aptic effects of Wnt-5a.

Wnt signalling in adult neurogenesis

In the adult brain, the generation of new neurons continuesmainly in two regions, the SVZ in the wall of the lateralventricles and the SGZ in the dentate gyrus of the hippocam-pus (Alvarez-Buylla and Garcia-Verdugo 2002; Zhao et al.2008). In these two neurogenic regions, neural stem cells(NSC) proliferate and differentiate into neuroblasts that inthe SVZ migrate through the rostral migratory stream to theolfactory bulb where they became interneurons, and in theSGZ mature into granule neurons that become integrated intothe granule cell layer (Ming and Song 2011; Zhao et al. 2008).

Many signalling cascades are involved in the regulation ofthe different steps of neurogenesis being important for theproper balance between the maintenance of the NSC pooland for the differentiation and maturation of newborn neurons(Faigle and Song 2013; Schwarz et al. 2012; Suh et al. 2009).During recent years, increasing evidence has supported a rolefor the Wnt/β-catenin pathways in the regulation of adult

Fig. 2 FZD5 in neuronal polarityand morphogenesis. FZD5 islocated in the peripheral zone ofaxonal growth cones. Gain- andloss-of-function experimentsdemonstrate that FZD5 isimportant for neuronal polarity.Overexpression of FZD5 inneurons that are already polarizeddecrease the total length of axons,a process partially prevented byinhibition of JNK, suggesting theWnt/PCP pathway modulatesaxon behavior

Cell Tissue Res (2015) 359:215–223 219

Page 6: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

neurogenesis (Varela-Nallar and Inestrosa 2013). In 2005, thegroup of Fred Gage determined in in vivo experiments thatoverexpression of Wnt-3 in the dentate gyrus increases cellproliferation and the generation of new neurons, while inhibi-tion of the Wnt signalling pathway reduces proliferation andneurogenesis (Lie et al. 2005). In that study, it was shown thatWnts derived from hippocampal astrocytes induce the differ-entiation of rat adult hippocampal progenitors (AHPs) and thatAHPs express Wnt signalling components. Later on, anotherstudy demonstrated in vitro that rat AHPs express several Wntligands and receptors, and also that there is an autocrine Wntstimulation that supports the proliferation and multipotency ofAHPs, and therefore it may be relevant for NSC maintenance(Wexler et al. 2009). More recently, it was shown that Wnt-7ais important for multiple steps of adult neurogenesis includingNSC self-renewal, neural progenitor cell proliferation, neuro-nal differentiation and maturation (Qu et al. 2013). Theseeffects of Wnt-7a involve the regulation of cyclin D1 andneurogenin 2 genes, involved in cell cycle control and neuro-nal differentiation, respectively (Qu et al. 2013).

In addition to Wnts, multiple components and regulators ofthe Wnt signalling pathway have been associated with adultneurogenesis. GSK-3β is involved in the regulation ofneurogenesis mediated by disrupted in schizophrenia 1(DISC1) (Inestrosa et al. 2012). This protein inhibits GSK-3β,and the impairment in progenitor cell proliferation caused byDISC1 knock-down can be rescued by overexpression of stabi-lizedβ-catenin (Mao et al. 2009) In vivo, the impairments in cellproliferation observed in the adult dentate gyrus by DISC1 lossof function can be rescued by administration of a GSK-3βinhibitor (Mao et al. 2009). In the SVZ, retrovirus-mediatedexpression of a stabilized β-catenin induces the proliferation oftype C cells (Adachi et al. 2007), and expression of Axin, a partof the protein complex that mediates the phosphorylation anddegradation of β-catenin, decreases cell proliferation (Qu et al.2010). Taken together, this evidence supports a role for thecanonical Wnt signalling pathway in NSC proliferation.

Soluble Wnt inhibitors have also been associated with adultneurogenesis. Dkk1 and sFRP3 are negative regulators of adultneurogenesis. Dkk1 prevents the activation of the canonicalWnt/β-catenin signalling cascade by preventing the formationof the FZD/LRP complex, while sFRP3 binds to Wntspreventing their interaction with their receptors (Cruciat andNiehrs 2013). Deletion of Dkk1 and sFRP3 increaseneurogenesis and dendrite complexity of newborn neurons(Jang et al. 2013; Seib et al. 2013). Interestingly, both Wntinhibitors are regulated by physiological stimuli that regulateneurogenesis, suggesting a physiological role for both proteinsand for the Wnt signalling pathway in the regulation ofneurogenesis in the adult brain. A reduction in sFRP3 is ob-served during exercise and electroconvulsive stimulation, sug-gesting that this reduction may be important for the increase inneurogenesis observed in both conditions (Jang et al. 2013). Onthe other hand, the expression of Dkk1 is increased duringaging (Seib et al. 2013), suggesting that it may be involved inthe decline of neurogenesis observed in different species duringlifespan (Knoth et al. 2010; Kuhn et al. 1996; Leuner et al.2007; Varela-Nallar et al. 2010b). During aging, a decline in thelevels of Wnt-3 and Wnt-3a in the dentate gyrus was alsoreported (Okamoto et al. 2011), which may also underlie thedecline in neurogenesis. In addition, canonicalWnt signalling isincreased in vivo by exposure to chronic hypoxia, concomi-tantly with an increase in cell proliferation and neurogenesis inthe SGZ of adult mice (Varela-Nallar et al. 2014). This suggeststhat the Wnt signalling pathway may also be involved in theeffect of mild hypoxia on neurogenesis.

The mechanisms involved in the regulation of adultneurogenesis by the Wnt signalling pathway may involvethe transcription of proneural Wnt target genes. It has beendetermined that the expression of the transcription factorsNeuroD1 and Prox1 is regulated by the canonical Wnt signal-ling pathway (Karalay et al. 2011; Kuwabara et al. 2009).NeuroD1 is important for neurogenesis in the SGZ and SVZ

Figure 3 ROR2 is involved in dendritic spine development. Wnt ligandsregulate spinogenesis in hippocampal neurons through activation of non-canonical Wnt pathways. Recently the tyrosine kinase receptor ROR2,which is located in dendritic spines, has been implicated in this process(unpublished results). In dendritic spines, ROR2 may act either as areceptor or co-receptor of FZD to activate non-canonical signallingpathways

220 Cell Tissue Res (2015) 359:215–223

Page 7: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

in the adult brain (Gao et al. 2009), and Prox1 is important forthe differentiation and survival of newborn granule cells(Karalay et al. 2011). Both transcription factors identified asWnt target genes might be important for the positive effect ofthe Wnt signalling pathway in adult neurogenesis.

Concluding remarks

The Wnt signalling pathway plays pivotal roles during theformation, maintenance and function of the CNS, having awide range of functions that include neuronal differentiation,development and maturation. There are many Wnt proteinsand severalWnt receptors and co-receptors that in conjunctionpaint a complex picture that might be relevant for the fineregulation of neuronal circuit formation and functioning.More recent evidence has shown that the Wnt signallingpathway also regulates the formation of new neurons in theadult brain, where it regulates the proliferation and differenti-ation of neural progenitor cells. Considering all the neuronalprocesses that are regulated by the Wnt signalling pathway inbrain neurons, it can be expected that additional roles for thispathway in the development, maturation and integration ofadult-born neurons might be unveiled in the near future.

We thank Felipe G. Serrano (CARE, Department of Cell and MolecularBiology, P. Catholic University of Chile) for artwork. This work wassupported by Grants from FONDECYT (No. 1120156) and the BasalCenter of Excellence in Science and Technology (CONICYT-PFB12/2007) to N.C.I. and by a Grant from FONDECYT (No. 11110012) toL.V.-N.

References

Adachi K, Mirzadeh Z, Sakaguchi M, Yamashita T, Nikolcheva T, GotohY, Peltz G, Gong L, Kawase T, Alvarez-Buylla A, Okano H,Sawamoto K (2007) Beta-catenin signaling promotes proliferationof progenitor cells in the adult mouse subventricular zone. StemCells 25:2827–2836

Ahmad-Annuar A, Ciani L, Simeonidis I, Herreros J, Fredj NB, RossoSB, Hall A, Brickley S, Salinas PC (2006) Signaling across thesynapse: a role forWnt andDishevelled in presynaptic assembly andneurotransmitter release. J Cell Biol 174:127–139

Alvarez AR, Godoy JA, Mullendorff K, Olivares GH, BronfmanM, Inestrosa NC (2004) Wnt-3a overcomes beta-amyloidtoxicity in rat hippocampal neurons. Exp Cell Res 297:186–196

Alvarez-Buylla A, Garcia-Verdugo JM (2002) Neurogenesis in adultsubventricular zone. J Neurosci 22:629–634

Arrazola MS, Varela-Nallar L, Colombres M, Toledo EM, Cruzat F,Pavez L, Assar R, Aravena A, Gonzalez M, Montecino M,Maass A, Martinez S, Inestrosa NC (2009) Calcium/calmodulin-dependent protein kinase type IV is a target geneof the Wnt/beta-catenin signaling pathway. J Cell Physiol 221:658–667

Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E (2011)Alzheimer’s disease. Lancet 377:1019–1031

Bielen H, Houart C (2014) The Wnt cries many: Wnt regulation ofneurogenesis through tissue patterning, proliferation, and asymmet-ric cell division. Neurobiol Dev (in press)

Bodmer D, Levine-Wilkinson S, Richmond A, Hirsh S, Kuruvilla R(2009) Wnt5a mediates nerve growth factor-dependent axonalbranching and growth in developing sympathetic neurons. JNeurosci 29:7569–7581

Bovolenta P, Rodriguez J, Esteve P (2006) Frizzled/RYK mediatedsignalling in axon guidance. Development 133:4399–4408

Cadigan KM, Liu YI (2006) Wnt signaling: complexity at the surface. JCell Sci 119:395–402

Carmon KS, Gong X, Lin Q, Thomas A, Liu Q (2011) R-spondinsfunction as ligands of the orphan receptors LGR4 and LGR5 toregulate Wnt/beta-catenin signaling. Proc Natl Acad Sci U S A 108:11452–11457

Carter M, Chen X, Slowinska B, Minnerath S, Glickstein S, Shi L,Campagne F, Weinstein H, Ross ME (2005) Crooked tail (Cd)model of human folate-responsive neural tube defects is mutatedin Wnt coreceptor lipoprotein receptor-related protein 6. Proc NatlAcad Sci U S A 102:12843–12848

Cerpa W, Farias GG, Godoy JA, Fuenzalida M, Bonansco C, InestrosaNC (2010) Wnt-5a occludes Abeta oligomer-induced depression ofglutamatergic transmission in hippocampal neurons. MolNeurodegener 5:3

Cerpa W, Gambrill A, Inestrosa NC, Barria A (2011) Regulation ofNMDA-receptor synaptic transmission by Wnt signaling. JNeurosci 31:9466–9471

Cerpa W, Godoy JA, Alfaro I, Farias GG, Metcalfe MJ, Fuentealba R,Bonansco C, Inestrosa NC (2008) Wnt-7a modulates the synapticvesicle cycle and synaptic transmission in hippocampal neurons. JBiol Chem 283:5918–5927

Chacon MA, Varela-Nallar L, Inestrosa NC (2008) Frizzled-1 is involvedin the neuroprotective effect of Wnt3a against Abeta oligomers. JCell Physiol 217:215–227

Chen J, Park CS, Tang SJ (2006) Activity-dependent synaptic Wntrelease regulates hippocampal long term potentiation. J Biol Chem281:11910–11916

ChennA,Walsh CA (2002) Regulation of cerebral cortical size by controlof cell cycle exit in neural precursors. Science 297:365–369

Ciani L, Boyle KA, Dickins E, Sahores M, Anane D, Lopes DM, GibbAJ, Salinas PC (2011) Wnt7a signaling promotes dendritic spinegrowth and synaptic strength through Ca2+/Calmodulin-dependentprotein kinase II. Proc Natl Acad Sci U S A 108:10732–10737

Clevers H, Nusse R (2012) Wnt/beta-catenin signaling and disease. Cell149:1192–1205

Cruciat CM, Niehrs C (2013) Secreted and transmembrane wnt inhibitorsand activators. Cold Spring Harb Perspect Biol 5:a015081

Curtin JA, Quint E, Tsipouri V, Arkell RM, Cattanach B, Copp AJ,Henderson DJ, Spurr N, Stanier P, Fisher EM, Nolan PM, SteelKP, Brown SD, Gray IC, Murdoch JN (2003) Mutation of Celsr1disrupts planar polarity of inner ear hair cells and causes severeneural tube defects in the mouse. Curr Biol 13:1129–1133

Davis EK, Zou Y, Ghosh A (2008) Wnts acting through canonical andnoncanonical signaling pathways exert opposite effects on hippo-campal synapse formation. Neural Dev 3:32

De Ferrari GV, Chacon MA, Barria MI, Garrido JL, Godoy JA, OlivaresG, Reyes AE, Alvarez A, Bronfman M, Inestrosa NC (2003)Activation of Wnt signaling rescues neurodegeneration and behav-ioral impairments induced by beta-amyloid fibrils. Mol Psychiatry8:195–208

De Ferrari GV, Inestrosa NC (2000) Wnt signaling function inAlzheimer’s disease. Brain Res Brain Res Rev 33:1–12

De Ferrari GV, Moon RT (2006) The ups and downs of Wnt signaling inprevalent neurological disorders. Oncogene 25:7545–7553

Esteve P,Morcillo J, Bovolenta P (2000) Early and dynamic expression ofcSfrp1 during chick embryo development. Mech Dev 97:217–221

Cell Tissue Res (2015) 359:215–223 221

Page 8: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

Faigle R, Song H (2013) Signaling mechanisms regulating adultneural stem cells and neurogenesis. Biochim Biophys Acta1830:2435–2448

Farias GG, Alfaro IE, Cerpa W, Grabowski CP, Godoy JA, Bonansco C,Inestrosa NC (2009) Wnt-5a/JNK signaling promotes the clusteringof PSD-95 in hippocampal neurons. J Biol Chem 284:15857–15866

Fradkin LG, Dura JM, Noordermeer JN (2009) Ryks: new partners forWnts in the developing and regenerating nervous system. TrendsNeurosci 33:84–92

Gao Z, Ure K, Ables JL, Lagace DC, Nave KA, Goebbels S, Eisch AJ,Hsieh J (2009) Neurod1 is essential for the survival and maturationof adult-born neurons. Nat Neurosci 12:1090–1092

Glinka A, Dolde C, Kirsch N, Huang YL, Kazanskaya O, Ingelfinger D,Boutros M, Cruciat CM, Niehrs C (2011) LGR4 and LGR5 are R-spondin receptors mediating Wnt/beta-catenin and Wnt/PCP signal-ling. EMBO Rep 12:1055–1061

Glinka A, Wu W, Delius H, Monaghan AP, Blumenstock C, Niehrs C(1998) Dickkopf-1 is a member of a new family of secreted proteinsand functions in head induction. Nature 391:357–362

GordonMD, Nusse R (2006) Wnt signaling: multiple pathways, multiplereceptors, and multiple transcription factors. J Biol Chem 281:22429–22433

Green JL, Kuntz SG, Sternberg PW (2008) Ror receptor tyrosine kinases:orphans no more. Trends Cell Biol 18:536–544

Grumolato L, Liu G, Mong P, Mudbhary R, Biswas R, Arroyave R,Vijayakumar S, Economides AN, Aaronson SA (2010) Canonicaland noncanonical Wnts use a common mechanism to activatecompletely unrelated coreceptors. Genes Dev 24:2517–2530

Hall AC, Lucas FR, Salinas PC (2000) Axonal remodeling and synapticdifferentiation in the cerebellum is regulated by WNT-7a signaling.Cell 100:525–535

Hart MJ, de los Santos R, Albert IN, Rubinfeld B, Polakis P (1998)Downregulation of beta-catenin by human Axin and its associationwith the APC tumor suppressor, beta-catenin and GSK3 beta. CurrBiol 8:573–581

Hart M, Concordet JP, Lassot I, Albert I, del los Santos R, Durand H,Perret C, Rubinfeld B, Margottin F, Benarous R, Polakis P (1999)The F-box protein beta-TrCP associates with phosphorylated beta-catenin and regulates its activity in the cell. Curr Biol 9:207–210

Hirabayashi Y, Itoh Y, Tabata H, Nakajima K, Akiyama T, Masuyama N,Gotoh Y (2004) The Wnt/beta-catenin pathway directs neuronaldifferentiation of cortical neural precursor cells. Development 131:2791–2801

Houart C, Caneparo L, Heisenberg C, Barth K, Take-Uchi M, Wilson S(2002) Establishment of the telencephalon during gastrulation bylocal antagonism of Wnt signaling. Neuron 35:255–265

Ikeda S, Kishida S, Yamamoto H,Murai H, Koyama S, Kikuchi A (1998)Axin, a negative regulator of the Wnt signaling pathway, forms acomplex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J 17:1371–1384

Inestrosa NC, Arenas E (2010) Emerging roles of Wnts in the adultnervous system. Nat Rev Neurosci 11:77–86

Inestrosa NC, Montecinos-Oliva C, Fuenzalida M (2012) Wnt signaling:role in Alzheimer disease and schizophrenia. J NeuroimmunePharmacol 7:788–807

Inestrosa NC, Varela-Nallar L (2014) Wnt signaling in the nervoussystem and in Alzheimer’s disease. J Mol Cell Biol 6:64–74

Jang MH, Bonaguidi MA, Kitabatake Y, Sun J, Song J, Kang E, Jun H,Zhong C, Su Y, Guo JU, Wang MX, Sailor KA, Kim JY, Gao Y,Christian KM, Ming GL, Song H (2013) Secreted frizzled-relatedprotein 3 regulates activity-dependent adult hippocampalneurogenesis. Cell Stem Cell 12:215–223

Karalay O, Doberauer K, Vadodaria KC, Knobloch M, Berti L,Miquelajauregui A, Schwark M, Jagasia R, Taketo MM,Tarabykin V, Lie DC, Jessberger S (2011) Prospero-related

homeobox 1 gene (Prox1) is regulated by canonical Wnt signalingand has a stage-specific role in adult hippocampal neurogenesis.Proc Natl Acad Sci U S A 108:5807–5812

Kawano Y, Kypta R (2003) Secreted antagonists of the Wnt signallingpathway. J Cell Sci 116:2627–2634

Kazanskaya O, Glinka A, Niehrs C (2000) The role of Xenopusdickkopf1 in prechordal plate specification and neural patterning.Development 127:4981–4992

Kiecker C, Niehrs C (2001) A morphogen gradient of Wnt/beta-cateninsignalling regulates anteroposterior neural patterning in Xenopus.Development 128:4189–4201

Kishida S, Yamamoto H, Ikeda S, Kishida M, Sakamoto I, Koyama S,Kikuchi A (1998) Axin, a negative regulator of the wnt signalingpathway, directly interacts with adenomatous polyposis coli andregulates the stabilization of beta-catenin. J Biol Chem 273:10823–10826

Knoth R, Singec I, Ditter M, Pantazis G, Capetian P, Meyer RP, Horvat V,Volk B, Kempermann G (2010) Murine features of neurogenesis inthe human hippocampus across the lifespan from 0 to 100 years.PLoS ONE 5:e8809

Kohn AD, Moon RT (2005) Wnt and calcium signaling: beta-catenin-independent pathways. Cell Calcium 38:439–446

Kuhn HG, Dickinson-Anson H, Gage FH (1996) Neurogenesis in thedentate gyrus of the adult rat: age-related decrease of neuronalprogenitor proliferation. J Neurosci 16:2027–2033

Kuwabara T, Hsieh J, Muotri A, Yeo G,Warashina M, Lie DC, Moore L,Nakashima K, Asashima M, Gage FH (2009) Wnt-mediated activa-tion of NeuroD1 and retro-elements during adult neurogenesis. NatNeurosci 12:1097–1105

Lee SM, Tole S, Grove E, McMahon AP (2000) A local Wnt-3a signal isrequired for development of the mammalian hippocampus.Development 127:457–467

Leuner B, Kozorovitskiy Y, Gross CG, Gould E (2007) Diminished adultneurogenesis in the marmoset brain precedes old age. Proc NatlAcad Sci U S A 104:17169–17173

Li G, Pleasure SJ (2005) Morphogenesis of the dentate gyrus: what weare learning from mouse mutants. Dev Neurosci 27:93–99

Li L, Hutchins BI, Kalil K (2009) Wnt5a induces simultaneous corticalaxon outgrowth and repulsive axon guidance through distinct sig-naling mechanisms. J Neurosci 29:5873–5883

Lie DC, Colamarino SA, Song HJ, Desire L, Mira H, Consiglio A, LeinES, Jessberger S, Lansford H, Dearie AR, Gage FH (2005) Wntsignalling regulates adult hippocampal neurogenesis. Nature 437:1370–1375

Liu C, Li Y, SemenovM, Han C, Baeg GH, Tan Y, Zhang Z, Lin X, He X(2002) Control of beta-catenin phosphorylation/degradation by adual-kinase mechanism. Cell 108:837–847

Lovestone S, Killick R, Di Forti M, Murray R (2007) Schizophrenia as aGSK-3 dysregulation disorder. Trends Neurosci 30:142–149

Lucas FR, Salinas PC (1997) WNT-7a induces axonal remodeling andincreases synapsin I levels in cerebellar neurons. Dev Biol 192:31–44

MachonO, BackmanM,Machonova O, Kozmik Z, Vacik T, Andersen L,Krauss S (2007) A dynamic gradient of Wnt signaling controlsinitiation of neurogenesis in the mammalian cortex and cellularspecification in the hippocampus. Dev Biol 311:223–237

Mao Y, Ge X, Frank CL, Madison JM, Koehler AN, Doud MK, Tassa C,Berry EM, Soda T, Singh KK, Biechele T, Petryshen TL, Moon RT,Haggarty SJ, Tsai LH (2009) Disrupted in schizophrenia 1 regulatesneuronal progenitor proliferation via modulation of GSK3beta/beta-catenin signaling. Cell 136:1017–1031

McMahon AP, Bradley A (1990) The Wnt-1 (int-1) proto-oncogene isrequired for development of a large region of the mouse brain. Cell62:1073–1085

Mikels AJ, Nusse R (2006) Purified Wnt5a protein activates or inhibitsbeta-catenin-TCF signaling depending on receptor context. PLoSBiol 4:e115

222 Cell Tissue Res (2015) 359:215–223

Page 9: Wnt signalling in neuronal differentiation and developmentNibaldo C. Inestrosa & Lorena Varela-Nallar Received: 2 May 2014/Accepted: 25 August 2014/Published online: 19 September 2014

Ming GL, Song H (2011) Adult neurogenesis in the mammalian brain:significant answers and significant questions. Neuron 70:687–702

Montcouquiol M, Crenshaw EB 3rd, Kelley MW (2006) NoncanonicalWnt signaling and neural polarity. Annu Rev Neurosci 29:363–386

Munji RN, Choe Y, Li G, Siegenthaler JA, Pleasure SJ (2011) Wntsignaling regulates neuronal differentiation of cortical intermediateprogenitors. J Neurosci 31:1676–1687

Mutch CA, Schulte JD, Olson E, Chenn A (2010) Beta-catenin signalingnegatively regulates intermediate progenitor population numbers inthe developing cortex. PLoS ONE 5:e12376

Niehrs C (2006) Function and biological roles of the Dickkopf family ofWnt modulators. Oncogene 25:7469–7481

Niehrs C, Acebron SP (2012) Mitotic and mitogenic Wnt signalling.EMBO J 31:2705–2713

Nusse R, VarmusH (2012) Three decades ofWnts: a personal perspectiveon how a scientific field developed. EMBO J 31:2670–2684

Okamoto M, Inoue K, Iwamura H, Terashima K, Soya H, AsashimaM, Kuwabara T (2011) Reduction in paracrine Wnt3 factorsduring aging causes impaired adult neurogenesis. FASEB J 25:3570–3582

Oliva CA, Vargas JY, Inestrosa NC (2013) Wnt signaling: role in LTP,neural networks and memory. Ageing Res Rev 12:786–800

Perry WL 3rd, Vasicek TJ, Lee JJ, Rossi JM, Zeng L, Zhang T, TilghmanSM, Costantini F (1995) Phenotypic and molecular analysis of atransgenic insertional allele of the mouse Fused locus. Genetics 141:321–332

Purro SA, Ciani L, Hoyos-Flight M, Stamatakou E, Siomou E, SalinasPC (2008) Wnt regulates axon behavior through changes in micro-tubule growth directionality: a new role for adenomatous polyposiscoli. J Neurosci 28:8644–8654

Qu Q, Sun G, Li W, Yang S, Ye P, Zhao C, Yu RT, Gage FH, Evans RM,Shi Y (2010) Orphan nuclear receptor TLX activates Wnt/beta-catenin signalling to stimulate neural stem cell proliferation andself-renewal. Nat Cell Biol 12:31-40; sup pp 31-39

Qu Q, Sun G, Murai K, Ye P, Li W, Asuelime G, Cheung YT, Shi Y(2013) Wnt7a regulates multiple steps of neurogenesis. Mol CellBiol 33:2551–2559

Rosso SB, Inestrosa NC (2013) WNT signaling in neuronal maturationand synaptogenesis. Front Cell Neurosci 7:103

Rosso SB, Sussman D, Wynshaw-Boris A, Salinas PC (2005) Wntsignaling through Dishevelled, Rac and JNK regulates dendriticdevelopment. Nat Neurosci 8:34–42

Sahores M, Gibb A, Salinas PC (2010) Frizzled-5, a receptor for thesynaptic organizer Wnt7a, regulates activity-mediated synaptogen-esis. Development 137:2215–2225

Sakanaka C, Weiss JB, Williams LT (1998) Bridging of beta-catenin andglycogen synthase kinase-3beta by axin and inhibition of beta-catenin-mediated transcription. Proc Natl Acad Sci U S A 95:3020–3023

Schwarz TJ, Ebert B, Lie DC (2012) Stem cell maintenance in the adultmammalian hippocampus: a matter of signal integration? DevNeurobiol 72:1006–1015

Seib DR, Corsini NS, Ellwanger K, Plaas C, Mateos A, Pitzer C, NiehrsC, Celikel T, Martin-Villalba A (2013) Loss of Dickkopf-1 restoresneurogenesis in old age and counteracts cognitive decline. Cell StemCell 12:204–214

Slater PG, Ramirez VT, Gonzalez-Billault C, Varela-Nallar L, InestrosaNC (2013) Frizzled-5 receptor is involved in neuronal polarity andmorphogenesis of hippocampal neurons. PLoS ONE 8:e78892

Slusarski DC, Corces VG, Moon RT (1997a) Interaction of Wnt and aFrizzled homologue triggers G-protein-linked phosphatidylinositolsignalling. Nature 390:410–413

Slusarski DC, Yang-Snyder J, Busa WB, Moon RT (1997b) Modulationof embryonic intracellular Ca2+ signaling by Wnt-5A. Dev Biol182:114–120

Suh H, Deng W, Gage FH (2009) Signaling in adult neurogenesis. AnnuRev Cell Dev Biol 25:253–275

Thomas KR, CapecchiMR (1990) Targeted disruption of themurine int-1proto-oncogene resulting in severe abnormalities in midbrain andcerebellar development. Nature 346:847–850

Toledo EM, Inestrosa NC (2010) Activation of Wnt signaling by lithiumand rosiglitazone reduced spatial memory impairment and neurode-generation in brains of an APPswe/PSEN1DeltaE9 mouse model ofAlzheimer’s disease. Mol Psychiatry 15:272–285

Valvezan AJ, Klein PS (2012) GSK-3 andWnt Signaling inNeurogenesisand Bipolar Disorder. Front Mol Neurosci 5:1

Varela-Nallar L, Alfaro IE, Serrano FG, Parodi J, Inestrosa NC (2010a)Wingless-type family member 5A (Wnt-5a) stimulates synapticdifferentiation and function of glutamatergic synapses. Proc NatlAcad Sci U S A 107:21164–21169

Varela-Nallar L, Aranguiz FC, Abbott AC, Slater PG, Inestrosa NC(2010b) Adult hippocampal neurogenesis in aging andAlzheimer’s disease. Birth Defects Res C 90:284–296

Varela-Nallar L, Grabowski CP, Alfaro IE, Alvarez AR, Inestrosa NC(2009) Role of the Wnt receptor Frizzled-1 in presynaptic differen-tiation and function. Neural Dev 4:41

Varela-Nallar L, Inestrosa NC (2013) Wnt signaling in the regulation ofadult hippocampal neurogenesis. Front Cell Neurosci 7:100

Varela-Nallar L, Rojas-Abalos M, Abbott AC, Moya EA, Iturriaga R,Inestrosa NC (2014) Chronic hypoxia induces the activation of theWnt/beta-catenin signaling pathway and stimulates hippocampalneurogenesis in wild-type and APPswe-PS1DeltaE9 transgenicmice in vivo. Front Cell Neurosci 8:17

Vargas JY, Fuenzalida M, Inestrosa NC (2014) In vivo activation of Wntsignaling pathway enhances cognitive function of adult mice andreverses cognitive deficits in an Alzheimer’s disease model. JNeurosci 34:2191–2202

Veeman MT, Axelrod JD, Moon RT (2003) A second canon. Functionsand mechanisms of beta-catenin-independent Wnt signaling. DevCell 5:367–377

Wayman GA, Impey S, Marks D, Saneyoshi T, Grant WF, Derkach V,Soderling TR (2006) Activity-dependent dendritic arborization me-diated by CaM-kinase I activation and enhanced CREB-dependenttranscription of Wnt-2. Neuron 50:897–909

Wen S, Zhu H, Lu W, Mitchell LE, Shaw GM, Lammer EJ, Finnell RH(2010) Planar cell polarity pathway genes and risk for spina bifida.Am J Med Genet A 152A:299–304

Wexler EM, Paucer A, KornblumHI, Palmer TD, Geschwind DH (2009)EndogenousWnt signalingmaintains neural progenitor cell potency.Stem Cells 27:1130–1141

Wu CI, Hoffman JA, Shy BR, Ford EM, Fuchs E, Nguyen H, Merrill BJ(2012) Function of Wnt/beta-catenin in counteracting Tcf3 repres-sion through the Tcf3-beta-catenin interaction. Development 139:2118–2129

Yu X, Malenka RC (2003) Beta-catenin is critical for dendritic morpho-genesis. Nat Neurosci 6:1169–1177

Zhao C, Deng W, Gage FH (2008) Mechanisms and functional implica-tions of adult neurogenesis. Cell 132:645–660

Cell Tissue Res (2015) 359:215–223 223