Open access

Wnt Signaling Roles on the Structure and Function of the Central Synapses: Involvement in Alzheimer’s Disease

Written By

Nibaldo C. Inestrosa and Lorena Varela-Nallar

Submitted: 30 April 2012 Published: 27 March 2013

DOI: 10.5772/54606

From the Edited Volume

Trends in Cell Signaling Pathways in Neuronal Fate Decision

Edited by Sabine Wislet-Gendebien

Chapter metrics overview

3,153 Chapter Downloads

View Full Metrics

1. Introduction

Wnts compromise a large family of secreted glycoproteins that have shown to be part of the signaling molecules that regulate several aspects of development such as axis formation and midbrain development [1, 2]. In mammals at least 19 Wnt members have been found. The interaction of a Wnt protein with members of the Frizzled (Fz) family of seven-pass transmembrane cell-surface receptors triggers the activation of the Wnt signaling pathway [3-5]. In human and mice, 10 members of the Fz family have been identified. In addition, receptor-like tyrosine kinase (Ryk) and receptor tyrosine kinase-like orphan receptor (Ror2) have been identified as alternative Wnt receptors [6-8]. Different Wnt signaling cascades are activated downstream the Wnt receptors, identified as Wnt/β-catenin or canonical pathway, and β-catenin-independent or non-canonical pathways. The canonical pathway involves the transcription of Wnt target genes, while activation of non-canonical Wnt pathways may induce either an increase in intracellular calcium concentration or activation of the c-Jun-N-terminal kinase (JNK) cascade [3, 9, 10].

The Wnt pathway participates in the development of the central nervous system (CNS) and growing evidence indicate that Wnts also regulates the function of the adult nervous system [11, 12]. In fact, most of the key components including Wnts and Fz receptors are expressed in the adult brain [13, 14]. Wnt ligands have shown to regulate synaptic assembly as well synaptic plasticity and neurotransmission [15-20], and more recently it has also been involved in the adult neurogenesis [21-25].

Deregulation of the Wnt signaling has been associated to several pathologies, been cancer the most widely documented [26-28]. More recently, altered Wnt signaling have been related to mental disorders, mood disorders and neurodegenerative diseases [12, 29-32].

In the first part of this chapter we will address what is currently known about the signaling cascades of canonical and non-canonical pathways. Then, we will review recent findings from our and other labs on the specific effects of different Wnt ligands on the structure of pre- and postsynaptic regions and on glutamatergic neurotransmission in hippocampal neurons. The synaptic role of some Fz receptors will also be reviewed. Finally, the neuroprotective effect of the Wnt signaling activation will be discussed mainly focused on the protection against the toxicity of Aβ-peptide aggregates associated to the pathogenesis of Alzheimer’s disease.

Advertisement

2. The Wnt signaling pathway: Canonical and non–canonical signaling cascades

The binding of Wnt ligands to Fz receptors can trigger the activation of different signaling cascades. In addition to Fz, other proteins have been described as alternative receptors or co-receptors, such as the low-density lipoprotein receptor-related protein 5 (LRP5), LRP6, Ror1, Ror2 and Ryk [3, 33-36], increasing the complexity of the Wnt signaling activation. It has been suggested that the binding of Wnts to specific receptors/co-receptors may selectively activate distinct signaling pathways.

The first Wnt signaling pathway identified was the canonical Wnt/β-catenin pathway (Figure 1). In the absence of Wnt stimulation, the levels of cytoplasmic β-catenin are low since it is ubiquitinated and constantly degraded in the proteasome [37]. β-catenin is phosphorylated by casein kinase 1α (CK1α) and glycogen synthase kinase-3β (GSK-3β) in a multiprotein complex composed also of the scaffold protein axin and adenomatous polyposis coli (APC) [38-42]. Phosphorylated β-catenin is recognized by β-TrCP, which is part of an E3 ubiquitin ligase complex, and is ubiquitinated and subsequently degraded [43]. Activation of the Wnt/β-catenin pathway initiated by the binding of a Wnt ligand to a Fz receptor and coreceptors LRP5/6 activates the protein Dishevelled (Dvl) usually by phosphorylation, and triggers the recruitment of axin to the phosphorylated tail of LRP, inhibiting the degradation pathway consequently inducing the cytoplasmic stabilization of β-catenin which enters the nucleus and regulates the transcription of Wnt target genes [28]. Recently, it was shown that when the destruction complex is associated with phosphorylated LRP, it may still capture and phosphorylates β-catenin, but ubiquitination is blocked (Figure 1, right panel) [44].

In the nucleus, β-catenin binds to members of the family of T-cell factor (Tcf) and lymphoid enhancer factor (Lef) [45-47]; this binding displaces Groucho, which is bound to Tcf/Lef and recruits histone deacetylases (HDAC) to repress the transcription of Wnt target genes [48-51]. Several Wnt target genes have been identified including c-Myc, cyclin D1, Axin2, Calcium/calmodulin-dependent protein kinase type IV (CamKIV) [52-55]. In addition, by using an in silico analysis based on multiple Classification and Regression Tree (CART), 89 new genes were predicted to be targets of the Wnt/β-catenin pathway [56].

Figure 1.

Canonical Wnt/β-catenin signaling pathway. (Left panel) In the absence of a Wnt protein, GSK-3β phosphorylates β-catenin which targets it for ubiquitination by β-TrCP and degradation in the proteasome. (Right panel) Activation of the signaling pathway by the binding of a Wnt ligand to Fz receptor and coreceptors LRP5/6 triggers the association of the destruction complex with phosphorylated LRP. In this condition, the complex may still capture and phosphorylate β-catenin, however the ubiquitination is blocked and it is stabilized in the cytoplasm and enters the nucleus to regulate the transcription of Wnt target genes.

There are at least two β-catenin-independent pathways: the planar cell polarity (PCP) pathway and the Ca2+ pathway (Figure 2). The PCP pathway was originally identified in Drosophila where it regulates tissue polarity and cell migration [10, 57]. This signaling pathway requires Fz receptors and Dvl and activates small GTPases including Rho and Rac and the protein kinase JNK. This pathway is also known as the Wnt/JNK pathway. The activation of the Wnt/Ca2+ pathway triggers the increase in intracellular Ca2+ levels and activates the protein kinases CamKII and protein kinase C (PKC) [10, 58]. It has been suggested that Wnt-mediated Ca2+ release involves heterotrimeric G proteins since it is inhibited by pertussis toxin [59]. As mentioned 10 Fz receptors are known in mammals. Fz receptors are seven-transmembrane-spanning receptors that belong to the G protein-coupled receptor (GPCR) list as a separate class [60]. Fz receptors have an extracellular amino-terminal region that contains a cysteine-rich domain (CRD) consisting of 120 to 125 residues with 10 conserved cysteines that is relevant for the binding of Wnt proteins [61]. Growing evidence indicate the involvement of G protein in the Wnt/Fz signaling. The first evidence came from inhibition of non-canonical Wnt effects by pertussis toxin [62]. Later on, many reports have indicated that heterotrimeric G protein participates of canonical and non-canonical Wnt signaling in Drosophila, Xenopus and mammals [63-69].

Figure 2.

β-catenin-independent Wnt signaling pathways. In the Wnt/JNK pathway or PCP pathway, a Wnt ligand through a Fz receptors and Dvl activates small GTPases including Rho and Rac and JNK, which in turns modulate cytoskeletal organization. The activation of the Wnt/Ca2+ pathway triggers an increase in intracellular Ca2+ levels which activates CamKII and PKC.

Advertisement

3. Roles of the Wnt signaling pathway at central synapses

The Wnt signaling pathway has different roles during development linked to neurite patterning and synaptogenesis. Different Wnt ligands have been linked to the presynaptic assembly. In 1997, Salinas and co-workers demonstrated in cerebellar neurons that Wnt-7a increases the levels of synapsin I, a protein associated to synaptic vesicles [70]. Moreover, Wnt-7a mutant mice show a delay in the accumulation of synapsin I [71]. In hippocampal neurons Wnt-7a as well as Wnt-3a and Wnt-7b increases the number of pre-synaptic puncta suggesting a role for these ligands in presynaptic assembly [18, 72, 73]. In addition, Wnt-7a was found to stimulate recycling and endocytosis of synaptic vesicles using FM dyes [74]. In hippocampal neurons, Wnt-7a was also able to increase the expression as well as the clustering of the α7- nicotinic acetylcholine receptor (α7-nAChR), indicating that the Wnt signaling regulates the clustering of presynaptic receptors [75]. Interestingly, all these ligands are able to modulate presynaptic differentiation by activation of the Wnt/β-catenin signaling pathway, suggesting that some of the components associated with this pathway may be involved in the presynaptic effect. On the other hand, the non-canonical ligand Wnt-5a decreases the number of presynaptic terminals [72], indicating that canonical and non-canonical signaling pathways may have promoting and inhibitory effects on presynaptic differentiation respectively. In accordance, electrophysiological recordings on adult rat hippocampal slices showed that Wnt-7a, but not Wnt-5a, increased neurotransmitter release in CA3-CA1 synapses by decreasing paired pulse facilitation and increasing the frequency of miniature excitatory postsynaptic currents (mEPSC) [73]. Also, Wnt-7a/Dvl1 double mutant mice exhibit decreased mEPSC frequency at the mossy fiber-granule cell synapse revealing a defect in neurotransmitter release [18].

The Wnt signaling also plays relevant roles in the postsynaptic structure. Wnt-5a, which activates non-canonical Wnt signaling cascades in hippocampal neurons [19, 76], modulates postsynaptic assembly by increasing the clustering of the postsynaptic density protein-95 (PSD-95) and increases spine morphogenesis in cultured hippocampal neurons [15, 19]. PSD-95 is a scaffold protein of the postsynaptic density (PSD), which is a multiprotein complex that interacts with key molecules involved in the regulation of glutamate receptor targeting and trafficking and regulatory proteins relevant for neurotransmission [77, 78]. In hippocampal neurons, Wnt-5a induces a fast increase in the number of clusters of PSD-95 without affecting total levels of PSD-95 protein or presynaptic protein clustering [19]. This postsynaptic effect is dependent on Wnt/JNK signaling pathway as demonstrated by using JNK inhibitors. In long-term experiments, we observed that Wnt-5a is also able to increase the total number of synapses [79]. When hippocampal neurons were incubated with the formylated hexapeptide Foxy-5, which is derived from the sequence of Wnt-5a and mimics the full Wnt-5a molecule action in neurons and other systems [19, 80], there was an increase in PSD-95 since 1 hour, but after 24 hours an increase in the synaptic vesicle protein 2 (SV2) clustering was also observed. In consequence, there was an increase in the total number of synaptic contacts [79].

Also, we determined that Wnt-5a induced a transient formation of dendrite protrusions that resulted in a net increase of mature dendrite spines. Videomicroscopy revealed that Wnt-5a induced de novo formation of dendritic spines and also increased the size of the preexisting ones [15]. Interestingly, treatment with the soluble CRD region of Fz2, acting as a Wnt scavenger, decreased spine density in cultured neurons, supporting the physiological relevance of this finding and supporting the implication of Wnt ligands in dendrite spine morphogenesis. Wnt-7a is also able to increase the density and maturity of dendritic spines through a CamKII-dependent mechanism [81]. Wnt-7a rapidly activates CaMKII in spines and inhibition of this kinase abolishes the effects of Wnt-7a on spine growth and excitatory synaptic strength. This finding implicates the Wnt/Ca2+ signaling cascade in synaptic effects of Wnt ligands. Interestingly, Wnt-5a and Wnt-7a induces an increase in intracellular Ca2+ concentration [15, 81], supporting the activation of this non-canonical Wnt pathway.

In addition to the structural effects of Wnt ligands at the excitatory synapse, different Wnts have shown modulatory effects on glutamatergic neurotransmission. Wnt-3a modulates the recycling of synaptic vesicles in hippocampal synapses [73, 82] and is able to induce an increase in the frequency of mEPSC [20]. In hippocampal slices, blockade of Wnt signaling impairs long-term potentiation (LTP), whereas activation of Wnt signaling facilitates LTP [17]. In the case of Wnt-5a, acute application of this ligand in hippocampal slices increases the amplitude of field excitatory postsynaptic potentials (fEPSP) and upregulates synaptic NMDA receptor currents facilitating induction of LTP [15, 16]. Interestingly, Wnt-5a produced a two-step increase in the amplitude of NMDAR responses [16]. The mechanisms involved in this two-step effect of Wnt-5a were investigated by the delivery of specific protein kinase inhibitors via the recording pipette. Specifically, the role of PKC and JNK was investigated, since these are two known downstream kinases of the non-canonical pathway. Inhibition of Ca22+-dependent PKC isoforms with Go6976 or the more general PKC inhibitor calphostin C eliminated the first step of potentiation of NMDAR currents and did not affect the second one. On the contrary, the slower developing increase in NMDAR currents was blocked by the JNK inhibitors TI-JIP153-163 and SP600125. This indicate that there are two mechanisms involved in in the potentiation of NMDAR by Wnt-5a. There is a fast PKC-dependent potentiation and a slower JNK-dependent potentiation that does not require previous activation of PKC [16].

Wnt-5a also regulates postsynaptically the hippocampal inhibitory synapses [76]. Wnt-5a induces surface expression and maintenance of GABAA receptor in the membrane of hippocampal neurons, increases the amplitude of GABA-currents due to a postsynaptic mechanisms, and induces the recycling of functional GABAA receptors through activation of CaMKII [76]. Therefore Wnt-5a is able to modulate both, excitatory and inhibitory synapses which must be relevant for neurotransmission.

The novel role for Wnt ligands in synaptic transmission provides a mechanism for Wnt signaling to acutely modulate synaptic plasticity and brain function in later stages of development and in the mature organism. Importantly, neuronal activity modulates the release and expression of Wnt ligands which may be relevant for the function of these ligands during neurotransmission. Activation of NMDA receptors increases the expression of Wnt-2 in hippocampal neurons which then stimulates dendritic arborization [83]. On the other hand, tetanic stimulation induce NMDA receptor-dependent synaptic Wnt3a release [17]. The role for endogenous Wnts was supported by incubation of hippocampal slices with secreted Wnt inhibitors, such as secreted Frizzled-related protein-2 (sFRP-2), which showed that endogenous Wnt ligands are modulators of glutamatergic neurotransmission being necessary to maintain basal NMDA receptor synaptic transmission [15, 16].

The in vivo relevance for the role of Wnt signaling in activity-mediated synaptic connectivity was revealed in mice exposed to an enriched environment (EE). These animals showed increased complexity and number of large mossy fiber terminals in the CA3 region [84]. EE increased Wnt7a/b levels in CA3 pyramidal neurons and inhibiting Wnt signaling through locally applied sFRP-1, suppressed the effects of EE on synapse numbers and further reduced synapse numbers in control mice.

These findings show that Wnt ligands are important regulators of the synaptic structure during development and in adult neurons, and that the Wnt pathway is one of the signaling cascades regulated by neuronal activity that is involved in the regulation of neurotransmission in adult nervous system.

In addition to the role of Wnts, Fz receptor have also been involved in synaptic structure and function. In the hippocampus, we have determined that different Fz receptors have very different patterns of expression during development, being some of them highly expressed in adulthood and others during early development [85]. In addition, the distribution of Fzs in hippocampal neurons is also very specific. Some receptors, are located in the synaptic region, while others are mainly located in the soma or in the growth cones of young neurons [85]. These findings suggest that these receptors could be important regulators for the specific activation of the Wnt signaling cascades during the development of hippocampal circuits. In fact, we determined an association of the distribution with specific functions. In hippocampal neurons, Fz1 is located in the synaptic region co-localizing with presynaptic proteins and with active synaptic vesicle recycling sites [82]. Interestingly, overexpression of Fz1 increased the number of clusters of Bassoon, a component of the active zone involved in the structural organization of neurotransmitter release sites that is recruited early during synapse formation [86], suggesting that Fz1 regulates synaptic differentiation. In agreement, treatment with the extracellular CRD of Fz1 decreased Bassoon clustering which was not observed with the CRD of Fz2, indicating a receptor specificity for the synaptic effect [82]. Fz5 also has a role in mature neurons where it modulates the synaptogenic effect of Wnt7a [87]. As well as Fz1, Fz5 is present in synaptosomes and colocalizes with synaptic markers, and changes in the expression of this receptor modulates the density of synaptic sites [87]. In addition to its function in mature neurons, Fz5 was shown to be in high levels in the growth cones of developing hippocampal neurons [85], and we have recently determined that this receptor is involved in neural polarization (unpublished results). We determined that overexpression of Fz5 triggers a mislocalization of axonal proteins such as Tau-1 and phosphorylated MAP1B (MAP1BP), which change their distribution to the whole cell suggesting altered polarization. When the expression of Fz5 is knocked-down by shRNA, MAP1BP is not polarized and is almost completely lost. These findings suggest that in developing hippocampal neurons Fz5 is relevant for neural polarization. These studies indicate that Fz receptors are relevant players in both the developing and the adult nervous system and support the notion that the Wnt signaling pathway is crucial for different aspects of the development and function of the CNS.

Advertisement

4. Role of Wnt signaling in adult neurogenesis

In the adult brain, there are two regions where there is a continuous generation of new neurons (Figure 3A), the subventricular zone (SVZ) of the lateral ventricles [88] and the subgranular zone (SGZ) in the hippocampal dentate gyrus [89]. In the SVZ, astrocyte-like neural stem cells (NSCs), called type B1 cells, generate type C cells that rapidly proliferate and give rise to type A neuroblasts (Figure 3B). These cells migrate through the rostral migratory stream to the olfactory bulb where they became interneurons [88] (Figure 3A). In the SGZ, radial and non-radial neural precursor cells give rise to transient amplifying progenitors that generate neuroblasts and then became immature neurons that extend dendrites toward the molecular layer and project their axons through the hilus toward the CA3 region [90] (Figure 3C). Newborn neurons then mature and fully integrate into the preexisting hippocampal circuitry.

Adult neurogenesis is highly regulated by intrinsic and extrinsic mechanisms. Many signaling pathways have been identified as regulators of different aspects of neurogenesis. Notch, Shh, BMPs, and Wnts are part of the signaling molecules of the niche that regulate the maintenance, activation and fate specification of neural precursor cells [91, 92].

In Wnt/β-catenin reporter mice (BATGAL) it was shown that this pathway is active in the SGZ and the dentate granule cell layer [23]. In that study, authors determined that Wnt3 is expressed in adult hippocampal astrocytes and that adult hippocampal progenitor (AHP) cells express key components of the Wnt/β-catenin signaling pathway. These findings suggested that the Wnt pathway may be involved in the regulation of adult neurogenesis. In vitro analysis in cultured cells revealed that Wnts derived from hippocampal astrocytes stimulate Wnt/β-catenin signaling in isolated AHPs inducing their neuronal commitment [23]. The effect of the Wnt signaling was supported in vivo using lentiviral vectors expressing Wnt3a or a secreted mutant Wnt1 protein that blocks Wnt signaling. Lentiviruses were stereotactically injected into the dentate gyrus of rats. As assessed by the incorporation of the nucleotide analog BrdU and immunodetection of the immature neuron protein doublecortin (DCX), blocking the Wnt signaling decreases adult hippocampal neurogenesis while stimulating this pathway has the opposite effect [23]. More recently, and by using the same lentiviral approach to block Wnt signaling in the dentate gyrus of adult rats it was shown that Wnt-mediated adult hippocampal neurogenesis contributes to learning and memory [93]. In the SVZ, β-catenin signaling also plays a role in the proliferation of progenitor cells in the adult mouse brain [94]. Retrovirus-mediated expression of a stabilized β-catenin promoted the proliferation of type C cells and inhibited their differentiation into neuroblasts. Also in the SVZ, transduction of the β-catenin inhibitor axin by intracranial lentiviral delivery decreased cell proliferation as revealed by decreased BrdU labeling [95], further supporting a role for Wnt/β-catenin signaling in neural stem cell proliferation in the neurogenic areas of adult brain.

Figure 3.

Neurogenesis in the adult brain. (A) Schematic representation of adult rodent brain highlighting the two neurogenic regions. The hippocampus and the SVZ (boxed). (B) Schematic of the SVZ in the wall of the lateral ventricles. Distinct stem/progenitor cell types (types B, C, and A) are shown. (C) Neurogenesis in the SGZ of the hippocampal dentate gyrus. The progression of radial type 1 cells to mature newborn granule neurons is schematized.

The Wnt-mediated effects in neurogenesis may be caused by the transcriptional activation of NeuroD1 which is dependent on the Wnt/β-catenin signaling activation [25]. NeuroD1 is a basic helix-loop-helix transcription factor important for the generation of granule cell and olfactory neuron in the embryonic and adult brain [96]. NeuroD1 gene promoter has overlapping DNA-binding site for Sox2 and TCF/LEF, then the activation of this gene implies activation of the canonical Wnt pathway and removal of Sox2 repression from the NeuroD1 gene promoter [25]. More recently, Prox1 was also determined as a target of the Wnt/β-catenin pathway relevant for neurogenesis [22]. Prox1 is expressed in newborn and mature granule cells and is required for the proper differentiation and survival of newborn granule cells, but not for the maintenance of granule cells after they have fully matured [22].

In addition, Wnts could indirectly modulate adult neurogenesis thorough their effects on neuronal activity. As previously described, different Wnts regulate glutamatergic neurotransmission, and evidence indicates that neural progenitor cells respond to neuronal activity as part of their differentiation program [97]. GABA is an important modulator of adult hippocampal neurogenesis being critical for the proper development and maturation of adult-born neurons [98-100]. Interestingly, Wnt-5a through activation of CaMKII, induces the recycling of functional GABAA receptors on hippocampal neurons and modulates inhibitory synapses [76].

As mentioned, in neurogenic niches Wnts are provided by astrocytes [23], and during aging it was reported that the levels of Wnt3 protein and the number of Wnt3-secreting astrocytes declines [101], which may be one of the factors underlying the impairment of neurogenesis that is observed in aging [102, 103]. On the contrary, running, that is a potent stimulator of adult neurogenesis in the SGZ [104] was found to significantly increase de novo expression of Wnt-3 [101], pointing to the Wnt pathway as one of the factors involved in running-mediated increase in neurogenesis. In addition to astrocytes-derived Wnts, an autocrine Wnt signaling activity has been observed in adult hippocampal progenitors (AHPs) derived from adult rat brains. Inhibiting this autocrine Wnt signaling increases the number of neurons formed and leads to a loss of multipotency among AHPs indicating that this autocrine pathway may preserve the balance between neural stem cell maintenance and differentiation [105].

The Wnt signaling has also been involved in the mechanism of the orphan nuclear receptor TLX (also known as NR2E1), which is an important regulator of neural stem cell maintenance and self-renewal in embryonic and adult brains [106, 107] and is involved in neurogenesis in the SVZ [108] and hippocampus [109]. To stimulate neural stem cell proliferation and self-renewal TLX activates the Wnt/β-catenin pathway in adult mouse neural stem cells by activating the expression of Wnt-7a, which expression was found to be downregulated in TLX-null mice, through binding to two TLX binding sites present in the Wnt-7a gene promoter [95]. Wnt-7a is important for adult neural stem cell proliferation in vivo since there is a decreased BrdU labeling in the SGZ and SVZ of adult Wnt7a knockout mice. In TLX-/- mice, intracranial lentiviral transduction of active β-catenin led to a considerable rescue of cell proliferation in the SVZ, suggesting that Wnt/β-catenin acts downstream of TLX to regulate neural stem cell proliferation in vivo [95].

It has been shown that low oxygen is associated with increased levels of β-catenin in vivo, and that hypoxia inducible factor-1α (HIF-1α) modulates the Wnt/β-catenin signaling in embryonic stem cells exposed to low oxygen [110]. Recently, we determined in vivo that hypoxia stimulates the activation of the Wnt/β-catenin signaling pathway in the hippocampus of adult mice (our unpublished results), and stimulates cell proliferation in the SGZ of 2 month old wild-type mice.

Altogether, these findings indicate that the Wnt pathway is relevant not only for the development of the nervous system but also for the development of new neurons in the adult brain, being important for the maintenance and self-renewal of the stem cell pool and for the commitment and proliferation of new neurons.

Advertisement

5. Wnt signaling in Alzheimer’s disease

Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a progressive deterioration of cognitive abilities, cerebral accumulation of extracellular amyloid plaques composed mainly of amyloid-β peptide (Aβ), and synaptic alterations [111]. In addition to the accumulation of Aβ aggregates, which is a product of the processing of the amyloid precursor protein (APP), cytoskeletal alterations associated to the abnormal phosphorylation of the microtubule associated protein tau [112, 113] are early manifestations that lead to aberrant remodeling of dendrites and axons, the appearance of dystrophic neurites, synaptic loss [114], and eventually progressive loss of neuronal populations [112].

During more than a decade, a strong relationship between an impaired Wnt signaling pathway activity and neuronal damage in AD has been raised [31, 115-118]. Different studies have shown that Wnt signaling components are altered in AD [119-124], and in addition, the Wnt signaling pathway has been related to other neurodegenerative disorders such as autism and schizophrenia [30, 125]. Among the Wnt components that are affected in AD, it was shown that β-catenin levels are reduced in AD patients carrying presenilin-1 (PS-1)-inherited mutations [124], while the secreted Wnt antagonist Dickkopf-1 (Dkk1) is elevated in postmortem AD brains and brains from transgenic mouse models for AD [121, 126]. A variant of the LRP6 has been associated with late-onset AD, which confers low levels of Wnt signaling [119]. In addition, genetic studies show a link between Wnt signaling and AD. Epidemiological data show an increased risk for AD in populations where the allele 4 of apo-lipoprotein E (apoE4) is present. Interestingly apoE4 causes inhibition of the canonical Wnt signaling in PC12 cells upon stimulation with Wnt-7a as determined by luciferase activities and nuclear β-catenin levels [127]. Aβ directly binds to the extracellular CRD of Fz5 at or in close proximity to the Wnt-binding site inhibiting the canonical Wnt signaling pathway [128], linking directly Aβ to Wnt impairment. Moreover, the exposure of cultured rat hippocampal neurons to Aβ results in inhibition of canonical Wnt signaling as determined by destabilization of endogenous levels of β-catenin, increase in GSK-3β activity, and a decrease in the expression of some Wnt target genes [129]. Moreover, acute exposure to Aβ increases Dkk1 mRNA levels in hippocampal brain slices, which seems to be associated to synaptic loss induced by Aβ [130].

As mentioned, one of the hallmarks of AD brains is the abnormal phosphorylation of the tau protein which accumulates as intraneuronal neurofibrillary tangles [131]. Several kinases can phosphorylate tau in vitro; however, the bulk of the information supports that Cdk5, extracellular signal-related kinase 2, microtubule affinity-regulating kinase and GSK-3β, a key component of the Wnt cascade, are the most relevant kinases for tau phosphorylation in vivo [132, 133]. Cultured neurons exposed to Aβ show an increased GSK-3β activity [134, 135], and active GSK-3β has been found in brains staged for AD neurofibrillary changes, with a concomitant decrease in β-catenin levels and an increase in tau hyperphosphorylation [136]. Also, neurodegeneration and spatial learning deficits have been observed in GSK-3β conditional transgenic mice [137, 138]. Interestingly, a study shows that the phosphorylation of tau antagonizes apoptosis by stabilizing β-catenin; therefore, up-regulation of β-catenin during tau phosphorylation prevents the cell from going into apoptosis. Increasing levels of phosphorylated tau was correlated with increased levels of nuclear β-catenin, and the knockdown of β-catenin antagonizes the anti-apoptotic effects of tau [139]. These findings support a role of β-catenin as a survival element in AD.

Several studies have shown neuroprotective properties of the Wnt signaling activation against the toxicity of Aβ peptide. In cultured hippocampal neurons, exposure to Aβ aggregates causes a decrease in endogenous β-catenin levels, and this effect was overcome by direct activation of the pathway with Wnt-3a conditioned media [117, 129]. The protective effect of Wnt-3a against the toxicity of Aβ oligomers was shown to be mediated by Fz1 receptor, since this effect is modulated by the expression levels of Fz1 in both, PC12 cells and hippocampal neurons [14]. Overexpression of Fz1 significantly increased cell survival induced by Wnt-3a and diminished caspase-3 activation, while knocking-down the expression of the receptor by antisense oligonucleotides decreased the stabilization of β-catenin induced by Wnt-3a and decreased the neuroprotive effect elicited by this Wnt ligand [14].

In agreement with the effect of Wnt-3a, inhibition of GSK-3β by lithium protects hippocampal neurons from Aβ-induced damage. More importantly, in vivo lithium treatment of double transgenic APPswe/PSEN1ΔE9 mice, which is a well characterized in vivo model of AD that shows most hallmarks of the disease [140], reduced spatial memory impairment, decreased Aβ oligomers and the activation of astrocytes and microglia [141]. In vivo, lithium treatment activated the Wnt signaling as shown by the increase in β-catenin and by the inhibition of GSK-3β [141]. These studies suggest that the loss of normal Wnt/β-catenin signaling activity may be involved in the Aβ-dependent neurodegeneration observed in AD and that the activation of the pathway might have beneficial effects for the treatment of the disease [12].

APPswe/PSEN1ΔE9 mice show decreased levels of adult neurogenesis [142]. In these mice, we evaluated the effect of hypoxia on the generation of new neurons in the hippocampus. As previously mentioned hypoxia induces the activation of the Wnt/β-catenin signaling pathway in the hippocampus of wild-type mice. Mice were exposed to low oxygen and neurogenesis was evaluated by incorporation of BrdU and double staining with DCX. It was determined that hypoxia is a strong stimulator of neurogenesis in AD mice (our unpublished results). Currently we are evaluating whether this effect is related to the activation of the canonical Wnt pathway. Also, we have observed that voluntary wheel running strongly increased neurogenesis in APPswe/PSEN1ΔE9 mice and also decreased Aβ burden and tau phosphorylation (our unpublished results). As previously mentioned, voluntary running was found to increase de novo expression of Wnt-3 [101], suggesting that the effects observed in runner AD mice could involve the activation of the Wnt signaling pathway.

In addition to the role of the canonical Wnt signaling, we have studied whether Wnt-5a is able to protect neurons against Aβ oligomers synaptotoxicity [143]. Synaptic failure is an early event in AD, and soluble Aβ oligomers are proposed to be responsible for the synaptic pathology that occurs before the plaque deposition and neuronal death [74, 144]. Electrophysiological analysis of Schaffer collaterals-CA1 glutamatergic transmission in hippocampal slices demonstrated that Wnt-5a prevents the decrease in the amplitude of fEPSP and EPSCs induced by Aβ oligomers, indicating that Wnt-5a prevents the synaptic damage triggered by Aβ [143]. Moreover, Wnt-5a prevented the decrease in the postsynaptic density scaffold protein PSD-95 and synaptic loss in cultured hippocampal neurons [143], supporting that Wnt-5a improves synaptic function in the presence of Aβ.

Additionally, the activation of several signaling pathways that crosstalk with the Wnt pathway also supports the neuroprotective potential of the Wnt cascades in AD [12].

Advertisement

6. Conclusions

As we have discussed throughout this Chapter, the Wnt signaling pathway has fundamental roles in the development and function of the CNS. As discussed, the canonical and non-canonical Wnt signaling cascades have shown to be important for the formation and structure of central synapses, and in addition to the structural effects, Wnt ligands acutely modulate synaptic transmission and plasticity. Also, in the adult brain the Wnt pathway is one of the signaling cascades that regulates the generation of new neurons in neurogenic niches. Importantly, different stimuli that regulate neurogenesis involve the regulation of the Wnt signaling, implicating this pathway as a relevant player in the modulation of this physiological process.

Considering all the discussed roles of Wnts, it was expected that alterations in the Wnt cascades leads to diseases associated to the nervous system. In fact, deregulation of the Wnt pathway has been related to mental disorders, mood disorders and neurodegenerative diseases. As we have discussed, a bulk of evidence associate Wnt dysfunction to AD, and strongly point to a neuroprotective potential of the Wnt cascades as a therapeutic approach. Future work should focus on explore the therapeutic benefits of stimulating the Wnt signaling pathway in vivo.

Advertisement

Acknowledgments

We thank to Felipe G. Serrano for his contribution in the artwork. This work was supported by Grants from FONDECYT (N°1120156) and the Basal Center of Excellence in Science and Technology (CONICYT-PFB12/2007) to NCI and FONDECYT (N°11110012) and Insertion of Postdoctoral Researchers in the Academy (CONICYT-79090027) to LV-N.

References

  1. 1. Nusse R Varmus H Three decades of Wnts: a personal perspective on how a scientific field developed. EMBO J 2012 31 12 2670 84
  2. 2. Van Amerongen R Nusse R Towards an integrated view of Wnt signaling in development. Development (Cambridge, England) 2009 136 19 3205 14
  3. 3. Gordon M. D Nusse R Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem 2006 281 32 22429 33
  4. 4. Schulte G International Union of Basic and Clinical Pharmacology. LXXX. The class Frizzled receptors. Pharmacol Rev 2010 62 4 632 67
  5. 5. Wang H. Y Liu T Malbon C. C Structure-function analysis of Frizzleds. Cell Signal 2006 18 7 934 41
  6. 6. Keeble T. R Halford M. M Seaman C Kee N Macheda M Anderson R. B Stacker S. A Cooper H. M The Wnt receptor Ryk is required for Wnt5a-mediated axon guidance on the contralateral side of the corpus callosum. J Neurosci 2006 26 21 5840 8
  7. 7. Oishi I Suzuki H Onishi N Takada R Kani S Ohkawara B Koshida I Suzuki K Yamada G Schwabe G. C Mundlos S Shibuya H Takada S Minami Y The receptor tyrosine kinase Ror2 is involved in non-canonical Wnt5a/JNK signalling pathway. Genes Cells 2003 8 7 645 54
  8. 8. Ho H. Y Susman M. W Bikoff J. B Ryu Y. K Jonas A. M Hu L Kuruvilla R Greenberg M. E Wnt5a-Ror-Dishevelled signaling constitutes a core developmental pathway that controls tissue morphogenesis. Proc Natl Acad Sci U S A 2012 109 11 4044 51
  9. 9. Angers S Moon R. T Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol 2009 10 7 468 77
  10. 10. Veeman M. T Axelrod J. D Moon R. T A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Developmental cell 2003 5 3 367 77
  11. 11. Salinas P. C Zou Y Wnt signaling in neural circuit assembly. Annu Rev Neurosci 2008 31 339
  12. 12. Inestrosa N. C Arenas E Emerging roles of Wnts in the adult nervous system. Nature reviews 2010 11 2 77 86
  13. 13. Shimogori T Vansant J Paik E Grove E. A Members of the Wnt, Fz, and Frp gene families expressed in postnatal mouse cerebral cortex. J Comp Neurol 2004 473 4 496 510
  14. 14. Chacon M. A Varela-Nallar L Inestrosa N. C Frizzled-1 is involved in the neuroprotective effect of Wnt3a against Abeta oligomers. J Cell Physiol 2008 217 1 215 27
  15. 15. Varela-Nallar L Alfaro I. E Serrano F. G Parodi J Inestrosa N. C Wingless-type family member 5A (Wnt-5a) stimulates synaptic differentiation and function of glutamatergic synapses. Proc Natl Acad Sci U S A 2010 107 49 21164 9
  16. 16. Cerpa W Gambrill A Inestrosa N. C Barria A Regulation of NMDA-receptor synaptic transmission by Wnt signaling. J Neurosci 2011 31 26 9466 71
  17. 17. Chen J Park C. S Tang S. J Activity-dependent synaptic Wnt release regulates hippocampal long term potentiation. J Biol Chem 2006 281 17 11910 6
  18. 18. Ahmad-Annuar A Ciani L Simeonidis I Herreros J Fredj N. B Rosso S. B Hall A Brickley S Salinas P. C Signaling across the synapse: a role for Wnt and Dishevelled in presynaptic assembly and neurotransmitter release. J Cell Biol 2006 174 1 127 39
  19. 19. Farias G. G Alfaro I. E Cerpa W Grabowski C. P Godoy J. A Bonansco C Inestrosa N. C Wnt-5a/JNK signaling promotes the clustering of PSD-95 in hippocampal neurons. J Biol Chem 2009 284 23 15857 66
  20. 20. Avila M. E Sepulveda F. J Burgos C. F Moraga-Cid G Parodi J Moon R. T Aguayo L. G Opazo C De Ferrari G. V Canonical Wnt3a modulates intracellular calcium and enhances excitatory neurotransmission in hippocampal neurons. J Biol Chem 2010 285 24 18939 47
  21. 21. Clevers H Nusse R Wnt/beta-catenin signaling and disease. Cell 2012 149 6 1192 205
  22. 22. Karalay O Doberauer K Vadodaria K. C Knobloch M Berti L Miquelajauregui A Schwark M Jagasia R Taketo M. M Tarabykin V Lie D. C Jessberger S Prospero-related homeobox 1 gene (Prox1) is regulated by canonical Wnt signaling and has a stage-specific role in adult hippocampal neurogenesis. Proc Natl Acad Sci U S A (2011). , 108(14), 5807-12.
  23. 23. Lie D. C Colamarino S. A Song H. J Desire L Mira H Consiglio A Lein E. S Jessberger S Lansford H Dearie A. R Gage F. H Wnt signalling regulates adult hippocampal neurogenesis. Nature 2005 437 7063 1370 5
  24. 24. Zhang L Yang X Yang S Zhang J The Wnt /beta-catenin signaling pathway in the adult neurogenesis. Eur J Neurosci 2011 33 1 1 8
  25. 25. Kuwabara T Hsieh J Muotri A Yeo G Warashina M Lie D. C Moore L Nakashima K Asashima M Gage F. H Wnt-mediated activation of NeuroD1 and retro-elements during adult neurogenesis. Nat Neurosci 2009 12 9 1097 105
  26. 26. Polakis P The many ways of Wnt in cancer. Curr Opin Genet Dev 2007 17 1 45 51
  27. 27. Klaus A Birchmeier W Wnt signalling and its impact on development and cancer. Nat Rev Cancer 2008 8 5 387 98
  28. 28. Logan C. Y Nusse R The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol (2004). , 20, 781-810.
  29. 29. De Ferrari G. V Moon R. T The ups and downs of Wnt signaling in prevalent neurological disorders. Oncogene 2006 25 57 7545 53
  30. 30. Lovestone S Killick R Di Forti M,Murray R. Schizophrenia as a GSK-3 dysregulation disorder. Trends Neurosci 2007 30 4 142 9
  31. 31. De Ferrari G. V Inestrosa N. C Wnt signaling function in Alzheimer’s disease. Brain Res Brain Res Rev 2000 33 1 1 12
  32. 32. Williams R. S Harwood A. J Lithium therapy and signal transduction. Trends Pharmacol Sci 2000 21 2 61 4
  33. 33. Fradkin L. G Dura J. M Noordermeer J. N Ryks: new partners for Wnts in the developing and regenerating nervous system. Trends Neurosci 2009 33 2 84 92
  34. 34. Green J. L Kuntz S. G Sternberg P. W Ror receptor tyrosine kinases: orphans no more. Trends Cell Biol 2008 18 11 536 44
  35. 35. Kikuchi A Yamamoto H Kishida S Multiplicity of the interactions of Wnt proteins and their receptors. Cell Signal 2007 19 4 659 71
  36. 36. Cadigan K. M Liu Y. I Wnt signaling: complexity at the surface. J Cell Sci 2006Pt 3) 395-402.
  37. 37. Aberle H Bauer A Stappert J Kispert A Kemler R beta-catenin is a target for the ubiquitin-proteasome pathway. EMBO J 1997 16 13 3797 804
  38. 38. Ikeda S Kishida S Yamamoto H Murai H Koyama S Kikuchi A Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J 1998 17 5 1371 84
  39. 39. Kishida S Yamamoto H Ikeda S Kishida M Sakamoto I Koyama S Kikuchi A Axin, a negative regulator of the wnt signaling pathway, directly interacts with adenomatous polyposis coli and regulates the stabilization of beta-catenin. J Biol Chem 1998 273 18 10823 6
  40. 40. Hart M. J de los Santos R, Albert IN, Rubinfeld B,Polakis P. Downregulation of beta-catenin by human Axin and its association with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr Biol 1998 8 10 573 81
  41. 41. Itoh K Krupnik V. E Sokol S. Y Axis determination in Xenopus involves biochemical interactions of axin, glycogen synthase kinase 3 and beta-catenin. Curr Biol 1998 8 10 591 4
  42. 42. Sakanaka C Weiss J. B Williams L. T Bridging of beta-catenin and glycogen synthase kinase-3beta by axin and inhibition of beta-catenin-mediated transcription. Proc Natl Acad Sci U S A 1998 95 6 3020 3
  43. 43. Liu C Li Y Semenov M Han C Baeg G. H Tan Y Zhang Z Lin X He X Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002 108 6 837 47
  44. 44. Li V. S Ng S. S Boersema P. J Low T. Y Karthaus W. R Gerlach J. P Mohammed S Heck A. J Maurice M. M Mahmoudi T Clevers H Wnt signaling through inhibition of beta-catenin degradation in an intact Axin1 complex. Cell 2012 149 6 1245 56
  45. 45. Behrens J Von Kries J. P Kuhl M Bruhn L Wedlich D Grosschedl R Birchmeier W Functional interaction of beta-catenin with the transcription factor LEF-1. Nature 1996 382 6592 638 42
  46. 46. Eastman Q Grosschedl R Regulation of LEF-1/TCF transcription factors by Wnt and other signals. Curr Opin Cell Biol 1999 11 2 233 40
  47. 47. Huber O Korn R Mclaughlin J Ohsugi M Herrmann B. G Kemler R Nuclear localization of beta-catenin by interaction with transcription factor LEF-1. Mech Dev 1996 59 1 3 10
  48. 48. Roose J Molenaar M Peterson J Hurenkamp J Brantjes H Moerer P van de Wetering M, Destree O,Clevers H. The Xenopus Wnt effector XTcf-3 interacts with Groucho-related transcriptional repressors. Nature 1998 395 6702 608 12
  49. 49. Cavallo R. A Cox R. T Moline M. M Roose J Polevoy G. A Clevers H Peifer M Bejsovec A Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature 1998 395 6702 604 8
  50. 50. Hurlstone A Clevers H. T-cell factors: turn-ons and turn-offs. EMBO J 2002 21 10 2303 11
  51. 51. Chen G Fernandez J Mische S Courey A. J A functional interaction between the histone deacetylase Rpd3 and the corepressor groucho in Drosophila development. Genes Dev 1999 13 17 2218 30
  52. 52. Arrazola M. S Varela-Nallar L Colombres M Toledo E. M Cruzat F Pavez L Assar R Aravena A Gonzalez M Montecino M Maass A Martinez S Inestrosa N. C Calcium/calmodulin-dependent protein kinase type IV is a target gene of the Wnt/beta-catenin signaling pathway. J Cell Physiol 2009 221 3 658 67
  53. 53. Jho E. H Zhang T Domon C Joo C. K Freund J. N Costantini F Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol 2002 22 4 1172 83
  54. 54. Jung H. C Kim K Identification of MYCBP as a beta-catenin/LEF-1 target using DNA microarray analysis. Life Sci 2005 77 11 1249 62
  55. 55. Tetsu O Mccormick F Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 1999 398 6726 422 6
  56. 56. Hodar C Assar R Colombres M Aravena A Pavez L Gonzalez M Martinez S Inestrosa N. C Maass A Genome-wide identification of new Wnt/beta-catenin target genes in the human genome using CART method. BMC Genomics (2010). ,11:348
  57. 57. Adler P. N Planar signaling and morphogenesis in Drosophila. Developmental cell 2002 2 5 525 35
  58. 58. Kohn A. D Moon R. T Wnt and calcium signaling: beta-catenin-independent pathways. Cell Calcium (2005). , 38(3-4), 439-46
  59. 59. Slusarski D. C Yang-snyder J Busa W. B Moon R. T Modulation of embryonic intracellular Ca2+ signaling by Wnt-5A. Dev Biol 1997 182 1 114 20
  60. 60. Foord S. M Bonner T. I Neubig R. R Rosser E. M Pin J. P Davenport A. P Spedding M Harmar A. J International Union of Pharmacology. XLVI. G protein-coupled receptor list. Pharmacol Rev 2005 57 2 279 88
  61. 61. Dann C. E Hsieh J. C Rattner A Sharma D Nathans J Leahy D. J Insights into Wnt binding and signalling from the structures of two Frizzled cysteine-rich domains. Nature 2001 412 6842 86 90
  62. 62. Slusarski D. C Corces V. G Moon R. T Interaction of Wnt and a Frizzled homologue triggers G-protein-linked phosphatidylinositol signalling. Nature 1997 390 6658 410 3
  63. 63. Liu X Rubin J. S Kimmel A. R Rapid, Wnt-induced changes in GSK3beta associations that regulate beta-catenin stabilization are mediated by Galpha proteins. Curr Biol 2005 15 22 1989 97
  64. 64. Salmanian S Najafi S. M Rafipour M Arjomand M. R Shahheydari H Ansari S Kashkooli L Rasouli S. J Jazi M. S Minaei T Regulation of GSK-3beta and beta-Catenin by Galphaq in HEK293T cells. Biochem Biophys Res Commun 2010 395 4 577 82
  65. 65. Najafi S. M Activators of G proteins inhibit GSK-3beta and stabilize beta-Catenin in Xenopus oocytes. Biochem Biophys Res Commun 2009 382 2 365 9
  66. 66. Katanaev V. L Ponzielli R Semeriva M Tomlinson A Trimeric G protein-dependent frizzled signaling in Drosophila. Cell 2005 120 1 111 22
  67. 67. Katanaev V. L Tomlinson A Dual roles for the trimeric G protein Go in asymmetric cell division in Drosophila. Proc Natl Acad Sci U S A 2006 103 17 6524 9
  68. 68. Jernigan K. K Cselenyi C. S Thorne C. A Hanson A. J Tahinci E Hajicek N Oldham W. M Lee L. A Hamm H. E Hepler J. R Kozasa T Linder M. E Lee E Gbetagamma activates GSK3 to promote LRP6-mediated beta-catenin transcriptional activity. Sci Signal 2010ra37.
  69. 69. Liu X Liu T Slusarski D. C Yang-snyder J Malbon C. C Moon R. T Wang H Activation of a frizzled-2/beta-adrenergic receptor chimera promotes Wnt signaling and differentiation of mouse F9 teratocarcinoma cells via Galphao and Galphat. Proc Natl Acad Sci U S A 1999 96 25 14383 8
  70. 70. Lucas F. R Salinas P. C WNT-7a induces axonal remodeling and increases synapsin I levels in cerebellar neurons. Dev Biol 1997 192 1 31 44
  71. 71. Hall A. C Lucas F. R Salinas P. C Axonal remodeling and synaptic differentiation in the cerebellum is regulated by WNT-7a signaling. Cell 2000 100 5 525 35
  72. 72. Davis E. K Zou Y Ghosh A Wnts acting through canonical and noncanonical signaling pathways exert opposite effects on hippocampal synapse formation. Neural Dev (2008). , 3, 32.
  73. 73. Cerpa W Godoy J. A Alfaro I Farias G. G Metcalfe M. J Fuentealba R Bonansco C Inestrosa N. C Wnt-7a modulates the synaptic vesicle cycle and synaptic transmission in hippocampal neurons. J Biol Chem 2008 283 9 5918 27
  74. 74. Cerpa W Dinamarca M. C Inestrosa N. C Structure-function implications in Alzheimer’s disease: effect of Abeta oligomers at central synapses. Curr Alzheimer Res 2008 5 3 233 43
  75. 75. Farias G. G Valles A. S Colombres M Godoy J. A Toledo E. M Lukas R. J Barrantes F. J Inestrosa N. C Wnt-7a induces presynaptic colocalization of alpha 7-nicotinic acetylcholine receptors and adenomatous polyposis coli in hippocampal neurons. J Neurosci 2007 27 20 5313 25
  76. 76. Cuitino L Godoy J. A Farias G. G Couve A Bonansco C Fuenzalida M Inestrosa N. C Wnt-5a modulates recycling of functional GABAA receptors on hippocampal neurons. J Neurosci 2010 30 25 8411 20
  77. 77. Han K Kim E Synaptic adhesion molecules and PSD-95. Progress in neurobiology 2008 84 3 263 83
  78. 78. Li Z Sheng M Some assembly required: the development of neuronal synapses. Nat Rev Mol Cell Biol 2003 4 11 833 41
  79. 79. Varela-Nallar L Parodi J Farias G. G Inestrosa N. C Wnt-5a is a synaptogenic factor with neuroprotective properties against Abeta toxicity. Neurodegener Dis (2012). , 10(1-4), 23-6.
  80. 80. Safholm A Leandersson K Dejmek J Nielsen C. K Villoutreix B. O Andersson T A formylated hexapeptide ligand mimics the ability of Wnt-5a to impair migration of human breast epithelial cells. J Biol Chem 2006 281 5 2740 9
  81. 81. Ciani L Boyle K. A Dickins E Sahores M Anane D Lopes D. M Gibb A. J Salinas P. C Wnt7a signaling promotes dendritic spine growth and synaptic strength through Ca2+/Calmodulin-dependent protein kinase II. Proc Natl Acad Sci U S A (2011). , 108(26), 10732-7.
  82. 82. Varela-Nallar L Grabowski C. P Alfaro I. E Alvarez A. R Inestrosa N. C Role of the Wnt receptor Frizzled-1 in presynaptic differentiation and function. Neural Dev (2009). , 4, 41.
  83. 83. Wayman G. A Impey S Marks D Saneyoshi T Grant W. F Derkach V Soderling T. R Activity-dependent dendritic arborization mediated by CaM-kinase I activation and enhanced CREB-dependent transcription of Wnt-2. Neuron 2006 50 6 897 909
  84. 84. Gogolla N Galimberti I Deguchi Y Caroni P Wnt signaling mediates experience-related regulation of synapse numbers and mossy fiber connectivities in the adult hippocampus. Neuron 2009 62 4 510 25
  85. 85. Varela-Nallar L Ramirez V. T Gonzalez-billault C Inestrosa N. C Frizzled receptors in neurons: From growth cones to the synapse. Cytoskeleton (Hoboken) 2012 69 7 528 34
  86. 86. Zhai, R, Olias, G, Chung, W. J, Lester R. A, tom Dieck, S, Langnaese, K, Kreutz, M. R, Kindler, S, Gundelfinger, E. D, & Garner C. C. Temporal appearance of the presynaptic cytomatrix protein bassoon during synaptogenesis. Mol Cell Neurosci 2000 15 5 417 28
  87. 87. Sahores M Gibb A Salinas P. C Frizzled-5, a receptor for the synaptic organizer Wnt7a, regulates activity-mediated synaptogenesis. Development (Cambridge, England) 2010 137 13 2215 25
  88. 88. Alvarez-Buylla, A, & Garcia-Verdugo, J. M. Neurogenesis in adult subventricular zone. J Neurosci 2002 22 3 629 34
  89. 89. Gage F. H Mammalian neural stem cells. Science (New York, N.Y.) (2000). 287 5457 1433 8
  90. 90. Zhao, C, Teng, E. M, Summers, R. G Jr, Ming, G. L, & Gage, F. H. Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus. J Neurosci 2006 26 1 3 11
  91. 91. Suh H Deng W Gage F. H Signaling in adult neurogenesis. Annu Rev Cell Dev Biol (2009). , 25, 253-75.
  92. 92. Ming G. L Song H Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 2011 70 4 687 702
  93. 93. Jessberger, S, Clark, R. E, Broadbent, N. J, Clemenson, G. D Jr, Consiglio, A, Lie, D. C, Squire, L. R, & Gage, F. H. Dentate gyrus-specific knockdown of adult neurogenesis impairs spatial and object recognition memory in adult rats. Learn Mem 2009 16 2 147 54
  94. 94. Adachi K Mirzadeh Z Sakaguchi M Yamashita T Nikolcheva T Gotoh Y Peltz G Gong L Kawase T Alvarez-Buylla A Okano H Sawamoto K Beta-catenin signaling promotes proliferation of progenitor cells in the adult mouse subventricular zone. Stem Cells 2007 25 11 2827 36
  95. 95. Qu Q Sun G Li W Yang S Ye P Zhao C Yu R. T Gage F. H Evans R. M Shi Y Orphan nuclear receptor TLX activates Wnt/beta-catenin signalling to stimulate neural stem cell proliferation and self-renewal. Nat Cell Biol 2010sup 1 9
  96. 96. Gao Z Ure K Ables J. L Lagace D. C Nave K. A Goebbels S Eisch A. J Hsieh J Neurod1 is essential for the survival and maturation of adult-born neurons. Nat Neurosci 2009 12 9 1090 2
  97. 97. Deisseroth K Singla S Toda H Monje M Palmer T. D Malenka R. C Excitation-neurogenesis coupling in adult neural stem/progenitor cells. Neuron 2004 42 4 535 52
  98. 98. Ge S Goh E. L Sailor K. A Kitabatake Y Ming G. L Song H GABA regulates synaptic integration of newly generated neurons in the adult brain. Nature 2006 439 7076 589 93
  99. 99. Jagasia R Steib K Englberger E Herold S Faus-kessler T Saxe M Gage F. H Song H Lie D. C GABA-cAMP response element-binding protein signaling regulates maturation and survival of newly generated neurons in the adult hippocampus. J Neurosci 2009 29 25 7966 77
  100. 100. Tozuka Y Fukuda S Namba T Seki T Hisatsune T GABAergic excitation promotes neuronal differentiation in adult hippocampal progenitor cells. Neuron 2005 47 6 803 15
  101. 101. Okamoto M Inoue K Iwamura H Terashima K Soya H Asashima M Kuwabara T Reduction in paracrine Wnt3 factors during aging causes impaired adult neurogenesis. Faseb J 2011 25 10 3570 82
  102. 102. Kuhn H. G Dickinson-anson H Gage F. H Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci 1996 16 6 2027 33
  103. 103. Varela-Nallar L Aranguiz F. C Abbott A. C Slater P. G Inestrosa N. C Adult hippocampal neurogenesis in aging and Alzheimer’s disease. Birth Defects Res C Embryo Today 2010 90 4 284 96
  104. 104. Van Praag H Kempermann G Gage F. H Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 1999 2 3 266 70
  105. 105. Wexler E. M Paucer A Kornblum H. I Palmer T. D Geschwind D. H Endogenous Wnt signaling maintains neural progenitor cell potency. Stem Cells 2009 27 5 1130 41
  106. 106. Shi, Y. Chichung Lie, D, Taupin, P, Nakashima, K, Ray, J, Yu, R. T, Gage, F. H, & Evans, R. M. Expression and function of orphan nuclear receptor TLX in adult neural stem cells. Nature 2004 427 6969 78 83
  107. 107. Li W Sun G Yang S Qu Q Nakashima K Shi Y Nuclear receptor TLX regulates cell cycle progression in neural stem cells of the developing brain. Mol Endocrinol 2008 22 1 56 64
  108. 108. Liu H. K Belz T Bock D Takacs A Wu H Lichter P Chai M Schutz G The nuclear receptor tailless is required for neurogenesis in the adult subventricular zone. Genes Dev 2008 22 18 2473 8
  109. 109. Zhang C. L Zou Y He W Gage F. H Evans R. M A role for adult TLX-positive neural stem cells in learning and behaviour. Nature 2008 451 7181 1004 7
  110. 110. Mazumdar , J, O'Brien, W. T, Johnson, R. S, LaManna, J. C, Chavez, J. C, Klein, P. S, & Simon, M. C. O2 regulates stem cells through Wnt/beta-catenin signalling. Nat Cell Biol (2010). , 12(10), 1007-13.
  111. 111. Toledo E. M Colombres M Inestrosa N. C Wnt signaling in neuroprotection and stem cell differentiation. Progress in neurobiology 2008 86 3 281 96
  112. 112. Dickson D. W Apoptotic mechanisms in Alzheimer neurofibrillary degeneration: cause or effect? J Clin Invest 2004 114 1 23 7
  113. 113. Takashima A Noguchi K Sato K Hoshino T Imahori K Tau protein kinase I is essential for amyloid beta-protein-induced neurotoxicity. Proc Natl Acad Sci U S A 1993 90 16 7789 93
  114. 114. Tsai L. H Lee M. S Cruz J Cdk5, a therapeutic target for Alzheimer’s disease? Biochim Biophys Acta (2004). , 1697(1-2), 137-42
  115. 115. Garrido J. L Godoy J. A Alvarez A Bronfman M Inestrosa N. C Protein kinase C inhibits amyloid beta peptide neurotoxicity by acting on members of the Wnt pathway. Faseb J 2002 16 14 1982 4
  116. 116. Inestrosa N. C Alvarez A Godoy J Reyes A De Ferrari G. V Acetylcholinesterase-amyloid-beta-peptide interaction and Wnt signaling involvement in Abeta neurotoxicity. Acta Neurol Scand Suppl 2000 53 9
  117. 117. De Ferrari G. V Chacon M. A Barria M. I Garrido J. L Godoy J. A Olivares G Reyes A. E Alvarez A Bronfman M Inestrosa N. C Activation of Wnt signaling rescues neurodegeneration and behavioral impairments induced by beta-amyloid fibrils. Mol Psychiatry 2003 8 2 195 208
  118. 118. Inestrosa N De Ferrari G. V Garrido J. L Alvarez A Olivares G. H Barria M. I Bronfman M Chacon M. A Wnt signaling involvement in beta-amyloid-dependent neurodegeneration. Neurochem Int 2002 41 5 341 4
  119. 119. De Ferrari, G. V, Papassotiropoulos, A, Biechele, T, Wavrant De-Vrieze, F, Avila, M. E, Major, M. B, Myers, A, Sáez, K, Henríquez, J. P, Zhao, A, Wollmer, M. A, Nitsch, R. M, Hock, C, Morris, C. M, Hardy, J, & Moon, R. T. Common genetic variation within the low-density lipoprotein receptor-related protein 6 and late-onset Alzheimer’s disease. Proc Natl Acad Sci U S A 2007 104 22 9434 9
  120. 120. Alvarez G Munoz-montano J. R Satrustegui J Avila J Bogonez E Diaz-nido J Regulation of tau phosphorylation and protection against beta-amyloid-induced neurodegeneration by lithium. Possible implications for Alzheimer’s disease. Bipolar Disord 2002 4 3 153 65
  121. 121. Caricasole A Copani A Caraci F Aronica E Rozemuller A. J Caruso A Storto M Gaviraghi G Terstappen G. C Nicoletti F Induction of Dickkopf-1, a negative modulator of the Wnt pathway, is associated with neuronal degeneration in Alzheimer’s brain. J Neurosci 2004 24 26 6021 7
  122. 122. Ghanevati M Miller C. A Phospho-beta-catenin accumulation in Alzheimer’s disease and in aggresomes attributable to proteasome dysfunction. J Mol Neurosci 2005 25 1 79 94
  123. 123. Takashima A Murayama M Murayama O Kohno T Honda T Yasutake K Nihonmatsu N Mercken M Yamaguchi H Sugihara S Wolozin B Presenilin 1 associates with glycogen synthase kinase-3beta and its substrate tau. Proc Natl Acad Sci U S A 1998 95 16 9637 41
  124. 124. Zhang, Z, Hartmann, H, Do, V. M, Abramowski, D, Sturchler-Pierrat, C, Staufenbiel, M, Sommer, B, van de Wetering, M, Clevers, H, Saftig, P, De Strooper, B, He, X, & Yankner B. A. Destabilization of beta-catenin by mutations in presenilin-1 potentiates neuronal apoptosis. Nature 1998 395 6703 698 702
  125. 125. Moon R. T Kohn A. D De Ferrari G. V Kaykas A WNT and beta-catenin signalling: diseases and therapies. Nat Rev Genet 2004 5 9 691 701
  126. 126. Rosi M. C Luccarini I Grossi C Fiorentini A Spillantini M. G Prisco A Scali C Gianfriddo M Caricasole A Terstappen G. C Casamenti F Increased Dickkopf-1 expression in transgenic mouse models of neurodegenerative disease. J Neurochem 2010 112 6 1539 51
  127. 127. Caruso A Motolese M Iacovelli L Caraci F Copani A Nicoletti F Terstappen G. C Gaviraghi G Caricasole A Inhibition of the canonical Wnt signaling pathway by apolipoprotein E4 in PC12 cells. J Neurochem 2006 98 2 364 71
  128. 128. Magdesian M. H Carvalho M. M Mendes F. A Saraiva L. M Juliano M. A Juliano L Garcia-abreu J Ferreira S. T Amyloid-beta binds to the extracellular cysteine-rich domain of Frizzled and inhibits Wnt/beta-catenin signaling. J Biol Chem 2008 283 14 9359 68
  129. 129. Alvarez A. R Godoy J. A Mullendorff K Olivares G. H Bronfman M Inestrosa N. C Wnt-3a overcomes beta-amyloid toxicity in rat hippocampal neurons. Exp Cell Res 2004 297 1 186 96
  130. 130. Purro S. A Dickins E. M Salinas P. C The secreted Wnt antagonist Dickkopf-1 is required for amyloid beta-mediated synaptic loss. J Neurosci 2010 32 10 3492 8
  131. 131. Ballard C Gauthier S Corbett A Brayne C Aarsland D Jones E Alzheimer’s disease. Lancet 2011 377 9770 1019 31
  132. 132. Churcher I Tau therapeutic strategies for the treatment of Alzheimer’s disease. Curr Top Med Chem 2006 6 6 579 95
  133. 133. Hooper C Killick R Lovestone S The GSK3 hypothesis of Alzheimer’s disease. J Neurochem 2008 104 6 1433 9
  134. 134. Takashima A Noguchi K Michel G Mercken M Hoshi M Ishiguro K Imahori K Exposure of rat hippocampal neurons to amyloid beta peptide (25-35) induces the inactivation of phosphatidyl inositol-3 kinase and the activation of tau protein kinase I/glycogen synthase kinase-3 beta. Neurosci Lett 1996 203 1 33 6
  135. 135. Takashima A Honda T Yasutake K Michel G Murayama O Murayama M Ishiguro K Yamaguchi H Activation of tau protein kinase I/glycogen synthase kinase-3beta by amyloid beta peptide (25-35) enhances phosphorylation of tau in hippocampal neurons. Neurosci Res 1998 31 4 317 23
  136. 136. Pei J. J Braak E Braak H Grundke-iqbal I Iqbal K Winblad B Cowburn R. F Distribution of active glycogen synthase kinase 3beta (GSK-3beta) in brains staged for Alzheimer disease neurofibrillary changes. J Neuropathol Exp Neurol 1999 58 9 1010 9
  137. 137. Hernandez F Borrell J Guaza C Avila J Lucas J. J Spatial learning deficit in transgenic mice that conditionally over-express GSK-3beta in the brain but do not form tau filaments. J Neurochem 2002 83 6 1529 33
  138. 138. Lucas J. J Hernandez F Gomez-Ramos P Moran M. A Hen R Avila J Decreased nuclear beta-catenin, tau hyperphosphorylation and neurodegeneration in GSK-3beta conditional transgenic mice. EMBO J (2001). , 20(1-2), 27-39.
  139. 139. Li H. L Wang H. H Liu S. J Deng Y. Q Zhang Y. J Tian Q Wang X. C Chen X. Q Yang Y Zhang J. Y Wang Q Xu H Liao F. F Wang J. Z Phosphorylation of tau antagonizes apoptosis by stabilizing beta-catenin, a mechanism involved in Alzheimer’s neurodegeneration. Proc Natl Acad Sci U S A 2007 104 9 3591 6
  140. 140. Garcia-Alloza M Robbins E. M Zhang-nunes S. X Purcell S. M Betensky R. A Raju S Prada C Greenberg S. M Bacskai B. J Frosch M. P Characterization of amyloid deposition in the APPswe/PS1dE9 mouse model of Alzheimer disease. Neurobiol Dis 2006 24 3 516 24
  141. 141. Toledo E. M Inestrosa N. C Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer’s disease. Molecular psychiatry 2010 15 3 272 85
  142. 142. Hu Y. S Xu P Pigino G Brady S. T Larson J Lazarov O Complex environment experience rescues impaired neurogenesis, enhances synaptic plasticity, and attenuates neuropathology in familial Alzheimer’s disease-linked APPswe/PS1DeltaE9 mice. FASEB J 2010 24 6 1667 81
  143. 143. Cerpa W Farias G. G Godoy J. A Fuenzalida M Bonansco C Inestrosa N. C Wnt-5a occludes Abeta oligomer-induced depression of glutamatergic transmission in hippocampal neurons. Mol Neurodegener (2010). , 5, 3.
  144. 144. Selkoe D. J Alzheimer’s disease is a synaptic failure. Science (New York, N.Y.) (2002). 298 5594 789 91

Written By

Nibaldo C. Inestrosa and Lorena Varela-Nallar

Submitted: 30 April 2012 Published: 27 March 2013