Review
Wnt signaling: Role in LTP, neural networks and memory

https://doi.org/10.1016/j.arr.2013.03.006Get rights and content

Highlights

  • Wnt signaling components are expressed in adult brain. The brain distribution of Frizzled receptor is examined in detail.

  • The different effects of Wnt ligands in pre- and postsynaptic region are discussed.

  • Wnt ligands are released in spontaneous and activity dependent processes, and probably play a major role in cognition.

  • Wnts modulate oscillatory activity in neural networks.

  • Wnt signaling stimulate neuroprotection and might be used in the control of neurodegenerative and mental disorders.

Abstract

Wnt components are key regulators of a variety of developmental processes, including embryonic patterning, cell specification, and cell polarity. The Wnt signaling pathway participates in the development of the central nervous system and growing evidence indicates that Wnts also regulates the function of the adult nervous system. In fact, most of the key components including Wnts and Frizzled receptors are expressed in the adult brain. Wnt ligands have been implicated in the regulation of synaptic assembly as well as in neurotransmission and synaptic plasticity. Deregulation of Wnt signaling has been associated with several pathologies, and more recently has been related to neurodegenerative diseases and to mental and mood disorders. In this review, we focus our attention on the Wnt signaling cascade in postnatal life and we review in detail the presence of Wnt signaling components in pre- and postsynaptic regions. Due to the important role of Wnt proteins in wiring neural circuits, we discuss recent findings about the role of Wnt pathways both in basal spontaneous activities as well as in activity-dependent processes that underlie synaptic plasticity. Finally, we review the role of Wnt in vivo and we finish with the most recent data in literature that involves the effect of components of the Wnt signaling pathway in neurological and mental disorders, including a special emphasis on in vivo studies that relate behavioral abnormalities to deficiencies in Wnt signaling, as well as the data that support a neuroprotective role of Wnt proteins in relation to the pathogenesis of Alzheimer's disease.

Introduction

The proto-oncogene int-1 (Wnt-1) was the first member of the Wnt family identified as a common integration site for a mouse mammary tumor virus (Nusse and Varmus, 1982). The int-1 sequence protein proved to be identical to its homologue Drosophila, the Wingless gene, which corresponds to one of the first “segment polarity gene” involved in development as well as in cancer (Nusse and Varmus, 1982, Nusse and Varmus, 2012, Rijsewijk et al., 1987). Later, homologous gene targeting techniques confirmed that knocking down int-1/Wingless family gene causes several developmental impairments in a number of brain regions, corroborating that the Wnt gene is fundamental in determining a normal neural phenotype (McMahon and Moon, 1989b). Now the Wnt family includes 19 members present in mammals and it has been implicated in other major functions, from embryogenesis to different types of cancer, in stem cell development and also in some metabolic disorders such as diabetes mellitus, reflecting its relevance in fundamental biological processes that might be highly observed across species (Clevers and Nusse, 2012).

Although the knowledge about Wnt signaling components and how they are compromised in different phenotypes is vast, there are important aspects of the molecular machinery and how the specificity is established that remains unclear. Historically, Wnt proteins have been classified as either canonical (i.e. Wnt1 and Wnt3a), or non-canonical (i.e. Wnt4, Wnt5 and Wnt11) (Gordon and Nusse, 2006). However, the traditional differentiation between canonical and non-canonical ligands seems to be not as simple as was initially defined, and more aspects should be considered. One of the most important is the recognition that the amount of Wnt ligands and the receptors involved allows enormous possibilities of interactions which are difficult to predict. Moreover, the result of these interactions can differentially activate intracellular cascades, which in turn makes the cellular responses complexes and diverse (van Amerongen and Nusse, 2009). Furthermore, some literature indicates that activation of a canonical or non-canonical pathway in a cell to a given Wnt may depend on the context of how that Wnt couples the receptor and co-receptor, and not by the property of the ligand itself (Grumolato et al., 2010, Mikels and Nusse, 2006b). Finally, it has been shown that Wnt ligands can compete to bind to specific receptors, causing reciprocal pathway inhibition of both signaling pathways (Grumolato et al., 2010). Consequently, the effect of a Wnt ligand depends on a combination of several factors, including the combinatorial probability of binding different types of receptors and co-receptors present in the target cell, which in turn may trigger the activation of a specific intracellular signaling cascade. In the next section we review the details of those interactions.

The prototypical Wnt receptor is the seven transmembrane-receptor Frizzled (Fz). In its structure is a cystein-rich domain (CRD) which forms the Wnt-binding site (Bhanot et al., 1996), ten of which have been described in mice and humans. Despite the fact that the Fz gene shows many redundancies, Fz proteins show a high degree of affinity to every Wnt protein, resulting in a very specific interaction (Rulifson et al., 2000). In addition to Fz, other proteins have been described as alternative co-receptors for Wnt signaling (Fig. 1), such as the single transmembrane low-density lipoprotein receptor-related protein 5/6 (LRP5/6) (MacDonald et al., 2009, Mao et al., 2001, Tamai et al., 2000, Wehrli et al., 2000) and the single-pass transmembrane receptors tyrosine kinases (RTKs) Ror1, Ror2 and Ryk (Cadigan and Liu, 2006, Gordon and Nusse, 2006, Green et al., 2008, Lu et al., 2004, Oishi et al., 2003), increasing the complexity of Wnt signaling activation. But Wnt signaling has also secreted inhibitors or antagonists such as Dickkopf (Dkk) that binds LRP blocking the interaction of Wnt/Fz (Mao et al., 2001), and inhibitors such as the soluble Fz-related protein (sFRP) that bind directly to Wnt because of their similarity to Fz (Finch et al., 1997, Rattner et al., 1997). This natural antagonism could reflect an additional control of Wnt concentration in the extracellular media. This regulation can be crucial during a signal transduction mechanism.

Downstream of Wnt-receptor binding, a sequence of intracellular responses is elicited. The first Wnt signaling pathway identified, the so called “canonical pathway”, has been focus of intense research. The name “canonical” was given because induction of some embryonic structures in Xenopus (McMahon and Moon, 1989a) and the transformation of mouse mammary cells (Wong et al., 1994) were strongly correlated with the increment in β-catenin/TCF levels (Shimizu et al., 1997). The canonical pathway controls growth and cell fate specification (Cadigan and Nusse, 1997). Because dysregulation of β-catenin/TCF levels is present in a variety of cancers, this Wnt pathway has been the most studied to date. As we know now, Wnt1, Wnt3a, Wnt7a and Wnt8 bind the receptor Frizzled and the LRP5/6 co-receptors, activating the Wnt/β-catenin (MacDonald et al., 2009). Both Fz and LRP5/6 recruit the protein dishevelled (Dvl) usually by phosphorylation, which oligomerizes in the plasma membrane forming a platform for the allocation of the scaffold protein Axin and the glycogen synthase kinase-3β (GSK-3β) (Cliffe et al., 2003, Gao and Chen, 2010, Schwarz-Romond et al., 2007). This fact promotes the phosphorylation of LRP6 by casein kinase 1 (CD1) which is associated with each cluster of proteins to form the signalosome (Bilic et al., 2007). The phosphorylation of LRP5/6 causes the inhibition of the “destruction complex” that not only includes GSK-3β but also the tumor suppressor adenomatous polyposis coli (APC). The consequence of this inhibition is the cytoplasmic stabilization of β-catenin which enters the nucleus and regulates the transcription of Wnt target genes (Logan and Nusse, 2004) through the binding of the transcription factors T cell factor and lymphoid enhancer factor (TCF/LEF) (Clevers and Nusse, 2012). Several Wnt target genes are activated in this process, including c-Myc, cyclin D1, Axin2 and Calcium/calmodulin-dependent protein kinase type IV (CamKIV) (Arrazola et al., 2009, Hodar et al., 2010, Nusse and Varmus, 2012, Toledo et al., 2008). Thus, in this review we refer to the Wnt/β-catenin/TCF pathway when we mention the “canonical” pathway.

In the absence of Wnt stimulation, the cytoplasmic levels of β-catenin are low since it is ubiquitinated and constantly degraded in the proteasome (Aberle et al., 1997). β-catenin is also phosphorylated by casein kinase 1α (CK1α) and GSK-3β in a multiprotein complex composed also for Axin and APC (Nusse and Varmus, 2012). β-catenin phosphorylated by CK1α and GSK-3β also targets the armadillo protein for ubiquitination and subsequent degradation (Liu et al., 2002).

Another Wnt pathway identified is the β-catenin-independent pathway (also referred as “non-canonical pathway”). Wnt4, Wnt5a and Wnt11 have been identified as members of this non-canonical pathway because they bind to RTKs co-receptors (Green et al., 2008). There are at least two of them: the planar cell polarity (PCP) pathway and the Ca2+ pathway. The Wnt/PCP pathway was originally identified in Drosophila where it regulates tissue polarity and cell migration during development (Adler, 2002, Veeman et al., 2003). The PCP pathway signals through the JNK pathway to control cell polarity, which is why it is also known as the Wnt/JNK pathway (Boutros et al., 1998, Yamanaka et al., 2002). During activation, Wnt binds the receptor Fz on the membrane surface, and Dvl is followed by the activation of Rho/Rac small GTPase and c-Jun N-terminal kinase (JNK). Particularly, it is required that Wnt5a physically bridges Ror2 to Fz extracellular domains to activate the downstream pathway (Grumolato et al., 2010). Like Fz, Ror1/2 binds Wnt by its CRDs (Grumolato et al., 2010, Logan and Nusse, 2004). The downstream effect of activating this pathway is the regulation of cytoskeletal organization and gene expression (Fig. 1). Instead, the Wnt/Ca2+ pathway is mostly a G-protein dependent signaling pathway (Kohn and Moon, 2005). The activation of the Wnt/Ca2+ pathway requires binding of Wnt to Fz on the membrane surface to trigger stimulation of heterotrimeric G-proteins (Slusarski et al., 1997a, Slusarski et al., 1997b), which activate phospholipase-C (PLC). PLC causes the increment on intracellular release Ca2+, decreases cyclic guanosine monophosphate (cGMP) and activates the protein kinases Ca2+-Calmodulin-dependent Protein kinase II (CaMKII) and protein kinase-C (PKC) (Kohn and Moon, 2005, Montcouquiol et al., 2006, Veeman et al., 2003). This process activates the nuclear factor NFAT and transcription factors like cAMP Response Element-Binding Protein-1 (CREB). Therefore, the classification of individual Wnt proteins in canonical and non-canonical terms is used now to describe the activation of a signaling pathway, either β-catenin/TCF dependent or independent (Mikels and Nusse, 2006a, Mikels and Nusse, 2006b, van Amerongen and Nusse, 2009).

In this review we will only superficially refer to the importance that the Wnt/Frizzled signaling pathway plays in the development of the nervous system. Instead, the purpose of this review is to highlight the fact that much of the literature suggests that Wnt signaling also plays a key role during postnatal life (Chen et al., 2006, Gogolla et al., 2009, Li and Pleasure, 2005, Stranahan et al., 2010, Wayman et al., 2006). In following sections we will review the evidence, showing first that expression of Wnt ligands and proteins of the Wnt signaling machinery occurs in the mature nervous system. Because Shimogori and Cols published a very detailed paper describing the postnatal expression pattern of Wnt, Fz and FRP (Shimogori et al., 2004), we will focus this section mainly on their results.

The expression of Wnt ligands and proteins of the Wnt signaling machinery in the adult brain has been identified in the major subdivisions of the cerebral cortex, from olfactory bulb (OB), hippocampal formation, neocortex and thalamus (Shimogori et al., 2004). In situ hybridization has shown that expression of different Wnts is particularly high in those areas where the neurons are continuously renewed, such as OB and dentate gyrus (DG) in the hippocampus. It has been shown that Wnt5a, Wnt7a and Wnt1 are expressed at least from birth in OB and also in the piriform cortex and other olfactory related areas, and last as late as P20 (Shimogori et al., 2004). Wnt2b is also expressed in piriform, somatosensory and in the entorhinal cortex (EC) of postnatal and young adult mouse brains. Wnt5a has also been found in pre- and parasubiculum and EC until young adulthood (P20). The hippocampi of young adult rodents express several Wnt ligands, including Wnt1, Wnt2, Wnt4, Wnt5a, Wnt7a and b, Wnt8b and Wnt11 (Fig. 2) (Cerpa et al., 2008, Shimogori et al., 2004, Wayman et al., 2006). Particularly, Wnt2 is well expressed in CA1, CA2 and CA3 until the third week after birth, precisely during the period of dendrite development in the hippocampus (Wayman et al., 2006), while the expression of Wnt5a, Wnt7a and Wnt8b in specific layers of DG where postnatal neurogenesis occurs, may be related to their function in this area.

In the neocortex the expression is strikingly particular, with layer- and region-specific expression patterns for Wnt2b, Wnt5a and Wnt7b in the neocortex (Shimogori et al., 2004). Those regions include expression of Wnt2b in close relation to the whisker barrel pattern in layer 4 and 6 of the somatosensory cortex, and also in layer 6 in the visual and auditory cortex in young adulthood. Wnt5a and Wnt7a also appear in layer specific pattern expression in other cortices, including prefrontal, parietal and temporal neocortex (Shimogori et al., 2004).

The presence of Wnt signaling components in the thalamus is also very interesting. The thalamus brings most of the sensory information to the cortex. Gene expression of Wnt machinery appears in the postnatal thalamus with a very conspicuous pattern of specific Wnt along its different sections (Shimogori et al., 2004). Due to the fact that the sensory information is continuously modified during an animal's life, it is possible that Wnt signaling plays an important role in thalamocortical connectivity.

There is also evidence of postnatal expression of Fz receptors. Expression of Fz3 in the olfactory system includes expression from P0 to P7 and in the related areas including piriform cortex until adulthood. In the hippocampus, Fz3 expresses ubiquitously in the pyramidal cell layer and granular cells of DG until adulthood, and during this time is always expressed in EC (Shimogori et al., 2004). Instead, the pattern of Fz3 in the cortex is clear at P20 and ascribes to a layer pattern at different cortices, from prefrontal, cingulated, parietal and temporal, matching with Wnt2b in whisker barrel in the somatosensory cortex (Shimogori et al., 2004).

Fz7 is expressed in olfactory nerves through P20 and then decreases the expression in the piriform cortex. In the hippocampus, Fz7 expresses only in a pyramidal layer at P0, in CA1 and CA3 at P7 and expresses only at CA3 and the entorhinal cortex at P20. There appears to be no postnatal expression of Fz7 in the neocortex (Shimogori et al., 2004).

The remaining Fz receptors, including Fz1, Fz2, Fz8, Fz9 and Fz10, are mainly ubiquitously expressed in the neocortex and hippocampus until young adulthood. Fz1, Fz2 and Fz9 expression resembles Wnt5a expression in OB and DG. Fz4, Fz5 and Fz6 do not show postnatal expression except in the thalamus, showing – like Wnt – a striking cellular- and subregion-defined expression that allows us to speculate about its function (Shimogori et al., 2004).

The presence of Wnt components in many brain areas, including those that are actively participating in neurogenesis, or those involved in sensory processing and superior cognitive processes, suggests that Wnt pathway is involved in more than only structural functions. In the next sections we review the evidence that relates Wnt to active structural and functional processing in mature synapses, and we discuss the implications that this can have in modulating active dependent processes, such as synaptic plasticity.

Wnt ligands have been linked to the assembly of structural components in presynaptic compartments. In the cerebellum, Wnt7a is expressed in granular cells (GC) at the same time as the mossy fiber (MF) axon, which is the presynaptic contact (Lucas and Salinas, 1997). Several changes remodel the connectivity between both areas to increase the contact surface. Wnt7a induces axonal spreading and incremental growth of cone size and branching, leading to the accumulation of synaptic proteins (Hall et al., 2000, Lucas and Salinas, 1997). Wnt7a probably contributes to the formation of active zones because it increases the clustering of synapsin I, a protein located in the presynaptic membrane involved in synapse formation and function (Hall et al., 2000). Because this effect of Wnt7a is mimicked by lithium application, it seems to involve GSK-3β (Lucas and Salinas, 1997). This effect has been blocked by the Wnt antagonist sFRP and a mutant mice deficient in Wnt7a shows a delayed synaptic maturation (Hall et al., 2000). Then in the cerebellum, Wnt7a can act as a retrograde signal from GC to induce presynaptic differentiation in MF, working as a synaptogenic factor (Ahmad-Annuar et al., 2006, Hall et al., 2000). Like Wnt7a, Wnt7b and Wnt3a increase the number of pre-synaptic puncta suggesting a role for these ligands in presynaptic assembly (Ahmad-Annuar et al., 2006, Cerpa et al., 2008, Davis et al., 2008). Wnt7a also increases the clustering of presynaptic proteins such as synaptophysin, synaptotagmin and SV-2, but does not affect postsynaptic clustering of proteins like PSD-95 (Cerpa et al., 2008). Despite Wnt7a clustering induction correlates with β-catenin stabilization, this does not involve Wnt gene target expression – an effect that is also mimicked by Wnt3a. Unexpectedly, GSK-3β is also not required for presynaptic clustering induced by Wnt7a, suggesting that an upstream mechanism is involved (Cerpa et al., 2008). It has been suggested that Wnt7a requires Dvl1 to mediate the normal recycling rate of synaptic vesicles, and the deficiency of both proteins (double null mutant) significantly reduces miniature excitatory postsynaptic current (mEPSC) frequency, an indication of a defect in neurotransmitter release (Ahmad-Annuar et al., 2006). Additionally, the use of FM dyes has shown that Wnt7a stimulates recycling and accelerates exocytosis of synaptic vesicles (Ahmad-Annuar et al., 2006, Cerpa et al., 2008). Moreover, Wnt7a increases the frequency of mEPSCs, suggesting that Wnt7a increases the dynamic of neurotransmitter release (Ahmad-Annuar et al., 2006, Cerpa et al., 2008). Furthermore, Wnt7a/Dvl1 double null mutant mice exhibit reduced mEPSC frequency at the mossy fiber-granule cell synapses, revealing a defect in neurotransmitter release as a consequence of this mutation (Ahmad-Annuar et al., 2006). Electrophysiological recordings on hippocampal rat slices also show that, in the CA3-CA1 synapse Wnt7a, but not Wnt5a, increases the frequency of field excitatory postsynaptic potentials (fEPSP) and decreases the rate of paired pulse facilitation (PPF) (Cerpa et al., 2008), a protocol used to distinguish the involvement of the presynaptic from the postsynaptic terminal (Foster and McNaughton, 1991). In addition, a similar modulation has been shown with nanomolar concentrations of Wnt3a, which modulates the recycling and exocytosis of synaptic vesicles in hippocampal synapses, increasing the frequency of mEPSC through a mechanism that involves Ca2+ entrance from extracellular media (Avila et al., 2010, Cerpa et al., 2008). Most of the ligands that are able to modulate presynaptic differentiation have shown to activate the Wnt/β-catenin signaling pathway. Although Wnt3a has been designated as a classical “canonical” Wnt, the latest evidence suggests a cross-talk between Wnt3a and Wnt/Ca2+ (Avila et al., 2010), strongly supporting the idea that some of the components associated with the non-canonical pathway may be involved in the functionality of the presynaptic nerve terminal.

Wnt7a has been also involved in trafficking of receptors, increasing the number and size of co-clusters of presynaptic α7-nicotinic acetylcholine receptors (α7-nAChR) and APC in hippocampal neurons, as well as in the modulation of the α7-nAChR trafficking to the nerve terminal (Farias et al., 2007), indicating that Wnt pathway components are actively involved in the functional availability of receptors in the synaptic terminal.

Wnt signaling can actively modulate the assembly of the postsynaptic region. Our laboratory has shown strong evidence supporting the role of the β-catenin independent pathway in remodeling of mature synapses. Wnt signaling activation modulates excitatory and inhibitory synaptic transmission (Cerpa et al., 2010, Cerpa et al., 2011, Cuitino et al., 2010). Wnt5a increases the evoked glutamatergic synaptic transmission without changes in the presynaptic compartment, as shown by PPF (Cerpa et al., 2010). Instead, Wnt5a acutely and specifically up-regulates synaptic currents through NMDA receptors, not AMPA receptors, involving PKC and JNK, and facilitating the induction of excitatory LTP (Cerpa et al., 2011). Moreover, Wnt5a is actively modulating inhibitory synapses in the hippocampus, increasing the turnover of GABAA receptors (GABAAR) on the surface, increasing GABA currents by postsynaptic mechanisms that involve Wnt/Ca2+/CaMKII (Cuitino et al., 2010). More evidence indicates that Wnt5a can induce de novo dendrite spine formation and increase the volume and density of pre-existent spines, enhancing the efficacy of hippocampal glutamatergic synapses (Varela-Nallar et al., 2010), suggesting that Wnt5a has a role in synaptic structure and function. Wnt5a can transiently induce formation of dendritic protrusions, which results in a net increase in mature dendrite spines (Varela-Nallar et al., 2010). Interestingly, the treatment with the soluble CRD region of Fz2, which act as a Wnt scavenger, decreases spine density in cultured neurons, supporting the idea that Wnt ligands participates in dendrite spine morphogenesis (Varela-Nallar et al., 2010). Wnt5a also increases calcium recruitment to synaptic puncta of culture cells, suggesting the activation of the Wnt/Ca2+ signaling pathway in cultured hippocampal neurons (Varela-Nallar et al., 2010) through a mechanism that involves fast phosphorylation of CaMKII induced by Wnt5a, as we demonstrated previously (Farias et al., 2009). As it does with GABAAR, Wnt5a modulates postsynaptic assembly increasing the clustering of the postsynaptic density protein-95 (PSD-95) via Wnt/JNK signaling pathway (Farias et al., 2009), inducing a fast increase in the number of PSD-95 clusters without affecting total levels of PSD-95 protein or presynaptic protein clustering in hippocampal cultured neurons (Farias et al., 2009). However, Wnt5a does not only induce protein clustering. It has also been shown that Wnt3a is able to induce recruitment of AChRs in motoneurons at the moment of neuromuscular innervations (Henriquez et al., 2008). This effect requires Dvl1 and Agrin, a protoglycan released by motoneurons, but does not involve the Wnt/β-catenin pathway. Instead, aggregation is induced through activation of Rac1 (Henriquez et al., 2008). However, Wnt3a inhibits Agrin-induced AChR clusters through the activation of the Wnt/β-catenin pathway, suggesting that Wnt signaling dynamically regulates the interaction between postsynaptic components during the establishment of neuromuscular junctions (Wang et al., 2008).

Besides its role in presynaptic functions, Wnt7a has also been shown to regulate the postsynaptic compartment stimulating the morphogenesis and function of excitatory dendritic spines, without affecting the inhibitory connections in the hippocampus (Ciani et al., 2011). To do this, Wnt7a requires Dvl and activation of CamKII at dendritic spines (Ciani et al., 2011). Interestingly, Dvl expressed only in postsynaptic spines and not in innervating presynaptic axons is enough to induce spine growth, suggesting that it is the activation of postsynaptic Wnt signaling which induces spine maturation (Ciani et al., 2011). Moreover, Dvl promotes the assembly of pre- and postsynaptic structures at pre-existing spines because this does not change the number of spines (Ciani et al., 2011). This evidence supports the idea that an extracellular signal such as Wnt7a can generate a divergent intracellular product, using a common molecule such as Dvl to support processes like synaptic differentiation (Gao and Chen, 2010), and a new role for Wnt7a inducing the formation and function of excitatory synapses through CaMKII.

In addition, Wnt7b and Dvl can activate Rac, which in turn stimulates downstream JNK to increase dendritic length and branching in immature hippocampal neurons (Rosso et al., 2005). This effect is independent of Wnt/GSK-3β and is mimicked by over-expression of Dvl. The use of a dominant-negative Rac or dominant-negative JNK also blocks Dvl-dependency of dendritic growth (Rosso et al., 2005). This confirms the activation of the β-catenin independent pathway by Wnt7b, demonstrating that the non-canonical Wnt pathway, the Wnt/JNK, participates in dendrite development (Rosso et al., 2005).

Previous studies showed that β-catenin enhances dendritic arborization and this effect does not require Wnt/β-catenin/TCF dependent transcription (Yu and Malenka, 2003). This effect could be explained depending on the requirements during synaptic formation and maturation. Wnt/β-catenin dependent pathways might be relevant during developmental processes and synapse formation processes, while β-catenin independent signaling might be more relevant during activity-dependent processes, such as synaptic remodeling during plasticity.

Besides the role of Wnt in synaptic differentiation and function, is the role of Fz receptors. It has been shown that Fz1 clusters are co-localized with presynaptic proteins like synapsin-1 (Varela-Nallar et al., 2012) in functional synapses of cultured hippocampal neurons and also with the postsynaptic PSD-95 scaffold (Varela-Nallar et al., 2009). Over-expression of Fz1 increases the number of Bassoon clusters, a protein component of the active zone. The treatment with Wnt3a, a Fz1 ligand, also induces presynaptic clustering, increasing presynaptic recycling sites and the kinetics of vesicle release. These effects are prevented by incubation with CRD of Fz1 that acts as a scavenger (Varela-Nallar et al., 2009). Thus, Wnt acting through Fz1 modulates synaptic differentiation and function (Varela-Nallar et al., 2009). A more detailed study has shown that different Fz expresses differently along hippocampal development, from E18 to P60 (Varela-Nallar et al., 2012) and some but not all have synaptic distribution in culture. In fact, some of them, like Fz7 and Fz 9, localize in soma and all the processes, while Fz9 and Fz5 also concentrate in the growth cone (Varela-Nallar et al., 2012). The particular temporal and location pattern of Fz could amplify the range of possible interactions during development and later during maturation, but also brings specificity to the interaction of each different Wnt. More studies are required to elucidate how and when these pathways become relevant for the system. In the next section, we address some of these questions by studying the physiological processes involved in the interaction of multiple cells during activity and non-activity dependent processes.

Section snippets

Wnt signaling in basal neuronal activity

The evidence we detailed in previous sections has related Wnt signaling pathways to the regulation of synaptic activity, increasing dendritic arborization and morphogenesis in the hippocampus (Ciani and Salinas, 2005, Varela-Nallar et al., 2010, Wayman et al., 2006). In this section we focus on the evidence that involves endogenous Wnt signaling pathways having a role in basal synaptic neurotransmission. In the next section we will focus on the evidence of Wnt involvement in activity-dependent

Wnt signaling in brain functions: From plasticity to cognitive processes

Wnt modulates synaptic and morphological plasticity and its receptors are expressed in areas of the brain that undergo plasticity. As we observed, Wnt is spontaneously released by cells but activity may also control its secreted levels, regulating synaptic transmission and neuronal growth. It is not surprising that dendrites are one of the targets for Wnt remodeling. Dendritic spines have been found to be one of the first visible effectors of plasticity (experience)-induced changes (Bosch and

Wnt signaling dysfunction in neurodegenerative and mental disorders

Increasing evidence suggests that disease progression is a consequence of a cascade of dysfunction effects at multiple levels of the network organization which in many cases precedes cell degeneration or death (Hermann et al., 2009, Hickey et al., 2008, Palop and Mucke, 2010a). Major neurological diseases such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD) are all progressive disorders with common symptoms: a range of neuropsychiatric features, massive

Concluding remarks

In this review we presented evidence that supports a strong modulatory effect of Wnt signaling pathways in mature synapses. The intracellular signaling cascades are activated as a consequence of a specific Wnt/Fz/co-receptor interaction, causing specific cellular responses not only in the spontaneous interaction among neurons but also during activity-dependient processes. Finally, we showed that there is a fine equilibrium that has common elements, such as GSK-3β or Dvl, key factors for several

Conflicts of interest

The authors declare that they have no conflicts of interest.

Acknowledgments

This work was supported by the Basal Center of Excellence in Science and Technology CONICYT-CARE (PFB 12/2007) and Fondecyt No. 11201 to NCI. A postdoctoral fellowship from MECESUP to CAO is also thanked.

References (204)

  • M. Boutros et al.

    Dishevelled activates JNK and discriminates between JNK pathways in planar polarity and wingless signaling

    Cell

    (1998)
  • V. Budnik et al.

    Wnt signaling during synaptic development and plasticity

    Current Opinion in Neurobiology

    (2011)
  • J. Busciglio et al.

    Beta-amyloid fibrils induce tau phosphorylation and loss of microtubule binding

    Neuron

    (1995)
  • G. Buzsaki

    Theta oscillations in the hippocampus

    Neuron

    (2002)
  • W. Cerpa et al.

    Wnt-7a modulates the synaptic vesicle cycle and synaptic transmission in hippocampal neurons

    Journal of Biological Chemistry

    (2008)
  • J. Chen et al.

    Activity-dependent synaptic Wnt release regulates hippocampal long term potentiation

    Journal of Biological Chemistry

    (2006)
  • M.J. Chen et al.

    Exercise activates the phosphatidylinositol 3-kinase pathway

    Brain Research Molecular Brain Research

    (2005)
  • H. Clevers et al.

    Wnt/beta-catenin signaling and disease

    Cell

    (2012)
  • A. Cliffe et al.

    A role of Dishevelled in relocating Axin to the plasma membrane during wingless signaling

    Current Biology

    (2003)
  • R.S. Duman et al.

    Signaling pathways underlying the pathophysiology and treatment of depression: novel mechanisms for rapid-acting agents

    Trends in Neurosciences

    (2012)
  • G.G. Farias et al.

    Wnt-5a/JNK signaling promotes the clustering of PSD-95 in hippocampal neurons

    Journal of Biological Chemistry

    (2009)
  • C. Gao et al.

    Dishevelled: the hub of Wnt signaling

    Cellular Signalling

    (2010)
  • N. Gogolla et al.

    Wnt signaling mediates experience-related regulation of synapse numbers and mossy fiber connectivities in the adult hippocampus

    Neuron

    (2009)
  • M.D. Gordon et al.

    Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors

    Journal of Biological Chemistry

    (2006)
  • J.L. Green et al.

    Ror receptor tyrosine kinases: orphans no more

    Trends in Cell Biology

    (2008)
  • C.A. Grimes et al.

    The multifaceted roles of glycogen synthase kinase 3beta in cellular signaling

    Progress in Neurobiology

    (2001)
  • A.C. Hall et al.

    Axonal remodeling and synaptic differentiation in the cerebellum is regulated by WNT-7a signaling

    Cell

    (2000)
  • C. Han et al.

    Shifted from Wnt to Hedgehog signaling pathways

    Molecular Cell

    (2005)
  • D. Hermann et al.

    Synaptic transmission is impaired prior to plaque formation in amyloid precursor protein-overexpressing mice without altering behaviorally-correlated sharp wave-ripple complexes

    Neuroscience

    (2009)
  • M.A. Hickey et al.

    Extensive early motor and non-motor behavioral deficits are followed by striatal neuronal loss in knock-in Huntington's disease mice

    Neuroscience

    (2008)
  • N. Inestrosa et al.

    Wnt signaling involvement in beta-amyloid-dependent neurodegeneration

    Neurochemistry International

    (2002)
  • R.S. Jope et al.

    The glamour and gloom of glycogen synthase kinase-3

    Trends in Biochemical Sciences

    (2004)
  • F. Kamenetz et al.

    APP processing and synaptic function

    Neuron

    (2003)
  • A.D. Kohn et al.

    Wnt and calcium signaling: beta-catenin-independent pathways

    Cell Calcium

    (2005)
  • J. Larson et al.

    Patterned stimulation at the theta frequency is optimal for the induction of hippocampal long-term potentiation

    Brain Research

    (1986)
  • H. Aberle et al.

    Beta-catenin is a target for the ubiquitin-proteasome pathway

    EMBO Journal

    (1997)
  • E. Abramov et al.

    Amyloid-beta as a positive endogenous regulator of release probability at hippocampal synapses

    Nature Neuroscience

    (2009)
  • A. Ahmad-Annuar et al.

    Signaling across the synapse: a role for Wnt and Dishevelled in presynaptic assembly and neurotransmitter release

    Journal of Cell Biology

    (2006)
  • H. Alimohamad et al.

    The effects of antipsychotics on beta-catenin, glycogen synthase kinase-3 and dishevelled in the ventral midbrain of rats

    Journal of Neurochemistry

    (2005)
  • J.C. Amatniek et al.

    Incidence and predictors of seizures in patients with Alzheimer's disease

    Epilepsia

    (2006)
  • M.S. Arrazola et al.

    Calcium/calmodulin-dependent protein kinase type IV is a target gene of the Wnt/beta-catenin signaling pathway

    Journal of Cellular Physiology

    (2009)
  • J.M. Beaulieu et al.

    Akt/GSK3 signaling in the action of psychotropic drugs

    Annual Review of Pharmacology and Toxicology

    (2009)
  • J.M. Beaulieu et al.

    Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade

    Proceedings of the National Academy of Sciences of the United States of America

    (2004)
  • J.M. Beaulieu et al.

    Role of GSK3 beta in behavioral abnormalities induced by serotonin deficiency

    Proceedings of the National Academy of Sciences of the United States of America

    (2008)
  • Y. Ben-Ari et al.

    Giant synaptic potentials in immature rat CA3 hippocampal neurones

    Journal of Physiology

    (1989)
  • P. Bhanot et al.

    A new member of the frizzled family from Drosophila functions as a Wingless receptor

    Nature

    (1996)
  • J. Bilic et al.

    Wnt induces LRP6 signalosomes and promotes dishevelled-dependent LRP6 phosphorylation

    Science

    (2007)
  • E.M. Blalock et al.

    Incipient Alzheimer's disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses

    Proceedings of the National Academy of Sciences of the United States of America

    (2004)
  • C.L. Busceti et al.

    Induction of the Wnt inhibitor, Dickkopf-1, is associated with neurodegeneration related to temporal lobe epilepsy

    Epilepsia

    (2007)
  • K.M. Cadigan et al.

    Wnt signaling: complexity at the surface

    Journal of Cell Science

    (2006)
  • Cited by (78)

    • Role of Wnt signaling in synaptic plasticity and memory

      2022, Neurobiology of Learning and Memory
      Citation Excerpt :

      These findings are reinforced by the fact that Dvl levels are increased in the presynaptic terminals of olfactory sensor neurons (Rodriguez-Gil et al., 2013). Additionally, Wnt-7a levels in the presynaptic terminal increase clustering of synaptotagmin, synaptophysin and SV-2 (Cerpa et al., 2008), which are involved in synaptic stabilization (Oliva et al., 2013a). Alternatively, Wnt-5a activation of the noncanonical Wnt/Ca++ pathway, results in postsynaptic alterations (Oliva et al., 2013b).

    • Post-translational modifications: Regulators of neurodegenerative proteinopathies

      2021, Ageing Research Reviews
      Citation Excerpt :

      In this way, the β-catenin destruction complex maintains a low level of β-catenin in the cytoplasm during the absence of Wnt ligands, thus promoting transcriptional inactivation of Wnt signal-related genes. However, when Wnt ligands bind to its seven-transmembrane Frizzled receptors and low-density lipoprotein receptor-related protein 5/6 (LRP 5/6) co-receptors, it activates an intracellular signal cascade, which deactivates the β-catenin destruction complex via interaction with Dishevelled (Dsh) protein, leading to stabilization and accumulation of β-catenin in the cytoplasm (Oliva et al., 2013). β-catenin then moves into the nucleus where it interacts with T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors leading to transcription/expression of Wnt target genes (Berwick and Harvey, 2012).

    • Wnt signaling and dementia

      2020, Genetics, Neurology, Behavior, and Diet in Dementia: The Neuroscience of Dementia, Volume 2
    View all citing articles on Scopus
    View full text