You are on page 1of 11

Neuropharmacology 62 (2012) 78e88

Contents lists available at SciVerse ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Review

The dynamic role of beta-catenin in synaptic plasticity


Kimberly A. Maguschaka, Kerry J. Resslerb, c, *
a
Massachusetts Institute of Technology, USA
b
Howard Hughes Medical Institute, USA
c
Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Yerkes National Primate Research Center, USA

a r t i c l e i n f o a b s t r a c t

Article history: In addition to its role in development and cell proliferation, b-catenin has been implicated in neuronal
Received 10 May 2011 synapse regulation and remodeling. Here we review basic molecular and structural mechanisms of
Received in revised form synaptic plasticity, followed by a description of the structure and function of b-catenin. We then describe
20 August 2011
a role for b-catenin in the cellular processes underlying synaptic plasticity. We also review recent data
Accepted 22 August 2011
demonstrating that b-catenin mRNA and protein phosphorylation are dynamically regulated during fear
memory consolidation in adult animals. Such alterations are correlated with a change in the association
Keywords:
of b-catenin with cadherin, and deletion of the b-catenin gene prevents fear learning. Overall, the extant
Fear
Amygdala
data suggest that b-catenin may function in mediating the structural changes associated with memory
Synaptic plasticity formation. This suggests a general role for b-catenin in synaptic remodeling and stabilization underlying
PTSD long-term memory in adults, and possible roles for dysfunction in the b-catenin pathway in disorders of
Gene memory impairment (e.g. Alzheimer’s Disease) and in disturbances in which emotional memories are too
Anxiety strong or resistant to inhibition (e.g. fear learning in Posttraumatic Stress Disorder). Further under-
standing of the b-catenin pathway may lead to better appreciation for the structural mechanisms
underlying learning and memory as well as provide novel therapeutic approaches in memory related
disorders.
This article is part of a Special Issue entitled ‘Anxiety and Depression’.
Ó 2011 Elsevier Ltd. All rights reserved.

1. Introduction Understanding molecular mechanisms of learning, memory and


synaptic plasticity promise to further our understanding and
The search to understand the mechanisms underlying learning treatment approaches for disorders of decreased memory such as
and memory has been a topic of interest for more than a century. Alzheimer’s and other dementias, as well as disorders of dysregu-
Dating back to the nineteenth century, Cajal proposed that learning lated emotional memory such as posttraumatic stress disorder,
requires the formation of new neuronal connections (Ramon y depression and bipolar disorders.
Cajal, 1894). Later, based on these and other early speculations,
Konorski and Hebb independently suggested that alterations 1.1. Molecular mechanisms of synaptic plasticity
in synaptic strength, as well as formation of new synapses, are
responsible for information storage (Hebb, 1949; Konorski, 1948). Synaptic plasticity in mature neurons is often initiated by neural
The ability of connections between neurons to change as a function activity and an influx of calcium. Calcium influx can lead to alterations
of learning is commonly referred to as synaptic plasticity, and this is in synapse structure and function through a process involving post-
accepted today as a mechanism underlying memory formation. translational modification, protein synthesis and gene transcription.
Such alterations in synaptic connections can be produced through Following presynaptic activity and postsynaptic depolarization,
artificial processes, such as long-term potentiation (LTP), or calcium enters the postsynaptic neuron through N-methyl-D-aspartic
more natural processes, such as behavioral learning in animals. acid (NMDA)-type glutamate receptors (NMDARs) and voltage-gated
calcium channels (VGCCs) (Collingridge and Bliss, 1995; Magee and
Johnston, 1995; Sabatini et al., 2001). Elevated intracellular calcium
levels then activate additional signaling pathways, including
* Corresponding author. Department of Psychiatry and Behavioral Sciences,
Yerkes Research Center, Emory University, 954 Gatewood Dr, Atlanta, GA 30329,
calmodulin (CaM)-dependent protein kinases (CaMKs) (Lisman et al.,
USA. Tel.: þ1 404 727 7739; fax: þ1 404 727 8070. 2002; Tanaka and Nishizuka, 1994). Activation of these protein
E-mail address: kressle@emory.edu (K.J. Ressler). kinases, subsequently leads to the phosphorylation of AMPA-type

0028-3908/$ e see front matter Ó 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.neuropharm.2011.08.032
K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88 79

glutamate receptors (AMPARs), along with the activation of protein Takeichi, 2008) as illustrated in Fig. 1. This cadherinecatenin
synthesis, and initiation of new gene transcription. These cellular complex is localized in synaptic junctions, and alterations in this
processes, which contribute to synaptic plasticity, begin within a few complex are thought to influence not only synapse development,
minutes following neural activity, but can persist for several hours. but also synaptic connectivity and activity (Takeichi and Abe, 2005).
Although a-catenin links to the actin cytoskeleton, the role of
1.2. Morphological changes associated with synaptic plasticity b-catenin in this cadherinecatenin complex is more pronounced
and has been shown to be a prerequisite for adhesion.
Rapid structural changes, involving the rearrangement of the
cytoskeleton at the synapse, can also occur with synaptic plasticity. 2. b-Catenin structure and function
Most excitatory synapses in the brain terminate on dendritic spines
(Yuste and Bonhoeffer, 2004). Dendritic spines are tiny protrusions 2.1. Structure of b-catenin
on the shaft of dendrites, that represent sites where new contacts
between cells can be created, and existing contacts strengthened. b-Catenin belongs to the armadillo family of proteins, and is
The cytoskeleton of dendritic spines contains high concentrations composed of three domains: an N-terminal domain, a central
of filamentous actin (F-actin) (Fifkova and Delay, 1982). The domain, and a C-terminal domain (Pokutta and Weis, 2007). The
dynamic nature of the actin permits the spine to change shape central domain is the core region and contains 12 copies of a 42
within seconds to minutes, thereby contributing to synaptic plas- amino acid sequence motif known as an armadillo repeat. Origi-
ticity. The resulting change in spine shape can last for hours or even nally identified in Drosophila, this armadillo repeat domain is
days, and may influence synaptic transmission. specialized for proteineprotein binding, and forms a superhelix of
In addition to actin, spines also contain a multi-protein complex, helices that features a long, positively charged groove (Huber et al.,
the postsynaptic density (PSD), which includes receptors, channels, 1997). b-catenin’s binding partners, including cadherins, adeno-
cell adhesion proteins, and other signaling molecules (Okabe, matosis polyposis coli (APC), and T-cell factor (TCF), are negatively
2007). All of these components play a role in mediating spine charged, and are proposed to interact with this groove.
changes. Such changes may be characterized by an increase in the
number of spines, or in the overall shape of the spine (Steiner et al., 2.2. Functional roles of b-catenin
2008). Changes in spine number and form have been observed
following many different experimental and behavioral conditions. As mentioned earlier, b-catenin provides an essential, structural
For example, induction of LTP in hippocampal slice cultures has component of the cadherin/catenin adhesion complex. It is neces-
been shown to produce new spine formation, increase the size of sary to prevent the rapid degradation of the cadherin cytoplasmic
the spine head, and shorten the length of the spine neck (Engert domain (Huber et al., 2001) and to recruit a-catenin to sites of
and Bonhoeffer, 1999; Fifkova and Anderson, 1981; Yang et al., cellecell contact (Drees et al., 2005; Yamada et al., 2005). Without
2008). Similarly, changes in spine morphology have been shown the interaction between b-catenin and cadherin, cellecell adhesion
in vivo with trace eyeblink conditioning (Leuner et al., 2003) and would be compromised (For recent review see: Heuberger and
fear conditioning (Ostroff et al., 2010). Birchmeier (2010) and Nelson (2008)).
In addition to b-catenin’s role in cell adhesion, it also is a central
1.3. Cell adhesion molecules and synaptic plasticity player in the Wnt/b-catenin signal transduction pathway (For
recent review see: MacDonald et al. (2009); Rao and Kühl (2010);
Along with structural changes in dendritic spines, synaptic Verkaar and Zaman (2011)). Wnts are highly conserved secreted
plasticity also results in the formation of new synaptic contacts. glycoproteins that regulate cellecell communication, and are
During this process, the adhesion between the pre- and post- involved in a diverse array of cellular processes including devel-
synaptic neurons is altered. The strengthening and weakening of opment. When Wnt proteins bind to Frizzled (Fz) and low-density
these contacts can be modulated by cell adhesion molecules. These
molecules are bound to the membrane and contain an extracellular
domain that engages in either homophilic or heterophilic interac-
tions with similar cell adhesion molecules, or other proteins in the
extracellular matrix, respectively. They also contain an intracellular
domain that interacts with the cytoskeleton and triggers signaling
pathways that can regulate spine and synapse formation (Dityatev
et al., 2004). Examples of such molecules include neurexins and
neuroligins, the SALM family of adhesion molecules, synaptic cell
adhesion molecules (SynCAM), neural cell adhesion molecules
(NCAM), and cadherins (see Giagtzoglou et al., 2009 for review). In
this review, we will focus on the role of the cadherin, and more
specifically, its partner b-Catenin, in synapse formation and
organization.
Cadherins are homophilic, calcium-dependent cell adhesion
molecules that have been shown to play a role in synapse assembly,
synaptic plasticity, and memory formation (Bruses, 2006; Takeichi,
2007). They contain an extracellular domain which provides a link
between opposing cells, promoting structural stability, and an
intracellular domain which provides a link to the actin cytoskel- Fig. 1. b-catenin at the synapse. Schematic diagram showing that the intracellular
eton, promoting spine dynamics. Considerable attention has been domain of cadherin binds to b-Catenin, along with a number of intermediate proteins,
which then links cadherin to the actin cytoskeleton via a-catenin. Please note that this
given to the latter. The intracellular domain of cadherins binds to is an extremely over-simplified diagram. The cadherin adhesion complex’s interaction
b-Catenin which then links cadherins, along with an intermediate with the actin cytoskeleton is a dynamic structure that includes other intermediate
protein complex, to the actin cytoskeleton via a-catenins (Abe and proteins that are outside of the scope of this review.
80 K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88

lipoprotein-related protein (LRP) receptors, disheveled (Dsh) is Kimelman, 2007). Since Wnt signaling generates a folded-back
recruited to the membrane. Activation of Dsh by Fz inhibits form of b-catenin, it is possible that some of the repeats may no
glycogen synthase kinase-3b (GSK-3b), a kinase that phosphory- longer be exposed, thus blocking the binding site for cadherin.
lates b-catenin and marks it for degradation by the proteasome Although many of the details of these differential interactions and
pathway. Through the inhibition of GSK-3b, b-catenin is stabilized, pathways remain unknown, it is clear that b-catenin plays a critical
enters the nucleus, and then forms a complex with the TCF/ function as a ‘hub’ of neuronal plasticity, mediating intracellular
lymphocyte enhancer factor (LEF) family of transcription factors to signaling that results both in structural changes underlying
regulate the expression of Wnt target genes (Logan and Nusse, synaptic strength and in regulating expression of activity-related
2004; Moon et al., 2004). This process is important for cell prolif- genes.
eration, survival, migration, and differentiation, and as suggested
by recent research, modulation of synaptic plasticity (Chen et al., 2.3. Post-translational modifications regulate b-catenin function
2006). Recent work also suggests a role for b-catenin in learning
and memory processes in animals (Maguschak and Ressler, 2008). In addition to structural changes in b-catenin, post-translational
Fig. 2 illustrates these different functional roles of b-catenin. modifications have also been reported to affect the interaction
There has been much discussion as to whether there is crosstalk between b-catenin and its binding partners. For example, phos-
between cadherin-mediated cell adhesion and canonical Wnt phorylation of b-catenin at tyrosine 654 (located on the 12th
signaling through b-Catenin. Experiments involving genetic armadillo repeat) by src and EGFR, decreases its affinity for cad-
modification or overexpression in embryos have suggested that the herin binding, and reduces its adhesive functions (Hoschuetzky
same pool of b-catenin is involved in both cell adhesion at the et al., 1994; Roura et al., 1999; Takahashi et al., 1997). Cadherin/
plasma membrane and signal transduction in the nucleus (Reiss b-catenin binding may also be influenced by kinases involved in the
et al., 2005). However, others have shown that there are different signaling form of b-catenin, including casein kinase 1 (CK1; Dupre-
pools of b-Catenin: 1) a monomeric, intramolecularly folded-back Crochet et al., 2007), GSK-3b (Maher et al., 2009), and CK2 (Wu
form that is generated by Wnt signaling and binds only to TCF et al., 2009; Bek and Kemler, 2002).
transcriptional complexes, and 2) a separate pool that exists as Aside from being bound to cadherin at the membrane, b-catenin
a heterodimer with a-catenin, and preferentially binds to cadherins can also be found in the cytosol. This less stable pool of b-catenin is
(Gottardi and Gumbiner, 2004). One explanation for this potential continuously phosphorylated by a dual kinase mechanism. First,
selectivity may be different requirements for cadherin and TCF to CK1a phosphorylates b-catenin at serine 45. This event primes
bind b-catenin. X-ray crystallography studies have shown that b-catenin for further phosphorylation by GSK-3b at residues serine
cadherins require all 12 armadillo repeats of b-catenin, while 33, serine 37, and threonine 41 (Liu et al., 2002; Yost et al., 1996).
TCF only requires the central eight armadillo repeats (Xu and Upon phosphorylation at these sites, b-catenin is ubiquitinated and

Fig. 2. b-catenin in Wnt signaling and the cadherin complex. In the resting state, b-catenin is phosphorylated by GSK-3b and rapidly degraded by the proteasome pathway. Upon
activation of Wnt signaling, b-catenin is stabilized through the inhibition of GSK-3b and translocates to the nucleus to regulate the expression of Wnt target genes. b-catenin also
associates with the cytoplasmic domain of cadherin and directly links to the actin cytoskeleton through a-catenin. Note that the cadherin adhesion complex is a dynamic structure
that also includes other intermediate proteins (such as EPLIN, Abe and Takeichi, 2008).
K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88 81

rapidly degraded by the 26S proteasome. As mentioned earlier, Table 1


Wnt signaling can inhibit this phosphorylation-dependent degra- Effects of loss of function or overexpression of b-catenin on presynaptic and post-
synaptic function.
dation by inhibiting GSK-3b activity. Consequently, there is an
increase in the unphosphorylated form of b-catenin, which has Manipulation Effect Reference
been shown to accumulate more readily in the nucleus, and is Presynaptic Role
associated with increased transcriptional activity. LOF Increase in diffusion of vesicles Bamji et al., 2003
along synapse
Once b-catenin enters the nucleus, it binds to TCF, and recruits
LOF Decrease in number of synaptic Bamji et al., 2003
complexes that promote transcriptional activation. These complexes vesicles per synapse
can be recruited by both the N- and C-termini of b-catenin. Phos- LOF Impairment in response to prolonged Bamji et al., 2003
phorylation of b-catenin by the Met receptor at N-terminal residue repetitive stimulation
tyrosine 142 (Y142), promotes the association between BCL9-2 and LOF No change in PSD-95 Bamji et al., 2003
Postsynaptic Role
b-catenin. BCL9-2 has been shown to increase nuclear location of LOF Increase in thin, elongated spines Okuda et al., 2007
b-catenin, and as a consequence, increase signaling (Brembeck et al., LOF Decrease in dendritic arborization Yu and Malenka, 2003;
2004). Since BCL9-2 cannot co-localize with the stable pool of Gao et al., 2007
b-catenin bound to cadherin at the plasma membrane, BCL9-2 may LOF Decrease in mEPSC amplitude Okuda et al., 2007
O/E Increase in dendritic arborization Krichmar et al., 2006;
play a role in determining whether b-catenin molecules favor the
Peng et al., 2009;
cell adhesion or Wnt signaling pathways. Yu and Malenka, 2003
Phosphorylation of b-catenin at the C-terminal can also affect O/E Decrease in mEPSC amplitude Peng et al., 2009
transcriptional activation. It has been shown that AKT phosphory- O/E Decrease in AMPAR density Peng et al., 2009
lates b-catenin at residue serine 552, resulting in its dissociation LOF: Loss of function, O/E: Overexpression.
from cellecell contacts and an accumulation of b-catenin in both
the cytosol and nucleus (Fang et al., 2007). Similarly, PKA-mediated
phosphorylation of b-catenin at residue serine 675 has been shown effect does not appear to be due to postsynaptic components since
to enhance the transcriptional activity of b-catenin (Taurin et al., examination of the shape and distribution of postsynaptic density
2006). (PSD)-95, a marker of postsynaptic densities, remains unaffected
The aforementioned studies suggest that post-translational (Bamji et al., 2003). The decrease in the number of synaptic vesicles
modifications can impact the interaction of b-catenin with both seems to be specific to undocked vesicles, those vesicles in the
cadherin and TCF, or other transcriptional co-activators. In turn, reserved/resting pool, as opposed to docked vesicles, those corre-
alterations in the affinity of b-catenin for these binding partners sponding to the readily releasable pool. A decrease in the reserved/
may influence b-catenin’s ability to take part in cell adhesion, Wnt resting pool is complemented by an impaired response to pro-
signaling, or both. Nevertheless, both processes require b-catenin, longed repetitive stimulation, and corresponds to a dispersion of
and have been shown to modulate synaptic plasticity, thus sug- vesicles along the axon. The C-terminal domain of b-catenin, which
gesting that b-catenin may be a candidate molecule to study contains a PDZ target sequence, may be responsible for presynaptic
learning and memory. vesicle localization (Bamji et al., 2003). Evidence suggests that the
PDZ-binding motif of b-catenin allows it to act as a scaffolding
2.4. Role of b-catenin in presynaptic structure and function protein to link cadherins to PDZ domain-containing proteins,
retaining vesicles at discrete sites. One protein that has recently
b-catenin is expressed in the developing and adult CNS. It can be been shown to interact with the PDZ-binding motif of b-catenin is
found in both pre- and postsynaptic cells and appears to be present scribble (Sun et al., 2009). The interaction between scribble and
before a synapse becomes functional. Due to the localization of this b-catenin, whether it is direct or indirect, may mediate b-catenin’s
protein prior to synapse formation, b-catenin stands out as role in synaptic vesicle localization.
a protein that may play a role in synapse assembly. Over the past In addition to the PDZ-binding motif, the phosphorylation state
several years, there has been much evidence suggesting that of b-catenin at tyrosine 654 (Y654) may also affect synaptic vesicle
b-catenin may be acting both pre- and postsynaptically to regulate localization. As stated earlier, phosphorylation of b-catenin at Y654
synapse formation and function (see Table 1 for an overview). decreases its affinity for cadherin (Roura et al., 1999). Recent
The presynaptic axon contains an active zone, where synaptic evidence has shown that phosphorylation of b-catenin at the Y654
vesicles dock and fuse to the plasma membrane (Zhai et al., 2001). residue can be promoted by application of brain-derived neuro-
Presynaptic molecules associated with synaptic vesicle proteins are trophic factor (BDNF) to cultured hippocampal neurons (Bamji
also recruited to the active zone, and aid in the transformation of et al., 2006). BDNF is a neurotrophin that is well known to func-
nascent presynaptic sites to functional presynaptic structures (Ziv tion in synaptic plasticity and regulate synaptic morphology
and Garner, 2004). Originally synaptic proteins were thought to (Greenberg et al., 2009). Upon application of BDNF, the cadherineb-
be relatively stable in mature synapses; however, more recent catenin complex is disrupted, and an enhancement in synaptic
studies have suggested that these proteins are highly mobile and vesicle mobility is observed. The dispersal of synaptic vesicles into
shuffle into and out of individual synapses (Bamji et al., 2006; perisynaptic regions can be abolished by preventing the phos-
Krueger et al., 2003). The cadherin/b-catenin complex has previ- phorylation at this residue by a b-catenin point mutation (Bamji
ously been shown to play a role in the recruitment and localization et al., 2006).
of synaptic vesicles to synapses (Iwai et al., 2002; Togashi et al., An effect on synaptic vesicle localization by phosphorylation of
2002; Stan et al., 2010). More recently, it has been determined b-catenin at Y654 can also be observed following manipulation of
that interfering with b-catenin itself can affect synapse assembly Fer (Lee et al., 2008). Fer is a cytoplasmic tyrosine kinase that is
(Bamji et al., 2006, 2003). known to act in several signaling pathways, including cell adhesion
b-Catenin is important for controlling the size and localization molecule-regulated signaling (Xu et al., 2004). Depletion of Fer
of vesicle clusters. Deletion of b-catenin in hippocampal pyramidal using small hairpin RNAs (shRNAs) in cultured hippocampal
neurons has been associated with a decrease in the number of neurons results in an increase in the motility of presynaptic clus-
synaptic vesicles per synapse, and an increase in the diffusion of ters, along with an increase in tyrosine phosphorylated b-catenin
these vesicles along the synapse (Bamji et al., 2006, 2003). This (Lee et al., 2008). This dispersion of synaptic vesicle clusters could,
82 K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88

once again, be prevented by overexpressing a mutant form of transcription factors, along with the C-terminal PDZ-binding
b-catenin that prevents phosphorylation at Y654. Overall, these motif, are important for regulating synaptic AMPARs (Okuda
results suggest that b-catenin acts presynaptically to control et al., 2007).
synaptic vesicle localization. Overexpression of b-catenin also reduces mEPSC amplitudes
and is accompanied by a decrease in surface AMPAR cluster size
2.5. Role of b-catenin in postsynaptic structure and function and density. However, the decrease in AMPA receptor density does
not appear to coincide with a decrease in synapse density (Peng
The role of b-catenin in postsynaptic shape and function has also et al., 2009). This incongruity can be explained by an observed
been studied. As mentioned earlier, the dendritic spines on the increase in the NMDAR/AMPAR ratio, which is indicative of silent,
postsynaptic neuron are the sites where most excitatory synapses or inactive, synapses. Furthermore, the physiological changes
take place. Spines can generally be classified by their shape and occur in parallel with the aforementioned increases in dendritic
volume as thin, stubby, or mushroom-like. The different shapes of arborization following neural activity (Peng et al., 2009). These
dendritic spines are thought to represent strength and maturity, results suggest that changes in dendritic morphology may coor-
where thin spines are immature, and mushroom shaped are dinate with excitatory synaptic strength to regulate synaptic
mature. b-catenin has been shown to be important in regulating scaling. Altogether, these results show that b-catenin may act
spine shape and size. Without postsynaptic b-catenin, there is an postsynaptically to couple the structure and function of excitatory
increase in thin, elongated spines and subsequent decrease in short synapses.
mushroom-shaped spines (Okuda et al., 2007). Although ablation
of postsynaptic b-catenin alters spine morphology, no changes in 3. b-Catenin, learning and memory
density of presynaptic markers were observed. Thus, these results
suggest that postsynaptic b-catenin ablation does not prevent 3.1. Activity-dependent regulation of b-catenin
neurons from maintaining presynaptic inputs.
Alterations in the dendrites themselves have also been observed Neural activity alters the localization of b-catenin at synapses,
following b-catenin manipulation. Overexpression of b-catenin in thus providing more evidence that b-catenin plays a role in
hippocampal neuronal cultures increases dendritic growth and synaptic regulation. Following depolarization with a solution con-
arborization, while decreasing endogenous b-catenin prevents taining a high concentration of KCl, there is an NMDA-dependent
dendritic morphogenesis (Peng et al., 2009; Yu and Malenka, 2003). redistribution of preexisting b-catenin from dendritic shafts to
This function of b-catenin in regulating dendritic growth appears to spines in cultured hippocampal neurons. This redistribution of
be due to its role in cell adhesion, and is not dependent on its b-catenin to the spines coincides with an increase in the association
transcriptional actions. of b-catenin with cadherin, and can be mimicked or prevented by
The above changes have been reported at baseline; however, application of a tyrosine kinase or phosphatase inhibitor, respec-
a similar effect can be observed following neural activation. Neural tively (Murase et al., 2002).
activity is known to induce changes in dendrite morphology, and Similar results were obtained by studying b-catenin point
this remodeling is critical for neural circuit formation and synaptic mutations at site, tyrosine 654 (Y654). Point mutations that prevent
function. The effects of neural activity can be mimicked by chron- phosphorylation of b-catenin at this specific site result in a redis-
ically depolarizing neurons with extracellular potassium, and has tribution of b-catenin to the spine, and an increase in the size and
been shown to increase dendritic arborization (Peng et al., 2009; Yu density of synapsin-1 and PSD-95, markers of presynaptic and
and Malenka, 2003). postsynaptic proteins. In addition to changes in synaptic proteins,
Interestingly, the effect reported following overexpression preventing phosphorylation at Y654 alters synaptic function by
of b-catenin is remarkably similar to the observed increase in increasing the frequency of mEPSCs (Murase et al., 2002). An
dendritic arborization following treatment with high potassium, increase in the frequency of mEPSCs, with no change in the
thus suggesting that the two treatments may function through amplitude, may reflect an increase in the probability of neuro-
a common signaling pathway. Indeed, depolarization induced by transmitter release, or a conversion of silent, or inactive synapses to
elevated potassium increases Wnt secretion, and one way to active synapses (Choi et al., 2000; Gasparini et al., 2000). None-
increase intracellular b-catenin is through activation of the Wnt theless, manipulating tyrosine phosphorylation and dephosphor-
pathway. Therefore, it may be that neuronal depolarization ylation of b-catenin produces changes in synaptic size and strength.
increases Wnt activity, which then stabilizes the intracellular pool More recent evidence suggests that cyclin-dependent kinase 5
of b-catenin, ultimately leading to an enhancement in dendritic (Cdk5) activity is responsible for this activity-dependent phos-
arborization. phorylation of b-catenin at Y654 (Schuman and Murase, 2003).
b-catenin has also been shown to regulate postsynaptic Together, these results suggest that activity-induced changes in the
strength. Miniature excitatory postsynaptic currents (mEPSCs) are localization of b-catenin, along with the regulation of tyrosine
events generated in dendrites and generally arise from the spon- phosphorylation, are important for synaptic regulation.
taneous release of single vesicles. Measuring mEPSCs in b-catenin- b-catenin regulation has also been shown to play a role in
ablated neurons gives insight into the effect of b-catenin loss on activity-dependent gene expression, an important component of
glutamatergic quantal responses. Loss of b-catenin in hippocampal synaptic plasticity (Cohen and Greenberg, 2008; Kandel, 2001). In
neurons results in a decrease in the amplitude of mEPSCs, which is response to NMDAR-dependent activation of calpain, b-catenin is
largely dependent on AMPA receptors, without affecting the cleaved at the N terminus, making it resistant to GSK-3b mediated
frequency of mEPSCs, which is dependent on the density of func- degradation (Abe and Takeichi, 2007). This results in an increase in
tional presynaptic boutons (Okuda et al., 2007). Following rescue the stabilized form of b-catenin which then translocates to the
with transfection of the wild-type b-catenin gene in these neurons, nucleus to regulate gene transcription. Fosl1 has been identified as
the mean mEPSC amplitude is restored to levels comparable to one gene that is upregulated following the NMDAR-mediated
control neurons, suggesting that b-catenin plays a role in modu- b-catenin signaling cascade, and interestingly, Fosl1 has also been
lating AMPA-mediated synaptic currents. Similar to the presyn- shown to be upregulated following behavioral learning in rats
aptic role of b-catenin in controlling synaptic vesicle localization, (Faure et al., 2006). Therefore, b-catenin regulation may be
the central armadillo repeats, which bind to cadherins and TCF/LEF important for the transcription of genes following synaptic activity.
K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88 83

3.2. b-Catenin and memory related pathology 4.1. b-Catenin is required for consolidation of fear memory
formation
Recent evidence points toward a potential role for b-catenin in
pathological states of neurotoxicity such as Huntington’s Although it was initially identified for its role in development,
disease (Godin et al., 2010) and Alzheimer’s disease (AD). Alz- b-catenin has also been shown to play a role in neuronal synapse
heimer’s disease is a neurodegenerative disorder characterized regulation and plasticity. Since alterations in synapse regulation
by progressive memory loss and cognitive impairment, and at the and plasticity are thought to underpin long-term memory
molecular level, by the presence of neurofibrillary tangles (NFTs) formation, b-catenin may play a critical role in this process.
and senile plaques comprised of the insoluble b-amyloid peptide However, there has been a scarcity of data exploring this possi-
(Ab) (Hardy, 2006; Hardy and Selkoe, 2002; Lee et al., 1991). bility. Thus, we examined the role of b-catenin in amygdala-
Three mutations identified in families affected by Familial Alz- dependent fear memory. We found that b-catenin is highly
heimer’s disease (FAD), presenilin-1 (PS-1), presenilin-2 (PS-2), expressed in the adult mouse amygdala and is dynamically
and the amyloid precursor protein (APP), result in dysfunctional regulated at both the transcriptional and post-translational
Ab production (Duff et al., 1996; Price and Sisodia, 1998). There is levels with fear learning (Fig. 3) (Maguschak and Ressler, 2008).
indirect evidence from AD brains which also supports a role for We then found that pharmacological stabilization of b-catenin
b-catenin in disease. AD patients with PS-1 mutations have with lithium chloride resulted in enhanced learning, while
reduced levels of b-catenin (Zhang et al., 1998). PS-1 is a trans- genetic deletion of the gene that encodes b-catenin, Ctnnb1, in
membrane protein located at synaptic cellecell contact sites the amygdala resulted in impaired learning. In both cases,
which forms complexes with b-catenin (Kang et al., 2002; the manipulation affected the consolidation, but not acquisition,
Murayama et al., 1998; Yu et al., 1998; Zhou et al., 1997). This of the fear memory. Notably, Ctnnb1 deletion did not
interaction is thought to increase b-catenin stability (Zhang et al., affect a number of other behaviors, including locomotor, anxiety-
1998). Mutations in PS-1 decrease b-catenin stability and are related behavior, or hippocampal-dependent memory
associated with an overproduction and aggregation of Ab peptide (Maguschak and Ressler, 2008).
(Fraser et al., 2000). Memory formation is thought to involve the weakening and
PS-1 is also thought to inactivate GSK-3, a negative regulator of strengthening of synapses, and this process can be modulated by
b-catenin. GSK-3 expression has been shown to be upregulated in the adhesion between pre- and postsynaptic neurons. One example
the hippocampus of AD patients (Blalock et al., 2004). Similarly, an of this phenomenon involves the role of Aplysia cell adhesion
increase in phosphorylated GSK-3 has also been reported in the molecules (apCAM’s) in the long-term sensitization of the gill- and
frontal cortex in AD (Leroy et al., 2007). Since increased GSK-3 siphon-withdrawal reflex, a form of learning in the marine mollusk.
activity is associated with a decrease in b-catenin stability, these Following long-term sensitization with application of serotonin to
early findings suggest that impairments in b-catenin regulation siphon sensory neurons, apCAMs become rapidly down-regulated
may be linked to AD pathology. in the sensory neurons. This observed dynamic regulation of cell
Ab neurotoxicity can be produced by the addition of Ab to adhesion molecules may be responsible for long-term synaptic
neuronal cultures (De Ferrari et al., 2003), conditions which also plasticity (Mayford et al., 1992). Since b-catenin is important for cell
lower levels of cytoplasmic b-catenin. Lithium, which acts as adhesion, and deletion of Ctnnb1 in the amygdala produces deficits
a positive regulator in the Wnt signaling pathway, by inactivating in the consolidation of memory, we propose the following model of
GSK-3b, can prevent the cytotoxic effects of Ab (De Ferrari and how b-catenin may function in memory formation (Fig. 4).
Inestrosa, 2000; Toledo and Inestrosa, 2010). Lithium has also b-catenin can be found in a complex with cadherin at the
been shown to prevent neurodegeneration and behavioral plasma membrane of dendritic spines. Immediately following
impairments induced by injections of Ab fibrils into the dorsal a learning event, b-catenin becomes phosphorylated at site Y654.
hippocampus of rats (De Ferrari et al., 2003). Lithium also increases Similar to previous findings, we found that the increase in phos-
b-catenin levels, thus suggesting that the mechanism by which phorylated b-catenin coincided with a decrease in the interaction
lithium treatment may be acting is due, in part, to the stabilization between b-catenin and cadherin (Maguschak and Ressler, 2008).
of b-catenin (De Ferrari et al., 2003). This decrease may be required to weaken the bond between the
It has been suggested that AD manifests itself as a perturbation pre- and postsynaptic neurons, allowing for synaptic remodeling to
of neuroplasticity prior to overt Ab-dependent neural degeneration take place. Following a period of b-cateninecadherin destabiliza-
and toxicity (Mesulam, 1999; Selkoe, 2002). b-catenin appears to be tion, b-catenin relocates to the spine and once again forms
required for synaptic plasticity; thus, understanding the physio- a complex with cadherin, thereby stabilizing the synapse, and
logical role of b-catenin may be critical to inform new treatment strengthening the memory. A change in the total amount of
and prevention approaches in AD. b-catenin does not appear to be necessary for the stabilization of
the synapse. Instead, it may require rapid dynamic changes in the
4. b-Catenin and learning and memory in the adult brain breakdown, redistribution, and replacement of the protein. This
proposed model is consistent with previous in vitro work showing
The above section suggests a role for b-catenin in adult learning that depolarization of hippocampal neurons with KCl causes
and memory and in disorders of memory function. Thus far, a redistribution of b-catenin from dendritic shafts to spines,
there has been substantial evidence suggesting that b-catenin is without changing the total amount of the protein (Murase et al.,
involved in neuronal synapse regulation and plasticity; however, 2002). One difference between the findings in the above study
the majority of this work has been done in vitro. Note that Bamji and ours is the rapid destabilization in the b-cateninecadherin
et al. (2003) used a CaMKII-Cre to specifically remove b-catenin complex prior to the redistribution of b-catenin. This may reflect
in the hippocampus and found changes in synapse number and a difference between studying neuronal cultures and whole animal
synaptic responses. However, there were previously no studies on or may be due to several other factors including the means of
the role of this intriguing protein in standard behavioral learning inducing neural activity or time course for analysis.
and memory assays. The below studies describe the recent In conditions where b-catenin function may be impaired, the
examination of a role for b-catenin in long-term memory forma- initial labile phase may remain unaffected; however, the stable
tion in adults. phase may be comprised. If b-catenin is not present, it will not be
84 K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88

Fig. 4. Schematic representation of the role of b-catenin in producing the labile and
stable phases of memory formation. (A) b-catenin is located in a complex with cad-
herin. Following a learning event, b-catenin becomes phosphorylated and shifts to the
dendritic shaft, allowing for synaptic remodeling to take place (labile phase). At some
point later, b-catenin redistributes to the dendritic spine, and re-associates with cad-
herin to strengthen the memory (stable phase). (Thickness of cadherin bar represents
relative synaptic structural stability. Note that the cadherin adhesion complex is
a dynamic structure that also includes other intermediate proteins, and is modulated
by a number of protein kinases, including Fer, Shp2, LAR, and CK1 among others
(Piedra et al., 2003; Xu et al., 2004; Grinnell et al., 2010; Dunah et al., 2005; Kypta et al.,
1996; Del Valle-Pérez et al., 2011)).

4.2. Dynamic regulation of cadherin/b-catenin interaction

The findings reported above suggest that b-catenin is required


for normal consolidation, but not acquisition, of memory. The
evidence presented thus far suggests that the decrease and
subsequent increase in the interaction between b-catenin and
cadherin may be critical for the labile and stable phases of memory
formation. Such dynamic regulation has been proposed previously
when examining its cellular regulation in hippocampal cultures
(Bamji et al., 2006; Tai et al., 2007), but it has not previously been
demonstrated in vivo or in behavioral learning and memory
paradigms.
Neural activity increases the synthesis and secretion of brain-
derived neurotrophic factor (BDNF), which has been shown to
play a critical role in synaptic plasticity (Lu et al., 2008). Treatment
with BDNF induces synaptic vesicle dispersion, which is associated
with an increase in b-catenin tyrosine phosphorylation and
a decrease in b-cateninecadherin interaction (Bamji et al., 2006).
Within 30 min after the dispersion, phosphorylation decreases, and
the b-cateninecadherin interaction is restored (Bamji et al., 2006).
This finding suggests that the disruption and re-stabilization of
b-cateninecadherin complexes may be required for new synapse
formation. BDNF has been demonstrated to be important in both
amygdala (Rattiner et al., 2004) and hippocampal-dependent
memory formation (Heldt et al., 2007). These new findings that
b-catenin regulation is also involved in these processes raise the
question of whether the specific effects of BDNF on memory
consolidation are, in part, via the b-catenin pathway.
Fig. 3. b-catenin and Amygdala-dependent fear conditioning. (A) b-catenin gene Similarly, NMDAR-dependent neural activity has also been
expression in the amygdala is increased following fear conditioning. Qualitative in situ shown to induce changes in the interaction between b-catenin and
hybridization analysis of b-catenin mRNA in the amygdala in context exposed animals. cadherin (Tai et al., 2007). NMDAR activation decreases the rate of
(B) Additionally, post-translational regulation (phosphorylation state) of b-catenin and
GSK-3b are altered following fear conditioning. Mice were exposed to five tone-shock
cadherin endocytosis, increasing the accumulation of cadherin in
pairings and then sacrificed 0, 0.5, 2, 4 h after training. Phospho-GSK-3b levels, the plasma membrane. In addition the level of tyrosine phos-
determined by western blot, are significantly changed across timepoints (p < .001). (C) phorylated b-catenin is decreased, leading to an increase in the
b-catenin (phospho-Y654) levels, determined by western blot, are significantly interaction between b-catenin and cadherin in dendritic spines (Tai
changed across timepoints (p < .05). (D) Immunoprecipitation results from a cadherin
et al., 2007). Furthermore, prolonged stability of the cadherin at the
immunoblot after b-catenin immunoprecipitation. Cadherin interaction with b-catenin
is significantly changed across timepoints (p < .05). ‘con’ ¼ context control group, plasma membrane blocks NMDAR-dependent synaptic plasticity,
‘unp’ ¼ unpaired shock control group; Bars indicate mean  sem; * denotes p < .05 for suggesting that the dynamic behavior of b-catenin and cadherin is
the different comparisons identified. (adapted from Maguschak and Ressler, 2008). important for this process.

able to bind to cadherin and stabilize the synapse. This proposed 4.3. NMDA receptors, BDNF, and b-catenin
model suggests that dynamic regulation of b-catenin may be
involved in the structural conversion of short-term labile to long- There have been previous studies showing that BDNF may
term stable memory traces (Fig. 4). modulate NMDAR activity. Upon BDNF binding to and activating
K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88 85

the tyrosine receptor kinase TrkB, there is an enhancement in vivo inhibition of GSK-3 produces antidepressive-like behavior
glutamatergic synaptic transmission (Levine et al., 1995) and an (Kaidanovich-Beilin et al., 2004). Among the more direct lines of
increase in the phosphorylation of the NMDAR (Suen et al., 1997). evidence linking the b-catenin/GSK-3 pathway with lithium’s effect
In vitro studies have shown that the phosphorylation of NMDAR on mood stabilization was the finding that b-catenin overexpression
occurs within 5 min of exposure to BDNF (Suen et al., 1997). Since in the mouse brain mimics lithium-sensitive behaviors (Gould et al.,
BDNF activation of TrkB receptors functions to transiently disso- 2007). This study demonstrated that b-catenin transgenic mice
ciate the b-catenin from the cadherin, while glutamatergic activa- exhibited behaviors identical to those observed in lithium-treated
tion of NMDA receptors functions to increase the association, it is mice. These data are consistent with the hypothesis that the
possible that the two systems may interact to produce the transient behavioral effects of lithium are mediated through its direct inhi-
destabilization and re-stabilization of synapses. bition of GSK-3 and the consequent increase in b-catenin. Addi-
Interestingly, both BDNF and NMDA are required for the consoli- tionally, recent data implicate the Wnt pathway inhibitor, Dkk-1, in
dation of memories. Therefore, we propose that similar mechanisms stress-induced hippocampal damage that has been previously
are taking place in vitro and in vivo to stabilize and strengthen associated with affective disorders (Matrisciano et al., 2011). Most
synapses (Fig. 5). BDNF is released following a learning event, which recently is a finding connecting DISC1 to this pathway. Numerous
phosphorylates b-catenin, decreasing the b-cateninecadherin inter- studies have associated DISC1 polymorphisms with affective
action. This decrease in the b-cateninecadherin interaction increases disorders (e.g. Hodgkinson et al., 2004; Hennah et al., 2009;
synaptic vesicle mobility, allowing for synaptic plasticity. At about the Schosser et al., 2010). Recently it was shown that DISC1 inhibits
same time, BDNF may increase the phosphorylation of NMDAR. The GSK-3 activity through direct physical interaction, which stabilizes
activation of the NMDAR then helps to re-stabilize the synapse, by b-catenin. These results implicate DISC1 in GSK-3/b-catenin
decreasing the rate of cadherin endocytosis and redistributing signaling pathways and provide a framework for understanding
b-catenin into spines. Once the synapse is stabilized, the memory how alterations in this pathway may contribute to the etiology of
becomes strengthened. Future studies directly examining this inter- psychiatric disorders (Mao et al., 2009).
action, in vivo, would be important and interesting.
5. Conclusions and translational implications

4.4. b-Catenin, depression, and mood regulation


b-catenin is essential for normal embryonic development of the
central nervous system as well as normal neuronal functioning in
The focus of this review has been the role of b-catenin pathways
adulthood. Alterations in b-catenin signaling lead to detrimental
in learning and memory; however, there is increasing awareness
effects throughout the lifespan. As mentioned earlier, b-catenin
that many affective disorders, such as depression and bipolar
knockouts are embryonic lethal (Machon et al., 2003). In contrast,
disorder, may also represent states of dysregulated synaptic plas-
embryonic transgenic mice that express stabilized b-catenin in
ticity. A number of lines of evidence now implicate the b-catenin/
neural precursors develop gross enlargements of the cerebral
GSK-3 pathways in these disorders (Gould et al., 2006; Wada, 2009).
cortex, hippocampus, and amygdala (Chenn and Walsh, 2002,
This connection was initially made through the recognition of
2003). Thus, both down- and up-regulation of b-catenin can alter
lithium as a potent regulator of the b-catenin/GSK-3 pathway as
developmental processes. Such tight regulation of b-catenin func-
described above. One series of studies demonstrated that targeted in
tion is also important for synapse assembly. Perturbations in
b-catenin regulation produce deficits in both pre- and postsynaptic
structure and function. Understanding how deregulated b-catenin
function interferes with homeostasis of the healthy adult verte-
brate brain may provide insight into the etiology of neurodegen-
erative conditions.
In summary, b-catenin is present from development into
adulthood. It plays critical roles in many of the cellular and
molecular functions that take place during all aspects of life.
Understanding how b-catenin may function not only during
development, but also during synapse remodeling in adulthood,
may help to understand how alterations in its normal regulation
can lead to disease. b-catenin has been shown to be involved in
synaptic plasticity, particularly involving emotional learning and
memory processes. b-catenin appears to be involved in disorders
related to decreased memory function (e.g. Alzheimer’s disease) as
well as overly strong memory formation (such as fear learning in
PTSD). Further understanding of the b-catenin pathway in adult-
hood may lead to better mechanistic appreciation for the structural
mechanisms underlying emotional and declarative learning and
memory as well as provide novel therapeutic approaches in
Fig. 5. Schematic representation showing a possible interaction between BDNF memory related disorders.
signaling and NMDAR activation. (A) b-catenin is located in a complex with cadherin at
the plasma membrane of the synapse. (B) Activation of TrkB receptor by BDNF, along
with enhanced synaptic activity, results in the phosphorylation of b-catenin at Y654, Financial disclosure statement
thus causing a dissociation of b-catenin from cadherin. (C) BDNF activation also results There were no commercial sponsors or commercial relation-
in the phosphorylation of NMDARs. During this time, phosphorylated b-catenin shifts ships related to the current work. Within the last 3 years, Dr. Ressler
to the dendritic shaft, and synaptic vesicles disperse. (D) The activation of NMDARs, has received awards and/or funding support related to other
along with the internalization of TrkB re-stabilizes the synapse by bringing b-catenin
and cadherin together at the membrane. (Thickness of cadherin bar represents relative
studies from Burroughs Wellcome Foundation, NARSAD, NIMH,
synaptic structural stability. Note that the cadherin adhesion complex is a dynamic NIDA, and is a cofounder of Extinction Pharmaceuticals for use of
structure that also includes other intermediate proteins). NMDA-based therapeutics with psychotherapy.
86 K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88

Acknowledgments Fifkova, E., Delay, R.J., 1982. Cytoplasmic actin in neuronal processes as a possible
mediator of synaptic plasticity. J. Cell Biol. 95, 345e350.
Fraser, P.E., Yang, D.S., Yu, G., Levesque, L., Nishimura, M., Arawaka, S., Serpell, L.C.,
This work was primarily supported by National Institutes of Rogaeva, E., St George-Hyslop, P., 2000. Presenilin structure, function and role in
Mental Health (MH071537) and the Burroughs Wellcome Fund Alzheimer disease. Biochim. Biophys. Acta 1502, 1e15.
(KJR). Gao, X., Arlotta, P., Macklis, J.D., Chen, J., 2007. Conditional knock-out of beta-catenin
in postnatal-born dentate gyrus granule neurons results in dendritic malfor-
mation. J. Neurosci. 27, 14317e14325.
Gasparini, S., Saviane, C., Voronin, L.L., Cherubini, E., 2000. Silent synapses in the
References developing hippocampus: lack of functional AMPA receptors or low probability
of glutamate release? Proc. Natl. Acad. Sci. U S A 97, 9741e9746.
Abe, K., Takeichi, M., 2007. NMDA-receptor activation induces calpain-mediated Giagtzoglou, N., Ly, C.V., Bellen, H.J., 2009. Cell adhesion, the backbone of the
beta-catenin cleavages for triggering gene expression. Neuron 53, 387e397. synapse: “vertebrate” and “invertebrate” perspectives. Cold Spring Harb. Per-
Abe, K., Takeichi, M., 2008. EPLIN mediates linkage of the cadherin catenin complex spect. Biol. 1 (4), a003079.
to F-actin and stabilizes the circumferential actin belt. Proc. Natl. Acad. Sci. Godin, J.D., Poizat, G., Hickey, M.A., Maschat, F., Humbert, S., 2010. Mutant
U S A 105 (1), 13e19. huntingtin-impaired degradation of beta-catenin causes neurotoxicity in
Bamji, S.X., Shimazu, K., Kimes, N., Huelsken, J., Birchmeier, W., Lu, B., Reichardt, L.F., Huntington’s disease. EMBO J. 29 (14), 2433e2445.
2003. Role of beta-catenin in synaptic vesicle localization and presynaptic Gottardi, C.J., Gumbiner, B.M., 2004. Distinct molecular forms of beta-catenin are
assembly. Neuron 40, 719e731. targeted to adhesive or transcriptional complexes. J. Cell Biol. 167, 339e349.
Bamji, S.X., Rico, B., Kimes, N., Reichardt, L.F., 2006. BDNF mobilizes synaptic vesi- Gould, T.D., Picchini, A.M., Einat, H., Manji, H.K., 2006. Targeting glycogen synthase
cles and enhances synapse formation by disrupting cadherinebeta-catenin kinase-3 in the CNS: implications for the development of new treatments for
interactions. J. Cell Biol. 174, 289e299. mood disorders. Curr. Drug Targets 7 (11), 1399e1409.
Bek, S., Kemler, R., 2002. Protein kinase CKII regulates the interaction of beta-catenin Gould, T.D., Einat, H., O’Donnell, K.C., Picchini, A.M., Schloesser, R.J., Manji, H.K.,
with alpha-catenin and its protein stability. J. Cell Sci. 115 (Pt 24), 4743e4753. 2007. Beta-catenin overexpression in the mouse brain phenocopies lithium-
Blalock, E.M., Geddes, J.W., Chen, K.C., Porter, N.M., Markesbery, W.R., sensitive behaviors. Neuropsychopharmacology 32 (10), 2173e2183.
Landfield, P.W., 2004. Incipient Alzheimer’s disease: microarray correlation Greenberg, M.E., Xu, B., Lu, B., Hempstead, B.L., 2009. New insights in the biology of
analyses reveal major transcriptional and tumor suppressor responses. Proc. BDNF synthesis and release: implications in CNS function. J. Neurosci. 29,
Natl. Acad. Sci. U S A 101, 2173e2178. 12764e12767.
Brembeck, F.H., Schwarz-Romond, T., Bakkers, J., Wilhelm, S., Hammerschmidt, M., Grinnell, K.L., Casserly, B., Harrington, E.O., 2010. Role of protein tyrosine phos-
Birchmeier, W., 2004. Essential role of BCL9-2 in the switch between beta- phatase SHP2 in barrier function of pulmonary endothelium. Am. J. Physiol.
catenin’s adhesive and transcriptional functions. Genes Dev. 18, 2225e2230. Lung Cell Mol. Physiol. 298 (3), L361eL370.
Bruses, J.L., 2006. N-cadherin signaling in synapse formation and neuronal physi- Hardy, J., Selkoe, D.J., 2002. The amyloid hypothesis of Alzheimer’s disease: progress
ology. Mol. Neurobiol. 33, 237e252. and problems on the road to therapeutics. Science 297, 353e356.
Chen, J., Park, C.S., Tang, S.J., 2006. Activity-dependent synaptic Wnt release regu- Hardy, J., 2006. A hundred years of Alzheimer’s disease research. Neuron 52, 3e13.
lates hippocampal long term potentiation. J. Biol. Chem. 281, 11910e11916. Hebb, D.O., 1949. The Organization of Behavior: A Neuropsychological Theory.
Chenn, A., Walsh, C.A., 2002. Regulation of cerebral cortical size by control of cell Wiley, New York.
cycle exit in neural precursors. Science 297, 365e369. Heldt, S.A., Stanek, L., Chhatwal, J.P., Ressler, K.J., 2007. Hippocampus-specific
Chenn, A., Walsh, C.A., 2003. Increased neuronal production, enlarged forebrains deletion of BDNF in adult mice impairs spatial memory and extinction of
and cytoarchitectural distortions in beta-catenin overexpressing transgenic aversive memories. Mol. Psychiatry 12, 656e670.
mice. Cereb. Cortex 13, 599e606. Hennah, W., Thomson, P., McQuillin, A., Bass, N., Loukola, A., Anjorin, A.,
Choi, S., Klingauf, J., Tsien, R.W., 2000. Postfusional regulation of cleft glutamate Blackwood, D., Curtis, D., Deary, I.J., Harris, S.E., Isometsä, E.T., Lawrence, J.,
concentration during LTP at ‘silent synapses’. Nat. Neurosci. 3, 330e336. Lönnqvist, J., Muir, W., Palotie, A., Partonen, T., Paunio, T., Pylkkö, E.,
Cohen, S., Greenberg, M.E., 2008. Communication between the synapse and the Robinson, M., Soronen, P., Suominen, K., Suvisaari, J., Thirumalai, S., St Clair, D.,
nucleus in neuronal development, plasticity, and disease. Annu. Rev. Cell Dev. Gurling, H., Peltonen, L., Porteous, D., 2009. DISC1 association, heterogeneity
Biol. 24, 183e209. and interplay in schizophrenia and bipolar disorder. Mol. Psychiatry 14 (9),
Collingridge, G.L., Bliss, T.V., 1995. Memories of NMDA receptors and LTP. Trends 865e873.
Neurosci. 18, 54e56. Heuberger, J., Birchmeier, W., 2010. Interplay of cadherin-mediated cell adhesion
De Ferrari, G.V., Inestrosa, N.C., 2000. Wnt signaling function in Alzheimer’s disease. and canonical Wnt signaling. Cold Spring Harb. Perspect. Biol. 2 (2), a002915.
Brain Res. Brain Res. Rev. 33, 1e12. Hodgkinson, C.A., Goldman, D., Jaeger, J., Persaud, S., Kane, J.M., Lipsky, R.H.,
De Ferrari, G.V., Chacon, M.A., Barria, M.I., Garrido, J.L., Godoy, J.A., Olivares, G., Malhotra, A.K., 2004. Disrupted in schizophrenia 1 (DISC1): association with
Reyes, A.E., Alvarez, A., Bronfman, M., Inestrosa, N.C., 2003. Activation of Wnt schizophrenia, schizoaffective disorder, and bipolar disorder. Am. J. Hum. Genet.
signaling rescues neurodegeneration and behavioral impairments induced by 75 (5), 862e872.
beta-amyloid fibrils. Mol. Psychiatry 8, 195e208. Hoschuetzky, H., Aberle, H., Kemler, R., 1994. Beta-catenin mediates the interaction
Del Valle-Pérez, B., Arqués, O., Vinyoles, M., de Herreros, A.G., Duñach, M., 2011 Jul. of the cadherinecatenin complex with epidermal growth factor receptor. J. Cell
Coordinated action of CK1 isoforms in canonical Wnt signaling. Mol. Cell Biol. Biol. 127, 1375e1380.
31 (14), 2877e2888. Huber, A.H., Nelson, W.J., Weis, W.I., 1997. Three-dimensional structure of the
Dityatev, A., Dityateva, G., Sytnyk, V., Delling, M., Toni, N., Nikonenko, I., Muller, D., armadillo repeat region of beta-catenin. Cell 90, 871e882.
Schachner, M., 2004. Polysialylated neural cell adhesion molecule promotes Huber, A.H., Stewart, D.B., Laurents, D.V., Nelson, W.J., Weis, W.I., 2001. The cad-
remodeling and formation of hippocampal synapses. J. Neurosci. 24, herin cytoplasmic domain is unstructured in the absence of beta-catenin.
9372e9382. A possible mechanism for regulating cadherin turnover. J. Biol. Chem. 276,
Drees, F., Pokutta, S., Yamada, S., Nelson, W.J., Weis, W.I., 2005. Alpha-catenin is 12301e12309.
a molecular switch that binds E-cadherinebeta-catenin and regulates actin- Iwai, Y., Hirota, Y., Ozaki, K., Okano, H., Takeichi, M., Uemura, T., 2002. DN-cadherin
filament assembly. Cell 123, 903e915. is required for spatial arrangement of nerve terminals and ultrastructural
Duff, K., Eckman, C., Zehr, C., Yu, X., Prada, C.M., Perez-Tur, J., Hutton, M., Buee, L., organization of synapses. Mol. Cell Neurosci. 19, 375e388.
Harigaya, Y., Yager, D., Morgan, D., Gordon, M.N., Holcomb, L., Refolo, L., Zenk, B., Kaidanovich-Beilin, O., Milman, A., Weizman, A., Pick, C.G., Eldar-Finkelman, H.,
Hardy, J., Younkin, S., 1996. Increased amyloid-beta42(43) in brains of mice 2004. Rapid antidepressive-like activity of specific glycogen synthase kinase-3
expressing mutant presenilin 1. Nature 383, 710e713. inhibitor and its effect on beta-catenin in mouse hippocampus. Biol. Psychiatry
Dunah, A.W., Hueske, E., Wyszynski, M., Hoogenraad, C.C., Jaworski, J., Pak, D.T., 55 (8), 781e784.
Simonetta, A., Liu, G., Sheng, M., 2005. LAR receptor protein tyrosine phos- Kandel, E.R., 2001. The molecular biology of memory storage: a dialogue between
phatases in the development and maintenance of excitatory synapses. Nat. genes and synapses. Science 294, 1030e1038.
Neurosci. 8 (4), 458e467. Kang, D.E., Soriano, S., Xia, X., Eberhart, C.G., De Strooper, B., Zheng, H., Koo, E.H.,
Dupre-Crochet, S., Figueroa, A., Hogan, C., Ferber, E.C., Bialucha, C.U., Adams, J., 2002. Presenilin couples the paired phosphorylation of beta-catenin indepen-
Richardson, E.C., Fujita, Y., 2007. Casein kinase 1 is a novel negative regulator of dent of axin: implications for beta-catenin activation in tumorigenesis. Cell 110,
E-cadherin-based cellecell contacts. Mol. Cell Biol. 27 (10), 3804e3816. 751e762.
Engert, F., Bonhoeffer, T., 1999. Dendritic spine changes associated with hippo- Konorski, J., 1948. Conditioned Reflexes and Neuron Organization. Cambridge
campal long-term synaptic plasticity. Nature 399, 66e70. University Press, Cambridge.
Fang, D., Hawke, D., Zheng, Y., Xia, Y., Meisenhelder, J., Nika, H., Mills, G.B., Krichmar, J.L., Velasquez, D., Ascoli, G.A., 2006. Effects of beta-catenin on dendritic
Kobayashi, R., Hunter, T., Lu, Z., 2007. Phosphorylation of beta-catenin by AKT morphology and simulated firing patterns in cultured hippocampal neurons.
promotes beta-catenin transcriptional activity. J. Biol. Chem. 282, 11221e11229. Biol. Bull. 211, 31e43.
Faure, A., Conde, F., Cheruel, F., el Massioui, N., 2006. Learning-dependent activation Krueger, S.R., Kolar, A., Fitzsimonds, R.M., 2003. The presynaptic release apparatus is
of Fra-1: involvement of ventral hippocampus and SNc/VTA complex in learning functional in the absence of dendritic contact and highly mobile within isolated
and habit formation. Brain Res. Bull. 68, 233e248. axons. Neuron 40, 945e957.
Fifkova, E., Anderson, C.L., 1981. Stimulation-induced changes in dimensions of Kypta, R.M., Su, H., Reichardt, L.F., 1996. Association between a transmembrane
stalks of dendritic spines in the dentate molecular layer. Exp. Neurol. 74, protein tyrosine phosphatase and the cadherinecatenin complex. J. Cell Biol.
621e627. 134 (6), 1519e1529.
K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88 87

Lee, V.M., Balin, B.J., Otvos Jr., L., Trojanowski, J.Q., 1991. A68: a major subunit of paired Rattiner, L.M., Davis, M., French, C.T., Ressler, K.J., 2004. Brain-derived neurotrophic
helical filaments and derivatized forms of normal Tau. Science 251, 675e678. factor and tyrosine kinase receptor B involvement in amygdala-dependent fear
Lee, S.H., Peng, I.F., Ng, Y.G., Yanagisawa, M., Bamji, S.X., Elia, L.P., Balsamo, J., conditioning. J. Neurosci. 24, 4796e4806.
Lilien, J., Anastasiadis, P.Z., Ullian, E.M., Reichardt, L.F., 2008. Synapses are Reiss, K., Maretzky, T., Ludwig, A., Tousseyn, T., de Strooper, B., Hartmann, D.,
regulated by the cytoplasmic tyrosine kinase Fer in a pathway mediated by Saftig, P., 2005. ADAM10 cleavage of N-cadherin and regulation of
p120catenin, Fer, SHP-2, and beta-catenin. J. Cell Biol. 183, 893e908. cellecell adhesion and beta-catenin nuclear signalling. EMBO J. 24 (4),
Leroy, K., Yilmaz, Z., Brion, J.P., 2007. Increased level of active GSK-3beta in Alz- 742e752.
heimer’s disease and accumulation in argyrophilic grains and in neurones at Roura, S., Miravet, S., Piedra, J., Garcia de Herreros, A., Dunach, M., 1999. Regulation
different stages of neurofibrillary degeneration. Neuropathol. Appl. Neurobiol. of E-cadherin/catenin association by tyrosine phosphorylation. J. Biol. Chem.
33, 43e55. 274, 36734e36740.
Leuner, B., Falduto, J., Shors, T.J., 2003. Associative memory formation increases the Sabatini, B.L., Maravall, M., Svoboda, K., 2001. Ca(2þ) signaling in dendritic spines.
observation of dendritic spines in the hippocampus. J. Neurosci. 23, 659e665. Curr. Opin. Neurobiol. 11, 349e356.
Levine, E.S., Dreyfus, C.F., Black, I.B., Plummer, M.R., 1995. Brain-derived neuro- Schosser, A., Gaysina, D., Cohen-Woods, S., Chow, P.C., Martucci, L., Craddock, N.,
trophic factor rapidly enhances synaptic transmission in hippocampal neurons Farmer, A., Korszun, A., Gunasinghe, C., Gray, J., Jones, L., Tozzi, F., Perry, J.,
via postsynaptic tyrosine kinase receptors. Proc. Natl. Acad. Sci. U S A 92, Muglia, P., Owen, M.J., Craig, I.W., McGuffin, P., 2010. Association of DISC1 and
8074e8077. TSNAX genes and affective disorders in the depression caseecontrol (DeCC)
Lisman, J., Schulman, H., Cline, H., 2002. The molecular basis of CaMKII function in and bipolar affective caseecontrol (BACCS) studies. Mol. Psychiatry 15 (8),
synaptic and behavioural memory. Nat. Rev. Neurosci. 3, 175e190. 844e849.
Liu, C., Li, Y., Semenov, M., Han, C., Baeg, G.H., Tan, Y., Zhang, Z., Lin, X., He, X., 2002. Schuman, E.M., Murase, S., 2003. Cadherins and synaptic plasticity: activity-dependent
Control of beta-catenin phosphorylation/degradation by a dual-kinase mecha- cyclin-dependent kinase 5 regulation of synaptic beta-cateninecadherin interac-
nism. Cell 108, 837e847. tions. Philos. Trans. R Soc. Lond B. Biol. Sci. 358, 749e756.
Logan, C.Y., Nusse, R., 2004. The Wnt signaling pathway in development and Selkoe, D.J., 2002. Alzheimer’s disease is a synaptic failure. Science 298,
disease. Annu. Rev. Cell Dev. Biol. 20, 781e810. 789e791.
Lu, Y., Christian, K., Lu, B., 2008. BDNF: a key regulator for protein synthesis- Stan, A., Pielarski, K.N., Brigadski, T., Wittenmayer, N., Fedorchenko, O., Gohla, A.,
dependent LTP and long-term memory? Neurobiol. Learn. Mem. 89, 312e323. Lessmann, V., Dresbach, T., Gottmann, K., 2010. Essential cooperation of
MacDonald, B.T., Tamai, K., He, X., 2009. Wnt/beta-catenin signaling: components, N-cadherin and neuroligin-1 in the transsynaptic control of vesicle accumula-
mechanisms, and diseases. Dev. Cell. 17 (1), 9e26. tion. Proc. Natl. Acad. Sci. U S A. 107 (24), 11116e11121.
Machon, O., van den Bout, C.J., Backman, M., Kemler, R., Krauss, S., 2003. Role of Steiner, P., Higley, M.J., Xu, W., Czervionke, B.L., Malenka, R.C., Sabatini, B.L., 2008.
beta-catenin in the developing cortical and hippocampal neuroepithelium. Destabilization of the postsynaptic density by PSD-95 serine 73 phosphoryla-
Neuroscience 122, 129e143. tion inhibits spine growth and synaptic plasticity. Neuron 60, 788e802.
Magee, J.C., Johnston, D., 1995. Synaptic activation of voltage-gated channels in the Suen, P.C., Wu, K., Levine, E.S., Mount, H.T., Xu, J.L., Lin, S.Y., Black, I.B., 1997. Brain-
dendrites of hippocampal pyramidal neurons. Science 268, 301e304. derived neurotrophic factor rapidly enhances phosphorylation of the post-
Maguschak, K.A., Ressler, K.J., 2008. Beta-catenin is required for memory consoli- synaptic N-methyl-D-aspartate receptor subunit 1. Proc. Natl. Acad. Sci. U S A
dation. Nat. Neurosci. 11 (11), 1319e1326. 94, 8191e8195.
Maher, M.T., Flozak, A.S., Stocker, A.M., Chenn, A., Gottardi, C.J., 2009. Activity of the Sun, Y., Aiga, M., Yoshida, E., Humbert, P.O., Bamji, S.X., 2009. Scribble interacts with
beta-catenin phosphodestruction complex at cell-cell contacts is enhanced by beta-catenin to localize synaptic vesicles to synapses. Mol. Biol. Cell. 20 (14),
cadherin-based adhesion. J. Cell Biol. 186 (2), 219e228. 3390e3400.
Mao, Y., Ge, X., Frank, C.L., Madison, J.M., Koehler, A.N., Doud, M.K., Tassa, C., Tai, C.Y., Mysore, S.P., Chiu, C., Schuman, E.M., 2007. Activity-regulated N-cadherin
Berry, E.M., Soda, T., Singh, K.K., Biechele, T., Petryshen, T.L., Moon, R.T., endocytosis. Neuron 54, 771e785.
Haggarty, S.J., Tsai, L.H., 2009. Disrupted in schizophrenia 1 regulates neuronal Takahashi, K., Suzuki, K., Tsukatani, Y., 1997. Induction of tyrosine phosphorylation
progenitor proliferation via modulation of GSK3beta/beta-catenin signaling. and association of beta-catenin with EGF receptor upon tryptic digestion of
Cell 136 (6), 1017e1031. quiescent cells at confluence. Oncogene 15, 71e78.
Matrisciano, F., Busceti, C.L., Bucci, D., Orlando, R., Caruso, A., Molinaro, G., Takeichi, M., Abe, K., 2005. Synaptic contact dynamics controlled by cadherin and
Cappuccio, I., Riozzi, B., Gradini, R., Motolese, M., Caraci, F., Copani, A., catenins. Trends Cell Biol. 15, 216e221.
Scaccianoce, S., Melchiorri, D., Bruno, V., Battaglia, G., Nicoletti, F., 2011. Takeichi, M., 2007. The cadherin superfamily in neuronal connections and inter-
Induction of the Wnt antagonist Dickkopf-1 is involved in stress-induced actions. Nat. Rev. Neurosci. 8, 11e20.
hippocampal damage. PLoS One. 6 (1), e16447. Tanaka, C., Nishizuka, Y., 1994. The protein kinase C family for neuronal signaling.
Mayford, M., Barzilai, A., Keller, F., Schacher, S., Kandel, E.R., 1992. Modulation of an Annu. Rev. Neurosci. 17, 551e567.
NCAM-related adhesion molecule with long-term synaptic plasticity in Aplysia. Taurin, S., Sandbo, N., Qin, Y., Browning, D., Dulin, N.O., 2006. Phosphorylation of
Science 256 (5057), 638e644. beta-catenin by cyclic AMP-dependent protein kinase. J. Biol. Chem. 281,
Mesulam, M.M., 1999. Neuroplasticity failure in Alzheimer’s disease: bridging the 9971e9976.
gap between plaques and tangles. Neuron 24, 521e529. Togashi, H., Abe, K., Mizoguchi, A., Takaoka, K., Chisaka, O., Takeichi, M., 2002.
Moon, R.T., Kohn, A.D., De Ferrari, G.V., Kaykas, A., 2004. WNT and beta-catenin Cadherin regulates dendritic spine morphogenesis. Neuron 35, 77e89.
signalling: diseases and therapies. Nat. Rev. Genet. 5, 691e701. Toledo, E.M., Inestrosa, N.C., 2010. Activation of Wnt signaling by lithium and
Murase, S., Mosser, E., Schuman, E.M., 2002. Depolarization drives beta-Catenin into rosiglitazone reduced spatial memory impairment and neurodegeneration in
neuronal spines promoting changes in synaptic structure and function. Neuron brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer’s disease. Mol.
35, 91e105. Psychiatry 15, 272e285, 228.
Murayama, M., Tanaka, S., Palacino, J., Murayama, O., Honda, T., Sun, X., Yasutake, K., Verkaar, F., Zaman, G.J., 2011. New avenues to target Wnt/b-catenin signaling. Drug
Nihonmatsu, N., Wolozin, B., Takashima, A., 1998. Direct association of Discov. Today 16 (1e2), 35e41.
presenilin-1 with beta-catenin. FEBS Lett. 433, 73e77. Wada, A., 2009. Lithium and neuropsychiatric therapeutics: neuroplasticity via
Nelson, W.J., 2008. Regulation of cellecell adhesion by the cadherin-catenin glycogen synthase kinase-3beta, beta-catenin, and neurotrophin cascades.
complex. Biochem. Soc. Trans. 36 (Pt 2), 149e155. J. Pharmacol. Sci. 110 (1), 14e28.
Okabe, S., 2007. Molecular anatomy of the postsynaptic density. Mol. Cell Neurosci. Wu, H., Symes, K., Seldin, D.C., Dominguez, I., 2009. Threonine 393 of beta-catenin
34, 503e518. regulates interaction with Axin. J. Cell Biochem. 108 (1), 52e63.
Okuda, T., Yu, L.M., Cingolani, L.A., Kemler, R., Goda, Y., 2007. Beta-catenin regulates Xu, W., Kimelman, D., 2007. Mechanistic insights from structural studies of beta-
excitatory postsynaptic strength at hippocampal synapses. Proc. Natl. Acad. Sci. catenin and its binding partners. J. Cell Sci. 120, 3337e3344.
U S A 104, 13479e13484. Xu, G., Craig, A.W., Greer, P., Miller, M., Anastasiadis, P.Z., Lilien, J., Balsamo, J., 2004.
Ostroff, L.E., Cain, C.K., Bedont, J., Monfils, M.H., Ledoux, J.E., 2010. Fear and safety Continuous association of cadherin with beta-catenin requires the non-receptor
learning differentially affect synapse size and dendritic translation in the lateral tyrosine-kinase Fer. J. Cell Sci. 117, 3207e3219.
amygdala. Proc. Natl. Acad. Sci. U S A 107, 9418e9423. Yamada, S., Pokutta, S., Drees, F., Weis, W.I., Nelson, W.J., 2005. Deconstructing the
Peng, Y.R., He, S., Marie, H., Zeng, S.Y., Ma, J., Tan, Z.J., Lee, S.Y., Malenka, R.C., Yu, X., cadherinecatenineactin complex. Cell 123, 889e901.
2009. Coordinated changes in dendritic arborization and synaptic strength Yang, Y., Wang, X.B., Frerking, M., Zhou, Q., 2008. Spine expansion and stabilization
during neural circuit development. Neuron 61, 71e84. associated with long-term potentiation. J. Neurosci. 28, 5740e5751.
Piedra, J., Miravet, S., Castaño, J., Pálmer, H.G., Heisterkamp, N., García de Yost, C., Torres, M., Miller, J.R., Huang, E., Kimelman, D., Moon, R.T., 1996. The axis-
Herreros, A., Duñach, M., 2003. p120 catenin-associated Fer and Fyn tyrosine inducing activity, stability, and subcellular distribution of beta-catenin is
kinases regulate beta-catenin Tyr-142 phosphorylation and beta-cat- regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev. 10,
eninealpha-catenin interaction. Mol. Cell Biol. 23 (7), 2287e2297. 1443e1454.
Pokutta, S., Weis, W.I., 2007. Structure and mechanism of cadherins and catenins in Yu, X., Malenka, R.C., 2003. Beta-catenin is critical for dendritic morphogenesis. Nat.
cellecell contacts. Annu. Rev. Cell Dev. Biol. 23, 237e261. Neurosci. 6, 1169e1177.
Price, D.L., Sisodia, S.S., 1998. Mutant genes in familial Alzheimer’s disease and Yu, G., Chen, F., Levesque, G., Nishimura, M., Zhang, D.M., Levesque, L., Rogaeva, E.,
transgenic models. Annu. Rev. Neurosci. 21, 479e505. Xu, D., Liang, Y., Duthie, M., St George-Hyslop, P.H., Fraser, P.E., 1998. The pre-
Ramon y Cajal, S., 1894. La fine structure des centres nerveux. Proc. R. Soc. Lond. 55, senilin 1 protein is a component of a high molecular weight intracellular
444e468. complex that contains beta-catenin. J. Biol. Chem. 273, 16470e16475.
Rao, T.P., Kühl, M., 2010. An updated overview on Wnt signaling pathways: Yuste, R., Bonhoeffer, T., 2004. Genesis of dendritic spines: insights from ultra-
a prelude for more. Circ. Res. 106 (12), 1798e1806. structural and imaging studies. Nat. Rev. Neurosci. 5, 24e34.
88 K.A. Maguschak, K.J. Ressler / Neuropharmacology 62 (2012) 78e88

Zhai, R.G., Vardinon-Friedman, H., Cases-Langhoff, C., Becker, B., mutations in presenilin-1 potentiates neuronal apoptosis. Nature 395,
Gundelfinger, E.D., Ziv, N.E., Garner, C.C., 2001. Assembling the presynaptic 698e702.
active zone: a characterization of an active one precursor vesicle. Neuron 29, Zhou, J., Liyanage, U., Medina, M., Ho, C., Simmons, A.D., Lovett, M., Kosik, K.S., 1997.
131e143. Presenilin 1 interaction in the brain with a novel member of the Armadillo
Zhang, Z., Hartmann, H., Do, V.M., Abramowski, D., Sturchler-Pierrat, C., family. Neuroreport 8, 2085e2090.
Staufenbiel, M., Sommer, B., van de Wetering, M., Clevers, H., Saftig, P., De Ziv, N.E., Garner, C.C., 2004. Cellular and molecular mechanisms of presynaptic
Strooper, B., He, X., Yankner, B.A., 1998. Destabilization of beta-catenin by assembly. Nat. Rev. Neurosci. 5, 385e399.

You might also like