You are on page 1of 16

Thehttp://nro.sagepub.

com/
Neuroscientist

The Wnt Signaling Pathway in Bipolar Disorder


Todd D. Gould and Husseini K. Manji
Neuroscientist 2002 8: 497
DOI: 10.1177/107385802237176

The online version of this article can be found at:


http://nro.sagepub.com/content/8/5/497

Published by:

http://www.sagepublications.com

Additional services and information for The Neuroscientist can be found at:

Email Alerts: http://nro.sagepub.com/cgi/alerts

Subscriptions: http://nro.sagepub.com/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

Citations: http://nro.sagepub.com/content/8/5/497.refs.html

>> Version of Record - Oct 1, 2002

What is This?

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


PROGRESS IN CLINICAL NEUROSCIENCE ■

The Wnt Signaling Pathway in Bipolar Disorder


TODD D. GOULD and HUSSEINI K. MANJI
Laboratory of Molecular Pathophysiology
National Institute of Mental Health
Bethesda, Maryland

The Wnt signaling pathway is a highly conserved pathway critical for proper embryonic development.
However, recent evidence suggests that this pathway and one of its key enzymes, glycogen synthase
kinase 3β, may play important roles in regulating synaptic plasticity, cell survival, and circadian rhythms in
the mature CNS—all of which have been implicated in the pathophysiology and treatment of bipolar disor-
der. Furthermore, two structurally highly dissimilar medications used to treat bipolar disorder, lithium and
valproic acid, exert effects on components of the Wnt signaling pathway. Together, these data suggest that
the Wnt signaling pathway may play an important role in the treatment of bipolar disorder. Here, the authors
review the modulation of the Wnt/GSK-3β signaling pathway by mood-stabilizing agents, focusing on two
therapeutically relevant aspects: neuroprotection and modulation of circadian rhythms. The future devel-
opment of selective GSK-3β inhibitors may have considerable utility not only for the treatment of bipolar
disorder but also for a variety of classical neurodegenerative disorders. NEUROSCIENTIST 8(5):497–511,
2002: DOI: 10.1177/107385802237176
KEY WORDS Wnt signaling, Glycogen synthase kinase, Lithium, Valproic acid, Neuroprotection, Circadian rhythms, Bipolar disorder

Bipolar disorder (BD, also referred to as manic-depressive the United States alone (Begley and others 2001). It is
illness) is a common, chronic, and often life-threatening thus not altogether surprising that the Global Burden of
illness (Goodwin and Jamison 1990). The disease is Disease Study has identified BD as one of the leading
equally prevalent in men and women and can occur at causes of disability worldwide (Murray and Lopez
any age, with the early twenties being the median age of 1996).
onset (Goodwin and Jamison 1990; Angst and Sellaro Despite the devastating impact BD has on the lives of
2000). Two almost diametrically opposite mood states millions worldwide, little is known about its etiology or
characterize BD: mania and depression. Mania is char- pathophysiology. As is the case with most psychiatric ill-
acterized by a hyperaroused state (either euphoric or nesses, the etiology of BD was once thought to be due
dysphoric), increases in motor activity, racing thoughts, solely to environmental and social causes; however,
impaired judgment, decreased sleep, and an apparent there is now incontrovertible evidence that genetic fac-
decreased need for sleep. The depressive phases of the tors play a major role. Whereas BD has an overall life-
illness, which occur in the same individuals, are charac- time incidence of about 1%, first-degree relatives and
terized by depressed mood, suicidal ideation, anhedonia, monozygotic twin siblings of a patient with BD develop
impaired sleep, reduced psychomotor activity, and the disorder about 7% and 60% percent of the time,
impaired memory and concentration. Although exact respectively (Goodwin and Jamison 1990; Craddock and
figures are difficult to obtain, suicide is estimated to be Jones 1999; Potash and DePaulo 2000). Although bio-
the cause of death in upwards of 15% of individuals with logical factors portend a predisposition for the develop-
BD, and many other deleterious health-related effects are ment of BD, environmental factors also affect the out-
increasingly being recognized (Jamison 1986; Goodwin come. For example, sleep deprivation, drug use, and
and Jamison 1990). The lifetime cost estimate of dis- changes in hormonal levels (i.e., during the postpartum
ability and general medical care of persons with onset of period) are all documented precipitants of mania in sus-
bipolar disorder in 1998 is projected to be $24 billion in ceptible individuals (Goodwin and Jamison 1990). In a
consolidated view, current models for BD—and other
diseases with complex genetics—suggest that an inter-
Peter S. Klein, University of Pennsylvania, provided invaluable com- play between genetic, environmental, and epigenetic fac-
ments regarding the substance and style of this review. Sarah K. Tsou tors causes the disease (Craddock and Jones 2001;
provided excellent editorial assistance. The National Institute of
Mental Heath and the Theodore and Vada Stanley Foundation support Petronis 2001).
our research. Space limitations necessitated the citing of review papers The brain systems that have heretofore received the
in many cases; a full reference list upon which this article was based greatest attention in neurobiologic studies of BD have
is available upon request. been the monoaminergic neurotransmitter systems,
Address correspondence to: Husseini K. Manji, Laboratory of which were implicated by discoveries that effective anti-
Molecular Pathophysiology, Bldg 49, Room B1EE16, NIMH, NIH, 49 depressant drugs exerted their initial biochemical effects
Convent Dr., Bethesda, MD 20892 (email: manjih@intra.nimh.nih.gov). by regulating the intrasynaptic concentrations of sero-

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 497


Copyright © 2002 Sage Publications
ISSN 1073-8584

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


tonin and norepinephrine. Antihypertensives that deplet- that the two forms may have very similar—though not
ed these monoamines sometimes precipitated depressive identical—biological properties (Plyte and others 1992;
episodes in susceptible individuals (Delgado and others Ali and others 2001).
1999). Furthermore, the monoaminergic systems are GSK-3β is a serine/threonine kinase that is believed to
extensively distributed throughout the network of limbic, be, in general, constitutively active in cells. Phosphory-
striatal, and prefrontal cortical neuronal circuits thought lation of GSK-3β by other kinases (including PKC and
to support the behavioral and visceral manifestations of Akt) generally results in a decrease in its activity. Thus,
mood disorders (Drevets 2000b). Thus, clinical studies the activity of GSK-3β can be regulated by the action of
over the past 40 years have utilized assessments of cere- a variety of other cellular signaling mechanisms, which
brospinal fluid chemistry, neuroendocrine responses to may serve to fine-tune this constitutively active
pharmacological challenge, and neurotransmitter recep- enzyme’s activity (Woodgett 2001). One of GSK-3β’s
tor and transporter binding, demonstrating a number of major substrates is β-catenin, which is found in two
abnormalities of the serotonergic, noradrenergic, and functional reservoirs within the cell—a cytoplasmic and
other neurotransmitter and neuropeptide systems in BD a membrane-associated pool. The cytoplasmic pool is
(Maes and Meltzer 1995; Schatzberg and Schildkraut involved in transmitting the Wnt signal to the nucleus,
1995; Garlow and others 1999). Although such investi- whereas the membrane-associated pool interacts with
gations have been heuristic over the years, they have cadherin to provide structural support in cell adhesions.
been of limited value in elucidating the unique neurobi- Phosphorylation of β-catenin by GSK-3β is regulated by
ology of BD. More recently, there has been a growing a complex that includes axin, adenomatous polyposis
appreciation that although dysfunctions within the coli (APC), frequently rearranged in T-cell lymphoma
monoaminergic neurotransmitter systems are likely to (FRAT1), disheveled, casein kinase Iε, and protein phos-
play important roles in mediating many facets of the phatase-2A (Peifer and Polakis 2000) (Fig. 1).
pathophysiology of BD, they likely represent the down- Phosphorylation of β-catenin by GSK-3β results in its
stream effects of other, more primary abnormalities degredation in a ubiqitin-dependent manner, thereby
(Bowden 1997; Manji and Lenox 2000). Consequently, terminating/inhibiting the actions of this important
recent evidence demonstrating that impairments of neu- mediator of the Wnt signaling pathway (Peifer and
roplasticity and cellular resilience may underlie the Polakis 2000; Sharpe and others 2001). β-catenin binds
pathophysiology of BD and that mood stabilizing agents with the T-cell factor/lymphoid enhancer factor (tcf/lef)
exert major effects on signaling pathways that regulate transcription factor that translocates to the nucleus and
critical neuroplastic events and cell survival has generat- acts as a transcription factor at lef/tcf sites in the pro-
ed considerable excitement among the clinical neuro- moter of a number of genes (Novak and Dedhar 1999;
science community and is reshaping views about the Harwood 2001). Activation of the Wnt pathway in early
neurobiological underpinnings of these disorders. In this development modulates organizers, which are groups of
perspectives paper, we review the emerging data sug- cells that provide instructions for early development and
gesting that the Wnt signaling pathway and one of its key organization of the surrounding tissue, and thereby reg-
intermediaries, GSK-3β, may play an important role in ulates cell fate determination and body patterning
the pathophysiology and treatment of BD and that strate- (Cadigan and Nusse 1997; Wodarz and Nusse 1998).
gic targeting of this pathway may assist in the develop- Although this linear description of Wnt signaling—
ment of improved therapeutics for this devastating neu- the canonical pathway—has received the most attention,
ropsychiatric disorder. recent evidence suggests that Wnt signal may interact
with other signaling pathways. For example, at least in
The Wnt Signaling Pathway: An Overview certain systems, phosphatidyl inositol and Ca2+ signaling
appear to be activated by Wnt-stimulation of the frizzled
The Wnt signaling pathway is highly conserved and family of receptors (Kuhl and others 2000; Patapoutian
plays a critical role in normal development in diverse and Reichardt 2000).
species from Drosophila to human. In humans, the Wnt
protein family is composed of at least 15 secreted glyco-
Wnt Signaling in the Mature CNS
proteins (Wodarz and Nusse 1998). These proteins bind
to the frizzled family of extracellular receptors, resulting GSK-3β is highly expressed in the adult brain (Ali and
in a signal that is transduced via an intracellular protein, others 2001) and is known to phosphorylate a number of
disheveled. Signaling through disheveled results in inhi- important cytoskeletal proteins, including three major
bition of the enzyme glycogen synthase kinase (GSK), microtubule-associated proteins: Tau, MAP-1B, and
which is also evolutionarily conserved, is found in MAP-2 (Salinas and Hall 1999). Recent studies have
species ranging in diversity from Dictyostelium to suggested that changes in GSK-3β-mediated MAP-1B
humans, and is found in two forms in mammals: 3α and phosphorylation are associated with the loss and/or
3β. Both GSK isozymes were originally identified based unbundling of stable axonal microtubules (Lucas and
on their ability to phosphorylate—and thereby inacti- others 1998). Furthermore, GSK-3β inhibition results in
vate—glycogen synthase, resulting in stimulation of the the accumulation of synapsin I, a protein involved in
formation of glycogen. Although most research has synaptic vesicle docking and release, at growth cone-like
focused on the 3β isoform, available evidence suggests areas (Lucas and Salinas 1997). Wnt-7a may mimic

498 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Fig. 1. The canonical Wnt pathway. Signaling through Wnt glycoproteins and frizzled receptors activates disheveled. Disheveled acti-
vation results in inhibition of glycogen synthase kinase-3β (GSK-3β). Phosphorylation of β-catenin by GSK-3β results in its degrada-
tion by ubiquitin. Nondegraded (nonphosphorylated) β-catenin binds to lef/tcf transcription factors, targeting transcription of specific
genes. Lithium competes with Mg++ to inhibit GSK-3β (Ryves and Harwood 2001). Valproic acid (VPA) may be an inhibitor of GSK-3β.
Alternately, it may exert its action on Wnt signaling through inhibition of histone deacetylase, by its known actions on c-Jun, through
up-regulation of β-catenin mRNA, or by its action on the Ras/RSK pathway (Chen and others 1997, 1999; Phiel and others 2001; Yuan
and others 2001).

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 499

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


many of these actions, as this protein has recently been implicate the canonical Wnt signaling cascade per se in
shown to induce axon and growth cone remodeling (Hall mediating these effects (Jope and Bijur 2000). In an attempt
and others 2000). Although additional research is clear- to address the specific role of Wnt in the regulation of
ly required, these data suggest that Wnt signaling apoptosis, a recent study reported that Rat-1 cell lines
through GSK-3β plays important roles in axonal remod- stable expressing Wnt-1 were resistant to vincristine- and
eling and regulation of synaptic connectivity. vinblastine-regulated apoptosis and that these findings
In addition to its apparent role in regulating synapse appeared to be mediated by Tcf-mediated transcription
formation and axonal growth in developing systems, (Chen and others 2001). By contrast, another study sug-
there has been considerable recent excitement regarding gested pro-apoptotic effects of up-regulation of components
the role of GSK-3β in regulating cell death in mature of the Wnt pathway (van Gijn and others 2001). Although
neuronal tissue, and for the development of GSK-3β additional studies are clearly needed, these findings sug-
inhibitors as novel therapeutic agents for neurodegener- gest that the precise manner in which the Wnt signaling
ative diseases. Although it was initially reported in 1993 pathway regulates cell survival/cell death may be both
that GSK-3β activity was required for β-amyloid- development stage and cell line/cell type specific.
induced neurotoxicity in primary hippocampal neurons Perhaps the strongest evidence suggesting that the
(Takashima and others 1993), these observations were Wnt signaling pathway may play a major role in cell sur-
not followed up until very recently. Indeed, it is quite vival comes from the observations that mutations in Wnt
likely that the demonstration in 1996 that GSK-3β is a pathway genes are associated with a large number of
target for lithium’s actions (Klein and Melton 1996; tumors. For example, more than 80% of colorectal carci-
Phiel and Klein 2001) has greatly contributed to the nomas have mutations in the APC or β-catenin genes
resurgence in interest of GSK-3β as a potential thera- (Miyoshi and others 1992; Polakis 2000). APC and β-
peutic target. Recent studies have demonstrated that catenin mutations are also found in medulloblastomas
GSK-3β may regulate cell death beyond its role in β- and neuroectodermal tumors, albeit at a much lower per-
amyloid-induced toxicity. For example, GSK-3β overex- centage (Huang and others 2000; Koch and others 2001).
pression was found to induce apoptosis in Rat-1 and PC- Finally, Turcot’s syndrome, characterized clinically by
12 cells; additionally, dominant negative GSK-3β the co-occurrence of primary brain tumors and multiple
mutants prevented apoptosis following inhibition of PI colorectal adenomas, appears to be caused in most cases
3-kinase in these cells (Pap and Cooper 1998). by defects in the APC gene (Hamilton and others 1995).
Furthermore, the expression of FRAT-1, a protein that is At this point, it should be acknowledged that all the
thought to interact with the β-catenin/axin/GSK-3β evidence thus far regarding the neuroprotective proper-
complex and inhibit GSK-3β in a substrate-specific ties of the Wnt pathway has come from primary and
manner, also rescues primary sympathetic neurons from immortalized cell culture studies. Thus, it is not com-
PI 3-kinase inhibition-induced cell death (Crowder and pletely clear to what extent this signaling pathway exerts
Freeman 2000). A number of endogenous growth factors a neurotrophic/neuroprotective role in models of health
(e.g., nerve growth factor and brain-derived neurotroph- and disease in the adult CNS. Nevertheless, as we dis-
ic factor [BDNF]) utilize the PI 3-kinase signaling cas- cuss in greater detail later, the possibility that this system
cade as a major effector system. Thus, growth factors can be manipulated pharmacologically to provide neuro-
may bring about many of their neurotrophic/neuropro- protection is compelling and is an active area of
tective effects, at least in part, by GSK-3β inhibition. research.
Consistent with such a contention, serum deprivation or
PI 3-kinase-induced apoptosis is attenuated by either a
dominant negative form of GSK-3β or an inhibitory GSK-3β and Circadian Rhythms
GSK-3β binding protein (Hetman and others 2000). Another role for GSK-3β that may be of considerable
Neuronal apoptosis in all three of these experiments was importance in the pathophysiology and treatment of BD
decreased following exposure to either a dominant neg- is the regulation of circadian rhythms. Emerging work
ative form of GSK-3β or an inhibitory GSK-3β binding from Drosophila suggests that the ortholog of GSK-3β
protein. In this regard, as discussed later in this review, in this species, SHAGGY, may play a very important
lithium is neuroprotective in many preclinical models role in regulating circadian rhythms (Martinek and oth-
(Manji and others 1999b). ers 2001). Specifically, this group found that overex-
Although the study of the effects of selective small pression of SHAGGY results in a shortening of the peri-
molecule GSK-3β inhibitors is still in its infancy, the od of the Drosophila circadian locomotor activity cycle,
available data suggest that pharmacological inhibition of a predictable measure of circadian rhythmicity in this
GSK-3β also exerts neuroprotective effects. Two novel species. Furthermore, a reduction of SHAGGY activity
inhibitors of GSK-3β have been demonstrated to protect resulted in a lengthening of the period. SHAGGY
primary sensory and granule neurons from potassium appears to act by modulating phosphorylation of the
deprivation or phosphatidylinositol 3-kinase-induced TIMELESS protein, an action that advances the nuclear
cell death (Cross and others 2001). entry of the TIMELESS/PERIOD dimer, thus promoting
As discussed already, GSK-3β is the target of multiple the shortening of the Drosophila circadian cycle
signaling systems, and the demonstration of GSK-3β’s role (Martinek and others 2001) (Fig. 2). Knowledge regard-
in neuronal survival and apoptosis does not necessarily ing human circadian physiology is not as advanced as

500 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Fig. 2. The circadian cycle in Drosophila. The CYCLE/CLOCK dimer initiates transcription of both period and timeless genes in the
early part of the day, and mRNA levels reach their peak at the beginning of the night. PERIOD accumulates at a faster rate but earli-
er in the day and is marked for degradation when phosphorylated by DOUBLETIME. As levels of TIMELESS increase in the cytoplasm,
unphosphorylated PERIOD binds with the phosphorylated form of TIMELESS, resulting in this dimmer translocating to the nucleus
and inhibiting the transcription of TIMELESS and PERIOD (Williams and Sehgal 2001). SHAGGY/ZESTY WHITE, the Drosophila
ortholog of glycogen synthase kinase-3β (GSK-3β), appears to be the enzyme that phosphorylates TIMELESS (Martinek and others
2001). TIMELESS is light sensitive and breaks down at sunrise. This leads to the breakdown of unbound PERIOD, thus allowing the
cycle to renew itself (Williams and Sehgal 2001). Many patients with bipolar disorder have abnormalities in circadian physiology (Wehr
and others 1983). Furthermore, lithium lengthens the circadian cycle in diverse species (Klemfuss 1992). The action of lithium on GSK-
3β may help to explain this finding.

that in Drosophila; however, although a true TIMELESS 2001). Furthermore, CKIε, which we have previously
ortholog has not been identified, orthologs of many of mentioned as an enzyme that interacts with GSK-3β, is
the proteins are present in humans, likely operate in a mutated in the autosomal dominant circadian mutant in
very similar manner, and may therefore play important Syrian hamsters, is the doubletime circadian mutant in
roles in the regulation of human circadian rhythms flies, and also interacts with GSK-3β and the Wnt sig-
(Wager-Smith and Kay 2000; Reppert and Weaver naling pathway, suggesting that these occurrences may

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 501

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


be linked (Price and others 1998; Peters and others 1999; (Stambolic and others 1996; Hedgepeth and others
Sakanaka and others 1999; Lowrey and others 2000; 1997). Although some indirect in vivo effects should not
Vielhaber and Virshup 2001). be entirely excluded at this point (Chalecka-Franaszek
We now turn to a discussion of the Wnt signaling and Chuang 1999), lithium inhibits GSK-3β in vitro by
pathway in the pathophysiology and treatment of BD. direct competition with magnesium, an ion that has a
similar hydrated ionic radius to that of lithium (Ryves
The Wnt Signaling Pathway and Harwood 2001). The action is specific to lithium, as
Is a Target of Lithium other group I metal ions—sodium, potassium, cesium,
and rubidium—have no effect on GSK-3β in vitro activ-
The discovery of lithium’s efficacy as a mood-stabilizing ity (Klein and Melton 1996; Ryves and Harwood 2001).
agent revolutionized the treatment of patients with BD; Importantly, in addition to the in vitro findings, lithium
indeed, it is likely that the remarkable efficacy of lithium has also been demonstrated to inhibit the phosphoryla-
has, in fact, served to spark a revolution that has, over tion of GSK-3β substrates, tau, and MAP-1B in vivo
time, reshaped not only medical and scientific but also (Stambolic and others 1996; Hedgepeth and others
popular concepts of severe mental illnesses. After 3 1997; Lucas and others 1998).
decades of use in North America, lithium continues to be Lithium exerts profound developmental effects in
one of the mainstays of treatment for this disorder, both diverse organisms, and the identification of GSK-3β as
for the acute manic phase and as a prophylaxis for recur- a target for lithium’s actions has provided a clear cellular
rent manic and depressive episodes (Goodwin and basis for many of the observed phenotypes; indeed,
Jamison 1990; Manji and Lenox 2000). Adequate lithi- many of the phenotypes are replicated by activation of
um treatment, particularly in the context of a lithium the Wnt pathway (Phiel and Klein 2001). For example,
clinic, is also reported to reduce the excessive mortality treating Dictyostelium with lithium results in the diver-
observed in the illness (Vestergaard and Aagaard 1991; sion of presumptive spore cells to the stalk cell fate, and
Muller-Oerlinghausen and others 1992; Tondo and oth- loss of GSKA, the GSK-3β homologue in this species,
ers 1997), and it has been estimated that in most indus- results in a remarkably similar phenotype (Maeda 1970;
trialized countries, 0.1% of the population is undergoing Harwood and others 1995). In Xenopus and zebrafish,
lithium treatment (Schou 1991). The effect on the broad- lithium administration mimics the activation of the Wnt
er community is highlighted by the estimation that the pathway by ectopic expression of Wnt, β-catenin, domi-
use of lithium saved the United States $4 billion during nant negative GSK-3β administration, or inhibitory
the years 1969 to 1979 by reducing associated medical forms of axin with expansion of the dorsal mesoderm
costs and restoring productivity (Reifman and Wyatt and duplication of the dorsal axis (Kao and Elinson
1980). However, despite its role as one of psychiatry’s 1988; McMahon and Moon 1989; Yost and others 1996;
most important treatments, the biochemical basis for Zeng and others 1997; Yost and others 1998; Hedgepeth
lithium’s antimanic and mood-stabilizing actions and others 1999).
remains to be elucidated (Jope 1999; Manji and Lenox As expected from a direct GSK-3β inhibitor, lithium
2000). administration leads to the accumulation of β-catenin in
Until fairly recently, there had been only one clear diverse species such as Xenopus embryos, sea urchin
biochemical target of lithium at a concentration of ~1 embryos, and cultured mammalian epithelial cells and
mM (similar to the therapeutic plasma levels). It has neurons (Stambolic and others 1996; Hedgepeth and
been known for many years that lithium, at therapeuti- others 1997; Lucas and Salinas 1997; Emily-Fenouil and
cally relevant concentrations, inhibits inositol others 1998; Wikramanayake and others 1998; Chen and
monophosphatases (IMPases) (Berridge and others others 2000; Phiel and Klein 2001). Most relevant to the
1982, 1989), and other monoesterases; however, the potential therapeutic relevance of pharmacologic inhibi-
“inositol depletion hypothesis” of lithium’s actions has tion of GSK-3β, chronic (3-week) lithium administra-
received limited direct experimental support (Jope and tion at therapeutic concentrations increases β-catenin
Williams 1994; Jope 1999; Phiel and Klein 2001). Thus, expression in the rat hippocampus (Guang Chen and H.
there was considerable excitement generated 5 years ago K. Manji, unpublished observation). Finally, lithium also
by the identification of a novel target for lithium’s increases transcription from lef/tcf reporter constructs
actions. Klein and Melton (1996) were the first to report (Staal and others 1999; Vonica and others 2000; Phiel
that lithium was an inhibitor of GSK-3β in vitro. These and others 2001).
studies arose out of their observation that lithium’s In sum, as described by Phiel and Klein, the evidence
effects on the dorsalization of the Xenopus embryo are that GSK-3β is a target of lithium includes the follow-
similar to the effects following activation of the Wnt sig- ing: 1) in vitro inhibition, 2) in vivo inhibition, 3) lithi-
naling pathway. They further showed that the selective um phenocopying of GSK-3β loss of function, 4) alter-
inhibition of IMPase with other agents did not mimic native inhibitors of GSK-3β mimic lithium action, and
lithium’s effects in Xenopus, leading to their studies 5) elevations of the levels of GSK-3β reverse the pheno-
demonstrating that lithium, at therapeutically relevant typic effects of lithium (Phiel and Klein 2001) (Table 1).
concentrations, is a direct inhibitor of GSK3β (Klein and Although the evidence that lithium is an inhibitor of
Melton 1996). This original study was followed by addi- GSK-3β is compelling, a major problem inherent in BD
tional reports confirming and extending these findings research is the ability to ascribe therapeutic relevance to

502 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Table 1. Effects of lithium and valproate on the Wnt pathway.

Lithium Valproate

Directly inhibits GSK-3β by competition with Magnesium Inhibits GSK-3β in some in vitro experiments, but not in
Inhibits the phosphorylation of GSK-3β substrates in cell others.
culture Increases back-phosphorylation of a putative GSK-3β
Mimics the effects of Wnt stimulation on development of substrate (GSK-3β enzyme is added to lysate after
diverse species incubation with valproate)
Increases β-catenin protein levels in cell culture Increases β-catenin protein levels in cell culture
Increases β-catenin protein levels in rat hippocampus Increases β-catenin mRNA expression in cell culture
Inhibits GSK-3β mediated potentiation of staurosporine- Increases β-catenin protein levels in rat hippocampus
induced apoptosis in cell culture Inhibits GSK-3β mediated potentiation of staurosporine-
Decreases platelet activating factor mediated increases induced apoptosis in cell culture
in GSK-3β activity in cell culture Decreases platelet activating factor mediated increases
Increases transcription from lef/tcf reporter constructs in GSK-3β activity in cell culture
Increases transcription from lef/tcf reporter constructs

Both lithium and valproic acid have effects on the Wnt pathway. See text and the following references: Maeda (1970); Kao and Elinson
(1988); McMahon and Moon (1989); Harwood and others (1995); Klein and Melton (1996); Stambolic and others (1996); Yost and oth-
ers (1996); Hedgepeth and others (1997); Lucas and Salinas (1997); Emily-Fenouil and others (1998); Lucas and others (1998);
Wikramanayake and others (1998); Yost and others (1998); Chen and others (1999); Hedgepeth and others (1999); Staal and others
(1999); Bijur and others (2000); Chen and others (2000); Vonica and others (2000); Grimes and Jope (2001); Phiel and Klein (2001);
Phiel and others (2001); Ryves and Harwood (2001); Tong and others (2001); Li and others (in press).

any observed biochemical finding. There are not clear recombinant GSK-3β mediated 32P incorporation into
phenotypic changes associated with treatment response, two putative GSK-3β substrates (Chen and others 1999).
and the available animal models are less than ideal. Furthermore, 1-day (and longer) treatment of SH-SY5Y
Indeed, numerous extracellular and intracellular effects cells with a therapeutically relevant concentration of
have been documented for lithium (Wang and others VPA (0.6 mM) resulted in a significant increase in both
1997; Manji and others 1999a). Whereas some of these nuclear and cytoplasmic β-catenin protein levels.
effects are likely related to the treatment efficacy of Additionally, similar to the effects of lithium, VPA also
these drugs, others may cause the numerous side effects, increased β-catenin levels in the rat hippocampus after
or simply be epiphenomenon, with no clinical signifi- chronic (3-week) VPA treatment at therapeutic concen-
cance. An approach some laboratories have undertaken trations (Guang Chen and H. K. Manji, unpublished
to locate potentially therapeutic targets involves identi- observation). Further supporting an action of VPA on
fying targets that are common to structurally dissimilar Wnt signaling, VPA has recently been demonstrated to
agents belonging to the same therapeutic class (e.g., increase lef/tcf-mediated reporter gene expression in
mood stabilizers) (Chen and others 1994; Lenox and Neuro 2A cells (Phiel and others 2001). Additionally,
others 1996; Manji and others 1996). Thus, although both VPA and lithium attenuate a platelet-activating-
chemically distinct agents likely do not work through factor-mediated increase in GSK-3β activity (Tong and
identical mechanisms, identification of targets that they others 2001) (Table 1).
regulate in concert in appropriate paradigms may great- Although the mechanism of lithium’s effect on the
ly serve to validate their therapeutic relevance. In this Wnt pathway appears to be due in large part (if not
context, it is noteworthy that the two most efficacious exclusively) to direct inhibition of GSK-3β, it is less
and most widely used agents in the treatment of BD, clear precisely how VPA inhibits this pathway—both
lithium and valproic acid (VPA), are structurally highly direct and indirect mechanisms may be operative. Thus,
dissimilar. It is thus not altogether surprising that recent Chen and others (1999) reported in vitro inhibition of
studies have begun to explore the possibility that the Wnt GSK-3β and –3α mediated phosphorylation of a CREB
signaling pathway may represent a convergent pathway peptide. Furthermore, a recent finding by Grimes and
for the treatment of BD by investigating the effects of Jope supports a direct inhibitory effect of VPA on GSK-
VPA on this signaling pathway. 3; they found that VPA inhibits the ability of immuno-
precipitated GSK-3β to phosphorylate recombinant
The Wnt Signaling Pathway Is Also a Target human tau in vitro (Grimes and Jope 2001). By contrast,
for the Actions of Valproic Acid Phiel and others found that VPA did not inhibit the in
vitro phosphorylation of glycogen synthase peptide or
Chen and others (1999) first described VPA as an acti- the phosphorylation of tau in cell culture (Phiel and oth-
vator of the Wnt pathway. They observed that incubation ers 2001). VPA also inhibits the phosphorylation of
of human neuroblastoma SH-SY5Y cells with 0.6 mM MAP-1B by GSK-3β in vivo, but not in vitro, suggest-
VPA caused an increase in the subsequent in vitro ing that VPA inhibits the GSK-3β-mediated phosphory-

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 503

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Table 2. Neuroprotective and neurotrophic effects of lithium.

Cultured Cells (human and rodent) Rodent Brain In Vivo Human Effects

Protects cells against a number of Reduces behavioral deficits and Increases N-acetyl asparate (NAA)
toxic stimuli including choline acetyltransferase activity levels in the brains of BD
induced by cholinergic lesions patients
Glutamate Protects against radiation injury Increases gray matter volume in the
NMDA Protects against cellular and behav- brains of BD patients
Calcium ioral changes after middle cereb- BD patients chronically
MPP+ ral artery occlusion treated with either lithium or val-
Thapsigarin Protects against quinolinic acid proate do not show reduced
β-amyloid toxicity PFCx volumes
Ouabain Enhances hippocampal neurogenesis
Age
Phenytoin
Carbamazepine
KCI deprivation
Serum deprivation
NGF deprivation
GSK-3β + staurosporin
or heat shock

Both preclinical (in cell culture and in intact animals) and clinical evidence suggests that lithium has neuroprotective effects. See Manji
and others (1999b, 2000a, 2000b) and the references found within.

lation of MAP-1B indirectly in intact neurons (Patricia ilar effects (at therapeutically relevant concentrations)
Salinas, personal communication). Thus, VPA inhibition on this important signaling pathway known to regulate
of GSK-3β may be substrate specific, or possibly due to synaptic plasticity, circadian rhythms, and neuronal sur-
differences in assay conditions (Phiel and Klein 2001). vival (Table 1).
At present, the degree to which VPA directly inhibits
GSK-3β, and its potential substrate specificity, requires Preclinical and Clinical Studies Suggest That
further delineation. Nevertheless, it is clear that VPA Mood Stabilizers Have Neurotrophic Effects
increases a downstream target of Wnt signaling, β-
catenin. Further recent data have identified an addition- The evidence reviewed above suggests that GSK-3β and
al pathway—isolated from GSK-3β—by which VPA may β-catenin are targets for the actions of both lithium and
activate the Wnt signaling cascade. VPA is an inhibitor VPA. Do these effects, in fact, have the potential to
of histone deacetylase (HDAC) (IC50 = 0.4 mM) and translate into therapeutic effects in patients? There is
increases β-catenin mRNA expression in Neuro 2A cell now a large body of data that shows that lithium, at con-
lines (Phiel and others 2001). It is noteworthy that other centrations similar to its IC50 for GSK-3 inhibition,
HDAC inhibitors—butyrate (like VPA, a small fatty exerts neurotrophic and neuroprotective effects in a vari-
acid) and trichostatin-A—also increase the transcription ety of preclinical and clinical paradigms (Table 2)
of tcf reporter constructs in cell culture (Bordonaro and (Chuang and others 2002; Jope and Bijur 2002). For
others 1999); thus, VPA’s effects on β-catenin mRNA, β- example, lithium protects cerebellar granule cells
catenin protein levels, and tcf transcriptional activity against glutamate-, NMDA receptor-, low potassium-,
may be mediated in part by HDAC inhibition. and toxic level of anticonvulsant-induced cell death
A final mechanism by which VPA may indirectly (Nonaka and others 1998a, 1998b; Manji and others
inhibit GSK-3β and regulate β-catenin is through the 2000b). In addition to the demonstration of protective
ERK/MAP kinase/RSK pathway. This pathway has effects in vitro, a number of studies have also investigat-
recently been demonstrated to be robustly activated by ed lithium’s neuroprotective effects in vivo. In this con-
VPA (Yuan and others 2001). Furthermore, GSK-3β can text, lithium has been shown to protect against a variety
be phosphorylated by Rsk, suggesting a mechanistic link of excitotoxic and ischemic insults (Pascual and
between the actions of VPA and the regulation of GSK- Gonzalez 1995; Nonaka and Chuang 1998; Arendt and
3β and β-catenin (Sutherland and others 1993; others 1999; Manji and others 1999b, 2000b). Please see
Stambolic and Woodgett 1994; Eldar-Finkelman and Table 2 and references contained within Manji and oth-
others 1995; Kleijn and Proud 2000; Desbois-Mouthon ers (2000b) and Manji and others (1999b) for a complete
and others 2001). review of lithium’s neuroprotective effects.
Overall, although they may regulate the Wnt signaling Although not as well studied, VPA also exerts neuro-
pathway by nonidentical mechanisms, it is striking that protective actions in several paradigms (Bruno and oth-
these two structurally highly dissimilar agents exert sim- ers 1995; Mark and others 1995; Mora and others 1999;

504 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Fig. 3. Neuroprotective effects of lithium and valproic acid (VPA) in human SH-SY5Y neuroblastoma cells. Cells were incubated with
either lithium (1.0 mM) or VPA (1.0 mM) for 3 days and then exposed to two different toxins—thapsigarin (which mobilizes intracellu-
lar calcium; 0.5 mM for 16 h) or MPP+ (25 uM for 16 h). The mitochondrial dehydrogenase activity that cleaves 3-(4,5-dimethylthiazol-
2-yl)-2,5-diphenyl tetrazolium bromide (MTT) was used to determine cell survival in a quantitative colormetric assay. Three days of
lithium or VPA treatment exerted significant protective effects against both toxins. *P < 0.05. (Copied with permission from Manji and
others 2000a.)

Manji and others 2000b). In a direct comparison, Manji Are the Neurotrophic and Neuroprotective Effects
and others (2000a) reported the neuroprotective effects of Mood Stabilizers Possibly Therapeutically
of lithium and VPA. Lithium and VPA protected Relevant in the Long-Term Treatment of BD?
human neuroblastoma SH-SY5Y cells from both thapsi-
garin- and MPP+-induced apoptosis at therapeutically Mood disorders have traditionally been conceptualized
relevant concentrations. Furthermore, at therapeutic as neurochemical disorders, but there is now evidence
concentrations, the degree of protection from both drugs from a variety of sources demonstrating regional reduc-
was similar (Fig. 3). VPA and lithium also protect SH- tions in CNS volume, as well as reductions in the num-
SY5Y cells from staurosporine-induced apoptosis in bers and/or size of glia and neurons in discrete brain
cells stably transfected with GSK-3β in a model that areas (Manji and others 2000b). Although the precise
has been shown to have the same results after treatment cellular mechanisms underlying these morphometric
with lithium, thus linking the neuroprotective effects of changes remain to be elucidated, the data suggest that
these medications with the Wnt/GSK-3β pathway mood disorders are associated with impairment of struc-
(Bijur and others 2000; Li and others 2002). Inhibition tural plasticity and cellular resilience. Structural imag-
of GSK-3β and/or the activation of the Wnt signaling ing studies have demonstrated reduced gray matter vol-
pathway is an attractive mechanism to explain these ume in areas of the orbital and medial prefrontal cortex
findings. (PFC), ventral striatum and hippocampus, and enlarge-

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 505

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


ment of third ventricles in mood disordered relative to metric MRI study demonstrated that 4 weeks of lithium
healthy control samples (Drevets and others 1999; Manji treatment also significantly increased total gray matter
and Duman 2001). Complementary postmortem neu- content in the human brain (Moore and others 2000b)
ropathological studies have shown abnormal reductions (Fig. 5). Taken together, these clinical data support the
in cortex volume, glial cell counts, and/or neuron size in hypothesis that mood stabilizers may exert hitherto
the subgenual PFC, orbital cortex, dorsal anterolateral underappreciated neurotrophic/neuroprotective effects
PFC, and amygdala (Manji and others 2000b; in humans, effects that are likely due, at least in part, to
Rajkowska in press). Finally, a recent study found a their effects on GSK-3β and/or Wnt pathway regulation.
decrease in N-acetyl-aspartate (NAA, a putative marker
of neuronal viability [Tsai and Coyle 1995]), in the dor- Does Lithium Also Regulate Circadian Rhythms
solateral prefrontal cortex in patients with BD compared via Its Effects on GSK-3β?
to healthy control subjects (Winsberg and others 2000).
At present, it is not known whether these deficits consti- Abnormalities in circadian physiology are common in
tute developmental abnormalities that may confer vul- patients with BD (Wehr and others 1983) and have been
nerability to abnormal mood episodes, compensatory hypothesized to reflect an endophenotype of the illness
changes to other pathogenic processes, or the sequelae of (Lenox and others 2002). Many patients have changes in
recurrent affective episodes. Understanding these issues the circadian period of temperature, melatonin, and
will partly depend upon experiments that delineate the other hormones, with unclear clinical significance
onset of such abnormalities within the illness course and (Souetre and others 1988; Healy and Waterhouse 1995;
determination if they antedate depressive episodes in Pacchierotti and others 2001). Most patients with BD
individuals at high familial risk for mood disorders have sleep cycle abnormalities and other changes asso-
(Drevets and others 1999). The hypothesis that dysfunc- ciated with circadian physiology during both the depres-
tion of these regions may contribute to the development sive and manic stages of the illness (Kasper and Wehr
of at least some symptoms of BD is consistent with evi- 1992). Furthermore, alteration of biological rhythms
dence that lesions (e.g., strokes or tumors) involving through interventions like sleep deprivation exerts a
either the PFC or the striatum (a major target of efferent major effect in BD. Partial or total sleep deprivation is
projections from the PFC), and degenerative diseases capable of inducing a rapid antidepressant effect in
affecting the striatum (e.g., Parkinson’s and Huntington’s ~40% to 60% of depressed BD patients (Wu and Bunney
diseases), are associated with increased risk for develop- 1990; Wirz-Justice and Van den Hoofdakker 1999). The
ing depression (Drevets and others 1999). Overall, what- antidepressant effect is transient and reverses after a full
ever the precise underlying mechanisms, the impairment night of sleep, thereby making it impractical as a routine
of structural plasticity in regions of the brain most asso- therapeutic intervention. Although sleep deprivation
ciated with the pathophysiology of BD suggests that neu- clearly has to be considered as more than just a circadi-
rotrophic and neuroprotective effects of mood disorders an rhythm manipulation, the striking and rapid effect
may play a major role in the long-term treatment of BD. (much faster than pharmacologic interventions) points to
Although the body of preclinical data demonstrating the potential critical role of circadian rhythms in the
neurotrophic and neuroprotective effects of lithium and pathophysiology and treatment of BD. In further support
VPA is impressive, considerable caution must clearly be of such a contention, there is considerable evidence that
exercised in extrapolating to the clinical situation with sleep deprivation is capable of inducing mania in many
humans. In a retrospective analysis, Drevets and associ- patients with BD, leading to the hypothesis that sleep
ates reanalyzed their data demonstrating ~40% reduc- deprivation and mania affect each other in a reciprocal
tions in subgenual PFC volumes in familial mood disor- fashion, with one increasing the intensity of the other
der subjects (Drevets and others 1997). Consistent with (Wehr and others 1987).
neurotrophic/neuroprotective effects of lithium and These clinical findings add important substance to the
VPA, they found that the patients treated with chronic observation that lithium increases free-running (organ-
lithium or VPA exhibited subgenual PFC volumes that ism is without external time cues) circadian period
were significantly higher than the volumes in nonlithi- length in humans and many other species including uni-
um- or VPA-treated patients, and not significantly dif- cellular organisms, plants, insects, and mice (Klemfuss
ferent from controls (Drevets 2000a). To prospectively 1992). Hitherto, a biological target for this action has
investigate if lithium also exerts neurotrophic/neuropro- been lacking (Klemfuss 1992; Ikonomov and Manji
tective effects in the human brain in vivo, a longitudinal 1999). However, as discussed already, the Drosophila
clinical study was undertaken utilizing proton magnetic ortholog of GSK-3β, SHAGGY, was recently identified
resonance spectroscopy to quantitate NAA levels (Moore as a mediator of circadian rhythms in this species
and others 2000a). Chronic lithium administration was (Martinek and others 2001). Tissue-specific overexpres-
found to significantly increase NAA concentration, and sion of SHAGGY resulted in a shortening of the period
furthermore, a striking ~0.97 correlation between lithi- of the Drosophila circadian locomotor activity cycle.
um-induced NAA increases and regional voxel gray mat- Furthermore, a reduction of SHAGGY activity resulted
ter content was observed, thus suggesting that lithium in a lengthening of the period (Martinek and others 2001),
may be exerting these effects by targeting gray matter precisely the effect that lithium has on free-running cir-
(Fig. 4). Further-more, a follow-up high-resolution volu- cadian length in most organisms studied (Fig. 2). Thus,

506 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Fig. 4. Brain N-Acetyl-Aspartate (NAA) is increased following 4 weeks of lithium administration at therapeutic levels. Quantitative pro-
ton (1H) magnetic resonance spectroscopy (MRS) was utilized to measure brain NAA level at baseline and after 4 weeks of lithium
administration. Voxel placement for each region investigated a) frontal lobe, b) parietal lobe, c) occipital lobe, d) temporal lobe. Chronic
lithium increases brain NAA content in the regions investigated (left). ■, occipital; ●, temporal; ▲, frontal; ◆, parietal. Four weeks of
lithium adminstration resulted in a small (5%) but significant (P = 0.02) increase of total brain NAA concentration. NAA increases cor-
relate with percentage of gray matter content (right). (Modified and reproduced with permission from Moore and others 2000a.)

lithium’s inhibition of GSK-3β represents a putative changes and modulation of circadian rhythms. As dis-
cellular mechanism for the ability of this simple monova- cussed in this review, increasing preclinical and clinical
lent cation to alter circadian physiology in diverse organ- evidence implicates changes in neuroplasticity and cel-
isms. This also raises the possibility that GSK-3β inhibitors lular resiliency in both the pathophysiology and treat-
may have broad utility in the treatment of disorders ment of mood disorders (Jope 1999; Manji and others
associated with abnormalities of circadian rhythms. 2000b). Similarly, evidence accumulated over the past 3
decades suggests that changes in, or the modulation of,
Conclusions and Prospects circadian rhythms may underlie both the underlying
cause of BD and an explanation for why some treatments
Although some indirect in vivo effects should not be for the disorder are efficacious (Kasper and Wehr 1992).
excluded (Chalecka-Franaszek and Chuang 1999), lithi- Although both these areas of focus are certainly strong-
um does directly target GSK-3β—thereby stimulating ly correlated with BD, it remains to be seen with cer-
the Wnt pathway—at therapeutic concentrations (Klein tainty what the true underlying causes are.
and Melton 1996). Similarly, VPA targets the Wnt path- To date, although there is strong suggestive evidence
way and may also have more specific effects on GSK-3β that Wnt/GSK-3β regulation may be related to BD treat-
(Chen and others 1999). Although GSK-3β’s role as a ment efficacy, there is no direct evidence that abnormal-
mediator of many cellular processes leaves the quest for ities of Wnt signaling are present in BD. Although post-
putative downstream physiological targets wide open, mortem brain studies are fraught with methodological
we have focused on two targets that have been a focus of problems—including long postmortem delays, ante-
BD research in the past: regulation of neuroplastic mortem medication and drug use, causes of death that

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 507

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Fig. 5. Brain gray matter volume is increased following 4 weeks of lithium administration at therapeutic levels in patients with bipolar
disorder (BD). a) A slice of three-dimensional volumetric magnetic resonance imaging (MRI) data that was segmented by tissue type
using quantitative methodology to determine tissue volumes at each scan time point. Brain tissue volumes were examined using high-
resolution three-dimensional MRI (124 1.5 mm-thick coronal T1-weighted spoiled gradient echo images) and validated quantitative
brain tissue segmentation methodology to identify and quantify the various components by volume, including total brain white and
gray matter content. Measurements were made at baseline (medication free, after a minimum 14-day washout) and then repeated after
4 weeks of lithium at therapeutic doses. b and c, Chronic lithium significantly increases total gray matter content in the human brain
of patients with BD. No significant changes were observed in brain white matter volume or in quantitative measures of regional cere-
bral water. (Modified and reproduced with permission from Moore and others 2000b.)

have variable effects on brain tissue, and the inability to be alterations in other mediators of the Wnt pathway in
adequately study posttranslational modifications—two patients with BD.
studies have recently quantitated GSK-3β protein levels Recently, selective small molecule GSK inhibitors have
in the frontal cortex in BD patients, finding no differ- been developed (Coghlan and others 2000; Cross and oth-
ences from controls (Lesort and others 1999; Kozlovsky ers 2001; Smith and others 2001); these molecules have
and others 2000). However, studies have found altered been used successfully to protect cerebellar granule cells
levels of GSK-3—or other molecules in the Wnt from toxic stimuli (Cross and others 2001). Future research
pathway—in postmortem brains or peripheral cells from may examine these molecules for efficacy in the treatment
patients with another neuropsychiatric disorder, schizo- of BD. Even more promising may be the development of
phrenia (Yang and others 1995; Cotter and others 1998; drugs that, similar to FRAT1, inhibit GSK-3β-mediated
Miyaoka and others 1999; Kozlovsky and others 2000; phosphorylation of specific substrates (e.g., unprimed
Beasley and others 2001). In this regard, it is noteworthy substrates such as β-catenin and axin) but not substrates
that a leading hypothesis posits that early neurodevelop- requiring pre-phosphorylation, such as glycogen syn-
mental changes predispose to the development of schiz- thase, thus being specific for the Wnt-mediated effects
ophrenia in adulthood (Weinberger 1997). Clearly addi- of this multitalented enzyme (Thomas and others 1999).
tional studies are required to investigate these findings This approach may make it possible to discern which
further, as well as to pursue the possibility that there may GSK-3β pathway is relevant for the treatment of BD.

508 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Lithium and VPA have numerous effects on G-proteins Chen RH, Ding WV, McCormick F. 2000. Wnt signaling to beta-
and second messenger systems with undetermined clini- catenin involves two interactive components. Glycogen synthase
kinase-3beta inhibition and activation of protein kinase C. J Biol
cal significance (Gould and Manji 2002; Manji and oth- Chem 275:17894–9.
ers 1996; Wang and others 1997). These studies make it Chen S, Guttridge DC, You Z, Zhang Z, Fribley A, Mayo MW, and oth-
clear that lithium does not target only GSK-3β in cells. ers. 2001. Wnt-1 signaling inhibits apoptosis by activating beta-
It therefore remains to be seen if the effects of lithium catenin/T cell factor-mediated transcription. J Cell Biol 152:87–96.
Chuang DM, Chen RW, Chalecka-Franaszek E, Ren M, Hashimoto R,
and VPA on the Wnt signaling pathway and GSK-3β are Senatorov V, and others. 2002. Neuroprotective effects of lithium in
truly important for the treatment and/or the pathophysi- cultured cells and animal models of disease. Bipolar Disord 4:129–36.
ology of BD. The findings presented in this review could Coghlan MP, Culbert AA, Cross DA, Corcoran SL, Yates JW, Pearce
be related to the numerous side effects of these medica- NJ, and others. 2000. Selective small molecule inhibitors of glyco-
tions. However, the putative roles are intriguing and sug- gen synthase kinase-3 modulate glycogen metabolism and gene
transcription. Chem Biol 7:793–803.
gest that future research into the role of the Wnt pathway Cotter D, Kerwin R, al-Sarraji S, Brion JP, Chadwich A, Lovestone S,
in BD could provide valuable clues both for understand- and others. 1998. Abnormalities of Wnt signalling in schizophre-
ing the underlying pathophysiology of the disorder and nia—evidence for neurodevelopmental abnormality. Neuroreport
for developing valuable potential therapies. 9:1379–83.
Craddock N, Jones I. 1999. Genetics of bipolar disorder. J Med Genet
36:585–94.
References Craddock N, Jones I. 2001. Molecular genetics of bipolar disorder. Br
J Psychiatry 178:S128–33.
Ali A, Hoeflich KP, Woodgett JR. 2001. Glycogen synthase kinase-3: Cross DA, Culbert AA, Chalmers KA, Facci L, Skaper SD, Reith AD.
properties, functions, and regulation. Chem Rev 101:2527–40. 2001. Selective small-molecule inhibitors of glycogen synthase
Angst J, Sellaro R. 2000. Historical perspectives and natural history of kinase-3 activity protect primary neurones from death. J
bipolar disorder. Biol Psychiatry 48:445–57. Neurochem 77:94–102.
Arendt T, Lehmann K, Seeger G, Gartner U. 1999. Synergistic effects Crowder RJ, Freeman RS. 2000. Glycogen synthase kinase-3 beta
of tetrahydroaminoacridine and lithium on cholinergic function activity is critical for neuronal death caused by inhibiting phos-
after excitotoxic basal forebrain lesions in rat. Pharmacopsychiatry phatidylinositol 3-kinase or Akt but not for death caused by nerve
32:242–7. growth factor withdrawal. J Biol Chem 275:34266–71.
Beasley C, Cotter D, Khan N, Pollard C, Sheppard P, Varndell I, and Delgado PL, Miller HL, Salomon RM, Licinio J, Krystal JH, Moreno
others. 2001. Glycogen synthase kinase-3beta immunoreactivity is FA, and others. 1999. Tryptophan-depletion challenge in depressed
reduced in the prefrontal cortex in schizophrenia. Neurosci Lett patients treated with desipramine or fluoxetine: implications for the
302:117–20. role of serotonin in the mechanism of antidepressant action. Biol
Begley CE, Annegers JF, Swann AC, Lewis C, Coan S, Schnapp WB, Psychiatry 46:212–20.
and others. 2001. The lifetime cost of bipolar disorder in the US: Desbois-Mouthon C, Cadoret A, Blivet-Van Eggelpoel MJ, Bertrand F,
an estimate for new cases in 1998. Pharmacoeconomics Cherqui G, Perret C, and others. 2001. Insulin and IGF-1 stimulate
19:483–95. the beta-catenin pathway through two signalling cascades involv-
Berridge MJ, Downes CP, Hanley MR. 1982. Lithium amplifies ago- ing GSK-3beta inhibition and Ras activation. Oncogene 20:252–9.
nist-dependent phosphatidylinositol responses in brain and salivary Drevets WC. 2000a. Functional anatomical abnormalities in limbic
glands. Biochem J 206:587–95. and prefrontal cortical structures in major depression. Prog Brain
Berridge MJ, Downes CP, Hanley MR. 1989. Neural and developmen- Res 126:413–31.
tal actions of lithium: a unifying hypothesis. Cell 59:411–9. Drevets WC. 2000b. Neuroimaging studies of mood disorders. Biol
Bijur GN, De Sarno P, Jope RS. 2000. Glycogen synthase kinase-3beta Psychiatry 48:813–29.
facilitates staurosporine- and heat shock-induced apoptosis. Drevets WC, Gadde K, Krishnan R. 1999. Neuroimaging studies of
Protection by lithium. J Biol Chem 275:7583–90. depression. In: Charney DS, Nester EJ, Bunney BS, editors.
Bordonaro M, Mariadason JM, Aslam F, Heerdt BG, Augenlicht LH. Neurobiology of mental illness. New York: Oxford University
1999. Butyrate-induced apoptotic cascade in colonic carcinoma Press. p 394–418.
cells: modulation of the beta-catenin-Tcf pathway and concordance Drevets WC, Price JL, Simpson JR Jr, Todd RD, Reich T, Vannier M,
with effects of sulindac and trichostatin A but not curcumin. Cell Raichle ME. 1997. Subgenual prefrontal cortex abnormalities in
Growth Differ 10:713–20. mood disorders. Nature 386:824–7.
Bowden CL. 1997. Toward an integrated biological model of bipolar Eldar-Finkelman H, Seger R, Vandenheede JR, Krebs EG. 1995.
disorder. In: Young LT, Joffe RT, editors. Bipolar disorder: biolog- Inactivation of glycogen synthase kinase-3 by epidermal growth
ical models and their clinical application. New York: Marcel factor is mediated by mitogen-activated protein kinase/p90 riboso-
Dekker. p 235–54. mal protein S6 kinase signaling pathway in NIH/3T3 cells. J Biol
Bruno V, Sortino MA, Scapagnini U, Nicoletti F, Canonico PL. 1995. Chem 270:987–90.
Antidegenerative effects of Mg(2+)-valproate in cultured cerebel- Emily-Fenouil F, Ghiglione C, Lhomond G, Lepage T, Gache C. 1998.
lar neurons. Funct Neurol 10:121–30. GSK3beta/shaggy mediates patterning along the animal-vegetal
Cadigan KM, Nusse R. 1997. Wnt signaling: a common theme in ani- axis of the sea urchin embryo. Development 125:2489–98.
mal development. Genes Dev 11:3286–305. Garlow SJ, Musselman DL, Nemeroff CB. 1999. The neurochemistry
Chalecka-Franaszek E, Chuang DM. 1999. Lithium activates the ser- of mood disorders clinical studies. In: Charney DS, Nester EJ,
ine/threonine kinase Akt-1 and suppresses glutamate-induced inhi- Bunney BS, editors. Neurobiology of mental illness. New York:
bition of Akt-1 activity in neurons. Proc Natl Acad Sci U S A Oxford University Press. p 348–64.
96:8745–50. Goodwin FK, Jamison KR. 1990. Manic-depressive illness. New York:
Chen G, Huang LD, Jiang YM, Manji HK. 1999. The mood-stabilizing Oxford University Press.
agent valproate inhibits the activity of glycogen synthase kinase-3. Gould TD, Manji HK. 2002. Signaling networks in the pathophysiolo-
J Neurochem 72:1327–30. gy and treatment of mood disorders. J Pyschosom Res 53:687–95.
Chen G, Manji HK, Hawver DB, Wright CB, Potter WZ. 1994. Grimes AC, Jope RS. 2001. CREB DNA binding activity is inhibited
Chronic sodium valproate selectively decreases protein kinase C by glycogen synthase kinase-3beta and facilitated by lithium. J
alpha and epsilon in vitro. J Neurochem 63:2361–4. Neurochem 78:1–15.
Chen G, Yuan P, Hawver DB, Potter WZ, Manji HK. 1997. Increase in Hall AC, Lucas FR, Salinas PC. 2000. Axonal remodeling and synap-
AP-1 transcription factor DNA binding activity by valproic acid. tic differentiation in the cerebellum is regulated by WNT-7a sig-
Neuropsychopharmacology 16:238–45. naling. Cell 100:525–35.

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 509

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell SM, Maeda Y. 1970. Influence of ionic conditions on cell differentiation
and others. 1995. The molecular basis of Turcot’s syndrome. N and morphogenesis of the cellular slime molds. Dev Growth Differ
Engl J Med 332:839–47. 12:217–27.
Harwood AJ. 2001. Regulation of GSK-3: a cellular multiprocessor. Maes M, Meltzer HY. 1995. The serotonergic hypothesis of depres-
Cell 105:821–4. sion. In: Bloom FE, Kupfer DJ, editors. Psychopharmacology: the
Harwood AJ, Plyte SE, Woodgett J, Strutt H, Kay RR. 1995. Glycogen fourth generation of progress. New York: Raven. p 921–32.
synthase kinase 3 regulates cell fate in Dictyostelium. Cell 80:139–48. Manji HK, Bebchuk JM, Moore GJ, Glitz D, Hasanat KA, Chen G.
Healy D, Waterhouse JM. 1995. The circadian system and the thera- 1999a. Modulation of CNS signal transduction pathways and gene
peutics of the affective disorders. Pharmacol Ther 65:241–63. expression by mood-stabilizing agents: therapeutic implications. J
Hedgepeth CM, Conrad LJ, Zhang J, Huang HC, Lee VM, Klein PS. Clin Psychiatry 60:27–39; discussion 40–1, 113–6.
1997. Activation of the Wnt signaling pathway: a molecular mech- Manji HK, Chen G, Hsiao JK, Risby ED, Masana MI, Potter WZ. 1996.
anism for lithium action. Dev Biol 185:82–91. Regulation of signal transduction pathways by mood-stabilizing
Hedgepeth CM, Deardorff MA, Rankin K, Klein PS. 1999. Regulation agents: implications for the delayed onset of therapeutic efficacy.
of glycogen synthase kinase 3beta and downstream Wnt signaling J Clin Psychiatry 57:34–46; discussion 47–8.
by axin. Mol Cell Biol 19:7147–57. Manji HK, Duman RS. 2001. Impairments of neuroplasticity and cel-
Hetman M, Cavanaugh JE, Kimelman D, Xia Z. 2000. Role of glyco- lular resilience in severe mood disorders: implications for the devel-
gen synthase kinase-3beta in neuronal apoptosis induced by troph- opment of novel therapeutics. Psychopharmacol Bull 35:5–49.
ic withdrawal. J Neurosci 20:2567–74. Manji HK, Lenox RH. 2000. Signaling: cellular insights into the
Huang H, Mahler-Araujo BM, Sankila A, Chimelli L, Yonekawa Y, pathophysiology of bipolar disorder. Biol Psychiatry 48:518–30.
Kleihues P, and others. 2000. APC mutations in sporadic medul- Manji HK, Moore GJ, Chen G. 1999b. Lithium at 50: have the neuro-
loblastomas. Am J Pathol 156:433–7. protective effects of this unique cation been overlooked? Biol
Ikonomov OC, Manji HK. 1999. Molecular mechanisms underlying Psychiatry 46:929–40.
mood stabilization in manic-depressive illness: the phenotype chal- Manji HK, Moore GJ, Chen G. 2000a. Clinical and preclinical evi-
lenge. Am J Psychiatry 156:1506–14. dence for the neurotrophic effects of mood stabilizers: implications
Jamison KR. 1986. Suicide and bipolar disorders. Ann N Y Acad Sci for the pathophysiology and treatment of manic-depressive illness.
487:301–15. Biol Psychiatry 48:740–54.
Jope RS. 1999. Anti-bipolar therapy: mechanism of action of lithium. Manji HK, Moore GJ, Rajkowska G, Chen G. 2000b. Neuroplasticity
Mol Psychiatry 4:117–28. and cellular resilience in mood disorders. Mol Psychiatry
Jope RS, Bijur GN. 2002. Mood stabilizers, glycogen synthase kinase- 5:578–93.
3beta and cell survival. Mol Psychiatry 7(Suppl 1):S35–45. Mark RJ, Ashford JW, Goodman Y, Mattson MP. 1995. Anticonvulsants
Jope RS, Williams MB. 1994. Lithium and brain signal transduction attenuate amyloid beta-peptide neurotoxicity, Ca2+ deregulation,
systems. Biochem Pharmacol 47:429–41. and cytoskeletal pathology. Neurobiol Aging 16:187–98.
Kao KR, Elinson RP. 1988. The entire mesodermal mantle behaves as Martinek S, Inonog S, Manoukian AS, Young MW. 2001. A role for the
Spemann’s organizer in dorsoanterior enhanced Xenopus laevis segment polarity gene shaggy/GSK-3 in the Drosophila circadian
embryos. Dev Biol 127:64–77.
clock. Cell 105:769–79.
Kasper S, Wehr TA. 1992. The role of sleep and wakefulness in the
McMahon AP, Moon RT. 1989. Ectopic expression of the proto-oncogene
genesis of depression and mania. Encephale 18(Spec No 1):45–50.
int-1 in Xenopus embryos leads to duplication of the embryonic
Kleijn M, Proud CG. 2000. The activation of eukaryotic initiation fac-
axis. Cell 58:1075–84.
tor (eIF)2B by growth factors in PC12 cells requires MEK/ERK
Miyaoka T, Seno H, Ishino H. 1999. Increased expression of Wnt-1 in
signalling. FEBS Lett 476:262–5.
schizophrenic brains. Schizophr Res 38:1–6.
Klein PS, Melton DA. 1996. A molecular mechanism for the effect of
lithium on development. Proc Natl Acad Sci U S A 93:8455–9. Miyoshi Y, Nagase H, Ando H, Horii A, Ichii S, Nakatsuru S, and oth-
Klemfuss H. 1992. Rhythms and the pharmacology of lithium. ers. 1992. Somatic mutations of the APC gene in colorectal tumors:
Pharmacol Ther 56:53–78. mutation cluster region in the APC gene. Hum Mol Genet 1:229–33.
Koch A, Waha A, Tonn JC, Sorensen N, Berthold F, Wolter M, and others. Moore GJ, Bebchuk JM, Hasanat K, Chen G, Seraji-Bozorgzad N,
2001. Somatic mutations of WNT/wingless signaling pathway com- Wilds IB, and others. 2000a. Lithium increases N-acetyl-aspartate
ponents in primitive neuroectodermal tumors. Int J Cancer 93:445–9. in the human brain: in vivo evidence in support of bcl-2’s neu-
Kozlovsky N, Belmaker RH, Agam G. 2000. Low GSK-3beta rotrophic effects? Biol Psychiatry 48:1–8.
immunoreactivity in postmortem frontal cortex of schizophrenic Moore GJ, Bebchuk JM, Wilds IB, Chen G, Manji HK. 2000b.
patients. Am J Psychiatry 157:831–3. Lithium-induced increase in human brain grey matter. Lancet
Kuhl M, Sheldahl LC, Park M, Miller JR, Moon RT. 2000. The 356:1241–2.
Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes Mora A, Gonzalez-Polo RA, Fuentes JM, Soler G, Centeno F. 1999.
shape. Trends Genet 16:279–83. Different mechanisms of protection against apoptosis by valproate
Lenox RH, Gould TD, Manji HK. 2002. Endophenotypes in bipolar and Li+. Eur J Biochem 266:886–91.
disorder. Am J Med Genet 114:391–406. Muller-Oerlinghausen B, Ahrens B, Grof E, Grof P, Lenz G, Schou M,
Lenox RH, McNamara RK, Watterson JM, Watson DG. 1996. and others. 1992. The effect of long-term lithium treatment on the
Myristoylated alanine-rich C kinase substrate (MARCKS): a mortality of patients with manic-depressive and schizoaffective ill-
molecular target for the therapeutic action of mood stabilizers in ness. Acta Psychiatr Scand 86:218–22.
the brain? J Clin Psychiatry 57:23–31; discussion 32–3. Murray CJ, Lopez AD. 1996. Evidence-based health policy—lessons
Lesort M, Greendorfer A, Stockmeier C, Johnson GV, Jope RS. 1999. from the Global Burden of Disease Study. Science 274:740–3.
Glycogen synthase kinase-3beta, beta-catenin, and tau in post- Nonaka S, Chuang DM. 1998. Neuroprotective effects of chronic lithi-
mortem bipolar brain. J Neural Transm 106:1217–22. um on focal cerebral ischemia in rats. Neuroreport 9:2081–4.
Li X, Bijur GN, Jope RS. 2002. Glycogen synthase kinase 3-beta, Nonaka S, Hough CJ, Chuang DM. 1998a. Chronic lithium treatment
mood stabilizers, and neuroprotection. Bipolar Disord 4:137–44. robustly protects neurons in the central nervous system against
Lowrey PL, Shimomura K, Antoch MP, Yamazaki S, Zemenides PD, excitotoxicity by inhibiting N-methyl-D-aspartate receptor-
Ralph MR, and others. 2000. Positional syntenic cloning and func- mediated calcium influx. Proc Natl Acad Sci U S A 95:2642–7.
tional characterization of the mammalian circadian mutation tau. Nonaka S, Katsube N, Chuang DM. 1998b. Lithium protects rat cere-
Science 288:483–92. bellar granule cells against apoptosis induced by anticonvulsants,
Lucas FR, Goold RG, Gordon-Weeks PR, Salinas PC. 1998. Inhibition phenytoin and carbamazepine. J Pharmacol Exp Ther 286:539–47.
of GSK-3beta leading to the loss of phosphorylated MAP-1B is an Novak A, Dedhar S. 1999. Signaling through beta-catenin and Lef/Tcf.
early event in axonal remodelling induced by WNT-7a or lithium. Cell Mol Life Sci 56:523–37.
J Cell Sci 111:1351–61. Pacchierotti C, Iapichino S, Bossini L, Pieraccini F, Castrogiovanni P.
Lucas FR, Salinas PC. 1997. WNT-7a induces axonal remodeling and 2001. Melatonin in psychiatric disorders: a review on the melatonin
increases synapsin I levels in cerebellar neurons. Dev Biol 192:31–44. involvement in psychiatry. Front Neuroendocrinol 22:18–32.

510 THE NEUROSCIENTIST Wnt and Bipolar Disorder

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014


Pap M, Cooper GM. 1998. Role of glycogen synthase kinase-3 in the Thomas GM, Frame S, Goedert M, Nathke I, Polakis P, Cohen P. 1999.
phosphatidylinositol 3- Kinase/Akt cell survival pathway. J Biol A GSK3-binding peptide from FRAT1 selectively inhibits the
Chem 273:19929–32. GSK3- catalysed phosphorylation of axin and beta-catenin. FEBS
Pascual T, Gonzalez JL. 1995. A protective effect of lithium on rat Lett 458:247–51.
behaviour altered by ibotenic acid lesions of the basal forebrain Tondo L, Jamison KR, Baldessarini RJ. 1997. Effect of lithium main-
cholinergic system. Brain Res 695:289–92. tenance on suicidal behavior in major mood disorders. Ann N Y
Patapoutian A, Reichardt LF. 2000. Roles of Wnt proteins in neural Acad Sci 836:339–51.
development and maintenance. Curr Opin Neurobiol 10:392–9. Tong N, Sanchez JF, Maggirwar SB, Ramirez SH, Guo H, Dewhurst S,
Peifer M, Polakis P. 2000. Wnt signaling in oncogenesis and embryo- and others. 2001. Activation of glycogen synthase kinase 3 beta (GSK-
genesis—a look outside the nucleus. Science 287:1606–9. 3beta) by platelet activating factor mediates migration and cell
Peters JM, McKay RM, McKay JP, Graff JM. 1999. Casein kinase I death in cerebellar granule neurons. Eur J Neurosci 13:1913–22.
transduces Wnt signals. Nature 401:345–50. Tsai G, Coyle JT. 1995. N-acetylaspartate in neuropsychiatric disor-
Petronis A. 2001. Human morbid genetics revisited: relevance of epi- ders. Prog Neurobiol 46:531–40.
genetics. Trends Genet 17:142–6. van Gijn ME, Snel F, Cleutjens JP, Smits JF, Blankesteijn WM. 2001.
Phiel CJ, Klein PS. 2001. Molecular targets of lithium action. Annu Overexpression of components of the Frizzled-Dishevelled cas-
Rev Pharmacol Toxicol 41:789–813. cade results in apoptotic cell death, mediated by beta-catenin. Exp
Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Cell Res 265:46–53.
2001. Histone deacetylase is a direct target of valproic acid, a potent Vestergaard P, Aagaard J. 1991. Five-year mortality in lithium-treated
anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 25:25. manic-depressive patients. J Affect Disord 21:33–8.
Plyte SE, Hughes K, Nikolakaki E, Pulverer BJ, Woodgett JR. 1992. Vielhaber E, Virshup DM. 2001. Casein kinase I: from obscurity to
Glycogen synthase kinase-3: functions in oncogenesis and devel- center stage. IUBMB Life 51:73–8.
opment. Biochim Biophys Acta 1114:147–62. Vonica A, Weng W, Gumbiner BM, Venuti JM. 2000. TCF is the
Polakis P. 2000. Wnt signaling and cancer. Genes Dev 14:1837–51. nuclear effector of the beta-catenin signal that patterns the sea
Potash JB, DePaulo JR Jr. 2000. Searching high and low: a review of urchin animal-vegetal axis. Dev Biol 217:230–43.
the genetics of bipolar disorder. Bipolar Disord 2:8–26. Wager-Smith K, Kay SA. 2000. Circadian rhythm genetics: from flies
Price JL, Blau J, Rothenfluh A, Abodeely M, Kloss B, Young MW. to mice to humans. Nat Genet 26:23–7.
1998. double-time is a novel Drosophila clock gene that regulates Wang J-F, Young LT, Li PP, Warsh JJ. 1997. Signal transduction abnor-
PERIOD protein accumulation. Cell 94:83–95. malities in bipolar disorder. In: Young LT, Joffe RT, editors. Bipolar
Rajkowska G. In press. Cell pathology in bipolar disorder. Bipolar Disord. disorder: biological models and their clinical application. New
Reifman A, Wyatt RJ. 1980. Lithium: a brake in the rising cost of men- York: Dekker. p 41–79.
tal illness. Arch Gen Psychiatry 37:385–8. Wehr TA, Sack D, Rosenthal N, Duncan W, Gillin JC. 1983. Circadian
Reppert SM, Weaver DR. 2001. Molecular analysis of mammalian cir- rhythm disturbances in manic-depressive illness. Fed Proc
cadian rhythms. Annu Rev Physiol 63:647–76. 42:2809–14.
Ryves WJ, Harwood AJ. 2001. Lithium inhibits glycogen synthase Wehr TA, Sack DA, Rosenthal NE. 1987. Sleep reduction as a final com-
kinase-3 by competition for magnesium. Biochem Biophys Res mon pathway in the genesis of mania. Am J Psychiatry 144:201–4.
Commun 280:720–5. Weinberger DR. 1997. The biological basis of schizophrenia: new
Sakanaka C, Leong P, Xu L, Harrison SD, Williams LT. 1999. Casein directions. J Clin Psychiatry 58:22–7.
kinase iepsilon in the wnt pathway: regulation of beta-catenin func- Wikramanayake AH, Huang L, Klein WH. 1998. beta-Catenin is
tion. Proc Natl Acad Sci U S A 96:12548–52. essential for patterning the maternally specified animal-vegetal
Salinas PC, Hall AC. 1999. Lithium and synaptic plasticity. Bipolar axis in the sea urchin embryo. Proc Natl Acad Sci U S A
Disord 1:87–90. 95:9343–8.
Schatzberg AF, Schildkraut JJ. 1995. Recent studies on norepinephrine Williams JA, Sehgal A. 2001. Molecular components of the circadian
systems in mood disorders. In: Bloom FE, Kupfer DJ, editors. system in Drosophila. Annu Rev Physiol 63:729–55.
Psychopharmacology: the fourth generation of progress. New Winsberg ME, Sachs N, Tate DL, Adalsteinsson E, Spielman D, Ketter
York: Raven. p 911–20. TA. 2000. Decreased dorsolateral prefrontal N-acetyl aspartate in
Schou M. 1991. Lithium and electroconvulsive therapy: adversaries, bipolar disorder. Biol Psychiatry 47:475–81.
competitors, allies? Acta Psychiatr Scand 84:435–8. Wirz-Justice A, Van den Hoofdakker RH. 1999. Sleep deprivation in
Sharpe C, Lawrence N, Martinez Arias A. 2001. Wnt signalling: a depression: what do we know, where do we go? Biol Psychiatry
theme with nuclear variations. Bioessays 23:311–8. 46:445–53.
Smith DG, Buffet M, Fenwick AE, Haigh D, Ife RJ, Saunders M, and Wodarz A, Nusse R. 1998. Mechanisms of Wnt signaling in develop-
others. 2001. 3-Anilino-4-arylmaleimides: potent and selective ment. Annu Rev Cell Dev Biol 14:59–88.
inhibitors of glycogen synthase kinase-3 (GSK-3). Bioorg Med Woodgett JR. 2001. Judging a protein by more than its name: gsk-3.
Chem Lett 11:635–9. Sci STKE RE12.
Souetre E, Salvati E, Wehr TA, Sack DA, Krebs B, Darcourt G. 1988. Wu JC, Bunney WE. 1990. The biological basis of an antidepressant
Twenty-four-hour profiles of body temperature and plasma TSH in response to sleep deprivation and relapse: review and hypothesis.
bipolar patients during depression and during remission and in nor- Am J Psychiatry 147:14–21.
mal control subjects. Am J Psychiatry 145:1133–7. Yang SD, Yu JS, Lee TT, Yang CC, Ni MH, Yang YY. 1995.
Staal FJ, Burgering BM, van de Wetering M, Clevers HC. 1999. Tcf-1- Dysfunction of protein kinase FA/GSK-3 alpha in lymphocytes of
mediated transcription in T lymphocytes: differential role for patients with schizophrenic disorder. J Cell Biochem 59:108–16.
glycogen synthase kinase-3 in fibroblasts and T cells. Int Immunol Yost C, Farr GH 3rd, Pierce SB, Ferkey DM, Chen MM, Kimelman D.
11:317–23. 1998. GBP, an inhibitor of GSK-3, is implicated in Xenopus devel-
Stambolic V, Ruel L, Woodgett JR. 1996. Lithium inhibits glycogen opment and oncogenesis. Cell 93:1031–41.
synthase kinase-3 activity and mimics wingless signalling in intact Yost C, Torres M, Miller JR, Huang E, Kimelman D, Moon RT. 1996.
cells. Curr Biol 6:1664–8. The axis-inducing activity, stability, and subcellular distribution of
Stambolic V, Woodgett JR. 1994. Mitogen inactivation of glycogen beta-catenin is regulated in Xenopus embryos by glycogen syn-
synthase kinase-3 beta in intact cells via serine 9 phosphorylation. thase kinase 3. Genes Dev 10:1443–54.
Biochem J 303:701–4. Yuan PX, Huong LD, Jiang YM, Gutkind JS, Manji HK, Chen G. 2001.
Sutherland C, Leighton IA, Cohen P. 1993. Inactivation of glycogen The mood-stabilizer valproic acid activates mitogen-activated pro-
synthase kinase-3 beta by phosphorylation: new kinase connec- tein kinases and promotes neurite growth. J Biol Chem 19:19.
tions in insulin and growth-factor signalling. Biochem J 296:15–9. Zeng L, Fagotto F, Zhang T, Hsu W, Vasicek TJ, Perry WL 3rd, and oth-
Takashima A, Noguchi K, Sato K, Hoshino T, Imahori K. 1993. Tau ers. 1997. The mouse Fused locus encodes Axin, an inhibitor of the
protein kinase I is essential for amyloid beta-protein-induced neu- Wnt signaling pathway that regulates embryonic axis formation.
rotoxicity. Proc Natl Acad Sci U S A 90:7789–93. Cell 90:181–92.

Volume 8, Number 5, 2002 THE NEUROSCIENTIST 511

Downloaded from nro.sagepub.com at Umea University Library on October 13, 2014

You might also like