You are on page 1of 11

Pharmacology & Therapeutics 114 (2007) 222 – 232

www.elsevier.com/locate/pharmthera

Circadian genes, rhythms and the biology of mood disorders


Colleen A. McClung ⁎
Department of Psychiatry and Center for Basic Neuroscience, University of Texas Southwestern Medical Center,
5323 Harry Hines Boulevard, Dallas, TX 75390-9070, United States

Abstract

For many years, researchers have suggested that abnormalities in circadian rhythms may underlie the development of mood disorders such as bipolar
disorder (BPD), major depression and seasonal affective disorder (SAD). Furthermore, some of the treatments that are currently employed to treat mood
disorders are thought to act by shifting or “resetting” the circadian clock, including total sleep deprivation (TSD) and bright light therapy. There is also
reason to suspect that many of the mood stabilizers and antidepressants used to treat these disorders may derive at least some of their therapeutic efficacy by
affecting the circadian clock. Recent genetic, molecular and behavioral studies implicate individual genes that make up the clock in mood regulation. As
well, important functions of these genes in brain regions and neurotransmitter systems associated with mood regulation are becoming apparent. In this
review, the evidence linking circadian rhythms and mood disorders, and what is known about the underlying biology of this association, is presented.
© 2007 Elsevier Inc. All rights reserved.

Keywords: Circadian; Mood; Bipolar; Depression; Seasonal affective disorder; Sleep deprivation; Light therapy

Abbreviations: BMAL1, brain and muscle ARNT-like protein 1; BPD, bipolar disorder; CK1, casein kinase 1; CLOCK, circadian locomotor output cycles kaput protein;
GSK3β, glycogen synthase kinase 3β; MDD, major depressive disorder; NPAS2, neuronal PAS domain protein 2; SAD, seasonal affective disorder; SCN,
suprachiasmatic nucleus; SNP, single nucleotide polymorphism; SSRI, selective serotonin reuptake inhibitor; TSD, total sleep deprivation; VTA, ventral tegmental area.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
2. The molecular clock. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3. A circadian basis of mood disorders? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3.1. A generally disrupted clock . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3.2. The inability to properly adapt. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3.3. Seasonal affective disorder . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224
3.4. Influence of the molecular clock on mood-related neurotransmitter systems . . . . . . . 224
4. Treating mood disorders by altering the circadian cycle . . . . . . . . . . . . . . . . . . . . . 225
4.1. Sleep deprivation therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225
4.2. Bright light therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225
4.3. Pharmacological treatments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
5. Evidence linking specific circadian genes with mood disorders. . . . . . . . . . . . . . . . . . 226
5.1. Human genetic studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
5.2. Gene expression studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
5.3. Behavioral studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
6. Summary and conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228

⁎ Tel.: 214 648 4129; fax: 214 648 1293.


E-mail address: Colleen.McClung@utsouthwestern.edu.

0163-7258/$ - see front matter © 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.pharmthera.2007.02.003
C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232 223

1. Introduction and negatively affect its transcription. Selectively in forebrain


regions, neuronal PAS domain protein 2 (NPAS2), a protein very
Daily rhythms are prominent in everything from sleep/wake similar to CLOCK, can bind BMAL1 and induce Per and Cry
cycles, body temperature, hormone levels, and even cognition, gene expression (Reick et al., 2001). NPAS2 may also function in
attention and mood (Bunney & Bunney, 2000; Reppert & Weaver, the place of CLOCK in the SCN if the CLOCK protein is
2001). Disruptions in biological rhythms are known to be strongly genetically disrupted (Debruyne et al., 2006). Though the central
associated with mood disorders. Indeed some of the major circadian pacemaker is located in the SCN, all of these genes are
hallmarks of diseases like major depressive disorder (MDD) and expressed throughout the brain and in other organs where they
bipolar disorder (BPD) are abnormal sleep/wake, appetite, and function as peripheral clocks that respond to nonphotic stimuli,
social rhythms (Boivin, 2000; Bunney & Bunney, 2000; Lenox and likely in other processes unrelated to circadian rhythms (Abe
et al., 2002; Grandin et al., 2006). Depression symptoms are also et al., 2001; Stokkan et al., 2001; Iijima et al., 2002; Granados-
diurnal with the most severe symptoms occurring typically in the Fuentes et al., 2006; McDearmon et al., 2006; Mieda et al.,
morning (Rusting & Larsen, 1998), and depression is more 2006).
prevalent in areas of the world that receive little sunlight for
extended periods of time (Booker et al., 1991). In addition, one of 3. A circadian basis of mood disorders?
the most common mood disorders, affecting some 2–5% of the
population in temperate climates, is seasonal affective disorder 3.1. A generally disrupted clock
(SAD), a syndrome where depressive symptoms occur only in
the winter months when there are shorter days and a later dawn Mood disorders such as MDD and BPD may be more
(Lam & Levitan, 2000; Magnusson & Boivin, 2003). Thus, it has prevalent in individuals that are born with an abnormally shifted
long been hypothesized that abnormalities in the molecular clock or arrhythmic clock. Indeed, blunted or abnormal circadian
underlie the development of these disorders. In addition, nearly all rhythms in a variety of bodily functions including body
of the successful treatments for mood disorders seem to affect temperature, plasma cortisol, norepinephrine, thyroid stimulat-
circadian rhythms, and it appears that the shifts, resetting and ing hormone, blood pressure, pulse, and melatonin have been
stabilization of these rhythms produced by these treatments are found in depressed and bipolar patients (Atkinson et al., 1975;
important for therapeutic efficacy. Though these associations Kripke et al., 1978; Souetre et al., 1989). Interestingly, these
have been known for many years, we are only now starting to rhythms seem to return to normal with antidepressant or mood
understand the biology that underlies this connection. With the stabilizer treatment and patient recovery. Furthermore, genetic
cloning and characterization of the individual genes that make up sleep disorders such as familial advanced phase sleep syndrome
the molecular clock, researchers now have the opportunity to (FASPS) in which individuals have shifted circadian rhythms
explore the molecular mechanisms that underlie this association, where they fall asleep and wake up much earlier than desired or
and determine the importance of circadian rhythms in mood delayed sleep phase syndrome (DSPS) which has the opposite
disorders. phenotype are both highly comorbid with depression and
anxiety (Shirayama et al., 2003; Xu et al., 2005; Hamet &
2. The molecular clock Tremblay, 2006). Even individuals that are genetically predis-
posed towards “eveningness” (a preference for the evening)
The primary molecular clock is located in the suprachiasmatic versus “morningness” (a preference for the morning) are more
nucleus (SCN) in the hypothalamus, and consists of a tran- likely to develop depression (Drennan et al., 1991; Chelminski
scriptional feedback loop which cycles over the course of ∼ 24 hr et al., 1999). Genetic variations in the circadian genes have been
in the absence of environmental input (Reppert & Weaver, 2001; found to associate with these sleep disorders and diurnal
Ko & Takahashi, 2006). The major transcriptional activator preference measures including an association between certain
consists of a dimer between the circadian locomotor output variants of Per2, and CK1δ with FASPS; Per3, Clock, and
cycles kaput protein (CLOCK) and brain and muscle ARNT-like CK1ε with DSPS; and Per1, Per2, Per3 and Clock with diurnal
protein 1 (BMAL1, also known as ARNTL or MOP3). This preference (Katzenberg et al., 1998; Iwase et al., 2002; Archer
complex binds to E-box sequences in the promoters of many et al., 2003; Johansson et al., 2003; Takano et al., 2004; Carpen
genes including the Period (Per) and Cryptochrome (Cry) genes. et al., 2005; Mishima et al., 2005; Xu et al., 2005; Carpen et al.,
The PER and CRY proteins are translated in the cytoplasm, and 2006; Vanselow et al., 2006). This suggests a connection
are phosphorylated by casein kinase 1 (CK1) ε and δ and between proper mood regulation and a normal functioning
glycogen synthase kinase 3β (GSK3β), leading to changes in circadian clock. Indeed, nearly all individuals that suffer from
their stability, association and nuclear entry (Harms et al., 2003; mood disorders benefit greatly from a strict sleep/wake cycle
Iitaka et al., 2005; Knippschild et al., 2005; Kurabayashi et al., where they rise in the morning and go to sleep at night at the
2006). Upon entering the nucleus, they can repress the actions of same time every day (Leibenluft & Suppes, 1999).
CLOCK/BMAL1, thus creating a negative feedback loop. In
addition, there is an adjoining loop in which CLOCK/BMAL1 3.2. The inability to properly adapt
activates the transcription of Rev-erbα and Rorα (Sato et al.,
2004; Guillaumond et al., 2005). Once translated, these proteins Though the central molecular clock and peripheral clocks are
can bind to the promoter of the Bmal1 gene and both positively preset and have an endogenous rhythm, the timing of these
224 C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232

rhythms can be altered due to environmental influences. For conducted. For example, studies done in the arctic which have
example, locomotor activity rhythms and molecular rhythms in extreme seasonal differences in day/night light intensity and
mice change with alterations in the light/dark cycle (Redlin, duration find strong seasonal rhythms in melatonin (Yoneyama
2001). Animals also shift their activity rhythms and sleep/wake et al., 1999). Studies done in areas of the world where there are
cycle in response to restricted food availability at a certain time less extreme seasonal differences may not find pronounced
of day (Stephan, 2002). This entrainment to food, or other seasonal rhythms, particularly in individuals who are exposed to
nonphotic stimuli, is controlled by SCN-independent pace- self-selected cycles of artificial light. Additional abnormalities
makers located in various regions of the brain and other organs in melatonin secretion in SAD patients, such as phase delays
(Stokkan et al., 2001; Stephan, 2002; Mieda et al., 2006). The over 24 hr, have also been reported; however, other studies have
ability of these clocks to adapt to the environment is highly reported no differences in circadian melatonin levels or rhythms
beneficial for the survival of many species, including our own. in SAD patients (Checkley et al., 1993; Srinivasan et al., 2006).
The effect of having a clock that is out of sync with the Therefore the link between abnormal or exaggerated melatonin
environment is evident to anyone who has experienced jet lag rhythms and SAD is still up for debate.
after traveling (Herxheimer, 2005). Indeed, the ability of these Similar to the melatonin hypothesis, another one of the
clocks to adapt may be important in the regulation of mood in central hypotheses put forth to explain SAD is called the
response to changes in seasons, stress levels, sleep schedules, circadian phase shift hypothesis. The idea behind this
and time zones. For some individuals, these types of changes hypothesis is that the later dawn in winter leads to a delay in
bring on severe depressive or manic episodes. This is seen in circadian rhythms and a disconnection between the molecular
shift workers where often a certain population of them will rhythms of the SCN and the sleep/wake cycle in SAD patients.
develop mood disorders over time (Scott, 2000). This This hypothesis is largely based on research demonstrating that
development of mood disturbances in response to circadian early morning bright light therapy is the most effective in
disruptions is called the social zeitgeber theory. This theory treating SAD, whereas evening light therapy is often not
proposes that depressive or manic episodes are brought on by effective (Lewy et al., 1998b; Terman & Terman, 2005).
life stresses because they disrupt normal social routines and the Therefore, the early morning light is leading to a phase advance
sleep/wake cycle, in turn leading to changes in biological in the circadian system putting it back on track with the sleep/
rhythms and mood (Grandin et al., 2006). Thus, it is possible wake cycle. In addition, there have been reports that melatonin
that individuals that suffer from these types of mood disorders therapy in the evening can also be effective in producing this
have a molecular clock that is not able to properly adapt to same phase advance and may help with the treatment of SAD
certain types of environmental or other changes. (Lewy et al., 1998a, 2006). Though this theory has a lot of
support, it still remains controversial. If this hypothesis is true,
3.3. Seasonal affective disorder then evening light exposure should make symptoms worse by
further delaying the circadian rhythm. However, in some
SAD is perhaps the most common disorder that arises from a studies, evening light exposure has an equally strong antide-
failure to properly adapt to changes in the environment. SAD is pressant effect as morning exposure and several studies find no
characterized by depressive symptoms that occur only in the therapeutic effects of melatonin treatment (Wirz-Justice et al.,
winter months (Magnusson & Boivin, 2003). It has been 1990, 1993; Terman et al., 2001). Other theories suggest that
hypothesized that the circadian hormone, melatonin, is centrally SAD patients actually fall into 2 categories, some with a phase
involved in the development of SAD (Pandi-Perumal et al., delay in rhythms and some with a phase advance in rhythms
2006). Melatonin is released primarily by the pineal gland and (Boivin, 2000; Lewy et al., 2006). The timing of light therapy
can bind to the G-protein coupled receptors, MT1 and MT2 and perhaps melatonin therapy would then need to be adjusted
(Pandi-Perumal et al., 2006). These receptors are expressed at for each group separately to produce the desired shift in rhythms
high levels in the SCN, and upon stimulation, modulate SCN and therapeutic effects.
transmission and circadian activity. Melatonin is suppressed by
light, participates in sleep, and varies seasonally in many 3.4. Influence of the molecular clock
mammals (Pandi-Perumal et al., 2006). In some studies, SAD on mood-related neurotransmitter systems
sufferers have been found to have either a pronounced seasonal
melatonin rhythm, or more daytime melatonin specifically in The biology that underlies the association between circadian
the winter, whereas healthy control subjects have no seasonal rhythms and mood disorders is still unknown, but may come
alterations in their melatonin rhythms (Danilenko et al., 1994; from the influence of the molecular clock on certain
Wehr et al., 2001). However, other studies have found neurotransmitters and their receptors. Indeed some of the
significant seasonal alterations in melatonin rhythms in healthy, major neurotransmitters that have been implicated in mood
nonaffected individuals, or even melatonin levels that are regulation, including serotonin, norepinephrine and dopamine,
greater in the summer than the winter (Touitou et al., 1984; have a circadian rhythm in their levels, release, and synthesis-
Kivela et al., 1988; Honma et al., 1992; Morera & Abreu, 2006). related enzymes (Weiner et al., 1992; Shieh et al., 1997; Aston-
Much of this variability in melatonin measurements likely Jones et al., 2001; Barassin et al., 2002; Khaldy et al., 2002;
comes from the variations in seasonal differences between Castaneda et al., 2004; Weber et al., 2004; Malek et al., 2005).
different areas of the world in which these studies were There are also circadian rhythms in the expression and activity
C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232 225

of several of the receptors that bind these neurotransmitters, HT1A autoreceptor and is used to augment the antidepressant
suggesting that these entire circuits are under circadian control effects of serotonin reuptake inhibitors, was shown to potentiate
(Kafka et al., 1983; Wesemann & Weiner, 1990; Witte & the effects of TSD (Smeraldi et al., 1999). In contrast,
Lemmer, 1991; Coon et al., 1997; Akhisaroglu et al., 2005). It pretreatment with the dopamine agonist, amineptine, prevents
seems likely that disruptions in the normal rhythms in these the antidepressant effects of TSD (Benedetti et al., 1996). TSD
circuits (either continuous or abrupt) could have major effects also seems to activate limbic dopaminergic pathways which can
on mood and motivational states. How these circuits are be measured in increased limbic blood flow, increased D2
controlled in a circadian fashion is still uncertain. Some of this dopamine receptor occupancy and increased eye-blink rates
modulation seems to occur through connections between the after TSD treatment (Ebert et al., 1994a,b, 1996). These results,
SCN and other brain regions. For example, an indirect pro- and the significant similarities between the feelings induced by
jection from the SCN to the locus coeruleus appears to regulate TSD and psychostimulant use, have implicated the limbic
the circadian rhythm in noradrenergic neuronal activity (Aston- dopaminergic system in the therapeutic actions of TSD (Ebert &
Jones et al., 2001). Furthermore, circadian gene expression Berger, 1998). How TSD leads to an activation of this pathway,
outside of the SCN, in these specific regions, may contribute to however, is currently unknown.
their rhythmic activity. Circadian activity rhythms in rodents A few studies using rodent models have begun to look at the
can be entrained to daytime methamphetamine injections, even mechanism that underlies TSD's effects on depression. One
in SCN lesioned animals (Iijima et al., 2002). This treatment night of TSD in rats decreases immobility in the forced swim
shifts the expression of the period genes in striatal regions test, similar to the actions of antidepressant drugs (Lopez-
typically associated with movement control in a manner that Rodriguez et al., 2004). Furthermore, like antidepressant
matches the shift in activity rhythms (Iijima et al., 2002). This treatment, TSD in rats stimulates hippocampal neurogenesis
same shift in period gene expression does not occur in the SCN (Grassi Zucconi et al., 2006). It has also been shown in hamsters
with methamphetamine treatment, thus there is a disconnect that TSD affects serotonin levels in the SCN and other brain
between the SCN, molecular rhythms in the striatum and regions, and results in a strong phase advance in activity
locomotor activity rhythms. This suggests that the period gene rhythms (Asikainen et al., 1995; Grossman et al., 2000). These
expression and rhythms in striatal regions is important in pro- studies are just beginning, and are important in determining the
ducing rhythms in locomotor activity. Therefore, the circadian mechanism underlying the therapeutic effects of TSD as an
genes both in the SCN and in these specific circuits may be antidepressant.
involved in regulating this rhythmic activity in neurotransmis-
sion. Future studies are needed to determine exactly how these 4.2. Bright light therapy
rhythms in dopamine, serotonin and other neurotransmitters are
involved in mood regulation. Bright light therapy has been used for more than 20 years to
treat SAD with comparable efficacy to antidepressant medica-
4. Treating mood disorders by altering the circadian cycle tions (Lam et al., 2006). Several studies also indicate that light
therapy can be equally effective in treating nonseasonal
4.1. Sleep deprivation therapy depression, as well as many other mood disorders (Terman &
Terman, 2005). It is thought that light therapy works by shifting
Total sleep deprivation (TSD) is a rapid and effective short- the circadian clock, and indeed light therapy given in the morning
term treatment for depression. It improves depressive symptoms will produce a phase advance in rhythms, whereas light in the
in some 40–60% of patients (Wirz-Justice & Van den evening produces a phase delay (Wirz-Justice et al., 2005). The
Hoofdakker, 1999; Giedke & Schwarzler, 2002). Partial sleep potential for light therapy as an effective treatment for a whole
deprivation (through the second half of the night) can also be spectrum of mood disorders is appealing because it is safe, has
effective, though usually not to the same degree as TSD (Wirz- few side effects, is relatively easy to use and is noninvasive. The
Justice et al., 2005). Sleep following treatment can lead to a standard light treatment involves exposure to a ∼ 10,000 lx light
relapse in symptoms, however, in some patients this is delayed box, typically in the morning after waking for 30–90 min
for several weeks. Concurrent treatment with antidepressant (Terman & Terman, 2005). Over time, this will produce a phase
medications or lithium may help prevent relapse (Wu & Bunney, shift in circadian body temperature rhythms of ∼ 1 hr (Burgess
1990; Benedetti et al., 2001). Furthermore, follow-up treatment et al., 2004). Similar to antidepressant treatment, it generally
with daily light therapy or a short phase advance in the sleep/ takes 2–4 weeks before the beneficial effects on mood are seen.
wake cycle can also prevent relapse (Berger et al., 1997; There are some indications that certain wavelengths of light
Riemann et al., 1999; Wirz-Justice et al., 2005). Melatonin are more effective than others in promoting a therapeutic
therapy, however, seems to be ineffective at preventing relapse response with the fewest side effects. This could be due to the
following TSD in SAD patients (Danilenko & Putilov, 2005). ability of certain wavelengths of light to more effectively control
The biological basis of TSD as a treatment for depression is circadian rhythms (Foster & Helfrich-Forster, 2001). Recent
poorly understood. Several circadian phase setting and sleep- studies have found that light in the blue spectrum (446–477)
phase hypotheses have been put forth in an attempt to explain its outperforms other wavelengths in melatonin suppression,
therapeutic action (Wirz-Justice & Van den Hoofdakker, 1999). circadian phase shifting, and antidepressant effects (Brainard
Interestingly, pindolol, a drug that blocks the serotonergic 5- et al., 2001; Thapan et al., 2001; Lockley et al., 2003; Glickman
226 C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232

et al., 2006). Whereas there have been several studies that have cortisol and other hormones in animal models and in humans,
investigated the mechanism by which light shifts the circadian which may underlie some of its therapeutic effects (Leproult
clock and the specific photoreceptors involved in this effect et al., 2005). Interestingly, agomelatine is much more effective
(Foster & Helfrich-Forster, 2001; Foster et al., 2003), few than melatonin in reversing depression-like behavioral responses
studies have examined the effects of light-induced circadian in animal models, suggesting that the therapeutic actions of
phase shifting as an antidepressant in animal models. agomelatine are not exclusively due to its actions at the melatonin
receptors (Delagrange & Boutin, 2006). However, the kinetics of
4.3. Pharmacological treatments agomelatine, and actions at the melatonin receptors, may differ
greatly from those of melatonin, so this action may still underlie
In bipolar patients, the mood stabilizers lithium and valproate at least part of its efficacy as an antidepressant (Hamon &
are commonly used for treatment. Interestingly, both of these Bourgoin, 2006).
drugs have been repeatedly shown to alter the circadian period, Though the latency to action is similar between agomelatine
leading to a long period in Drosophila, nonhuman primates, and the SSRIs, agomelatine seems to have no effect on central
rodents and humans (Johnsson et al., 1983; Welsh & Moore-Ede, serotonin transmission or the density and function of 5-HT(1A)
1990; Klemfuss, 1992; Hafen & Wollnik, 1994; Dokucu et al., receptors (Hanoun et al., 2004; Millan et al., 2005). However, its
2005). This effect on circadian rhythms could involve the actions at the 5-HT(2C) receptors enhances mesolimbic dopami-
inhibition of GSK3β which modifies multiple members of the nergic and noradrenergic transmission, an effect also seen with
molecular clock (Iwahana et al., 2004; Padiath et al., 2004; Gould SSRIs (Millan et al., 2003; Serretti et al., 2004). Furthermore,
& Manji, 2005; Iitaka et al., 2005; Yin et al., 2006). It is thought chronic, but not acute, treatment with agomelatine also induces
that this action of lithium on the circadian clock is important in its neurogenesis in the hippocampus similar to other antidepres-
therapeutic efficacy. Lithium is able to slow the abnormally fast sants (Banasr et al., 2006). Interestingly, specific antagonists at the
circadian rhythms found in many bipolar patients (Atkinson 5-HT(2C) receptor have potent anxiolytic-like activity in animal
et al., 1975; Kripke et al., 1978). Furthermore, patients that have models, but they seem to have no effect in models of depression
a shift in rhythms respond positively to lithium treatment in terms (Jenck et al., 1998). Therefore, the therapeutic actions of
of mood stabilization, whereas those few bipolar patients that agomelatine in the treatment of depression are still uncertain
begin with an abnormally slow clock do not respond to lithium and may involve both the 5-HT(2C) and melatonin receptors.
treatment. Furthermore, lithium treatment is able sustain and
enhance the phase-shifting and mood-altering effects of TSD 5. Evidence linking specific
(Benedetti et al., 2001). circadian genes with mood disorders
Similar to morning bright light therapy, the antidepressant,
fluoxetine, also affects circadian output by producing a phase 5.1. Human genetic studies
advance in the firing of SCN neurons in rat slice culture (Sprouse
et al., 2006). Indeed, serotonin neurons from the midbrain raphe Several human genetic studies have implicated specific
nuclei innervate the SCN, and local applications of 5-HT or 5-HT genes that make up the molecular clock in the manifestation of
1A and 7 receptor agonists to the SCN will also produce a phase mood disorders. For example, an amino acid substitution in
advance in circadian activity (Dudley et al., 1999; Ehlen et al., Npas2 (471 Leu/Ser) has been found to associate with the
2001). Thus antidepressants in the selective serotonin reuptake development of SAD (Johansson et al., 2003). Furthermore, in
inhibitor (SSRI) class may also exert some of their effects on bipolar patients, a single nucleotide polymorphism (SNP) in the
depression through modulation of the circadian clock. Interest- 3′ flanking region of the Clock gene (3111 T to C) associates
ingly, SSRI and mood stabilizers can have opposing therapeutic with a higher recurrence rate of bipolar episodes (Benedetti
actions in bipolar patients (Thase, 2005). This could be linked to et al., 2003). This SNP is also associated with greater insomnia
their opposing actions on rhythms since SSRI cause a phase and decreased need for sleep in bipolar patients, as well as
advance in rhythms while lithium can cause a phase delay greater insomnia in individuals undergoing antidepressant treat-
(Campbell et al., 1989; Sprouse et al., 2006). ment (Serretti et al., 2003, 2005). However, in patients with
Recently, agomelatine, a potent agonist of the melatonin MDD, there appears to be no general association between the
receptors and an antagonist at the serotonin 5-HT(2C) receptor, disease and this SNP (Desan et al., 2000). Since this SNP is in
has proven to be highly effective in animal models of depression, the 3′ flanking region, it could affect polyadenylation or RNA
and in several on-going clinical trials involving patients with stability; however, these possibilities have not been tested.
MDD (den Boer et al., 2006; Hamon & Bourgoin, 2006; In addition to the 3111 SNP in the Clock gene, 2 rare SNP in the
Zupancic & Guilleminault, 2006). Agomelatine also seems to same region (3117 G to T and 3125 A to G) have been identified by
produce fewer adverse side effects than some of the other anti- Pirovano et al. (2005), in 2 subjects with major depression, and not
depressant medications, and it alleviates many of the sleep in any of the healthy controls in their study. Interestingly, both of
problems associated with depression that are typically exacer- these individuals had the same pattern of sleep which consisted of
bated by SSRI treatment, making it a potentially valuable new alternating phases of good sleep and insomnia over the course of a
treatment for depression (Hamon & Bourgoin, 2006). As few days, which may be related to this change in the Clock gene.
expected by its pharmacologic profile, agomelatine has been In recent studies, other members of the molecular clock have
shown to resynchronize circadian rhythms in body temperature, been implicated in BPD. Haplotypes in Bmal1 and Per3 were
C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232 227

found to significantly associate with BPD in one study In addition to the fluoxetine study, a microarray study by
(Nievergelt et al., 2006). Furthermore, a SNP in Bmal1 and a Ogden et al. (2004) found that the mood stabilizer, valproate,
SNP in the Timeless gene have also been identified that decreased the expression of CK1δ and Cry2 in the amygdala, a
associate with BPD (Mansour et al., 2006). It should be noted region of the brain known to be important in anxiety and
that additional studies have found associations between emotional responses. These changes were prevented by
members of the molecular clock and other psychiatric disorders cotreatment with methamphetamine, which was given to induce
such as schizophrenia and alcoholism, suggesting that these manic-like symptoms, suggesting that they may be involved
genes are important in a range of psychiatric conditions in the treatment of mania (Ogden et al., 2004). Additional
(Spanagel et al., 2005; Mansour et al., 2006). However, in microarray studies have found expression changes in circadian
general, most of these studies only find modest associations, genes in striatal regions with psychostimulants such as cocaine,
and other studies that have examined SNP throughout the which suggest that expression of these genes in the striatum
sequence of some of the central members of the circadian clock may be important in controlling the hedonic state (Yuferov et al.,
have found no associations with these genes and any psychiatric 2003; McClung et al., 2005; Uz et al., 2005).
disorders (Shiino et al., 2003; Nievergelt et al., 2005, 2006;
Mansour et al., 2006). Therefore the functional importance of 5.3. Behavioral studies
these variations is still uncertain, and only certain members of
the circadian clock may be involved. A few studies using rodent models have examined the effects
One of the modulators of the circadian clock, GSK3β, is of an SCN lesion on measures of anxiety and depression to
perhaps the most well-characterized target of the mood stabilizer, determine if this central circadian pacemaker is involved in
lithium (Gould & Manji, 2005). Therefore it is somewhat surprising modulating these responses. A study by Tataroglu et al. (2004)
that most studies have not found general associations between found that bilateral SCN lesions in rats had an antidepressant-
variants in this gene and BPD, or the response to lithium. like effect in the forced swim test in that the animals showed
Two studies find that an identified SNP in the promoter of this less immobility time and more swimming. Immobility in this
gene (−50 T to C) does not appear to be related to the degree of task is regarded as “helpless” or “depressed” behavior, and it
prophylactic lithium response in bipolar patients (Michelon et al., can be reduced with antidepressant treatment. These results
2006; Szczepankiewicz et al., 2006a). Furthermore, one gene- would suggest that disruption of the SCN has a protective effect
tic study failed to find any significant association between 2 on depression-like behavior. However, a study by Tuma et al.
SNP (− 1727 A to T, and − 50 T to C) in Gsk3β with BPD or (2005) found that SCN lesions had no effect on the depression
schizophrenia in a Korean population (Lee et al., 2006). Similarly, and anxiety-like behaviors that are displayed after repeated
other studies failed to show a general association between these bouts of social defeat. The SCN does appear to play some role in
SNP and BPD, however, one of these SNP (− 50 T to C) is linked to this task, however, since the antidepressant, agomelatine, which
a later age of onset of BPD and a greater response to TSD and long- is normally effective in reducing the depression and anxiety-like
term lithium treatment (Benedetti et al., 2004, 2005). Additionally, behavior following social defeat, was not effective in the SCN
one study showed that this polymorphism may associate with the lesioned animals (Tuma et al., 2005). Thus, the SCN might be
development of bipolar II disorder specifically in female patients needed to produce the therapeutic effects of agomelatine. It is
(Szczepankiewicz et al., 2006b). Thus the effect of these not clear in this study whether the measurements performed are
polymorphisms, and perhaps others in Gsk3β, may be very more indicative of anxiety or depression, and more specific
specific. paradigms will have to be employed to differentiate the two. It
may seem in opposition to find that lesions of the SCN produce
5.2. Gene expression studies an antidepressant effect while SCN integrity is needed for
antidepressant action. However, agomelatine is known to inhibit
In addition to the human genetic studies, there have been a few SCN neuronal firing, which would suggest that reducing SCN
gene expression studies implicating circadian genes in either the function has an antidepressant effect (Ying et al., 1996). The
manifestation or treatment of mood disorders. One study found that finding that the SCN lesion on its own does not have any effect
the antidepressant, fluoxetine, altered the expression of Clock, on behavioral measures after social defeat suggests that other
Bmal1 and Npas2 in the mouse hippocampus (Uz et al., 2005). The brain regions are clearly involved in these responses.
hippocampus is thought to be particularly relevant to mood Behavioral studies aimed at investigating the role of
disorders since stress and antidepressant treatments have opposing individual circadian genes in mood regulation are just begin-
effects on neurogenesis in this region, and structural changes in the ning. Interestingly, transgenic mice overexpressing the circadi-
hippocampus have been observed in depressed patients (Campbell an modulator, GSK3β, are hyperactive and have reduced
& Macqueen, 2004). The same changes in circadian gene immobility in the forced swim test (indicative of lower
expression did not occur in striatal regions, indicating that they depression-like behavior) and an increased startle response
may be hippocampal specific. In addition, these genes were all (Prickaerts et al., 2006). These behaviors are reminiscent of
induced by chronic and not acute fluoxetine, suggesting that these bipolar patients in the manic state. This is perhaps not surprising
changes may be therapeutically relevant since fluoxetine needs to since lithium treatment is known to inhibit the actions of
be administered for days to weeks to see significant antidepressant GSK3β (Jope & Roh, 2006). Besides its role in circadian
effects in humans (Uz et al., 2005). rhythms, GSK3β is a widely expressed kinase that has many
228 C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232

functions in the brain (Jope & Roh, 2006). Furthermore, it is identification of individual members of the molecular clock,
unclear as to what brain region is involved in the manic-like studies examining the biology behind this association and the
behaviors since the transgene is expressed in several regions, or clock's influence on mood are now being conducted. These
if this overexpression affects circadian rhythms or the regulation studies should provide valuable information in terms of our
of circadian genes (Spittaels et al., 2002). One reason to suspect overall understanding of the development of mood disorders
that the manic-like phenotype seen in these mice may involve and the most appropriate ways to treat them. Treatments like
the circadian clock is that our laboratory has found that mice TSD and light therapy are now commonly used by many
harboring a mutation in the Clock gene also display a behavioral physicians. Though these treatments are effective for many
profile that is strikingly similar to human mania (Roybal et al., individuals, they still have limitations. Combinations of short-
in press). These mice have a point mutation in the Clock gene term treatments such as TSD combined with longer-term
caused by ENU mutagenesis that results in a dominant-negative pharmacological treatment may provide the quickest and most
protein (King et al., 1997). Their behavioral profile includes sustained relief of mood-related symptoms. An understanding
hyperactivity in response to novelty and over the light/dark of how these treatments alleviate symptoms and how shifts in
cycle, reduced depression-like behavior in the forced swim test circadian rhythms result in changes in mood will allow us to
and learned helplessness tests, reduced anxiety or increased risk design specific, less invasive, and more effective treatments for
taking behavior in several measures, and an increase in the these dehabilitating disorders.
reward value of cocaine, sucrose and intracranial self-
stimulation (McClung et al., 2005; Roybal et al., in press). Acknowledgments
Other laboratories have found that these mice sleep less and
have increased exploratory activity, adding to their overall I would like to thank Eric Nestler, John Enwright, Shibani
manic-like phenotype (Naylor et al., 2000; Easton et al., 2003). Mukherjee, Joseph Peevey and Addie Dickson for their helpful
Importantly, when we treat these mice with the mood stabilizer, comments on this manuscript. Results included from our
lithium, the majority of their behavioral responses return to laboratory were funded by NIDA, NIMH and NARSAD.
wild-type levels (Roybal et al., in press). Since alterations in the
midbrain dopaminergic system have been implicated in mania, References
we performed in vivo recordings from the dopaminergic
neurons of the Clock mutant mice and we found an increase Abe, H., Honma, S., Namihira, M., Masubuchi, S., Ikeda, M., Ebihara, S., et al.
in dopamine cell firing and bursting in the ventral tegmental (2001). Clock gene expressions in the suprachiasmatic nucleus and other
areas of the brain during rhythm splitting in CS mice. Brain Res Mol Brain
area (VTA) (McClung et al., 2005; Nestler, 2005; Nestler &
Res 87, 92−99.
Carlezon, 2006). CLOCK is expressed in the VTA, and it Akhisaroglu, M., Kurtuncu, M., Manev, H., Uz, T., et al. (2005). Diurnal
appears to regulate a number of genes that are important in rhythms in quinpirole-induced locomotor behaviors and striatal D2/D3
dopaminergic transmission (McClung et al., 2005). When we receptor levels in mice. Pharmacol Biochem Behav 80, 371−377.
expressed a functional CLOCK protein specifically in the VTA Archer, S. N., Robilliard, D. L., Skene, D. J., Smits, M., Williams, A., Arendt, J.,
et al. (2003). A length polymorphism in the circadian clock gene Per3 is
of the mutant mice using viral-mediated gene transfer, we are
linked to delayed sleep phase syndrome and extreme diurnal preference.
able to return several of their behavioral phenotypes to wild- Sleep 26, 413−415.
type levels, including locomotor activity and levels of anxiety Asikainen, M., Deboer, T., Porkka-Heiskanen, T., Stenberg, D., & Tobler, I.
(Roybal et al., in press). These results suggest that CLOCK is (1995). Sleep deprivation increases brain serotonin turnover in the
important in the development of mania, and that at least some of Djungarian hamster. Neurosci Lett 198, 21−24.
Aston-Jones, G., Chen, S., Zhu, Y., & Oshinsky, M. L. (2001). A neural circuit
the behavioral and mood related phenotypes seen in the mutant
for circadian regulation of arousal. Nat Neurosci 4, 732−738.
mice are a result of the loss of CLOCK in the dopamine-rich Atkinson, M., Kripke, D. F., & Wolf, S. R. (1975). Autorhythmometry in manic-
VTA. We have yet to determine how CLOCK function in other depressives. Chronobiologia 2, 325−335.
brain regions (including the SCN) is important in these Banasr, M., Soumier, A., Hery, M., Mocaer, E., & Daszuta, A. (2006).
behavioral measures. Since CLOCK is so widely expressed, it Agomelatine, a new antidepressant, induces regional changes in hippocam-
pal neurogenesis. Biol Psychiatry 59, 1087−1096.
is likely that we will find important functions for CLOCK in
Barassin, S., Raison, S., Saboureau, M., Bienvenu, C., Maitre, M., Malan, A., et al.
several regions. The finding that antidepressant treatment (2002). Circadian tryptophan hydroxylase levels and serotonin release in the
increases CLOCK expression in the hippocampus is particularly suprachiasmatic nucleus of the rat. Eur J Neurosci 15, 833−840.
interesting since the hippocampus has been widely implicated in Benedetti, F., Barbini, B., Campori, E., Colombo, C., & Smeraldi, E. (1996).
depression (Campbell & Macqueen, 2004). Additional behav- Dopamine agonist amineptine prevents the antidepressant effect of sleep
deprivation. Psychiatry Res 65, 179−184.
ioral studies with mice lacking other circadian genes are
Benedetti, F., Barbini, B., Campori, E., Fulgosi, M. C., Pontiggia, A., & Colombo,
ongoing. They will undoubtedly prove useful in determining C. (2001). Sleep phase advance and lithium to sustain the antidepressant effect
how these genes and the associated changes in rhythms regulate of total sleep deprivation in bipolar depression: new findings supporting the
mood. internal coincidence model? J Psychiatr Res 35, 323−329.
Benedetti, F., Serretti, A., Colombo, C., Barbini, B., Lorenzi, C., Campori, E., et al.
(2003). Influence of CLOCK gene polymorphism on circadian mood
6. Summary and conclusions
fluctuation and illness recurrence in bipolar depression. Am J Med Genet B
Neuropsychiatr Genet 123, 23−26.
The connection between mood disorders and circadian Benedetti, F., Serretti, A., Colombo, C., Lorenzi, C., Tubazio, V., & Smeraldi, E.
rhythms is becoming increasingly clear. With the cloning and (2004). A glycogen synthase kinase 3-beta promoter gene single nucleotide
C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232 229

polymorphism is associated with age at onset and response to total sleep Dudley, T. E., Dinardo, L. A., & Glass, J. D. (1999). In vivo assessment of the
deprivation in bipolar depression. Neurosci Lett 368, 123−126. midbrain raphe nuclear regulation of serotonin release in the hamster
Benedetti, F., Serretti, A., Pontiggia, A., Bernasconi, A., Lorenzi, C., Colombo, C., suprachiasmatic nucleus. J Neurophysiol 81, 1469−1477.
et al. (2005). Long-term response to lithium salts in bipolar illness is influenced Easton, A., Arbuzova, J., & Turek, F. W. (2003). The circadian Clock mutation
by the glycogen synthase kinase 3-beta-50 T/C SNP. Neurosci Lett 376, 51−55. increases exploratory activity and escape-seeking behavior. Genes Brain
Berger, M., Vollmann, J., Hohagen, F., Konig, A., Lohner, H., Voderholzer, U., Behav 2, 11−19.
et al. (1997). Sleep deprivation combined with consecutive sleep phase Ebert, D., & Berger, M. (1998). Neurobiological similarities in antidepressant
advance as a fast-acting therapy in depression: an open pilot trial in sleep deprivation and psychostimulant use: a psychostimulant theory of
medicated and unmedicated patients. Am J Psychiatry 154, 870−872. antidepressant sleep deprivation. Psychopharmacology (Berl) 140, 1−10.
Boivin, D. B. (2000). Influence of sleep–wake and circadian rhythm Ebert, D., Feistel, H., Barocka, A., & Kaschka, W. (1994). Increased limbic
disturbances in psychiatric disorders. J Psychiatry Neurosci 25, 446−458. blood flow and total sleep deprivation in major depression with melancholia.
Booker, J. M., Hellekson, C. J., Putilov, A. A., & Danilenko, K. V. (1991). Psychiatry Res 55, 101−109.
Seasonal depression and sleep disturbances in Alaska and Siberia: a pilot Ebert, D., Feistel, H., Kaschka, W., Barocka, A., & Pirner, A. (1994). Single
study. Arctic Med Res, 281−284 (Suppl). photon emission computerized tomography assessment of cerebral dopa-
Brainard, G. C., Hanifin, J. P., Greeson, J. M., Byrne, B., Glickman, G., Gerner, mine D2 receptor blockade in depression before and after sleep deprivation-
E., et al. (2001). Action spectrum for melatonin regulation in humans: preliminary results. Biol Psychiatry 35, 880−885.
evidence for a novel circadian photoreceptor. J Neurosci 21, 6405−6412. Ebert, D., Albert, R., Hammon, G., Strasser, B., May, A., & Merz, A. (1996).
Bunney, W. E., & Bunney, B. G. (2000). Molecular clock genes in man and Eye-blink rates and depression. Is the antidepressant effect of sleep
lower animals: possible implications for circadian abnormalities in deprivation mediated by the dopamine system? Neuropsychopharmacol-
depression. Neuropsychopharmacology 22, 335−345. ogy 15, 332−339.
Burgess, H. J., Fogg, L. F., Young, M. A., & Eastman, C. I. (2004). Bright light Ehlen, J. C., Grossman, G. H., & Glass, J. D. (2001). In vivo resetting of the
therapy for winter depression-is phase advancing beneficial? Chronobiol Int hamster circadian clock by 5-HT7 receptors in the suprachiasmatic nucleus.
21, 759−775. J Neurosci 21, 5351−5357.
Campbell, S., & Macqueen, G. (2004). The role of the hippocampus in the Foster, R. G., & Helfrich-Forster, C. (2001). The regulation of circadian clocks
pathophysiology of major depression. J Psychiatry Neurosci 29, 417−426. by light in fruitflies and mice. Philos Trans R Soc Lond B Biol Sci 356,
Campbell, S. S., Gillin, J. C., Kripke, D. F., Janowsky, D. S., & Risch, S. C. 1779−1789.
(1989). Lithium delays circadian phase of temperature and REM sleep in a Foster, R. G., Hankins, M., Lucas, R. J., Jenkins, A., Munoz, M., Thompson, S.,
bipolar depressive: a case report. Psychiatry Res 27, 23−29. et al. (2003). Non-rod, non-cone photoreception in rodents and teleost fish.
Carpen, J. D., Archer, S. N., Skene, D. J., Smits, M., & von Schantz, M. (2005). Novartis Found Symp 253, 3−23 (discussion 23-30, 52-25, 102-109).
A single-nucleotide polymorphism in the 5′-untranslated region of the Giedke, H., & Schwarzler, F. (2002). Therapeutic use of sleep deprivation in
hPER2 gene is associated with diurnal preference. J Sleep Res 14, 293−297. depression. Sleep Med Rev 6, 361−377.
Carpen, J. D., von Schantz, M., Smits, M., Skene, D. J., & Archer, S. N. (2006). Glickman, G., Byrne, B., Pineda, C., Hauck, W. W., & Brainard, G. C. (2006).
A silent polymorphism in the PER1 gene associates with extreme diurnal Light therapy for seasonal affective disorder with blue narrow-band light-
preference in humans. J Hum Genet 51(12), 1122−1125. emitting diodes (LEDs). Biol Psychiatry 59, 502−507.
Castaneda, T. R., de Prado, B. M., Prieto, D., & Mora, F. (2004). Circadian Gould, T. D., & Manji, H. K. (2005). Glycogen synthase kinase-3: a putative
rhythms of dopamine, glutamate and GABA in the striatum and nucleus molecular target for lithium mimetic drugs. Neuropsychopharmacology 30,
accumbens of the awake rat: modulation by light. J Pineal Res 36, 177−185. 1223−1237.
Checkley, S. A., Murphy, D. G., Abbas, M., Marks, M., Winton, F., Palazidou, Granados-Fuentes, D., Tseng, A., & Herzog, E. D. (2006). A circadian clock in
E., et al. (1993). Melatonin rhythms in seasonal affective disorder. Br J the olfactory bulb controls olfactory responsivity. J Neurosci 26,
Psychiatry 163, 332−337. 12219−12225.
Chelminski, I., Ferraro, F. R., Petros, T. V., & Plaud, J. J. (1999). An analysis of Grandin, L. D., Alloy, L. B., & Abramson, L. Y. (2006). The social zeitgeber
the “eveningness-morningness” dimension in “depressive” college students. theory, circadian rhythms, and mood disorders: review and evaluation. Clin
J Affect Disord 52, 19−29. Psychol Rev 26, 679−694.
Coon, S. L., McCune, S. K., Sugden, D., & Klein, D. C. (1997). Regulation of Grassi Zucconi, G., Cipriani, S., Balgkouranidou, I., & Scattoni, R. (2006). ‘One
pineal alpha1B-adrenergic receptor mRNA: day/night rhythm and beta- night’ sleep deprivation stimulates hippocampal neurogenesis. Brain Res
adrenergic receptor/cyclic AMP control. Mol Pharmacol 51, 551−557. Bull 69, 375−381.
Danilenko, K. V., & Putilov, A. A. (2005). Melatonin treatment of winter Grossman, G. H., Mistlberger, R. E., Antle, M. C., Ehlen, J. C., & Glass, J. D.
depression following total sleep deprivation: waking EEG and mood (2000). Sleep deprivation stimulates serotonin release in the suprachiasmatic
correlates. Neuropsychopharmacology 30, 1345−1352. nucleus. NeuroReport 11, 1929−1932.
Danilenko, K. V., Putilov, A. A., Russkikh, G. S., Duffy, L. K., & Ebbesson, S. O. Guillaumond, F., Dardente, H., Giguere, V., & Cermakian, N. (2005).
(1994). Diurnal and seasonal variations of melatonin and serotonin in women Differential control of Bmal1 circadian transcription by REV-ERB and
with seasonal affective disorder. Arctic Med Res 53, 137−145. ROR nuclear receptors. J Biol Rhythms 20, 391−403.
Debruyne, J. P., Noton, E., Lambert, C. M., Maywood, E. S., Weaver, D. R., & Hafen, T., & Wollnik, F. (1994). Effect of lithium carbonate on activity level and
Reppert, S. M. (2006). A clock shock: mouse CLOCK is not required for circadian period in different strains of rats. Pharmacol Biochem Behav 49,
circadian oscillator function. Neuron 50, 465−477. 975−983.
Delagrange, P., & Boutin, J. A. (2006). Therapeutic potential of melatonin Hamet, P., & Tremblay, J. (2006). Genetics of the sleep–wake cycle and its
ligands. Chronobiol Int 23, 413−418. disorders. Metabolism 55, S7−S12.
den Boer, J. A., Bosker, F. J., & Meesters, Y. (2006). Clinical efficacy of agomelatine Hamon, M., & Bourgoin, S. (2006). Pharmacological profile of antidepressants:
in depression: the evidence. Int Clin Psychopharmacol 21(Suppl 1), S21−S24. a likely basis for their efficacy and side effects? Eur Neuropsychopharmacol
Desan, P. H., Oren, D. A., Malison, R., Price, L. H., Rosenbaum, J., Smoller, J., 16(Suppl 5), S625−S632.
et al. (2000). Genetic polymorphism at the CLOCK gene locus and major Hanoun, N., Mocaer, E., Boyer, P. A., Hamon, M., & Lanfumey, L. (2004).
depression. Am J Med Genet 96, 418−421. Differential effects of the novel antidepressant agomelatine (S 20098) versus
Dokucu, M. E., Yu, L., & Taghert, P. H. (2005). Lithium- and valproate-induced fluoxetine on 5-HT1A receptors in the rat brain. Neuropharmacology 47,
alterations in circadian locomotor behavior in Drosophila. Neuropsycho- 515−526.
pharmacology 30, 2216−2224. Harms, E., Young, M. W., & Saez, L. (2003). CK1 and GSK3 in the Drosophila
Drennan, M. D., Klauber, M. R., Kripke, D. F., & Goyette, L. M. (1991). The and mammalian circadian clock. Novartis Found Symp 253, 267−277
effects of depression and age on the Horne-Ostberg morningness- (discussion 102-269, 277-284).
eveningness score. J Affect Disord 23, 93−98. Herxheimer, A. (2005). Jet lag. Clin Evid, 2178−2183.
230 C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232

Honma, K., Honma, S., Kohsaka, M., & Fukuda, N. (1992). Seasonal variation Leibenluft, E., & Suppes, T. (1999). Treating bipolar illness: focus on treatment
in the human circadian rhythm: dissociation between sleep and temperature algorithms and management of the sleep-wake cycle. Am J Psychiatry 156,
rhythm. Am J Physiol 262, R885−R891. 1976−1981.
Iijima, M., Nikaido, T., Akiyama, M., Moriya, T., & Shibata, S. (2002). Lenox, R. H., Gould, T. D., & Manji, H. K. (2002). Endophenotypes in bipolar
Methamphetamine-induced, suprachiasmatic nucleus-independent circadian disorder. Am J Med Genet 114, 391−406.
rhythms of activity and mPer gene expression in the striatum of the mouse. Leproult, R., Van Onderbergen, A., L'Hermite-Baleriaux, M., Van Cauter, E., &
Eur J Neurosci 16, 921−929. Copinschi, G. (2005). Phase-shifts of 24-h rhythms of hormonal release and
Iitaka, C., Miyazaki, K., Akaike, T., & Ishida, N. (2005). A role for glycogen body temperature following early evening administration of the melatonin
synthase kinase-3beta in the mammalian circadian clock. J Biol Chem 280, agonist agomelatine in healthy older men. Clin Endocrinol (Oxf) 63, 298−304.
29397−29402. Lewy, A. J., Bauer, V. K., Cutler, N. L., & Sack, R. L. (1998). Melatonin
Iwahana, E., Akiyama, M., Miyakawa, K., Uchida, A., Kasahara, J., Fukunaga, treatment of winter depression: a pilot study. Psychiatry Res 77, 57−61.
K., et al. (2004). Effect of lithium on the circadian rhythms of locomotor Lewy, A. J., Bauer, V. K., Cutler, N. L., Sack, R. L., Ahmed, S., Thomas, K. H.,
activity and glycogen synthase kinase-3 protein expression in the mouse et al. (1998). Morning vs. evening light treatment of patients with winter
suprachiasmatic nuclei. Eur J Neurosci 19, 2281−2287. depression. Arch Gen Psychiatry 55, 890−896.
Iwase, T., Kajimura, N., Uchiyama, M., Ebisawa, T., Yoshimura, K., Kamei, Y., Lewy, A. J., Lefler, B. J., Emens, J. S., & Bauer, V. K. (2006). The circadian
et al. (2002). Mutation screening of the human Clock gene in circadian basis of winter depression. Proc Natl Acad Sci U S A 103, 7414−7419.
rhythm sleep disorders. Psychiatry Res 109, 121−128. Lockley, S. W., Brainard, G. C., & Czeisler, C. A. (2003). High sensitivity of the
Jenck, F., Bos, M., Wichmann, J., Stadler, H., Martin, J. R., & Moreau, J. L. human circadian melatonin rhythm to resetting by short wavelength light.
(1998). The role of 5-HT2C receptors in affective disorders. Expert Opin J Clin Endocrinol Metab 88, 4502−4505.
Investig Drugs 7, 1587−1599. Lopez-Rodriguez, F., Kim, J., & Poland, R. E. (2004). Total sleep deprivation
Johansson, C., Willeit, M., Smedh, C., Ekholm, J., Paunio, T., Kieseppa, T., et al. decreases immobility in the forced-swim test. Neuropsychopharmacology
(2003). Circadian clock-related polymorphisms in seasonal affective 29, 1105−1111.
disorder and their relevance to diurnal preference. Neuropsychopharma- Magnusson, A., & Boivin, D. (2003). Seasonal affective disorder: an overview.
cology 28, 734−739. Chronobiol Int 20, 189−207.
Johnsson, A., Engelmann, W., Pflug, B., & Klemke, W. (1983). Period Malek, Z. S., Dardente, H., Pevet, P., & Raison, S. (2005). Tissue-specific
lengthening of human circadian rhythms by lithium carbonate, a expression of tryptophan hydroxylase mRNAs in the rat midbrain:
prophylactic for depressive disorders. Int J Chronobiol 8, 129−147. anatomical evidence and daily profiles. Eur J Neurosci 22, 895−901.
Jope, R. S., & Roh, M. S. (2006). Glycogen synthase kinase-3 (GSK3) in Mansour, H. A., Wood, J., Logue, T., Chowdari, K. V., Dayal, M., Kupfer, D. J.,
psychiatric diseases and therapeutic interventions. Curr Drug Targets 7, et al. (2006). Association study of eight circadian genes with bipolar I
1421−1434. disorder, schizoaffective disorder and schizophrenia. Genes Brain Behav 5,
Kafka, M. S., Wirz-Justice, A., Naber, D., Moore, R. Y., & Benedito, M. A. 150−157.
(1983). Circadian rhythms in rat brain neurotransmitter receptors. Fed Proc McClung, C. A., Sidiropoulou, K., Vitaterna, M., Takahashi, J. S., White, F. J.,
42, 2796−2801. Cooper, D. C., et al. (2005). Regulation of dopaminergic transmission and
Katzenberg, D., Young, T., Finn, L., Lin, L., King, D. P., Takahashi, J. S., et al. cocaine reward by the Clock gene. Proc Natl Acad Sci U S A 102, 9377−9381.
(1998). A CLOCK polymorphism associated with human diurnal prefer- McDearmon, E. L., Patel, K. N., Ko, C. H., Walisser, J. A., Schook, A. C.,
ence. Sleep 21, 569−576. Chong, J. L., et al. (2006). Dissecting the functions of the mammalian clock
Khaldy, H., Leon, J., Escames, G., Bikjdaouene, L., Garcia, J. J., & Acuna- protein BMAL1 by tissue-specific rescue in mice. Science 314, 1304−1308.
Castroviejo, D. (2002). Circadian rhythms of dopamine and dihydroxyphe- Michelon, L., Meira-Lima, I., Cordeiro, Q., Miguita, K., Breen, G., Collier, D.,
nyl acetic acid in the mouse striatum: effects of pinealectomy and of et al. (2006). Association study of the INPP1, 5HTT, BDNF, AP-2beta and
melatonin treatment. Neuroendocrinology 75, 201−208. GSK-3beta GENE variants and restrospectively scored response to lithium
King, D. P., Zhao, Y., Sangoram, A. M., Wilsbacher, L. D., Tanaka, M., Antoch, prophylaxis in bipolar disorder. Neurosci Lett 403, 288−293.
M. P., et al. (1997). Positional cloning of the mouse circadian clock gene. Mieda, M., Williams, S. C., Richardson, J. A., Tanaka, K., & Yanagisawa, M.
Cell 89, 641−653. (2006). The dorsomedial hypothalamic nucleus as a putative food-entrainable
Kivela, A., Kauppila, A., Ylostalo, P., Vakkuri, O., & Leppaluoto, J. (1988). circadian pacemaker. Proc Natl Acad Sci U S A 103, 12150−12155.
Seasonal, menstrual and circadian secretions of melatonin, gonadotropins Millan, M. J., Gobert, A., Lejeune, F., Dekeyne, A., Newman-Tancredi, A.,
and prolactin in women. Acta Physiol Scand 132, 321−327. Pasteau, V., et al. (2003). The novel melatonin agonist agomelatine (S20098)
Klemfuss, H. (1992). Rhythms and the pharmacology of lithium. Pharmacol is an antagonist at 5-hydroxytryptamine2C receptors, blockade of which
Ther 56, 53−78. enhances the activity of frontocortical dopaminergic and adrenergic
Knippschild, U., Gocht, A., Wolff, S., Huber, N., Lohler, J., & Stoter, M. (2005). pathways. J Pharmacol Exp Ther 306, 954−964.
The casein kinase 1 family: participation in multiple cellular processes in Millan, M. J., Brocco, M., Gobert, A., & Dekeyne, A. (2005). Anxiolytic
eukaryotes. Cell Signal 17, 675−689. properties of agomelatine, an antidepressant with melatoninergic and
Ko, C. H., & Takahashi, J. S. (2006). Molecular components of the mammalian serotonergic properties: role of 5-HT2C receptor blockade. Psychophar-
circadian clock. Hum Mol Genet 15(Suppl 2), R271−R277. macology (Berl) 177, 448−458.
Kripke, D. F., Mullaney, D. J., Atkinson, M., & Wolf, S. (1978). Circadian Mishima, K., Tozawa, T., Satoh, K., Saitoh, H., & Mishima, Y. (2005). The
rhythm disorders in manic-depressives. Biol Psychiatry 13, 335−351. 3111T/C polymorphism of hClock is associated with evening preference and
Kurabayashi, N., Hirota, T., Harada, Y., Sakai, M., & Fukada, Y. (2006). delayed sleep timing in a Japanese population sample. Am J Med Genet B
Phosphorylation of mCRY2 at Ser557 in the hypothalamic suprachiasmatic Neuropsychiatr Genet 133, 101−104.
nucleus of the mouse. Chronobiol Int 23, 129−134. Morera, A. L., & Abreu, P. (2006). Seasonality of psychopathology and
Lam, R. W., & Levitan, R. D. (2000). Pathophysiology of seasonal affective circannual melatonin rhythm. J Pineal Res 41, 279−283.
disorder: a review. J Psychiatry Neurosci 25, 469−480. Naylor, E., Bergmann, B. M., Krauski, K., Zee, P. C., Takahashi, J. S., Vitaterna,
Lam, R. W., Levitt, A. J., Levitan, R. D., Enns, M. W., Morehouse, R., M. H., et al. (2000). The circadian clock mutation alters sleep homeostasis in
Michalak, E. E., et al. (2006). The Can-SAD study: a randomized controlled the mouse. J Neurosci 20, 8138−8143.
trial of the effectiveness of light therapy and fluoxetine in patients with Nestler, E. J. (2005). Is there a common molecular pathway for addiction? Nat
winter seasonal affective disorder. Am J Psychiatry 163, 805−812. Neurosci 8, 1445−1449.
Lee, K. Y., Ahn, Y. M., Joo, E. J., Jeong, S. H., Chang, J. S., Kim, S. C, et al. Nestler, E. J., & Carlezon, W. A., Jr. (2006). The mesolimbic dopamine reward
(2006). No association of two common SNPs at position-1727 A/T,-50 C/T circuit in depression. Biol Psychiatry 59, 1151−1159.
of GSK-3 beta polymorphisms with schizophrenia and bipolar disorder of Nievergelt, C. M., Kripke, D. F., Remick, R. A., Sadovnick, A. D., McElroy, S. L.,
Korean population. Neurosci Lett 395, 175−178. Keck, P. E., Jr., et al. (2005). Examination of the clock gene cryptochrome 1 in
C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232 231

bipolar disorder: mutational analysis and absence of evidence for linkage or amplitude as the main chronobiological abnormality. Psychiatry Res 28,
association. Psychiatr Genet 15, 45−52. 263−278.
Nievergelt, C. M., Kripke, D. F., Barrett, T. B., Burg, E., Remick, R. A., Spanagel, R., Pendyala, G., Abarca, C., Zghoul, T., Sanchis-Segura, C., Magnone,
Sadovnick, A. D., et al. (2006). Suggestive evidence for association of the M. C., et al. (2005). The clock gene Per2 influences the glutamatergic system
circadian genes PERIOD3 and ARNTL with bipolar disorder. Am J Med and modulates alcohol consumption. Nat Med 11, 35−42.
Genet B Neuropsychiatr Genet 141, 234−241. Spittaels, K., Van den Haute, C., Van Dorpe, J., Terwel, D., Vandezande, K.,
Ogden, C. A., Rich, M. E., Schork, N. J., Paulus, M. P., Geyer, M. A., Lohr, J. B., Lasrado, R., et al. (2002). Neonatal neuronal overexpression of glycogen
et al. (2004). Candidate genes, pathways and mechanisms for bipolar (manic- synthase kinase-3 beta reduces brain size in transgenic mice. Neuroscience
depressive) and related disorders: an expanded convergent functional 113, 797−808.
genomics approach. Mol Psychiatry 9, 1007−1029. Sprouse, J., Braselton, J., & Reynolds, L. (2006). Fluoxetine modulates the
Padiath, Q. S., Paranjpe, D., Jain, S., & Sharma, V. K. (2004). Glycogen circadian biological clock via phase advances of suprachiasmatic nucleus
synthase kinase 3beta as a likely target for the action of lithium on circadian neuronal firing. Biol Psychiatry 60, 896−899.
clocks. Chronobiol Int 21, 43−55. Srinivasan, V., Smits, M., Spence, W., Lowe, A. D., Kayumov, L., Pandi-
Pandi-Perumal, S. R., Srinivasan, V., Maestroni, G. J., Cardinali, D. P., Perumal, S. R., et al. (2006). Melatonin in mood disorders. World J Biol
Poeggeler, B., & Hardeland, R. (2006). Melatonin: Nature's most versatile Psychiatry 7, 138−151.
biological signal? FEBS J 273, 2813−2838. Stephan, F. K. (2002). The “other” circadian system: food as a Zeitgeber. J Biol
Pirovano, A., Lorenzi, C., Serretti, A., Ploia, C., Landoni, S., Catalano, M., et al. Rhythms 17, 284−292.
(2005). Two new rare variants in the circadian “clock” gene may influence Stokkan, K. A., Yamazaki, S., Tei, H., Sakaki, Y., & Menaker, M. (2001).
sleep pattern. Genet Med 7, 455−457. Entrainment of the circadian clock in the liver by feeding. Science 291,
Prickaerts, J., Moechars, D., Cryns, K., Lenaerts, I., van Craenendonck, H., 490−493.
Goris, I., et al. (2006). Transgenic mice overexpressing glycogen synthase Szczepankiewicz, A., Rybakowski, J. K., Suwalska, A., Skibinska, M.,
kinase 3beta: a putative model of hyperactivity and mania. J Neurosci 26, Leszczynska-Rodziewicz, A., Dmitrzak-Weglarz, M., et al. (2006).
9022−9029. Association study of the glycogen synthase kinase-3beta gene polymor-
Redlin, U. (2001). Neural basis and biological function of masking by light in phism with prophylactic lithium response in bipolar patients. World J Biol
mammals: suppression of melatonin and locomotor activity. Chronobiol Int Psychiatry 7, 158−161.
18, 737−758. Szczepankiewicz, A., Skibinska, M., Hauser, J., Slopien, A., Leszczynska-
Reick, M., Garcia, J. A., Dudley, C., & McKnight, S. L. (2001). NPAS2: an Rodziewicz, A., Kapelski, P., et al. (2006). Association analysis of the GSK-
analog of clock operative in the mammalian forebrain. Science 293, 3beta T-50C gene polymorphism with schizophrenia and bipolar disorder.
506−509. Neuropsychobiology 53, 51−56.
Reppert, S. M., & Weaver, D. R. (2001). Molecular analysis of mammalian Takano, A., Uchiyama, M., Kajimura, N., Mishima, K., Inoue, Y., Kamei, Y., et al.
circadian rhythms. Annu Rev Physiol 63, 647−676. (2004). A missense variation in human casein kinase I epsilon gene that
Riemann, D., Konig, A., Hohagen, F., Kiemen, A., Voderholzer, U., Backhaus, induces functional alteration and shows an inverse association with circadian
J., et al. (1999). How to preserve the antidepressive effect of sleep rhythm sleep disorders. Neuropsychopharmacology 29, 1901−1909.
deprivation: a comparison of sleep phase advance and sleep phase delay. Eur Tataroglu, O., Aksoy, A., Yilmaz, A., & Canbeyli, R. (2004). Effect of lesioning
Arch Psychiatry Clin Neurosci 249, 231−237. the suprachiasmatic nuclei on behavioral despair in rats. Brain Res 1001,
Roybal, K., Theobold, D., Graham, A., DiNeri, J.A., Russo, S.J., Krishnan, V., 118−124.
et al., in press. Mania-like behavior induced by disruption of CLOCK. Proc Terman, M., & Terman, J. S. (2005). Light therapy for seasonal and nonseasonal
Natl Acad Sci U S A, (Electronic publication ahead of print, March 19). depression: efficacy, protocol, safety, and side effects. CNS Spectr 10,
Rusting, C. L., & Larsen, R. J. (1998). Diurnal patterns of unpleasant mood: 647−663 (quiz 672).
associations with neuroticism, depression, and anxiety. J Pers 66, 85−103. Terman, J. S., Terman, M., Lo, E. S., & Cooper, T. B. (2001). Circadian time of
Sato, T. K., Panda, S., Miraglia, L. J., Reyes, T. M., Rudic, R. D., McNamara, P., morning light administration and therapeutic response in winter depression.
et al. (2004). A functional genomics strategy reveals Rora as a component of Arch Gen Psychiatry 58, 69−75.
the mammalian circadian clock. Neuron 43, 527−537. Thapan, K., Arendt, J., & Skene, D. J. (2001). An action spectrum for melatonin
Scott, A. J. (2000). Shift work and health. Prim Care 27, 1057−1079. suppression: evidence for a novel non-rod, non-cone photoreceptor system
Serretti, A., Benedetti, F., Mandelli, L., Lorenzi, C., Pirovano, A., Colombo, C., in humans. J Physiol 535, 261−267.
et al. (2003). Genetic dissection of psychopathological symptoms: insomnia Thase, M. E. (2005). Bipolar depression: issues in diagnosis and treatment.
in mood disorders and CLOCK gene polymorphism. Am J Med Genet B Harv Rev Psychiatry 13, 257−271.
Neuropsychiatr Genet 121, 35−38. Touitou, Y., Fevre, M., Bogdan, A., Reinberg, A., De Prins, J., Beck, H., et al.
Serretti, A., Artioli, P., & De Ronchi, D. (2004). The 5-HT2C receptor as a target (1984). Patterns of plasma melatonin with ageing and mental condition:
for mood disorders. Expert Opin Ther Targets 8, 15−23. stability of nyctohemeral rhythms and differences in seasonal variations.
Serretti, A., Cusin, C., Benedetti, F., Mandelli, L., Pirovano, A., Zanardi, R., et al. Acta Endocrinol (Copenh) 106, 145−151.
(2005). Insomnia improvement during antidepressant treatment and CLOCK Tuma, J., Strubbe, J. H., Mocaer, E., & Koolhaas, J. M. (2005). Anxiolytic-like
gene polymorphism. Am J Med Genet B Neuropsychiatr Genet 137, 36−39. action of the antidepressant agomelatine (S 20098) after a social defeat
Shieh, K. R., Chu, Y. S., & Pan, J. T. (1997). Circadian change of dopaminergic requires the integrity of the SCN. Eur Neuropsychopharmacol 15, 545−555.
neuron activity: effects of constant light and melatonin. NeuroReport 8, Uz, T., Ahmed, R., Akhisaroglu, M., Kurtuncu, M., Imbesi, M., Dirim Arslan, A.,
2283−2287. et al. (2005). Effect of fluoxetine and cocaine on the expression of
Shiino, Y., Nakajima, S., Ozeki, Y., Isono, T., & Yamada, N. (2003). Mutation clock genes in the mouse hippocampus and striatum. Neuroscience 134,
screening of the human period 2 gene in bipolar disorder. Neurosci Lett 338, 1309−1316.
82−84. Vanselow, K., Vanselow, J. T., Westermark, P. O., Reischl, S., Maier, B., Korte, T.,
Shirayama, M., Shirayama, Y., Iida, H., Kato, M., Kajimura, N., Watanabe, T., et al. et al. (2006). Differential effects of PER2 phosphorylation: molecular basis for
(2003). The psychological aspects of patients with delayed sleep phase the human familial advanced sleep phase syndrome (FASPS). Genes Dev 20,
syndrome (DSPS). Sleep Med 4, 427−433. 2660−2672.
Smeraldi, E., Benedetti, F., Barbini, B., Campori, E., & Colombo, C. (1999). Weber, M., Lauterburg, T., Tobler, I., & Burgunder, J. M. (2004). Circadian
Sustained antidepressant effect of sleep deprivation combined with pindolol patterns of neurotransmitter related gene expression in motor regions of the
in bipolar depression. A placebo-controlled trial. Neuropsychopharmacology rat brain. Neurosci Lett 358, 17−20.
20, 380−385. Wehr, T. A., Duncan, W. C., Jr., Sher, L., Aeschbach, D., Schwartz, P. J., Turner,
Souetre, E., Salvati, E., Belugou, J. L., Pringuey, D., Candito, M., Krebs, B., et al. E. H., et al. (2001). A circadian signal of change of season in patients with
(1989). Circadian rhythms in depression and recovery: evidence for blunted seasonal affective disorder. Arch Gen Psychiatry 58, 1108−1114.
232 C.A. McClung / Pharmacology & Therapeutics 114 (2007) 222–232

Weiner, N., Clement, H. W., Gemsa, D., & Wesemann, W. (1992). Circadian Wu, J. C., & Bunney, W. E. (1990). The biological basis of an antidepres-
and seasonal rhythms of 5-HT receptor subtypes, membrane anisotropy and sant response to sleep deprivation and relapse: review and hypothesis.
5-HT release in hippocampus and cortex of the rat. Neurochem Int 21, 7−14. Am J Psychiatry 147, 14−21.
Welsh, D. K., & Moore-Ede, M. C. (1990). Lithium lengthens circadian period Xu, Y., Padiath, Q. S., Shapiro, R. E., Jones, C. R., Wu, S. C., Saigoh, N., et al.
in a diurnal primate, Saimiri sciureus. Biol Psychiatry 28, 117−126. (2005). Functional consequences of a CKIdelta mutation causing familial
Wesemann, W., & Weiner, N. (1990). Circadian rhythm of serotonin binding in advanced sleep phase syndrome. Nature 434, 640−644.
rat brain. Prog Neurobiol 35, 405−428. Yin, L., Wang, J., Klein, P. S., & Lazar, M. A. (2006). Nuclear receptor Rev-
Wirz-Justice, A., & Van den Hoofdakker, R. H. (1999). Sleep deprivation in erbalpha is a critical lithium-sensitive component of the circadian clock.
depression: what do we know, where do we go? Biol Psychiatry 46, Science 311, 1002−1005.
445−453. Ying, S. W., Rusak, B., Delagrange, P., Mocaer, E., Renard, P., & Guardiola-
Wirz-Justice, A., Graw, P., Krauchi, K., Gisin, B., Arendt, J., Aldhous, M., et al. Lemaitre, B. (1996). Melatonin analogues as agonists and antagonists in the
(1990). Morning or night-time melatonin is ineffective in seasonal affective circadian system and other brain areas. Eur J Pharmacol 296, 33−42.
disorder. J Psychiatr Res 24, 129−137. Yoneyama, S., Hashimoto, S., & Honma, K. (1999). Seasonal changes of human
Wirz-Justice, A., Graw, P., Krauchi, K., Gisin, B., Jochum, A., Arendt, J., et al. circadian rhythms in Antarctica. Am J Physiol 277, R1091−R1097.
(1993). Light therapy in seasonal affective disorder is independent of time of Yuferov, V., Kroslak, T., Laforge, K. S., Zhou, Y., Ho, A., & Kreek, M. J. (2003).
day or circadian phase. Arch Gen Psychiatry 50, 929−937. Differential gene expression in the rat caudate putamen after “binge” cocaine
Wirz-Justice, A., Benedetti, F., Berger, M., Lam, R. W., Martiny, K., Terman, M., administration: advantage of triplicate microarray analysis. Synapse 48,
et al. (2005). Chronotherapeutics (light and wake therapy) in affective 157−169.
disorders. Psychol Med 35, 939−944. Zupancic, M., & Guilleminault, C. (2006). Agomelatine: a preliminary review of
Witte, K., & Lemmer, B. (1991). Rhythms in second messenger mechanisms. a new antidepressant. CNS Drugs 20, 981−992.
Pharmacol Ther 51, 231−237.

You might also like