You are on page 1of 14

ª 2010 The Authors

Bipolar Disorders 2010: 12: 459–472 Journal compilation ª 2010 John Wiley & Sons A/S
BIPOLAR DISORDERS

Review Article

Circadian rhythms and sleep in bipolar


disorder
Murray G, Harvey A. Circadian rhythms and sleep in bipolar disorder. Greg Murraya and Allison Harveyb
Bipolar Disord 2010: 12: 459–472. ª 2010 The Authors. a
Faculty of Life and Social Sciences, Swinburne
Journal compilation ª 2010 John Wiley & Sons A ⁄ S. University of Technology, Hawthorn, Victoria,
Australia, bDepartment of Psychology,
Objective: Biological rhythm pathways are highlighted in a number of University of California, Berkeley, CA, USA
etiological models of bipolar disorder, and the management of circadian
instability appears in consensus treatment guidelines. There are,
however, significant conceptual and empirical limitations on our
understanding of a hypothesised link between circadian, sleep, and
emotion regulation processes in bipolar disorder. The aim of this article
is to articulate the limits of scientific knowledge in relation to this
hypothesis.

Methods: A critical evaluation of various literatures was undertaken.


The basic science of circadian and sleep processes, their involvement in doi: 10.1111/j.1399-5618.2010.00843.x
normal emotion regulation, and the types of evidence suggesting Key words: bipolar disorder – circadian – emotion
circadian ⁄ sleep involvement in bipolar disorder are reviewed. regulation – sleep

Results: Multiple lines of evidence suggest that circadian and sleep- Received 23 December 2009, revised and
wake processes are causally involved in bipolar disorder. These processes accepted for publication 25 May 2010
demonstrably interact with other neurobiological pathways known to be
important in bipolar disorder, but are unique in that they are open to Corresponding author:
behavioural manipulation. Greg Murray, Associate Professor
Faculty of Life and Social Sciences
Conclusion: Further research into biological rhythm pathways to Swinburne University of Technology
bipolar disorder is warranted. Person-environment feedback loops are Hawthorn, Victoria 3122, Australia
fundamental to circadian adaptation, and models of circadian Fax: + 61-3-9819-0574
pathogenesis (and treatment) should recognize this complexity. E-mail: gwm@swin.edu.au

critical role in the emotion dysregulation at the


Introduction and overview
heart of BD. Biological rhythm approaches are
Circadian rhythm hypotheses have been prominent compatible with other neurobiological explana-
in the explanation of bipolar disorder (BD) for tions of BD, and this article includes numerous
more than 20 years (1, 2). Changes in sleep are part examples of overlap between circadian ⁄ sleep path-
of diagnostic criteria (3), and stabilising daily ways and other targets of investigation.
rhythms is recognised as therapeutic in consensus The aim of this review article is to encourage
treatment guidelines (4, 5). Biological rhythmicity further research into these important pathways by
is also a priority for patients, who report significant tracing the limits of scientific knowledge about
concern about their sleep (6) and readily appreciate biological rhythms in BD. We first outline the
the importance of rhythm stability (7).1 In short, basics of circadian rhythms and sleep, highlighting
there is consensus that biological rhythms play a the challenge of empirically separating these two
factors. In the next section, literature on circadian
1 and sleep moderation of emotion is reviewed,
The term biological rhythms is used to encompass both
largely focusing on nonclinical populations. Evi-
circadian and sleep-wake processes.
dence for an association between biological
Neither GM nor AH has any commercial associations that rhythms and BD is detailed next, and causal
might pose a conflict of interest in connection with this inferences considered. The penultimate section
manuscript. critically considers potential mechanisms of
459
Murray and Harvey

biological rhythm action in BD, particularly the and dark due to the planetÕs rotation (15). To a
impact of clock genes and the putative role of rapid lesser degree, the SCN is also responsive to
eye movement (REM) sleep in emotion regulation. nonphotic cues, including arousal ⁄ locomotor activ-
Finally, we review clinical implications of the ity, social cues, feeding, sleep deprivation, and
fact that biological rhythmicity is modified by temperature (16).
behaviour. As shown in Figure 1, the circadian system is a
network including multiple feedback loops (14).
The peripheral clocks (extra-SCN oscillators in
Circadian rhythms and sleep many organs and tissues) that the SCN orches-
trates impact on the central clock. For example,
Circadian system
redox potentials within cells are affected by circa-
The endogenous circadian time-keeping system, dian modulation of the mitochondrial oxidative
strongly conserved across species, is adapted to phosphorylation pathway, but redox potential in
optimise engagement with the earthÕs cyclic envi- turn affects the concentrations of oscillator com-
ronment by predicting critical environmental ponents (including Clock, Mop3 and NPAS2) and
change (8). In mammals, the circadian pacemaker thereby the rate of reactions in the SCN (17, 18).
responsible for the temporal internal organisation The output rhythms themselves also affect the
and generation of endogenous rhythms of approx- clock. For example, one of the major functional
imately 24 hours is located in the hypothalamic targets of SCN output is the pineal gland and its
suprachiasmatic nucleus (SCN) (9). At the molec- release of melatonin. There is a processing loop
ular level, intrinsically rhythmic cells of the SCN from the SCN to the pineal gland and back,
generate self-sustained rhythmicity via an autoreg- supporting a process whereby when melatonin is
ulatory transcription-translation feedback loop synthesized (during the subjective night), it is
involving the genes: circadian locomotor output detected by receptors in the SCN (MT1 and
cycles kaput (Clock), Bmal1 (also known as Arntl), MT2), which moderate clock resetting at dusk
period homologue 1 (Per1), Per2, Cryptochrome 1 and dawn (19). The SCN also appears to modify its
(Cry1), and Cry2 (10). The SCN output rhythm own inputs (e.g., 20, 21). For example, lesion
arises from nonlinear dynamic interaction between studies in rats suggest that retinal circadian
the cells in the SCN (11, 12). rhythms in Per2 messenger RNA are dependent
The endogenous period generated in the SCN is on SCN functioning (22).
close to but generally not equal to 24 hours. The Functionally, the most important clock feedback
process by which the pacemaker is both set to a is behaviour itself. Behaviour influences the circa-
24-hour period and kept in appropriate phase with dian clock directly (arousal is a zeitgeber) (16), but
seasonally shifting astronomical day length is also indirectly via gating of light exposure. This is
called entrainment. An important property of the demonstrated clinically in the use of light treatment
circadian system, therefore, is its fundamentally for seasonal affective disorder (23, 24) and the use
open nature, and this open nature includes feed- of social rhythm stabilization to moderate relapse
back to the pacemaker from clock-controlled risk in BD (25, 26; see below). Given circadian
activity of the organism (13, 14). Entrainment involvement in the sleep-wake cycle, evidence for
occurs via zeitgebers (environmental events that the effectiveness of cognitive-behavioural interven-
can affect phase and period of the clock), the most tions for insomnia also speaks to this important
prominent of which is the daily alternation of light feedback loop (27).

Output
SCN Peripheral rhythms in
Light physiology
pacemaker oscillators
and
behaviour

Fig. 1. Schematic representation of the circadian system. SCN = suprachiasmatic nucleus.

460
Circadian rhythms and sleep in BD

Processes C and S are unsuitable for people


Sleep
diagnosed with BD. Under normal conditions,
Phylogenetically, sleep is a more recent and less Process C and Process S operate in synchrony (41),
ubiquitous adaptation than circadian rhythmicity and in the field we can only observe the sleep-wake
(28). The specific adaptive function of sleep is cycle, which is a complex endpoint of volition,
unknown, but sleep is widely believed to perform a SCN output, and the sleep homeostat (Fig. 2).
restorative function associated with the strength- Most of what we know about separate circadian
ening of immune function, somatic growth, and an and sleep processes, therefore, comes from labora-
anabolic metabolic state (29). Evidence is also tory studies. In the forced desynchrony (FD)
accumulating for sleepÕs role in particular types of protocol, a non-24-hour sleep-wake schedule (typ-
cognition (30, 31), memory consolidation, and ically 28 hours) is enforced. Under these condi-
brain plasticity (32–35). tions, the circadian oscillator continues to cycle at
Based on characteristic physiological patterns, approximately 24 hours and desynchronises from
normal sleep can be divided into two distinct the sleep-wake cycle which adopts the enforced
states: REM and non-REM (NREM). During 28-hour period, thereby enabling separate estimate
REM (or paradoxical) sleep, the brain is highly of circadian and sleep-homeostatic components
activated, the muscles of the body are paralyzed, (42). The sleep disruption and isolation of the
and dreaming occurs. In NREM, growth hormone multiday FD protocol make it unsuitable for
is preferentially secreted and protein synthesis is individuals vulnerable to emotion dysregulation,
increased, leading to the view that NREM may be and therefore fundamental questions about Process
critical for energy conservation and restoration. C and S involvement in BD cannot be directly
Characteristic electroencephalograph (EEG) pat- tested.
terns support NREM sleep being further catego- The neurobiology of the C · S interaction is not
rized into substages: 1 (drowsiness, transition to fully understood. Arousal is mediated by neurons
sleep, lowest arousal thresholds), 2 (light sleep), of the ascending reticular activating system which
and 3 and 4 (deep sleep, highest arousal thresh- project to the thalamus and basal forebrain (43,
olds). Together, stages 3 and 4 are often called 44). Research by Saper and others (45–47) suggests
delta or slow wave sleep (SWS) (36). that the mechanism by which arousal is curtailed to
On a typical night, sleep of a young adult begins in produce sleep involves the ventrolateral preoptic
stage 1 NREM and progresses through deeper nucleus (VLPO), a group of neurons in the
NREM stages. The first episode of REM sleep hypothalamus which, via release of galanin and
occurs approximately 80–100 min after sleep onset. GABA, inhibits activity of the locus coeruleus,
Thereafter, NREM sleep and REM sleep cycle with raphe nucleus, and the tuberomammillary nucleus.
a period of approximately 90 min. NREM stages 3 Mutual inhibition between arousal circuits and the
and 4 concentrate in the early cycles and REM sleep VLPO ensures extended periods of wake undis-
episodes lengthen across the night. Therefore, in a turbed by sleep and vice versa. Stability in sleep or
normal night of sleep, SWS predominates in the wake, and rapid switching between the two states,
early part of the sleep phase, while REM is more is achieved by a putative bistable switch (45). This
prevalent later in the night. Indeed, we are most
likely to wake through the gate of REM sleep.
Social time
and volition
The relationship between circadian and sleep processes
The sleep-wake cycle is regulated by two proc-
esses, one of which is the clock-like circadian
system described above (typically called Process
C). The second factor is sleepÕs (hourglass-like) Sleep-wake cycle
homeostatic self-modulation [Process S (37)]. Proc-
ess S is accumulated during wakefulness, dissipates
during sleep, and regulates duration and structure
of sleep on the basis of the history of sleep and
SCN Sleep
wakefulness. The bidirectional interaction between
pacemaker homeostat
Processes C and S predicts the propensity, timing,
and internal structure of sleep (38–40). Fig. 2. Interaction between circadian and sleep homeostatic
For what follows, it is important to note that mechanisms in modulating the 24-hour rhythm of activity.
protocols for investigating the interaction between SCN = suprachiasmatic nucleus.

461
Murray and Harvey

switch is modulated by a circadian alerting signal


Particular importance of REM sleep in emotion regulation
deriving from the SCN, and, on the other side of
the switch, the homeostatic sleep drive. The neural The sleep state most strongly regulated by the
mechanisms of the sleep homeostat are unknown, SCN, REM sleep, may be particularly important in
but there is growing evidence that the wake-linked emotional processing and emotion regulation. Key
accumulation of the endogenous neuromodulator findings include correlations between presleep
adenosine may be involved (48, 49). mood and REM parameters, prospective associa-
tions between dream content and psychological
outcomes, and REM-facilitated recall of emotional
Sleep, circadian function, and affect
information (73, 74). There is also converging
As in the study of other psychiatric disorders, it is evidence concerning the role of REM in emotional
not uncommon to conceptualize BD as the extreme processing (75–79).
clinical manifestation of a neurobehavioral trait The neurobiology of dreaming is an area of
that is distributed throughout the population. Spe- growing interdisciplinary interest. Stickgold and
cifically, BD can be characterised as a pathologically colleagues (33, 80) proposed that dreaming is the
elevated propensity to emotional dysregulation (50– conscious awareness of critical ÔofflineÕ cortical
52). Before reviewing the known associations generation functions occurring during sleep. In
between biological rhythms and BD, it is therefore this model, dreaming is distinguished by the
useful to review findings linking biological rhythms activation of neural networks supporting weak
to emotion regulation in normal populations. cortical associations in the absence of dorsolateral
prefrontal cortex or hippocampal feedback, but in
the presence of active error detection circuits (in
Circadian interactions with emotion
the anterior cingulate cortex) and affective evalu-
The circadian system modulates current mood ation of errors by the amygdala and orbitofrontal
state, particularly positive affect (53–56), and cortex (81, 82). Potential implications of REM
challenges to the circadian system, such as shift neurobiology for emotion regulation in BD are
work and jet lag, have negative consequences for considered below in the subsection ÔSleep-specific
mood (57–59). Under naturalistic conditions, emo- mechanismsÕ.
tional state also moderates circadian function via
its influence on social rhythms, light exposure,
Neurotransmitters and the sleep · circadian · emotion
arousal, and sleep (16, 60). Bidirectional influence
interaction
can also be traced at the neurobiologic level:
afferents from the SCN project via the paraven- At least two neurotransmitter circuits are critical
tricular thalamic nucleus to the mesolimbic dopa- in the link between biological rhythms and
minergic reward system (61), while emotion circuits emotion. First, dopaminergic pathways, especially
impact circadian (and homeostatic) aspects of sleep cells of the ventral tegmental area (VTA) and
regulation (62, 63). zona compacta of the substantia nigra (SNc), are
strongly implicated in reward motivation and
positive affects (83–85). Dopamine has been
Sleep disturbance and emotion regulation
called a key substance in the regulation of
Behavioural data support the popular assumption sleep-wake (86), and dopaminergic neurons of
that sleep disturbance strongly increases negative the VTA and SNc are again implicated (87),
mood, irritability, and affective volatility (e.g., 64– particularly in REM sleep (88). Dopaminergic
69). Sleep loss has been shown not only to increase pathways also have multiple interactions with the
negative emotional response to goal-thwarting SCN (e.g., 55, 61, 89, 90). Second, serotonergic
events, but also to decrease positive emotional pathways have pronounced (albeit complex)
responses to goal-enhancing events (70). High relationships with anxiety and stress responses
levels of emotional arousal can, in turn, disturb (91). Serotonergic pathways are critical in circa-
sleep, raising the possibility of a vicious cycle dian function (92) [and vice versa (93)], sleep per
between sleep disturbance and emotion dysregula- se (46, 94), and the interaction between stress and
tion (71). At the neural-systems level, sleep sleep (67, 95–98).
deprivation has been linked to decreased medial-
prefrontal cortical activity and increased amygdala
From normal populations to patients with BD
activation, a distribution of activation consistent
with impaired top-down regulation of emotional The above review presents strong behavioural
responses (72). evidence for the notion that emotion and emotion
462
Circadian rhythms and sleep in BD

regulation are associated with circadian function


Sleep, circadian function, and mood symptoms in BD
and sleep. Neuroanatomical pathways subserving
these associations have been outlined, and the A number of specific links have been demon-
interacting role of two critical neurotransmitters strated between BD and circadian and ⁄ or sleep
(dopamine and serotonin) has been described. It function. Our aim here is to consider critically the
seems reasonable to propose that the same rela- nature and implications of these known associa-
tionships hold in patients with BD, and indeed to tions, and it is useful to group findings as shown in
hypothesize that the symptoms of mood episodes Table 1.
in BD may be attributable, at least in part, to the The findings summarised in Table 1 indicate that
pathways described here. It is particularly note- most phases of BD (depressed, manic, episode
worthy that an independent literature argues for prodrome, and interepisode periods) are cross-
dopaminergic and serotonergic circuits as critical sectionally associated with sleep and ⁄ or circadian
pathways in BD (e.g., 99–102). rhythm abnormalities. Clinical research shows that
Until recently, there was little integration disturbed sleep affects quality of life in BD (107,
between research into biological rhythm and 108), and, as reviewed above, sleep disturbance is
neurotransmitter approaches to psychopathology. detrimental to emotion regulation. Evidence that
Early steps in this direction are captured in the BD is associated with sleep disturbance therefore
hypothetical model of Figure 3 [adapted from suggests that outcomes in BD can be improved by
Harvey et al. (102)], in which emotion regulation addressing sleep difficulties across phases of the
and sleep disturbance are proposed as the overt disorder. Such interventions are briefly introduced
manifestation of interactions between dopaminer- below in the section ÔPsychosocial interventions
gic, serotonergic, and circadian ⁄ sleep biology. targeting biological rhythms in BDÕ.
It is worth noting three caveats on generalizing Beyond the immediate clinical implications of
from nonclinical to BD populations. First, the poor sleep, a range of evidence also suggests that
multiplicity of BDÕs clinical manifestations is a biological rhythm disturbance is etiologically
challenge to generalization. For example, the involved in BD. Not only is there prospective data
sleep disturbance common in mania (decreased linking sleep to mood in BD, but sleep changes
need for sleep) contrasts with that in bipolar reliably precede episodes (especially mania) and
depression (insomnia and hypersomnia) (103). correlate with symptom load. Most strikingly,
Second, much of what we know about sleep and manipulation of sleep (sleep deprivation) improves
emotion regulation is based on sleep-deprivation bipolar depression and can induce hypoma-
studies, while hypersomnia is a common symptom nia ⁄ mania in some patients. This conclusion aligns
of bipolar depression. Finally, some provocative with current clinical practice, in which sleep
disjunctions need to be recognized: (i) in nonclin- monitoring is a central relapse prevention strategy
ical samples, sleep restriction may decrease (109).
reward sensitivity (70), while mania appears to The literature cited in Table 1 also provides
involve positive feedback between sleep loss and support for circadian dysfunction as a causal path-
reward seeking (104); and (ii) in nonclinical way to BD. First, relapse can be precipitated by
samples, REM sleep appears to support emotion zeitgeber challenge (particularly light manipula-
regulation (72, 105), while in mood disorder tion), and effective treatments for acute episodes
samples, increased density and decreased latency moderate circadian parameters. Second, a range of
of REM are associated with pathology (106; see data suggests that Ôcircadian instabilityÕ may act as
below). a traitlike vulnerability or diathesis to BD. For

Neurobiology Phenomenology Diagnosis

DA function
Sleep
disturbance
Genetic Sleep/circadian Bipolar
processes biology disorder

Emotion
dysregulation

5-HT function

Fig. 3. Emotion regulation and sleep disturbance as a final common pathway from neurobiological abnormality to bipolar disorder
[adapted with permission from Harvey et al. (102)]. DA = dopamine.

463
Murray and Harvey

Table 1. Associations between bipolar disorder (BD) and biological rhythm function

Neurobiology shared between BD and biological rhythms


• Circadian and sleep function are subserved by the same brain regions (153) and neurotransmitters (e.g., 99, 100) are implicated in
mood disorder.
• Polymorphisms in circadian genes have been associated with symptoms of BD in preclinical and human studies (129, 147, 162, 181).

Biological rhythms as diatheses to BD


• Sleep disturbance and instability of 24-hour rhythms continue when BD patients are not acutely ill (6, 182–185).
• Dysregulation of 24-hour activity rhythms has been reported in a familial high-risk sample (186) and in individuals at risk of
hypomania (187).
• Neuroticism, the primary (albeit nonspecific) temperamental predisposition to BD, may be associated with circadian instability (119).
• Biological sensitivity to light, as measured in nighttime suppression of the pineal hormone melatonin, has been proposed as a trait
marker of vulnerability to BD (188).
• Delayed circadian phase of melatonin secretion has been reported in euthymic BD patients (189), and BD patients self-report as more
evening type (190).

Cross-sectional associations
• Some clinical features of BD are timed phenomena: diurnal variation in mood and early-morning wakening in depression, as well as
seasonal and other cyclic variation in symptoms, are suggestive of circadian involvement (142).
• Circadian rhythms (including activity, body temperature, melatonin, cortisol and thyrotropin) are altered in episodes of BD
(see 111, 191 for a review).
• Mania is strongly associated with decreased need for sleep, and insomnia and hypersomnia are both reliably found in bipolar
depression (see 103 for a review).
• Within BD, short sleep duration is associated with more severe symptomatology, while both short and long sleep duration are
associated with poorer function and quality of life (192).
• Episodes of BD have been associated with sleep polysomnographic changes (e.g., 193–195). The most consistent finding is
abnormalities of rapid eye movement (REM) sleep in both mania and bipolar depression [typically shorter latency and increased
density (see 103)].

Predictive relationships
• Episodes of BD can be precipitated by zeitgeber challenges, including seasonal change and time-zone travel (see 196 for a review).
• Sleep disturbance is the most common prodrome of mania and a significant prodromal symptom of bipolar depression (197). Altered
sleep often precedes deterioration in clinical state and worsens further during an episode (197–199).
• Prospective evidence of a complex association between sleep length and mood change in BD has been found in a number of studies
(198, 200–203).

Experimental and treatment effects


• Deliberate sleep deprivation is a same-day powerful treatment for bipolar (and unipolar) depression (203, 204). Maintenance of the
therapeutic effect beyond the next sleep phase is a target of current research (171).
• In a recent study, combined sleep deprivation, sleep phase advance and timed light was an effective adjunctive intervention for
bipolar depression (205).
• Bright light can induce symptoms of mania (171) and has significant effects on bipolar depression (206).
• Experimentally induced sleep deprivation induces hypomania or mania in a proportion of patients (199, 203).
• Bright light can induce symptoms of mania, and Ôdark therapyÕ has been found to stabilize patients with rapid-cycling BD (171, 207).
• Effective treatments for BD (lithium, antidepressants, anxioloytics, electroconvulsive therapy) affect circadian function (208–210).
• Lithium and selective serotonin reuptake inhibitors impact genes involved in circadian function (92, 132, 211, 212).
• Future course in BD is predicted by sleep disruption and REM density (106).
• Effective psychosocial treatments for BD decrease REM density (213, 214).
• Improved social rhythmicity decreases relapse in BD (26).

example, disruption of activity and sleep cycles are


Mechanisms underpinning biological rhythm
found outside episodes and in children of BD
involvement in BD
patients. Further evidence for a circadian diathesis
comes from a possible link between circadian The mechanisms by which biological rhythms
function and the general vulnerability trait neurot- affect the development, course, and treatment of
icism, and from preclinical and human studies BD are not known. Early hypotheses emphasized
suggesting an association between specific clock gross abnormalities in circadian function, includ-
genes and BD (see below). This conclusion is also ing short intrinsic period of the oscillator (114),
consistent with existing approaches which empha- phase advance (115), phase delay (116), and
size circadian instability as an element of patho- attenuated circadian amplitude (117–119). The
genesis and a target for relapse prevention efforts relation between Processes C and S has also been
(e.g., 5, 110–113). implicated (120), and Process S itself has been
464
Circadian rhythms and sleep in BD

hypothesised to underpin the therapeutic effect the reward value of cocaine (146). Notably,
of sleep deprivation (121). Coupling to external CLOCK mutant mice also show mania-like behav-
zeitgebers is critical in some models (26, 118, 122, iour which is reversed by lithium treatment (147),
123). Physiological investigations of these broad and this reversal is mediated by altered regulation
hypotheses have been scarce over the past decade of dopamine release in the VTA (146).
(2), and the methodological issues flagged above Evidence for a BD-relevant pathway linking
are a barrier to progress. Promising foci of circadian genes to the dopaminergic reward system
contemporary research are circadian genes and is consistent with the strong comorbidity between
the role of REM sleep in emotional processing, as BD and substance abuse (148), the association
discussed next. between manic states and psychostimulant use
(149), and the action of atypical antipsychotic
medications for mania (134). A dopaminergic
Circadian genes
reward · clock gene circuit has been reliably dem-
As research into broad circadian hypotheses has onstrated in nonhuman species (e.g., 150, 151) and
waned, the study of the genetics of circadian is strongly implicated in human mood regulation
function in BD has expanded. Traditional candi- (55, 152–156).
date gene studies have found trends for an asso- It is important to situate these clock gene
ciation between BD and the CLOCK gene (124, findings within the broader genetics of BD. Like
125), but findings are inconsistent (126). A single other psychiatric disorders, BD is probably inher-
nucleotide polymorphism in the 3¢ flanking region ited polygenetically, with numerous genes adding
of CLOCK has also been associated with clinical small effects to overall risk (143). Evidence from
features of BD, namely, patterns of more activity human studies suggests that these genetic effects
in the evening, delayed sleep onset and less sleep are probabilistic, nonspecific, contingent on
(127), higher rates of initial, middle and early gene · gene · environment interactions, and caus-
insomnia (128), recurrence of illness episodes (128), ally remote from the phenotype (157). Given small
and changes in neural response (129). genetic effects on risk, it is not surprising that
Several studies show that the mood stabilizer failure to replicate associations is common, and
lithium affects circadian rhythms through the that researchers have sought to refine the pheno-
glycogen synthase kinase 3 beta gene (GSK-3b), type under investigation. In particular, endophe-
which is a central regulator of the circadian clock notypes [quantitative phenotypes intermediate
(130–132). Valproic acid may act via the same between the genotype and the disorder (158)], are
intracellular signalling pathway (133, 134). Inves- a major focus of contemporary study. Disturbance
tigations of associations between BD, its clinical of circadian function is commonly forwarded as a
features, and GSK-3b have not been consistent candidate endophenotype for BD (e.g., 142, 159–
(e.g., 135–137). Findings for an association with 163).
NR1D1 gene (regulation of which by GSK-3b is
important for clock function) are also mixed (138,
Sleep-specific mechanisms
139).
Other circadian genes for which some support The potential importance of REM sleep in normal
exists include PER3 (140, 141), ARNTL (141, 142) emotion regulation was reviewed above. Recently,
and TIMELESS (142). In addition, Shi et al. (124) this hypothesis has been extended to make predic-
reported an interaction between CLOCK and two tions about the role of REM in psychiatric
other genes of interest for their potential circadian disorders, including BD (105, 164). Walker and
function (BHLHB2, CSNK1E). In one genome- colleagues propose that the neurobiology of REM
wide association study, the gene VGCNL1 (the sleep is critical in processing episodic emotional
mouse homolog of which regulates circadian func- memories, and describe three features of REM
tion) had a significant association with caseness neurobiology that together support this role. First,
(143). Larger genome-wide studies have not iden- emotional experiences of the preceding day are
tified circadian gene associations (144, 145). reactivated by increased arousal in limbic and
Preclinical studies observing the effect of gene paralimbic structures. Second, integration of the
mutations have been a fruitful source of hypoth- emotional experiences with other aspects of seman-
eses. Mice with a mutation in the CLOCK gene tic memory are facilitated by dominant theta
show robust sensitization to cocaine and increases oscillations in subcortical and cortical nodes.
in the preference for cocaine (146, 147). These mice Finally, REM occurs in the absence of noradren-
also have increased dopaminergic activity in the ergic arousal, suggesting a brain state that supports
VTA which may be responsible for the increase in emotional processing unencumbered by high
465
Murray and Harvey

anxiety. In parallel, the REM state is dominated by is a largely behavioural psychotherapy aimed at
cholinergic neurochemistry, providing an ideal helping patients maintain stability in their social
substrate for neocortical consolidation of memory rhythms and so reduce the risk of relapse. In the
information. Taken together, REM sleep seems treatment of BD, social rhythm therapy is typically
adapted to progressively decouple emotions from integrated with principles from interpersonal psy-
the memory of emotional experiences, potentially chotherapy in a treatment known as interpersonal
improving next-day mood (164). and social rhythm therapy (IPSRT) (169). IPSRT
Under this model, the increased REM density has proven effective in two large studies (26, 170).
seen in unipolar and bipolar depression (106) has Interest in biological rhythm management is
two implications. First, it may represent a failed expanding alongside growing research into adjunc-
attempt to depotentiate negative emotional expe- tive psychosocial treatments for BD (112, 171–
riences across nights. Second, because emotional 173). Rhythm stabilization is a core component in
memories are preferentially encoded during REM most adjunctive psychosocial treatments for BD
sleep (34), increased REM density may patholog- (113), including cognitive behavioural therapy.
ically reinforce negative self-narratives, maintain- Improving sleep quality is an important compo-
ing negative moods postsleep (105). The nent of rhythm stabilization, and has functional
translational implications of this Ôsleep to forget, benefits in its own right (102, 103). Interestingly,
sleep to rememberÕ model have not been teased out, management of the sleep-wake cycle is a commonly
and there are some contradictory data (e.g., 165), reported well-being strategy amongst people with
but the model raises the exciting possibility that BD (174). Our qualitative investigations suggest
manipulating sleep architecture may therapeuti- that this strategy is appealing not only because it is
cally regulate the balance of emotion and memory judged effective, but also because it is empowering
of past experiences. Important questions for future for patients to understand that a core element of
research include comparisons of the REM-emotion BD biology is partly under their control (175).
regulation association in healthy controls versus Particularly interesting are treatments for bipo-
patients across the phases of BD. lar depression [the most common and most resis-
tant manifestation of BD (176)] using
combinations of zeitgeber manipulation and phar-
Psychosocial interventions targeting biological
macotherapy. For example, Benedetti and col-
rhythms in BD
leagues (177) have shown that progressive sleep
Interactions between biology and psychosocial phase advance across three days can sustain the
factors cannot be ignored in a biological rhythm antidepressant effects of total sleep deprivation,
approach to BD (166). The circadian system is and that the chronobiological mechanism is
adapted to be open to external cues, and environ- enhanced by lithium.
mental manipulations (particularly light exposure) Chronotherapeutic interventions are therefore
have predictable effects on amplitude and phase of an important target for further study. Future
the endogenous oscillator (167, 168). Under natu- research into these interventions should seek to
ralistic conditions, human exposure to zeitgebers is identify moderators (e.g., light sensitivity, severity
gated by behaviour, prominently the sleep-wake and diagnosis, temperament, gender) and media-
cycle (Circadian rhythms and sleep). The sleep-wake tors (e.g., light-dark cycle exposure, sleep quality,
cycle is in turn moderated by volitional and social sleep architecture, circadian rhythmicity, daytime
factors, and mediated partly through cognitive arousal) of treatment outcome. Our knowledge of
mechanisms (Sleep, circadian function, and affect). possible interactions between behavioural and
Biological rhythms therefore function as an open- pharmacological treatments is also limited (171).
loop system crossing multiple levels of persons and Finally, the recent discovery that lightÕs neurobio-
their environment (21). Psychosocial factors are logical effects are blue-shifted (178, 179) encour-
integral to a biological rhythm explanation of BD, ages a more sophisticated investigation of light
a fact which underpins the recent development of treatment for BD. One aspect of this investigation
some novel adjunctive psychosocial interventions must be the potential toxicity of blue-enhanced
for BD. light (see 180).
The social zeitgeber hypothesis proposes that
fundamental circadian instability in BD can be
Summary and conclusions
moderated by increased stabilization of daily
rhythms and zeitgeber exposure [which could Four main themes can be drawn from the present
include light itself (112)]. This hypothesis takes review. First, standard laboratory protocols for
clinical form in social rhythm therapy (169), which separating sleep and circadian function are not
466
Circadian rhythms and sleep in BD

feasible in vulnerable populations, so the biologi- implications for physiology and disease. Nat Rev Genet
cally important distinction between these processes 2008; 9: 764–775.
11. Hu K, Scheer FA, Buijs RM, Shea SA. The endogenous
is difficult to investigate. Fundamental questions circadian pacemaker imparts a scale-invariant pattern of
can instead be addressed by careful extrapolation heart rate fluctuations across time scales spanning minutes
from well populations, perhaps quantified by their to 24 hours. J Biol Rhythms 2008; 23: 265–273.
degree of risk for BD. Second, biological rhythm 12. Liu AC, Welsh DK, Ko CH et al. Intercellular coupling
pathways are interwoven with other pathways confers robustness against mutations in the SCN circa-
dian clock network. Cell 2007; 129: 605–616.
implicated in BD (e.g., monoamines, GSK-3b), 13. Mrosovsky N. Critical assessment of methods and con-
and future research should actively address this cepts in nonphotic phase shifting. Biol Rhythms Res 1999;
complexity. Third, the wide variety of evidence 30: 135–148.
demonstrating biological rhythm involvement in 14. Bechtel W. The downs and ups of mechanistic research:
BD is a rich source of hypotheses for future work circadian rhythm research as an exemplar. Erkenntnis, in
press.
in this burgeoning area. Research in humans has 15. Roennebert T, Foster RG. Twilight times: light and the
yet to discover mechanisms of large effect, but circadian system. Photochem Photobiol 1997; 66: 549–
preclinical studies suggest the reward · circadian 561.
interaction may be particularly fruitful. Finally, 16. Mistlberger RE, Antle MC, Glass JD, Miller JD. Behav-
interventions built on chronobiological principles ioral and serotonergic regulation of circadian rhythms.
Biol Rhythms Res 2000; 31: 240–283.
should be energetically investigated. These inter- 17. Rutter J, Reick M, Wu LC, McKnight SL. Regulation of
ventions are theoretically plausible, benign, eco- clock and NPAS2 DNA binding by the redox state of
nomical, and attractive to patients. NAD cofactors. Science 2001; 293: 510–514.
In conclusion, research into sleep and circadian 18. Panda S, Hogenesch JB. ItÕs all in the timing: many
function in BD is an exciting, challenging and clocks, many outputs. J Biol Rhythms 2004; 19: 374–
387.
rewarding avenue for the future. It has the 19. Hunt AE, Al-Ghoul WM, Gillette MU, Dubocovich ML.
potential to significantly broaden understanding Activation of MT(2) melatonin receptors in rat suprach-
of a particularly serious mental illness and to iasmatic nucleus phase advances the circadian clock. Am J
improve the quality of life and functioning of Physiol Cell Physiol 2001; 280: C110–C118.
patients. 20. Roenneberg T, Merrow M. The network of time: under-
standing the molecular circadian system. Curr Biol 2003;
13: R198–R207.
References 21. Bechtel W, Abrahamsen A. Decomposing, recomposing,
and situating circadian mechanisms: three tasks in devel-
1. Goodwin F, Jamison K. Manic-Depressive Illness. New oping mechanistic explanations. In: Leitgeb H, Hieke A
York: Oxford University Press, 1990. eds. Reduction and Elimination in Philosophy of Mind
2. Goodwin FK, Jamison KR. Manic-Depressive Illness: and Philosophy of Neuroscience. Frankfurt: Ontos Ver-
Bipolar Disorders and Recurrent Depression. New York: lag, 2009: 173–186.
Oxford University Press, 2007. 22. Sakamoto K, Oishi K, Shiraishi M et al. Two circadian
3. American Psychiatric Association. Diagnostic and statis- oscillatory mechanisms in the mammalian retina. Neuro-
tical manual of mental disorders: DSM-IV-TR. 4th Text report 2000; 11: 3995–3997.
Revision ed. Washington, DC: APA, 2000. 23. Murray G, Michalak EE, Levitt AJ et al. O sweet spot
4. Goodwin GM. Evidence-based guidelines for treating where art thou? Light treatment of Seasonal Affective
bipolar disorder: revised second edition. Recommenda- Disorder and the circadian time of sleep. J Affect Disord
tions from the British Association for Psychopharmacol- 2006; 90: 227–231.
ogy. J Psychopharmacol 2009; 23: 346–388. 24. Murray G, Michalak EE, Levitt AJ et al. Therapeutic
5. Yatham LN, Kennedy SH, OÕDonovan C et al. Canadian mechanism in Seasonal Affective Disorder: do fluoxetine
Network for Mood and Anxiety Treatments (CANMAT) and light operate through advancing circadian phase?
guidelines for the management of patients with bipolar Chronobiol Int 2005; 22: 937–943.
disorder: update 2007. Bipolar Disord 2006; 8: 721–739. 25. Frank E, Swartz HA, Boland E. Interpersonal and social
6. Harvey AG, Schmidt DA, Scarna A, Semler CN, Good- rhythm therapy: an intervention addressing rhythm dys-
win GM. Sleep-related functioning in euthymic patients regulation in bipolar disorder. Dialogues Clin Neurosci
with bipolar disorder, patients with insomnia, and sub- 2007; 9: 325–332.
jects without sleep problems. Am J Psychiatry 2005; 162: 26. Frank E, Kupfer DJ, Thase ME et al. Two-year outcomes
50–57. for interpersonal and social rhythm therapy in individuals
7. Suto M, Murray G, Hale S, Amari E, Michalak EE. What with bipolar I disorder. Arch Gen Psychiatry 2005; 62:
works for people with bipolar disorder? Tips from the 996–1004.
experts. J Affect Disord 2010; 124: 76–84. 27. Murray G, Harvey A. Circadian rhythms and sleep in
8. Moore-Ede MC. Physiology of the circadian timing bipolar disorder. In: Yatham LN, Maj M eds. Bipolar
system: predictive versus reactive homeostasis. Am J Disorder: Clinical and Neurobiological Foundations.
Physiol 1986; 250: R737–R752. New York: John Wiley & Sons, in press.
9. Reppert SM, Weaver DR. Coordination of circadian 28. Rial RV, Nicolau MC, Gamundi A et al. The trivial
timing in mammals. Nature 2002; 418: 935–941. function of sleep. Sleep Med Rev 2007; 11: 311–325.
10. Takahashi JS, Hong H-K, Ko CH, McDearmon EL. The 29. Maquet P. Sleep function(s) and cerebral metabolism.
genetics of mammalian circadian order and disorder: Behav Brain Res 1995; 69: 75–83.

467
Murray and Harvey

30. Durmer JS, Dinges DF. Neurocognitive consequences of 52. Murray G, Goldstone E, Cunningham E. Personality and
sleep deprivation. Semin Neurol 2005; 25: 117–129. the predisposition(s) to bipolar disorder: heuristic benefits
31. Wagner U, Gais S, Haider H, Verleger R, Born J. Sleep of a two-dimensional model. Bipolar Disord 2007; 9: 453–
inspires insight. Nature 2004; 427: 352–355. 461.
32. Stickgold R, Walker MP. Sleep-dependent memory con- 53. Murray G, Allen NB, Trinder J. Mood and the circadian
solidation and reconsolidation. Sleep Med 2007; 8: 331– system: investigation of a circadian component in positive
343. affect. Chronobiol Int 2002; 19: 1151–1169.
33. Stickgold R. Sleep-dependent memory consolidation. 54. Thayer RE. Problem perception, optimism, and related
Nature 2005; 437: 1272–1278. states as a function of time of day (diurnal rhythm) and
34. Hu P, Stylos-Allan M, Walker MP. Sleep facilitates moderate exercise: two arousal systems in interaction.
consolidation of emotional declarative memory. Psychol Motiv Emot 1987; 11: 19–36.
Sci 2006; 17: 891–898. 55. Murray G, Nicholas CL, Kleiman J et al. NatureÕs clocks
35. Frank MG, Benington JH. The role of sleep in memory and human mood: the circadian system modulates reward
consolidation and brain plasticity: dream or reality? motivation. Emotion 2009; 9: 705–716.
Neuroscientist 2006; 12: 477–488. 56. Boivin DB, Czeisler CA, Dijk D-J et al. Complex inter-
36. Shneerson JM. Handbook of Sleep Medicine. Oxford: action of the sleep-wake cycle and circadian phase
Blackwell, 2000. modulates mood in healthy subjects. Arch Gen Psychiatry
37. Borbely AA. Sleep: circadian rhythm versus recovery 1997; 54: 145–152.
process. In: Koukkou M, Lehmann D, Angst J eds. 57. Srinivasan V, Spence DW, Pandi-Perumal SR, Trakht I,
Functional States of the Brain: Their Determinants. Cardinali DP. Jet lag: therapeutic use of melatonin and
Amsterdam: Elsevier, 1980: 151–161. possible application of melatonin analogs. Travel Med
38. Dijk D-J, Franken P. Interaction of sleep homeostasis and Infect Dis 2008; 6: 17–28.
circadian rhythmicity: dependent or independent systems? 58. Reinberg A, Ashkenazi I. Internal desynchronization of
In: Kryger MH, Roth T, Dement WC eds. Principles and circadian rhythms and tolerance to shift work. Chrono-
Practice of Sleep Medicine. Philadelphia: Saunders, 2005: biol Int 2008; 25: 625–643.
418–434. 59. Meyrer R, Demling J, Kornhuber J, Nowak M. Effects of
39. Borbely AA. A two process model of sleep regulation. night shifts in bipolar disorders and extreme morningness.
Human Neurobiol 1982; 1: 195–204. Bipolar Disord 2009; 11: 897–899.
40. Edgar DM, Dement WC, Fuller CA. Effect of SCN 60. Ehlers CL, Frank E, Kupfer DJ. Social zeitgebers and
lesions on sleep in squirrel monkeys: evidence for oppo- biological rhythms. Arch Gen Psychiatry 1988; 45: 948–
nent processes in sleep-wake regulation. J Neurosci 1993; 952.
13: 1065–1079. 61. Sleipness EP, Sorg BA, Jansen HT. Diurnal differences in
41. Czeisler CA, Buxton OM, Khalsa SBS. The human dopamine transporter and tyrosine hydroxylase levels in
circadian timing system and sleep-wake regulation. In: rat brain: dependence on the suprachiasmatic nucleus.
Kryger MH, Roth T, Dement WC eds. Principles and Brain Res 2007; 1129: 34–42.
Practice of Sleep Medicine, 4th ed. Philadelphia: Elsevier, 62. Yoshida K, McCormack S, Espana RA, Crocker A,
2005: 375–394. Scammell TE. Afferents to the orexin neurons of the rat
42. Czeisler CA, Duffy JF, Shanahan TL et al. Stability, brain. J Comp Neurol 2006; 494: 845–861.
precision, and near-24-hour period of the human circa- 63. Saper CB, Cano G, Scammell TE. Homeostatic, circa-
dian pacemaker. Science 1999; 284: 2177–2181. dian, and emotional regulation of sleep. J Comp Neurol
43. Coull JT, Jones ME, Egan TD, Frith CD, Maze M. 2005; 493: 92–98.
Attentional effects of noradrenaline vary with arousal 64. Dinges DF, Pack F, Williams K et al. Cumulative
level: selective activation of thalamic pulvinar in humans. sleepiness, mood disturbance, and psychomotor vigilance
Neuroimage 2004; 22: 315–322. performance decrements during a week of sleep restricted
44. Aston-Jones G, Chen S, Zhu Y, Oshinsky ML. A neural to 4–5 hours per night. Sleep 1997; 20: 267–277.
circuit for circadian regulation of arousal. Nat Neurosci 65. Drake CL, Roehrs TA, Burduvali E, Bonahoom A,
2001; 4: 732–738. Rosekind M, Roth T. Effects of rapid versus slow
45. Fuller PM, Gooley JJ, Saper CB. Neurobiology of the sleep- accumulation of eight hours of sleep loss. Psychophysiol-
wake cycle: sleep architecture, circadian regulation, and ogy 2001; 38: 979–987.
regulatory feedback. J Biol Rhythms 2006; 21: 482–493. 66. Hamilton NA, Catley D, Karlson C. Sleep and the
46. Saper CB, Scammell TE, Lu J. Hypothalamic regulation affective response to stress and pain. Health Psychol 2007;
of sleep and circadian rhythms. Nature 2005; 437: 1257– 26: 288–295.
1263. 67. Novati A, Roman V, Cetin T et al. Chronically restricted
47. Gooley JJ, Saper CB. Anatomy of the mammalian sleep leads to depression-like changes in neurotransmitter
circadian system. In: Kryger MH, Roth T, Dement WC receptor sensitivity and neuroendocrine stress reactivity in
eds. Principles and Practice of Sleep Medicine, 4th ed. rats. Sleep 2008; 31: 1579–1585.
Philadelphia: Elsevier, 2005: 335–350. 68. Franzen PL, Siegle GJ, Buysse DJ. Relationships between
48. Landolt HP. Sleep homeostasis: a role for adenosine in affect, vigilance, and sleepiness following sleep depriva-
humans? Biochem Pharmacol 2008; 75: 2070–2079. tion. J Sleep Res 2008; 17: 34–41.
49. Heller HC. A global rather than local role for adenosine 69. El-Sheikh M, Buckhalt JA, Mark Cummings E, Keller P.
in sleep homeostasis. Sleep 2006; 29: 1382–1383. Sleep disruptions and emotional insecurity are pathways
50. Miklowitz DJ, Johnson SL. The psychopathology and of risk for children. J Child Psychol Psychiatry 2007; 48:
treatment of bipolar disorder. Ann Rev Clin Psychol 88–96.
2006; 2: 199–235. 70. Zohar D, Tzischinsky O, Epstein R, Lavie P. The effects
51. Phillips ML. The neural basis of mood dysregulation in of sleep loss on medical residentsÕ emotional reactions
bipolar disorder. Cogn Neuropsychiatry 2006; 11: 233– to work events: a cognitive-energy model. Sleep 2005; 28:
249. 47–54.

468
Circadian rhythms and sleep in BD

71. Dahl RE, Lewin DS. Pathways to adolescent health sleep 91. Charney DS. Psychobiological mechanisms of resilience
regulation and behavior. J Adolesc Health 2002; 31: 175– and vulnerability: implications for successful adaptation
184. to extreme stress. Am J Psychiatry 2004; 161: 195–216.
72. Yoo SS, Gujar N, Hu P, Jolesz FA, Walker MP. The 92. Sprouse J, Braselton J, Reynolds L. Fluoxetine modulates
human emotional brain without sleep–a prefrontal amyg- the circadian biological clock via phase advances of
dala disconnect. Curr Biol 2007; 17: R877–R878. suprachiasmatic nucleus neuronal firing. Biol Psychiatry
73. Harvey AG, Mullin BC, Hinshaw SP. Sleep and circadian 2006; 60: 896–899.
rhythms in children and adolescents with bipolar disorder. 93. Yuan Q, Lin F, Zheng X, Sehgal A. Serotonin modulates
Dev Psychopathol 2006; 18: 1147–1168. circadian entrainment in Drosophila. Neuron 2005; 47:
74. Cartwright R, Agargun MY, Kirkby J, Friedman JK. 115–127.
Relation of dreams to waking concerns. Psychiatry Res 94. Adrien J. Neurobiological bases for the relation between
2006; 141: 261–270. sleep and depression. Sleep Med Rev 2002; 6: 341–351.
75. Nishida M, Pearsall J, Buckner RL, Walker MP. REM 95. Brummett BH, Krystal AD, Ashley-Koch A et al. Sleep
Sleep, Prefrontal theta, and the consolidation of human quality varies as a function of 5-HTTLPR genotype and
emotional memory. Cereb Cortex 2009; 19: 1158–1166. stress. Psychosom Med 2007; 69: 621–624.
76. Wagner U, Born J. Memory consolidation during sleep: 96. Machado RB, Tufik S, Suchecki D. Chronic stress during
interactive effects of sleep stages and HPA regulation. paradoxical sleep deprivation increases paradoxical sleep
Stress 2008; 11: 28–41. rebound: association with prolactin plasma levels and
77. Cantero JL, Atienza M, Stickgold R, Kahana MJ, brain serotonin content. Psychoneuroendocrinology 2008;
Madsen JR, Kocsis B. Sleep-dependent theta oscillations 33: 1211–1224.
in the human hippocampus and neocortex. J Neurosci 97. Meerlo P, Sgoifo A, Suchecki D. Restricted and disrupted
2003; 23: 10897–10903. sleep: effects on autonomic function, neuroendocrine
78. Maquet P, Ruby P, Maudoux A et al. Human cognition stress systems and stress responsivity. Sleep Med Rev
during REM sleep and the activity profile within frontal 2008; 12: 197–210.
and parietal cortices: a reappraisal of functional neuro- 98. Roman V, Walstra I, Luiten PG, Meerlo P. Too little
imaging data. Prog Brain Res 2005; 150: 219–227. sleep gradually desensitizes the serotonin 1A receptor
79. Maquet P, Péters J-M, Aerts J et al. Functional neuro- system. Sleep 2005; 28: 1505–1510.
anatomy of human rapid-eye-movement sleep and dream- 99. Berk M, Dodd S, Kauer-SantÕanna M et al. Dopamine
ing. Nature 1996; 383: 163–166. dysregulation syndrome: implications for a dopamine
80. Stickgold R, Hobson JA, Fosse R, Fosse M. Sleep, hypothesis of bipolar disorder. Acta Psychiatr Scand
learning, and dreams: off-line memory reprocessing. Suppl 2007; 116: 41–49.
Science 2001; 294: 1052–1057. 100. Oquendo MA, Hastings RS, Huang YY et al. Brain
81. Stickgold R. Why we dream. In: Kryger MH, Roth T, serotonin transporter binding in depressed patients with
Dement WC eds. Principles and Practice of Sleep Med- bipolar disorder using positron emission tomography.
icine. Philadelphia: Elsevier, 2005: 579–587. Arch Gen Psychiatry 2007; 64: 201–208.
82. Hobson JA, Pace-Schott EF, Stickgold R. Dreaming and 101. Cousins DA, Butts K, Young AH. The role of dopamine
the brain: toward a cognitive neuroscience of conscious in bipolar disorder. Bipolar Disord 2009; 11: 787–806.
states. Behav Brain Sci 2000; 23: 793–842. 102. Harvey A, Murray G, Chandler RA, Soehner A. Sleep
83. Ashby FG, Isen AM, Turken AU. A neuropsychological disturbance as transdiagnostic: consideration of neurobi-
theory of positive affect and its influence on cognition. ological mechanisms. Clin Psychol Rev 2010; doi:
Psychol Rev 1999; 106: 529–550. 10.10161j.cpr.2010.04.003.
84. Barbano MF, Cador M. Opioids for hedonic experience 103. Harvey AG. Sleep and circadian rhythms in bipolar
and dopamine to get ready for it. Psychopharmacology disorder: seeking synchrony, harmony, and regulation.
2007; 191: 497–506. Am J Psychiatry 2008; 165: 820–829.
85. Bressan RA, Crippa JA. The role of dopamine in 104. Johnson SL. Mania and dysregulation in goal pursuit: a
reward and pleasure behaviour–review of data from review. Clin Psychol Rev 2005; 25: 241–262.
preclinical research. Acta Psychiatr Scand Suppl 2005; 105. Walker MP. The role of sleep in cognition and emotion.
111: 14–21. Ann NY Acad Sci 2009; 1156: 168–197.
86. Lima MM, Andersen ML, Reksidler AB et al. Blockage 106. Krystal AD, Thakur M, Roth T. Sleep disturbance in
of dopaminergic D(2) receptors produces decrease of psychiatric disorders: effects on function and quality of
REM but not of slow wave sleep in rats after REM sleep life in mood disorders, alcoholism, and schizophrenia.
deprivation. Behav Brain Res 2008; 188: 406–411. Ann Clin Psychiatry 2008; 20: 39–46.
87. Monti JM, Monti D. The involvement of dopamine in the 107. Giglio LM, Andreazza AC, Andersen M et al. Sleep in
modulation of sleep and waking. Sleep Med Rev 2007; 11: bipolar patients. Sleep Breath 2009; 13: 169–173.
113–133. 108. Michalak EE, Murray G, Young AH, Lam RW. Quality
88. Dahan L, Astier B, Vautrelle N, Urbain N, Kocsis B, of life impairment in bipolar disorder. In: Ritsner M,
Chouvet G. Prominent burst firing of dopaminergic Awad AG eds. Quality of Life Impairment in Schizo-
neurons in the ventral tegmental area during paradoxical phrenia, Mood and Anxiety Disorders: From Brain
sleep. Neuropsychopharmacology 2007; 32: 1232–1241. Functions to Clinical Practice. New York: Springer,
89. Yuferov V, Butelman ER, Kreek MJ. Biological clock: 2007: 253–274.
biological clocks may modulate drug addiction. Eur J 109. Horn M, Scharer L, Walser S, Scherer-Klabunde D,
Hum Genet 2005; 13: 1101–1103. Biedermann C, Walden J. Comparison of long-term
90. Yujnovsky I, Hirayama J, Doi M, Borrelli E, Sassone- monitoring methods for bipolar affective disorder. Neu-
Corsi P. Signaling mediated by the dopamine D2 ropsychobiology 2002; 45 (Suppl. 1): 27–32.
receptor potentiates circadian regulation by CLOCK: 110. Frank E, Gonzalez JM, Fagiolini A. The importance of
BMAL1. Proc Natl Acad Sci USA 2006; 103: 6386– routine for preventing recurrence in bipolar disorder. Am
6391. J Psychiatry 2006; 163: 981–985.

469
Murray and Harvey

111. Jones SH. Circadian rhythms, multilevel models of 129. Benedetti F, Radaelli D, Bernasconi A et al. Clock genes
emotion and bipolar disorder: an initial step towards beyond the clock: CLOCK genotype biases neural corre-
integration? Clin Psychol Rev 2001; 21: 1193–1209. lates of moral valence decision in depressed patients.
112. Grandin LD, Alloy LB, Abramson LY. The social Genes Brain Behav 2008; 7: 20–25.
zeitgeber theory, circadian rhythms, and mood disorders: 130. Beaulieu JM, Sotnikova TD, Yao WD et al. Lithium
review and evaluation. Clin Psychol Rev 2006; 26: 679– antagonizes dopamine-dependent behaviors mediated by
694. an AKT ⁄ glycogen synthase kinase 3 signaling cascade.
113. Miklowitz DJ, Goodwin GM, Bauer MS, Geddes JR. Proc Natl Acad Sci USA 2004; 101: 5099–5104.
Common and specific elements of psychosocial treatments 131. Gould TD, Manji HK. Glycogen synthase kinase-3: a
for bipolar disorder: a survey of clinicians participating in putative molecular target for lithium mimetic drugs.
randomized trials. J Psychiatr Pract 2008; 14: 77–85. Neuropsychopharmacology 2005; 30: 1223–1237.
114. Kripke DF, Mullaney DJ, Atkinson M, Wolf S. Circadian 132. Yin L, Wang J, Klein PS, Lazar MA. Nuclear receptor
rhythm disorders in manic-depressives. Biol Psychiatry Rev-ERB alpha is a critical lithium-sensitive component
1978; 13: 335–351. of the circadian clock. Science 2006; 311: 1002–1005.
115. Wehr TA, Wirz-Justice A, Goodwin FK. Phase advance 133. Li X, Bijur GN, Jope RS. Glycogen synthase kinase-
of the circadian sleep-wake cycle as an antidepressant. 3beta, mood stabilizers, and neuroprotection. Bipolar
Science 1979; 206: 710–713. Disord 2002; 4: 137–144.
116. Lewy AJ, Sack RL, Singer CM, White DM, Hoban TM. 134. Coyle JT. What can a clock mutation in mice tell us about
Winter depression and the phase-shift hypothesis for bipolar disorder? Proc Natl Acad Sci USA 2007; 104:
bright lightÕs therapeutic effects: history theory and 6097–6098.
experimental evidence. In: Rosenthal NE, Blehar MC 135. Yang S, Van Dongen HP, Wang K, Berrettini W, Bucan
eds. Seasonal Affective Disorders and Phototherapy. New M. Assessment of circadian function in fibroblasts of
York: Guilford Press, 1989: 295–310. patients with bipolar disorder. Mol Psychiatry 2009; 14:
117. Czeisler CA, Kronauer RE, Mooney JJ, Anderson JL, 143–155.
Allan JS. Biologic rhythm disorders, depression and 136. Benedetti F, Bernasconi A, Lorenzi C et al. A single
phototherapy: a new hypothesis. Psychiatr Clin North nucleotide polymorphism in glycogen synthase kinase
Am 1987; 10: 687–709. 3-beta promoter gene influences onset of illness in patients
118. Teicher MH, Glod CA, Harper D et al. Locomotor affected by bipolar disorder. Neurosci Lett 2004; 355:
activity in depressed children and adolescents, I: Circa- 37–40.
dian dysregulation. J Am Acad Child Adolesc Psychiatry 137. Benedetti F, Serretti A, Colombo C, Lorenzi C, Tubazio
1993; 32: 760–769. V, Smeraldi E. A glycogen synthase kinase 3-beta
119. Murray G, Allen NB, Trinder J, Burgess H. Is weakened promoter gene single nucleotide polymorphism is associ-
circadian rhythmicity a characteristic of neuroticism? ated with age at onset and response to total sleep
J Affect Disord 2002; 72: 281–289. deprivation in bipolar depression. Neurosci Lett 2004;
120. Boivin DB. Influence of sleep-wake and circadian rhythm 368: 123–126.
disturbances in psychiatric disorders. J Psychiatry Neu- 138. Severino G, Manchia M, Contu P et al. Association study
rosci 2000; 25: 446–459. in a Sardinian sample between bipolar disorder and the
121. Wirz-Justice A. Biological rhythm disturbances in mood nuclear receptor REV-ERB alpha gene, a critical compo-
disorders. Int Clin Psychopharmacol 2006; 21 (Suppl. 1): nent of the circadian clock system. Bipolar Disord 2009;
S11–S15. 11: 215–220.
122. Ehlers CL, Kupfer DJ, Frank E, Monk TH. Biological 139. Kishi T, Kitajima T, Ikeda M et al. Association analysis
rhythms and depression: the role of zeitgebers and of nuclear receptor Rev-erb alpha gene (NR1D1) with
zeitstorers. Depression 1993; 1: 285–293. mood disorders in the Japanese population. Neurosci Res
123. Healy D, Waterhouse JM. The circadian system and 2008; 62: 211–215.
affective disorders: clocks or rhythms? Chronobiol Int 140. Artioli P, Lorenzi C, Pirovano A et al. How do genes
1990; 7: 5–10. exert their role? Period 3 gene variants and possible
124. Shi J, Wittke-Thompson JK, Badner JA et al. Clock genes influences on mood disorder phenotypes. Eur Neuropsy-
may influence bipolar disorder susceptibility and dysfunc- chopharmacol 2007; 17: 587–594.
tional circadian rhythm. Am J Med Genet B Neuropsy- 141. Nievergelt CM, Kripke DF, Barrett TB et al. Suggestive
chiatr Genet 2008; 147B: 1047–1055. evidence for association of the circadian genes PERIOD3
125. Kripke DF, Nievergelt CM, Joo E, Shekhtman T, Kelsoe and ARNTL with bipolar disorder. Am J Med Genet B
JR. Circadian polymorphisms associated with affective Neuropsychiatr Genet 2006; 141B: 234–241.
disorders. J Circadian Rhythms 2009; 7: 2. 142. Mansour HA, Monk TH, Nimgaonkar VL. Circadian
126. Kishi T, Kitajima T, Ikeda M et al. Association study of genes and bipolar disorder. Ann Med 2005; 37: 196–205.
clock gene (CLOCK) and schizophrenia and mood 143. Baum AE, Akula N, Cabanero M et al. A genome-wide
disorders in the Japanese population. Eur Arch Psychiatry association study implicates diacylglycerol kinase eta
Clin Neurosci 2009; 259: 293–297. (DGKH) and several other genes in the etiology of
127. Benedetti F, Dallaspezia S, Fulgosi MC et al. Actimetric bipolar disorder. Mol Psychiatry 2008; 13: 197–207.
evidence that CLOCK 3111 T ⁄ C SNP influences sleep 144. Consortium WTCC. Genome-wide association study of
and activity patterns in patients affected by bipolar 14,000 cases of seven common diseases and 3,000 shared
depression. Am J Med Genet B Neuropsychiatr Genet controls. Nature 2007; 447: 661–678.
2007; 144B: 631–635. 145. Sklar P, Smoller JW, Fan J et al. Whole-genome associ-
128. Benedetti F, Serretti A, Colombo C et al. Influence of ation study of bipolar disorder. Mol Psychiatry 2008; 13:
CLOCK gene polymorphism on circadian mood fluctua- 558–569.
tion and illness recurrence in bipolar depression. Am J 146. McClung CA, Sidiropoulou K, Vitaterna M et al. Reg-
Med Genet B Neuropsychiatr Genet 2003; 123B: 23–26. ulation of dopaminergic transmission and cocaine reward

470
Circadian rhythms and sleep in BD

by the Clock gene. Proc Natl Acad Sci USA 2005; 102: 168. Czeisler CA, Wright KP Jr. Influence of light on circadian
9377–9381. rhythmicity in humans. In: Turek FW, Zee PC eds.
147. Roybal K, Theobold D, Graham A et al. Mania-like Regulation of Sleep and Circadian Rhythms. New York:
behavior induced by disruption of CLOCK. Proc Natl Marcel Dekker, 1999: 149–180.
Acad Sci USA 2007; 104: 6406–6411. 169. Frank E, Kupfer DJ, Ehlers CL et al. Interpersonal and
148. Falcon E, McClung CA. A role for the circadian genes in social rhythm therapy for bipolar disorder: Integrating
drug addiction. Neuropharmacology 2009; 56 (Suppl. 1): interpersonal and behavioural approaches. Behav Thera-
91–96. pist 1994; 17: 143–149.
149. Brown ES. Bipolar disorder and substance abuse. Psychi- 170. Miklowitz DJ, Otto MW, Frank E et al. Psychosocial
atr Clin North Am 2005; 28: 415–425. treatments for bipolar depression: a 1-year randomized
150. Abarca C, Albrecht U, Spanagel R. Cocaine sensitization trial from the Systematic Treatment Enhancement Pro-
and reward are under the influence of circadian genes and gram. Arch Gen Psychiatry 2007; 64: 419–426.
rhythm. Proc Natl Acad Sci USA 2002; 99: 9026–9030. 171. Benedetti F, Barbini B, Colombo C, Smeraldi E. Chro-
151. Andretic R, Chaney S, Hirsh J. Requirement of circadian notherapeutics in a psychiatric ward. Sleep Med Rev
genes for cocaine sensitization in Drosophila. Science 2007; 11: 509–522.
1999; 285: 1066–1068. 172. Wirz-Justice A, Benedetti F, Berger MA et al. Chrono-
152. Nestler EJ, Carlezon WA Jr. The mesolimbic dopamine therapeutics (light and wake therapy) in affective disor-
reward circuit in depression. Biol Psychiatry 2006; 59: ders. Psychol Med 2005; 35: 939–944.
1151–1159. 173. Wirz-Justice A, Benedetti F, Terman M. Chronothera-
153. Nestler EJ, Barrot M, DiLeone RJ, Eisch AJ, Gold SJ, peutics for Affective Disorders: A ClinicianÕs Manual for
Monteggia LM. Neurobiology of depression. Neuron Light & Wake Therapy. Basel: Karger, 2009.
2002; 34: 13–25. 174. Russell SJ, Browne JL. Staying well with bipolar disorder.
154. Lamont EW, Legault-Coutu D, Cermakian N, Boivin Aust NZ J Psychiatry 2005; 39: 187–193.
DB. The role of circadian clock genes in mental disorders. 175. Murray G, Suto M, Hole R, Hale S, Amari E, Michalak
Dialogues Clin Neurosci 2007; 9: 333–342. EE. Self-management strategies used by Ôhigh functioningÕ
155. Hampp G, Ripperger JA, Houben T et al. Regulation of individuals with bipolar disorder: From research to
monoamine oxidase A by circadian-clock components clinical practice. Clin Psychol Psychother 2010: doi:
implies clock influence on mood. Curr Biol 2008; 18: 678– 10.1002/cpp.710.
683. 176. Michalak EE, Murray G, Young AH, Lam RW. Burden
156. McClung CA. Circadian genes, rhythms and the biology of bipolar depression: impact of disease and medications
of mood disorders. Pharmacol Ther 2007; 114: 222–232. on quality of life. CNS Drugs 2008; 22: 389–406.
157. Kendler KS. ‘‘A gene for...’’: the nature of gene action in 177. Benedetti F, Barbini B, Campori E, Cigala Fulgosi M,
psychiatric disorders. Am J Psychiatry 2005; 162: 1243– Pontiggia A, Colombo C. Sleep phase advance and
1252. lithium to sustain the antidepressant effect of total sleep
158. Gottesman II, Gould TD. The endophenotype concept in deprivation in bipolar depression: new findings supporting
psychiatry: etymology and strategic intentions. Am J the internal coincidence model? J Psychiatr Res 2001; 35:
Psychiatry 2003; 160: 636–645. 323–329.
159. Lenox RH, Gould TD, Manji HK. Endophenotypes in 178. Vandewalle G, Balteau E, Phillips C et al. Daytime light
bipolar disorder. Am J Med Genet 2002; 114: 391–406. exposure dynamically enhances brain responses. Curr Biol
160. Papadimitriou GN, Calabrese JR, Dikeos DG, Christo- 2006; 16: 1616–1621.
doulou GN. Rapid cycling bipolar disorder: biology and 179. Anderson JL, Glod CA, Dai J, Cao Y, Lockley SW. Lux
pathogenesis. Int J Neuropsychopharmacol 2005; 8: 281– vs. wavelength in light treatment of Seasonal Affective
292. Disorder. Acta Psychiatr Scand 2009; 120: 203–212.
161. Hasler G, Drevets WC, Gould TD, Gottesman II, Manji 180. Terman M. Blue in the face. Sleep Med 2009; 10: 277–278.
HK. Toward constructing an endophenotype strategy for 181. Desan PH, Oren DA, Malison R et al. Genetic polymor-
bipolar disorders. Biol Psychiatry 2006; 60: 93–105. phism at the CLOCK gene locus and major depression.
162. Mitterauer B. Clock genes, feedback loops and their Am J Med Genet 2000; 96: 418–421.
possible role in the etiology of bipolar disorders: an 182. Sitaram N, Nurnberger JI Jr, Gershon ES, Gillin JC.
integrative model. Med Hypotheses 2000; 55: 155–159. Cholinergic regulation of mood and REM sleep: potential
163. Bunney WE, Bunney BG. Molecular clock genes in man model and marker of vulnerability to affective disorder.
and lower animals: possible implications for circadian Am J Psychiatry 1982; 139: 571–576.
abnormalities in depression. Neuropsychopharmacology 183. Knowles JB, Cairns J, MacLean AW et al. The sleep of
2000; 22: 335–345. remitted bipolar depressives: comparison with sex and
164. Walker MP, van der Helm E. Overnight therapy? The role age-matched controls. Can J Psychiatry 1986; 31: 295–
of sleep in emotional brain processing. Psychol Bull 2009; 298.
135: 731–748. 184. Millar A, Espie CA, Scott J. The sleep of remitted bipolar
165. Wagner U, Fischer S, Born J. Changes in emotional patients: a controlled naturalistic study using actigraphy.
responses to aversive pictures across periods rich in slow- J Affect Disord 2004; 80: 145–153.
wave sleep versus rapid eye movement sleep. Psychosom 185. Jones SH, Hare DJ, Evershed K. Actigraphic assessment
Med 2002; 64: 627–634. of circadian activity and sleep patterns in bipolar disorder.
166. Wehr TA, Goodwin FK. Circadian Rhythms in Psychi- Bipolar Disord 2005; 7: 176–186.
atry. Pacific Grove: Boxwood Press, 1983. 186. Jones SH, Tai S, Evershed K, Knowles R, Bentall R.
167. Indic P, Forger DB, St Hilaire MA et al. Comparison of Early detection of bipolar disorder: a pilot familial
amplitude recovery dynamics of two limit cycle oscillator high-risk study of parents with bipolar disorder and
models of the human circadian pacemaker. Chronobiol their adolescent children. Bipolar Disord 2006; 8: 362–
Int 2005; 22: 613–629. 372.

471
Murray and Harvey

187. Ankers D, Jones SH. Objective assessment of circadian Presented at the Australian Society for Bipolar Disorders
activity and sleep patterns in individuals at behavioural Conference 2007, Sydney, Australia, 20–22 September,
risk of hypomania. J Clin Psychol 2009; 65: 1071–1086. 2007.
188. Hallam KT, Olver JS, Chambers V, Begg DP, McGrath 201. Perlman CA, Johnson SL, Mellman TA. The prospective
C, Norman TR. The heritability of melatonin secretion impact of sleep duration on depression and mania.
and sensitivity to bright nocturnal light in twins. Psycho- Bipolar Disord 2006; 8: 271–274.
neuroendocrinology 2006; 31: 867–875. 202. Leibenluft E, Albert PS, Rosenthal NE, Wehr A.
189. Nurnberger JI Jr, Adkins S, Lahiri DK et al. Melatonin Relationship between sleep and mood in patients with
suppression by light in euthymic bipolar and unipolar rapid-cycling bipolar disorder. Psychiatry Res 1996; 63:
patients. Archiv Gen psychiatry 2000; 57: 572–579. 161–168.
190. Wood J, Birmaher B, Axelson D et al. Replicable differ- 203. Barbini B, Bertelli S, Colombo C, Smeraldi E. Sleep loss,
ences in preferred circadian phase between bipolar disor- a possible factor in augmenting manic episode. Psychiatry
der patients and control individuals. Psychiatry Res 2009; Res 1996; 65: 121–125.
166: 201–209. 204. Barbini B, Benedetti F, Colombo C et al. Dark therapy
191. Pandi-Perumal SR, Moscovitch A, Srinivasan V, Spence for mania: a pilot study. Bipolar Disord 2005; 7: 98–101.
DW, Cardinali DP, Brown GM. Bidirectional communi- 205. Wu JC, Kelsoe JR, Schachat C et al. Rapid and sustained
cation between sleep and circadian rhythms and its antidepressant response with sleep deprivation and chro-
implications for depression: lessons from agomelatine. notherapy in bipolar disorder. Biol Psychiatry 2009; 66:
Prog Neurobiol 2009; 88: 264–271. 298–301.
192. Gruber J, Harvey AG, Wang PW et al. Sleep functioning 206. Terman M. Evolving applications of light therapy. Sleep
in relation to mood, function, and quality of life at entry Med Rev 2007; 11: 497–507.
to the Systematic Treatment Enhancement Program for 207. Wirz-Justice A, Quinto C, Cajochen C, Werth E, Hock C.
Bipolar Disorder (STEP-BD). J Affect Disord 2009; 114: A rapid-cycling bipolar patient treated with long nights,
41–49. bedrest, and light. Biol Psychiatry 1999; 45: 1075–1077.
193. Riemann D, Voderholer U, Berger M. Sleep and sleep- 208. Padiath QS, Paranjpe D, Jain S, Sharma VK. Glycogen
wake manipulations in bipolar depression. Neuropsycho- synthase kinase 3b as a likely target for the action of
biology 2002; 45 (Suppl. 1): 7–12. lithium on circadian clocks. Chronobiol Int 2004; 21:
194. Hudson JI, Lipinski JF, Frankenburg FR, Grochocinski 43–55.
VJ, Kupfer DJ. Electroencephalographic sleep in mania. 209. Abe M, Herzog ED, Block GD. Lithium lengthens the
Arch Gen Psychiatry 1988; 45: 267–273. circadian period of individual suprachiasmatic nucleus
195. Hudson JI, Lipinski JF, Keck PE et al. Polysomnographic neurons. Neuroreport 2000; 11: 3261–3264.
characteristics of young manic patients. Arch Gen Psy- 210. Manji HK, Moore GJ, Chen G. Bipolar disorder: leads
chiatry 1992; 49: 378–383. from the molecular and cellular mechanisms of action of
196. Murray G. Seasonality, Personality and the Circadian mood stabilisers. Br J Psychiatry 2001; 178: S107–S119.
Regulation of Mood. New York: Nova Science, 2006. 211. Duncan JWC. Circadian rhythms and the pharmacology
197. Jackson A, Cavanagh J, Scott J. A systematic review of of affective illness. Pharmacol Ther 1996; 71: 253–312.
manic and depressive prodromes. J Affect Disord 2003; 212. Dokucu ME, Yu L, Taghert PH. Lithium- and valproate-
74: 209–217. induced alterations in circadian locomotor behavior in
198. Bauer M, Grof P, Rasgon N, Bschor T, Glenn T, Drosophila. Neuropsychopharmacology 2005; 30: 2216–
Whybrow PC. Temporal relation between sleep and mood 2224.
in patients with bipolar disorder. Bipolar Disord 2006; 8: 213. Buysse DJ, Frank E, Lowe KK, Cherry CR, Kupfer DJ.
160–167. Electroencephalographic sleep correlates of episode and
199. Colombo C, Benedetti F, Barbini B, Campori E, Smeraldi vulnerability to recurrence in depression. Biol Psychiatry
E. Rate of switch from depression into mania after 1997; 41: 406–418.
therapeutic sleep deprivation in bipolar depression. Psy- 214. Nofzinger EA, Schwartz RM, Reynolds CF III et al.
chiatry Res 1999; 86: 267–270. Affect intensity and phasic REM sleep in depressed men
200. Murray G, Judd F, Bullock B. Circadian and sleep ⁄ wake before and after treatment with cognitive-behavioral
variables as predictors of relapse in bipolar disorder. therapy. J Consult Clin Psychol 1994; 62: 83–91.

472

You might also like