You are on page 1of 16

Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

Contents lists available at ScienceDirect

Progress in Neuropsychopharmacology
& Biological Psychiatry
journal homepage: www.elsevier.com/locate/pnp

Depressive disorders: Processes leading to neurogeneration and potential T


novel treatments
Gregory M. Browna,⁎, Roger S. McIntyreb, Joshua Rosenblatc, Rüdiger Hardelandd
a
Department of Psychiatry, University of Toronto, Centre for Addiction and Mental Health, 250 College St. Toronto, ON M5T 1R8, Canada
b
Psychiatry and Pharmacology, University of Toronto, Mood Disorders Psychopharmacology Unit, University Health Network, 399 Bathurst Street, MP 9-325, Toronto,
ON M5T 2S8, Canada
c
Resident of Psychiatry, Clinician Scientist Stream, University of Toronto, Mood Disorders Psychopharmacology Unit, University Health Network, 399 Bathurst Street, MP
9-325, Toronto, ON M5T 2S8, Canada
d
Johann Friedrich Blumenbach Institut für Zoologie und Anthropologie, Universität Göttingen, Buergerstrasse 50, D-37073 Göttingen, Germany

A R T I C L E I N F O A B S T R A C T

Keywords: Mood disorders are wide spread with estimates that one in seven of the population are affected at some time in
Depression their life (Kessler et al., 2012). Many of those affected with severe depressive disorders have cognitive deficits
Cognition which may progress to frank neurodegeneration. There are several peripheral markers shown by patients who
Neurodegeneration have cognitive deficits that could represent causative factors and could potentially serve as guides to the
Mechanisms
prevention or even treatment of neurodegeneration. Circadian rhythm misalignment, immune dysfunction and
Inflammation
Chronotherapy
oxidative stress are key pathologic processes implicated in neurodegeneration and cognitive dysfunction in
depressive disorders. Novel treatments targeting these pathways may therefore potentially improve patient
outcomes whereby the primary mechanism of action is outside of the monoaminergic system. Moreover,
targeting immune dysfunction, oxidative stress and circadian rhythm misalignment (rather than primarily the
monoaminergic system) may hold promise for truly disease modifying treatments that may prevent neurode-
generation rather than simply alleviating symptoms with no curative intent. Further research is required to more
comprehensively understand the contributions of these pathways to the pathophysiology of depressive disorders
to allow for disease modifying treatments to be discovered.

1. Introduction Bipolar disorders are a group of disorders that present with both
episodes of major depression and episodes of mania or hypomania. A
1.1. Depressive disorders manic or hypomanic episode not only requires a persistently elevated or
irritable mood, but also a persistently increased level of goal directed
Despite numerous scientific advances, the diagnosis of psychiatric activity. In addition, transition from a depressed episode to a main or
disorders is still based on phenomenology (ie: the constellations of hypomanic episode during antidepressant treatment may be a diag-
symptoms displayed by the depressed individual). It had been pre- nostic criterion (Kaltenboeck et al., 2016). Lifetime prevalence has been
viously hoped that advances in a broad range of fields including brain estimated at about 1%.
imaging, genetics, epigenetics, non-coding RNAs, microRNAs and The diagnosis of Major Depressive Disorder (MDD, a prominent type
proteomics would lead to diagnostic markers but no markers have yet of Depressive Disorder) requires that there has been one or more Major
been shown to have clinical utility (Nemeroff et al., 2013). Currently Depressive Episodes (MDE) and the lifetime absence of mania or
psychiatric diagnosis is based on the Diagnostic and Statistical Manual of hypomania. A MDE requires a period of two weeks during which at
Mental Disorders (DSM-5), the latest version of the diagnostic manual least five of nine symptoms are present and completely excludes
published by the American Psychiatric Association (American episodes related to an exacerbation of a psychotic disorder such as
Psychiatric Association, 2013). Bipolar and related disorders have been schizophrenia (Uher et al., 2014).
separated from Depressive disorders in the DSM-5 because of some Despite the lack of diagnostic biological markers, biological findings
degree of phenomenological overlap between bipolar disorder and are frequently reported that are not yet accepted as diagnostic (Bilello
schizophrenia. et al., 2015; Rothschild, 2015). Several of these are found in MDD and


Corresponding author at: 100 Bronte Rd., Oakville, ON L6L 6L5, Canada.
E-mail addresses: gregm.brown@utoronto.ca (G.M. Brown), roger.mcintyre@uhn.ca (R.S. McIntyre), rhardel@gwdg.de (R. Hardeland).

http://dx.doi.org/10.1016/j.pnpbp.2017.04.023
Received 20 January 2017; Accepted 1 April 2017
Available online 20 April 2017
0278-5846/ © 2017 Elsevier Inc. All rights reserved.
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

in bipolar disorder and may cause or aggravate cognitive problems interfere with cognition e.g. hypothyroidism, diabetes, as well as
which in some cases could lead to neurodegeneration. Among the alcohol and other substance abuse (eg cannabis), is also warranted.
findings are alterations in circadian rhythms together with changes in Many antidepressants may also benefit measures of cognitive dysfunc-
melatonin profiles, immune dysfunction with increased cytokines and tion with relatively few demonstrating direct independent effects on
indicators of oxidative stress. The objective of this article is to review cognitive functions. A surfeit of other psychotropic agents e.g. stimu-
these findings, their possible pathophysiological roles and potential lants, anti-inflammatory agents, antioxidants, are currently being
treatment implications. explored as strategies for treatment. Behavioural approaches are
aerobic exercise as well as cognitive remediation therapy which are
2. Cognitive alterations also currently being evaluated (McIntyre et al., 2015b).

Cognitive dysfunction is a core dimension in MDD. In addition to 3. Neurodegenerative changes


being a criterion item e.g. difficulty in thinking and making decisions,
sub-domains and cognitive functions are implicated in anhedonia, Neurodegeneration is not a diagnostic criterion for MDD. However
psychomotor retardation, suicidality, and negativistic thought patterns there is evidence both from volumetric magnetic resonance imaging
e.g. hopelessness (McIntyre et al., 2013). Cognitive dysfunction is a (MRI) and functional MRI (fMRI), among others, for changes in brain
persistent feature in some individuals, and in sub-populations cognitive structure and function that indicate neurodegeneration in many
dysfunction is a progressive phenomenon. patients with MDD.
The common occurrence of both episodic and between-episode- It has been well documented in volumetric MRI studies that there
cognitive symptoms has heuristic as well as illness prognostication are decreases in prefrontal (especially orbital frontal) and anterior
implications. For example, depression can be conceptualized as a multi- cingulate cortex (ACC) volumes as well as in caudate nucleus, putamen
dimensional syndrome comprised of psychological disturbances across and hippocampus in patients with longstanding recurrent MDD (Bora
mood, cognitive and vegetative dimensions. In addition, disturbances in et al., 2012; Depping et al., 2016; Harrisberger et al., 2015; Jaworska
cognition are a core dimensional disturbance in depression (Insel et al., et al., 2016; Malykhin and Coupland, 2015; O'Connor and Agius, 2015;
2010). The multi-dimensional psychopathology of MDD is subserved by Sexton et al., 2013; Stratmann et al., 2014), although only women with
neurobiological changes that could be described as neural circuits and a first episode seemed to show evidence of decreased habenula volume
networks that are asynchronous in their reciprocity. (Carceller-Sindreu et al., 2015). These volumetric changes have been
From a clinical perspective, cognitive disturbances in depression are associated with histopathological abnormalities in post-mortem studies
a principle mediator in psychosocial impairment and work place (Price and Drevets, 2012). There is a decrease in glial cells and a
disability (McIntyre and Lee, 2016). For example, results from the corresponding increase in density of neurons in the same areas.
International Mood Disorders Collaborative Project indicate that cog- Evidence implicates that they form part of a functional circuit for
nitive disturbances account for more variability in workplace perfor- emotions in humans; hence it is theorized that these changes result in
mance than do the total depression symptoms (McIntyre et al., 2015a). abnormal functioning in MDD (Delvecchio et al., 2012; Price and
Further lines of evidence indicate that psychosocial recovery from an Drevets, 2010). Certain of these areas are also central to cognitive
indexed depressive episode is more strongly correlated with improve- dysfunction. The prefrontal cortex (PFC) is known to be important for
ment in cognitive measures rather than with changed depressive planning for or restraining from actions. Elevated activity in the PFC
symptom severity. and the ACC has been shown during non-affective cognitive tasks in
The domains of cognitive dysfunction that are affected in MDD MDD patients as compared to controls using fMRI (Kerestes et al.,
include executive function, memory and processing speed, as well as 2014). Emotional cognition, in contrast, seems to also involve area of
attention plus concentration (Roiser and Sahakian, 2013). Results from the amygdala and the hippocampus which show increased activation
a meta-analysis indicate that the magnitude of the deficit in cognitive with emotive negative visual challenges processing; effects which are
function in MDD is approximately 0.2–0.7 (Cohens d) commensurate typically reversed by antidepressants (Jaworska et al., 2014; Kerestes
with clinically significant cognitive impairment (Rock et al., 2014). et al., 2014). In addition poor performance on hippocampal-related
Conventional screening/rating instruments for depressive symptoms memory tasks in MDD has been reported to precede any changes in
[e.g. Patient health questionnaire 9 (PHQ-9)] do not provide sufficient hippocampal volume (Malykhin and Coupland, 2015). An optical
assessment of the cognitive symptoms of depression (Harrison et al., coherence tomographic study reported that the ganglion cell and inner
2016). More specifically, conventional depression methods rely on self- plexiform retinal layers were decreased in volume in those with
report of cognitive function which is not correlated with objective recurrent MDD as compared with first episode patients thus extending
measures of cognitive function and do not capture the complexity and findings to the retina (Kalenderoglu et al., 2016). The decreased
circumstances of how cognitive function affects patient-reported out- hippocampal volumes in MDD have been shown to be reversed by
comes (PROs) and individuals with MDD. The first sensitive, valid and antidepressant treatment leading to the suggestion that increasing
reliable screening tool for detecting cognitive dysfunction has just neurogenesis by antidepressants may play a role in increasing the
recently been made available: the THINC-it tool has been validated in hippocampal volume (Chi et al., 2015; Kalenderoglu et al., 2016).
adults 18–65 with MDD and is capable of detecting deficits in overall Several reports have indicated that antdepressants can normalize limbic
cognitive function. It is estimated that approximately 50% of adults PFC and ACC activity in MDD (Dusi et al., 2015; Qin et al., 2015; Wessa
with MDD presenting with single, as well as with multiple, episode and Lois, 2015).
depression have clinically significant cognitive impairment, as defined Studies of MDD patients using magnetic resonance diffusion tensor
by greater than 1 standard deviation (SD) below matched healthy imaging (DTI) have shown changes indicating altered connectivity in
control performance (THINC-it is available for a fee at http://thinc. several areas including the left superior longitudinal fasciculus, right
progress.im/en). caudate, corona radiate, genu of the corpus callusom, posterior
Potential treatments are reviewed in Sections 5 and 7. Mitigating thalamic radiations and brain stem (Choi et al., 2015; Han et al.,
cognitive deficits begins with prevention. For example, preventing 2017; Lener and Iosifescu, 2015; Liao et al., 2013; Murphy and Frodl,
episode recurrence and reducing episode severity and duration may 2011; Sexton et al., 2009; Tymofiyeva et al., 2017). Because of these
improve cognitive performance. Moreover, chronotherapeutic ap- numerous reports it has been proposed that depression may be a
proaches e.g. social rhythm therapy, and where appropriate, sedative syndrome with a disconnection between prefrontal and limbic areas
hypnotic agents may assist in some persons as sleep disruption is a well- (Liao et al., 2013).
known anti-cognitive behaviour. Targeting comorbid conditions that A recent positron emission tomography (PET) brain study of

190
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

patients with MDD has shown the first direct evidence of microglia intrinsically photosensitive ganglion cells that uniquely contain the
activation in MDD, activation which is known to be associated with photopigment melanopsin and coordinate the SCN rhythm with the
neurogeneration. Activation was found not only in the PFC, ACC and outside world (Berson, 2007; Gooley et al., 2003). In addition to
insula but in all brain areas studied (Setiawan et al., 2015). In the ACC entraining the SCN rhythm, light supresses synthesis of the hormone
activation was correlated with greater depression severity on the melatonin (Gooley et al., 2011; Lewy and Sack, 1989; Nathan et al.,
Hamilton rating scale. Reviews of magnetic resonance spectroscopy 1999). The SCN acts to synchronize rhythms in other tissues via neural,
(MRS) studies describe a variety of changes in metabolites which have autonomic and neuroendocrine pathways including the hormones
been associated with neuronal death and also abnormalities in neuro- cortisol and melatonin.
transmitter systems in some of the areas identified in MDD by MRI Depression has to be seen as a complex of several forms and
(Frodl and Amico, 2014; Lener and Iosifescu, 2015). Moreover circulat- subforms of disorders that differ with regard to their etiology.
ing inflammatory and antioxidant markers are associated with changes Therefore, an optimal therapy should not be reduced to a uniform
on MRI and MRS (Frodl and Amico, 2014; Lindqvist et al., 2014; Young symptomatic treatment, once the possible causes of the disorder
et al., 2014). Post-mortem neuropathological studies have shown become apparent. Consequently, a differential diagnosis that discrimi-
decreased neuronal cell size and quantity, synaptic density and glial nates between the forms of depressive disorders is of utmost impor-
cell quantity (Lener and Iosifescu, 2015). Whether these volumetric tance. This may not always be easy, with regard to some overlap of
changes represent a susceptibility to MDD, a result of or compensation symptoms, but there are, at least, several types of depression that can be
to MDD or even a response to treatment is not clearly established at this related to circadian dysfunction. Evidence for this conclusion is, in part,
time. based on the associations with mutations in genes of core and accessory
Studies using these and other techniques provide compelling elements of cellular circadian oscillators, such as Per2, Cry2, Bmal1
evidence of neurodegeneration, especially in patients with recurring, (= Arntl) and Npas2 in winter depression (Johansson et al., 2003;
longstanding MDD. A recent review describes the close relationship Lavebratt et al., 2010a, 2010b; Partonen et al., 2007; Rajendran and
between the neurodegenerative processes found in MDD and the Janakarajan, 2016), as well as Per3, Cry2, Bmal1 (Arntl), Bmal2 (Arntl2),
changes in the molecular processes involved in aging (Maurya et al., Clock, Dbp, Tim, CsnK1ε and NR1D1 (Rev-erbα; Ear1) in bipolar disorder
2016). Among other factors they include changes in telomere length, (Benedetti et al., 2008; Dallaspezia et al., 2016; Dallaspezia and
enzymatic antioxidant activities and inflammatory cytokines together Benedetti, 2015, 2011; Drago et al., 2015; Gonzalez et al., 2015;
with lower plasma concentrations of antioxidants. Kripke et al., 2009; Le-Niculescu et al., 2009; Mansour et al., 2006;
Nievergelt et al., 2006; Partonen, 2014; Sjöholm et al., 2010). While the
4. Circadian rhythm misalignment and melatonin alterations role of the circadian system was discovered relatively early in seasonal
affective and bipolar disorders, involvement in MDD appeared to be
Most body functions have a 24 h rhythm that is closely synchronized rather uncertain for quite some time. To date polymorphisms of Per3,
with the environment and allow optimal function in relation to the Cry1, Clock and Npas2 were reported to be associated with MDD (Hua
external world. These fluctuations are controlled by a master body et al., 2014; Shi et al., 2016; Soria et al., 2010a, b), whereas several
clock located in the suprachiasmatic nucleus of the hypothalamus other studies did not reveal significant findings. Moreover, time-of-
(SCN). Because of the ubiquity of this control it is not surprising that death expression levels of the oscillator genes Per1, Per2, Per3, Bmal1,
disrupted body rhythms would be present in most psychiatric disorders NR1D1, Dbp, Dec1 and Dec2 were shown to be changed in the brains of
(Boivin, 2000; Buzsáki and Watson, 2012; Byrne et al., 2014; Jones and MDD patients (Li et al., 2013). Another hint comes from an association
Benca, 2015). Circadian alterations in mood disorders are amply of a sirtuin 1 (Sirt1) variant with MDD (Kishi et al., 2010), a result of
documented and have been multiply reviewed very recently (Baron relevance because of the role of SIRT1 as an accessory oscillator
and Reid, 2014; Bellivier et al., 2015; Dallaspezia and Benedetti, 2015; component that is required for high circadian amplitudes (Bellet
Kripke et al., 2015; Martynhak et al., 2015; Nechita et al., 2015). MDD et al., 2011; Chang and Guarente, 2013; Nakahata et al., 2008).
has long been associated with alterations in body rhythms which range Nevertheless, the conclusions concerning MDD do not seem to be that
from major alterations in the sleep/ wake cycle and rhythmic changes firm as in the cases of seasonal affective and bipolar disorders. The
in activity and mood to alterations in rhythms of blood pressure, body reason may be sought in the divergence of MDD subforms with different
temperature, hormones (including cortisol and melatonin) and brain etiologies not all of which must be related to circadian dysfunction.
and body biochemistry (Albrecht, 2013; Jones and Benca, 2015; Uncertainties may also arise from the fact that alterations in the
McClung, 2013; Schnell et al., 2014). One of the diagnostic criteria of circadian oscillator system can be caused by deviations other than
MDD is various forms of insomnia or hypersomnia (Table 1). variant alleles. Among those which are not detected by polymorphism
Most body organs contain clocks that are synchronized to the screening, epigenetic modifications may play a significant role, as has
predominant cue, the light cycle, via the SCN while other lesser cues, been recently discussed (Liu and Chung, 2015). In fact, the circadian
including exercise and feeding, primarily affect muscles and liver system is modulated in manifold ways by epigenetic mechanisms
respectively. These clocks consist of some 10 or more clock genes (Hardeland, 2014a). The first evidence for epigenetic effects in the
which interact with each other to produce a rhythm with a period that circadian control by a microRNA (miR) was presented by Saus et al.
is close to 24 h (circadian) (Dibner et al., 2010; Hastings et al., 2014; (Saus et al., 2010), who described an abnormal processing of pre-miR-
Jin et al., 1999; Lowrey and Takahashi, 2004; Shearman et al., 2000). 182 in MDD. Recently, a deviating DNA methylation pattern was
Light cues reach the SCN via a pathway from a small population of discovered in the Bmal1 gene of patients with bipolar disorder
(Bengesser et al., 2016).
Table 1 Although such associations may only reveal the existence of risk
Circadian rhythm misalignment. factors and their genetic penetrance may sometimes be moderate, these
findings are insofar important, as they are not easily compatible with
• Rhythm desynchronization is frequent in depression
• Mutations of circadian rhythm genes are found in MDD and BD
the assumption put forward that mood disorders and circadian mal-
• Alterations in zeitgebers ie light cues, sleep schedule etc. may also cause
dysregulation
function have a common cause instead of resulting from circadian
disruption (Bechtel, 2015). In fact, the genetic argument that relates
• Altered
response
HPA regulation occurs with a recent focus on the cortisol awakening circadian malfunction to an affective disorder cannot be explained by
the same cause. Although the two deviations are, at first glance, only
• There are variable melatonin findings perhaps because light exposure varies
• Desynchronization of rhythms is pro-neurodegenerative in animals statistically associated, a mere correlation of changes in circadian
patterns with depressive disorders is insufficient for ruling out a causal

191
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

relationship. et al., 1985, 1984; Brown et al., 1985; Frazer et al., 1985; Khaleghipour
The cause or causes of circadian rhythm disruption in individual et al., 2012; Naismith et al., 2012; Souetre et al., 1989)although there
patients with MDD are stil not clear. And the extent to which circadian are also contrary reports (Bouwmans et al., 2015; Rubin et al., 1992;
misalignment is a cause or an effect of the mood disorder. That may Stewart and Halbreich, 1989; Thompson et al., 1988) plus a report of
well vary between individuals. It has been argued that disruption of increased am serum melatonin (Bumb et al., 2016). These differences
circadian rhythms aggravates MDD and that treatment of the disrup- may be related to the heterogeneity of MDD subforms.
tions may be helpful (Cardinali et al., 2011; Jones and Benca, 2015). Decreased levels of melatonin have been suggested to be a direct
Several factors, including the absence of, or a limited number of result of MDD, however it is also possible that the reported decrease
environmental zeitgebers, a delayed sleep schedule, absence of morning could be secondary to alterations in sleep patterns. In those patients
light, and receiving more light during the evening, especially when who spend less time in darkness because of late bedtime, being up at
these occur in combination, can disrupt the normal phase relationships night, or early wakening the presence of light would suppress melato-
between internal and external time keepers. Studies of sleep depriva- nin synthesis (Claustrat et al., 2005; Zawilska et al., 2009). It is clearly
tion, which keeps patients awake all or part of the night, have shown essential that exposure to light occurs in a healthy circadian way in
that it provides a rapid antidepressant action in 40 to 60% of patients order to maintain normal body rhythms including a normal nocturnal
with MDD which unfortunately may not be maintained (Bunney and melatonin rise (Bonmati-Carrion et al., 2014).
Bunney, 2013; Soria and Urretavizcaya, 2009). However, when supple- Circadian dysregulation is known to be a common feature of the
mented with both medication and sleep phase advance by bright light major neurodegenerative diseases: Alzheimer's, Huntington and
therapy (SPA) the response may be continued for up to several months. Parkinson disease. These disorders are all characterized by neurode-
In one study in post-mortem brain tissue it was shown that core clock generation and not by depression although they may result in depres-
gene expression shows robust 24 h rhythms in six brain regions in sive features. Alzheimer's disease is occasionally preceded by depres-
control subjects that were significantly disrupted in patients with MDD sion. Although it is not yet established whether the rhythm dysregula-
(Bunney et al., 2015). Most robust changes were seen in the ACC. The tion is cause or effect, it has been suggested that treatments targeting
authors speculate that the antidepressant response seen in MDD the sleep-wake cycle would be invaluable in these disorders (Videnovic
patients who respond to sleep deprivation is due to reset of the clock et al., 2014). All of these major neurodegenerative diseases are
genes to stabilize and synchronize body rhythms. characterized by aberrant protein aggregation. It has been postulated
Changes in the hypothalamic pituitary adrenal (HPA) axis regula- that several types of pro-neurodegenerative processes can be aggra-
tion in MDD have long been known. The SCN regulates this axis by vated by circadian rhythm disruption and that circadian treatments
synchronizing release of adrenocorticotrophic hormone (ACTH) from may be an important and novel approach (Barnard and Nolan, 2008;
the median eminence of the hypothalamus which in turn activates Hastings and Goedert, 2013).
synthesis and release of cortisol from the adrenal gland. As early as
1973 Sachar and colleagues reported on disrupted 24 h rhythms of 5. Potential treatments based on circadian rhythm
cortisol in patients with MDD (Sachar et al., 1973). In 1982 Carrol desynchronization
reported on abnormalities in the dexamethasone suppression test (DST)
in MDD speculating that it might be a diagnostic test (Carroll, 1982), The symptomatic relationship between circadian malfunction and
however numerous factors severely restrict its specificity (Herbert, mood disorders is a rather frequent phenomenon, which is not
2013). Serum cortisol levels, after correcting for time of day, are one surprising because of the circadian influence on sleep, since disturbed
part of a nine component test that has been proposed as a diagnostic sleep is a classic correlate of depression as well as of many other
procedure (Bilello et al., 2015; Papakostas et al., 2015), however it has psychiatric pathologies (Cardinali et al., 2011; Srinivasan et al., 2009b)
been questioned whether in its current form it would add any value to (Table 2).
the diagnosis of a trained physician (Rothschild, 2015). Recently On the background of these relationships between circadian dys-
attention has been focused on the cortisol awakening response, a rise function and, at least, subtypes of depressive disorders, several treat-
in cortisol that occurs 30 min after waking (Elder et al., 2014). This ment options can be designed. Since subtypes of depression with an
response has been shown to be exaggerated in those with MDD and etiology of circadian malfunction may be more easily treated than other
those at risk although exaggerated responses are also seen in a variety subforms and may, additionally, respond to relatively harmless inter-
of other conditions (Vreeburg et al., 2009; Vrshek-Schallhorn et al., ventions without or with rather mild side effects, it may be recom-
2013). This altered response been interpreted as a response to stress, a mended to first analyze whether circadian abnormalities are present. In
response that could affect the immune system. Alternatively it could the future, this may by done by screening for variants of clock genes.
represent an inherent change in HPA regulation as the 24 h rhythm of This should be possible on the basis of DNA in peripheral blood
cortisol would be altered by desynchronized body rhythms (Nicolaides mononuclear cells (PBMCs) obtained from blood samples, as is increas-
et al., 2014). ingly done in other polymorphism studies. Technological progress may
Synthesis of the hormone melatonin is controlled by the SCN via a allow these techniques to become an important diagnostic tool.
pathway which travels down through the brain stem and thence via the
sympathetic outflow to the pineal gland. Because it is regulated by Table 2
light, melatonin blood levels have been used as an index of the SCN Potential treatments based on rhythm dysregulation.
rhythm rising in the evening under dim light, being high only during
• Determine whether rhythm dysregulation is present
dark so that it peaks 6 to 8 h later and then falling to virtually
undetectable levels. Moreover melatonin serves as a cue to sleepiness;
• Three types of intervention are possible
o Light treatment in the morning
when administered in a low dose in the evening it will facilitate the o Blue blocking glasses in the evening
o Melatonin treatment in the evening

onset of sleep (Brzezinski et al., 2005; Kayumov et al., 2000).
Light treatment used successfully especially in winter depression and is also used
In bipolar patients nocturnal serum melatonin are reported as in combination treatments
decreased (Kennedy et al., 1996; Lam et al., 1990) although there is • Short acting melatonin useful to reset rhythms
some dissent (Whalley et al., 1991). There is also a report that morning • Long acting agonists ramelteon and agomelatine are very unlikely to reset
rhythms and normalize sleep
CSF melatonin is decreased (Bumb et al., 2016). An initial report of
• BB glasses were recently shown to be helpful additive treatment in mania
melatonin supersensitivity to light in bipolar patients (Lewy et al.,
1985) has been disputed (Whalley et al., 1991). Many groups have • Melatonin has variable effects on neuroinflammation and oxidative stress
depending on dose
reported decreased levels of nocturnal melatonin in MDD (Beck-Friis

192
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

Alternate tests on the basis of sleep deviations will be only meaningful those patients that exhibit a shortened rhythm as seen in a subpopula-
or sufficient, if they reveal substantial deviations in free-running period tion of bipolar disorder (Atkinson et al., 1975; Moreira and Geoffroy,
lengths or poor coupling to external time cues. If circadian rhythm sleep 2016). However, some bipolar patients display instead a lengthened
disorders (CRSDs) have been diagnosed, such as familial advanced sleep spontaneous period and are, therefore, nonresponders to lithium
phase syndrome (FASPS) or delayed sleep phase syndrome (DSPS), this therapy (Kripke et al., 1978). These differences shed light on the
should also suffice for trying a treatment of enforced circadian necessity of knowing details on the circadian deviations of a patient
entrainment. In all other cases, sleep difficulties cannot be taken as prior to treatment. Moreover, bright light treatment has also been used
an indication of an underlying circadian etiology, since the disturbance as an adjunctive therapy to enhance the effects by conventional
of sleep is a general phenomenon associated not only with affective as antidepressants. A recent meta-analysis of randomized trials revealed
well as several other disorders. efficacy of this approach in bipolar depression and MDD (Penders et al.,
In the case that a substantially aberrant circadian rhythm or 2016). An earlier systematic review had arrived at similar, but more
variants of oscillator genes have been detected, three modes of cautious conclusions, with regard to both light treatment as mono-
intervention are possible, by which the alignment of rhythms may be therapy and adjunctive therapy (Tuunainen et al., 2004). A retro-
improved. These three types of treatment may be even combined, respective meta-analysis of light therapy for the prevention of seasonal
because they may be applied in different phases of the circadian cycle. affective disorder revealed a relatively modest outcome (Nussbaumer
The first one is light treatment in the morning, the second one et al., 2015). However, the relatively poor demonstrable efficacy was
administration of melatonin or another melatonergic drug in the largely caused by methodological limits and also by the heterogeneity
evening or night and the third use of blue blocking glasses in the of cohorts.
evening or night. While light therapy is anyway devoid of pharmaco- If light therapy or BB glasses are capable of augmenting the efficacy
logical effects, melatonin is usually very well tolerated and side effects of other forms of chronotherapy as well as of antidepressant pharma-
or contraindications have to be only considered under certain specific cotherapy, this may also be possible with the use of melatonin or
conditions. synthetic melatonergic drugs. The phasing conditions of using both
Light therapy exists in several variations that are mostly conceived light and melatonin for entraining circadian rhythms have been worked
as methods of chronotherapy, to readjust the circadian system. Bright out (Skene, 2003). In fact, the combination of appropriately phased
light therapy is the most common procedure in this category. It has melatonin and bright light has already been used for symptomatic
been used especially for the treatment of seasonal affective disorder therapies in elderly subjects (der Lek et al., 2008), including Alzheimer
(Danilenko and Ivanova, 2015; Gordijn et al., 2012; Meesters et al., patients (Dowling et al., 2008), and for treating DSPS (Saxvig et al.,
2016, 2011; Oldham and Ciraulo, 2014; Pail et al., 2011; Prasko, 2008; 2014; Wilhelmsen-Langeland et al., 2013), whereas depressive disor-
Rastad et al., 2011), but also in bipolar disorder (Pail et al., 2011; ders still await respective studies on sufficiently large cohorts.
Prasko, 2008; Schwartz and Olds, 2015; Suzuki et al., 2016) and MDD The possible usefulness of melatonin, also as a monotherapy, is
(Al-Karawi and Jubair, 2016; Bais et al., 2016; Dridi et al., 2014; Eniola based on several considerations. Variants of the genes of melatonin
et al., 2016; Oldham and Ciraulo, 2014; Pail et al., 2011; Schwartz and biosynthesis, Aanat (aralkylamine N-acetyltransferase) and Asmt (N-
Olds, 2015), reportedly with a variable degree of therapeutic success. acetylserotonin O-methyltransferase) were reported to be associated
Light therapy as a nonpharmaceutical treatment has been recom- with MDD (Soria et al., 2010a) or recurrent depression (Galecki et al.,
mended and is being tested as a method avoiding any pharmacological 2010), respectively. However, decreases of melatonin levels were not
load during pregnancy and in perinatal depression (Bais et al., 2016; always found in the various forms of depressive disorders (Hardeland,
Crowley and Youngstedt, 2012). 2012a). Bipolar and seasonal affective disorders were reported to be
However, bright light therapy is perceived by some patients as associated with variants of the GPR50 gene (Delavest et al., 2012;
discomfort. Since the circadian pacemaker SCN receives photic infor- Macintyre et al., 2010; Thomson et al., 2005), which encodes a protein
mation mainly from melanopsin-containing retinal ganglion cells that with homology to melatonin receptors, but does not bind melatonin.
most strongly absorb in the visible blue range between 400 and 530 nm, However, it heterodimerizes with MT1 and thereby inhibits agonist
blue-enriched bright light or low-level blue light have been tested binding and Gi protein coupling (Hardeland, 2009a; Levoye et al.,
(Gordijn et al., 2012). Similar to white light, blue light causes phase 2006). Recently, the GPR50 gene was reported to be directly targeted
advances when administered in the early morning (Smith et al., 2009) by SIRT1 and to be, thus, associated with the modulation of circadian
and phase delays when given in the evening (Smith and Eastman, rhythms (Leheste and Torres, 2015).
2009). Although this concept seems to be chronobiologically well- An advantage of melatonin is the application in subjects that are not
founded, the use of blue light has given rise to concerns, because this susceptible to phase resetting by light, such as blind people without
light quality causes reductions in photoreceptor sensitivity as measured photoreception and others affected by degeneration of the SCN or its
in the ERG (Gagné et al., 2011). On the other hand blue blocking glasses neuronal connections. Moreover, one should take into account that
(BB) have revealed minimal concerns. BB glasses prevent the suppres- elderly persons frequently have a reduced sensitivity to blue light
sant effect of evening light on melatonin and improve sleep (Burkhart especially because of decreased crystalline lens transmission or pupil-
and Phelps, 2009; Kayumov et al., 2005; Sasseville et al., 2006). They lary miosis, changes that impair circadian photoreception by melanop-
have been used successfully in pilot studies in permanent night workers sin (Hardeland, 2012a). Circadian deviations such as dysphased
(Sasseville et al., 2009), shift workers (Rahman et al., 2013; Sasseville rhythmicity or absence of light entrainment have been demostrated in
and Hébert, 2010) and adolescents watching television (van der Lely a subgroup of blind individuals (Flynn-Evans et al., 2014; Lockley et al.,
et al., 2015). Recently BB glasses have been used as an additive 2007). In blind people and in other subjects with circadian disorders,
treatment of mania with a marked effect size beginning after three such as delayed sleep phase insomnia, melatonin was shown to be
days and no side effects differing from the placebo group (Henriksen effective in improving entrainment and sleep (Arendt et al., 1997;
et al., 2016). Other forms of chronotherapy such as dawn simulation Arendt and Skene, 2005; Lockley et al., 2000; Skene, 2003; Skene et al.,
(Terman and Terman, 2006), total sleep deprivation, or sleep phase 1996). In blind people, a low dose of 0.5 mg melatonin was found to be
advance, have been alternately used, but also chronotherapeutic sufficient (Hack et al., 2003).
combinations including bright light (Benedetti et al., 2014; Gottlieb Theoretically, there may be a category of nonresponders to light
and Terman, 2012; Wu et al., 2009). Additionally, combinations with therapy who are also insensitive or poorly sensitive to melatonin
lithium have been tested, procedures in which lithium also acts on the treatment, in terms of readjustment of circadian rhythms. This should
circadian system (Benedetti et al., 2014; Wu et al., 2009). Lithium has be assumed in three cases, (1) in advanced neurodegeneration of the
been long known to lenthen the circadian period, which is of value for SCN and/or its afferent pathways, (2) inborn dysfunction of circadian

193
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

oscillators because of severe mutations in oscillator genes, and (3) null phase of administration. The disregard of these fundamental chron-
mutations of both melatonin receptor genes. The first case is present in obiological aspects would only lead to inappropriate conclusions on
late stages of Alzheimer's disease, in which not only has melatonin inefficacy. A further point concerns the dose of melatonin. As outlined
strongly declined but also the precision of the circadian system is elsewhere (Hardeland, 2016), the conditions of circadian phase reset-
heavily impaired (Mishima et al., 1999). The two other cases have not ting do not follow the pharmacokinetic rules an average pharmacologist
been clearly demonstrated in humans. In this regard, one should take is familiar with. Especially, the area under curve (AUC) is not decisive
into account the multiplicity of parallel oscillators based on the in this case, where melatonin is rapidly taken up and soon attains a
alternate use of homologs and paralogs of oscillator components, which sufficient level of bioavailability. Notably, it is not an absolute long-
implies that the circadian system may not be completely lost in sustained level of the chronobiotic that conveys a synchronizing signal
mutants, but may persist in various cells and functions. Whether the but rather the velocity and extent of the change, in terms of the so-
third case may be either extemely rare or even lethal is unknown. Even called nonparametric resetting. In studies on entrainment of blind
if circadian rhythmicity is not corrected by melatonin, the pineal individuals, it has turned out that relatively small amounts of melatonin
hormone may not be entirely useless, but of some limited value, in (0.5 or 0.3 mg) are much more effective than higher doses such as
terms of improving symptoms and by providing some temporal 10 mg, which can fail to entrain (Lewy et al., 2006, 2005, 2002). Two
structure via direct actions not mediated by oscillators and through reasons can be responsible for the inefficacy of higher doses. Elevated
influencing peripheral oscillators. This was observed in Alzheimer levels of melatonin may either spill over into other parts of the PRC or
patients, in which melatonin was poorly effective on sleep, according may cause desensitization of oversaturated receptors. The latter possi-
to a large multicenter study (Singer et al., 2003), but still caused some bility has been studied in cell cultures, but there is some uncertainty
moderate improvements in cognitive functions as well as by reducing concerning its physiological or pharmacological relevance (Hardeland,
sundowning (Brusco et al., 1999; Cardinali et al., 2002; Srinivasan 2009a). The entraining capacity of very low melatonin levels is
et al., 2006). astonishing. In the extreme, even 0.025 mg melatonin have been shown
Melatonin monotherapy for treating depressive disorders requires to be sufficient for synchronizing a blind individual (Lewy, 2003). This
differentiated considerations. Although numerous publications have dose did not cause more than a brief spike of melatonin, a finding that
addressed the possibility of alleviating depressive symptoms in seasonal underlines the importance of the nonparametric, pulse-like and change-
affective disorder by readjusting the circadian system using melatonin, dependent characteristics of a well-operating sychronizing signal. These
the direct clinical evidence for this is remarkably scarce. Since the findings should be kept in mind when attempting a correction of
promising data of the early pilot study by Lewy et al. (Lewy et al., circadian rhythms to treat mood disorders with an etiology of circadian
1998), very little supporting evidence has been added. In patients of malfunction.
this category, melatonin was sometimes reported to mainly improve In bipolar disorder, in which circadian malfunction also seems to be
sleep parameters (Leppämäki et al., 2003) or to not be efficacious, in of relevance, the clinical basis for judging melatonin's efficacy is, again,
contrast to chronotherapy by sleep deprivation (Danilenko and Putilov, disappointingly small. Melatonin has been used in a couple of studies as
2005). There are several possible reasons for this poor outcome. First, add-on therapy, but these approaches cannot be taken for supporting
there have been difficulties in raising money for large state-of-the-art melatonin's general usefulness. Other studies not discussed here in
studies on the natural compound melatonin, which is not of sufficient detail concerned sleep improvements in these patients, effects that were
commercial interest to pharmaceutical companies. Second, a successful mainly restricted to sleep onset. In a case study (Robertson and
treatment aiming to correct circadian deviations has to first identify Tanguay, 1997), a child who was resistant to lithium, carbamazepine
these deviations, especially with regard to phasing and abnormal period and valproic acid responded well to melatonin, however, in combina-
length. As long as these alterations have not been assessed prior to tion with alprazolam. A small study on melatonin in patients with
treatment, a promising strategy to reentrain circadian rhythms may be rapid-cycling bipolar disorder arrived at the conclusion that melatonin
missed. Moreover, differences between patients concerning phase had no significant effects (Leibenluft et al., 1997). This finding is not
position and period length can completely obscure effects, since the surprising in the light of the dose dependence of circadian resetting, as
chronobiotic actions of melatonin follow a phase response curve (PRC), discussed above in the context of seasonal affective disorders, because
in which a silent zone without substantial phase shifts, a delay part, a those investigators applied a dose of 10 mg in the evening. Similar
transition phase and an advance part have to be distinguished (Lewy conclusions may be drawn for the use of melatonin in MDD. Three small
et al., 1992). Therefore, a schematic administration “shortly before add-on studies using between 5 and 10 mg slow-release melatonin did
bedtime” may completely miss the phase in which resetting is possible. not demonstrate substantial improvements (Dalton et al., 2000; Dolberg
It it recommended to determine a temporal marker within the circadian et al., 1998; Serfaty et al., 2010). It seems that the lack of chronobio-
cycle, using a measure such as the dim-light melatonin onset (DLMO). logical understanding is a major reason for unsuccessful treatments
Investigators should be aware that the baseline DLMO is typically found with melatonin. The high doses applied are poor in resetting, and slow-
in the silent zone (Lewy et al., 1992). Although melatonin may be release formulations may be additionally counterproductive, in terms of
capable of reducing sleep onset latency when given at that time, one inappropriately covering different parts of the PRC. If meaningful
should not expect to resynchronize circadian rhythms by melatonin in studies are to be conducted by aiming at readjustment of circadian
this phase. With regard to the extremely short half-life of melatonin rhythms, low doses of immediate-release melatonin have to be used in
(between 20 and 30, maximally 45 min), melatonin will have presum- suitable phases for either advancing or delaying the circadian rhythms,
ably disappeared from the circulation before a phase-shifting part of the depending on the deviations found in the individual. Especially in
PRC has been reached (Claustrat et al., 2005; Hardeland, 2009b). MDD, the prior assessment of presence or absence of circadian
Moreover, the investigator has to take into consideration whether the malfunction seems to be essential, since this complex of disorders is
pathologically disturbed rhythm exhibits an unusually extended or a not at all uniform in its etiology. Importantly, investigators have to
shortend period length, or whether the rhythms are phase-delayed or dismiss the idea that melatonin may be another directly acting
phase-advanced relative to the sleep/wake cycle; in other words, antidepressant drug. However, it may be useful by readjusting circadian
whether treatment requires phase advances or phase delays. Both clocks, if its indirect actions on subforms of depression are of relevance
changes have been observed in subgroups with winter depression. and considered in the design of studies.
The time points of melatonin administration, or bright light treatment, The same chronobiological limits of application have to be also
to achieve phase advances or phase delays have been determined (Lewy considered for synthetic melatonergic drugs. These other agonists have
et al., 2009). To assure reentrainment, it may be necessary to re- been mostly developed with regard to improving sleep and their design
determine the DLMO in the course of treatment and to re-adapt the aimed to extend the halflife compared to the rapidly catabolized

194
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

melatonin (Hardeland, 2016; Srinivasan et al., 2009a). However, such exists, since oscillator mutants have been shown to display enhanced
an extension does not seem to be an improvement with regard to short, molecular damage by free radicals (Hardeland et al., 2003), but
nonparametric signals required for circadian resetting, which is possi- oxidative stress can be also induced by numerous other pathological
ble with low doses of melatonin, whereas higher doses exhibit a processes. Some major sources of oxidants and reactive nitrogen
reduced resetting efficacy. The ramelteon metabolite M-II, which species, too, arise from inflammation, from mitochondrial dysfunction
displays melatonergic agonism, is extremely long-lived and attains and NADPH oxidase activation (Hardeland et al., 2015). Many pre-
blood concentrations much above those of the parent compound (Karim clinical data indicate that melatonin attenuates mitochondrial dysfunc-
et al., 2006). The receptor affinities of the approved synthetic drugs are tion and can downregulate NADPH oxidase activation, but the rele-
similar to those of melatonin or, especially in the case of ramelteon, vance to human pathophysiology, especially with regard to changes in
even higher (Hardeland, 2012b). Despite high receptor affinities and the depressive brain, are highly uncertain. Keeping in mind the usually
longer persistence in the blood, the recommended doses are usually high doses applied to suppress these sources of damage, one may be
much higher than the 2 or 3 mg present in melatonin pills, such as 8 or skeptic that the much lower amounts suitable for entrainment would
4 mg of ramelteon and 25 or 50 mg of agomelatine. These higher doses suffice. Perhaps, only damage caused by dysphased or misaligned
have been selected with regard to improvements of sleep and, in rhythms may be successfully treated that way, but presumably not that
agomelatine, for antidepressant actions. However, their suitability for by other, more severe causes. A further problem concerns the role of
phase resetting must be a matter of skepticism. Moreover, the question melatonin in the immune system. Although one can frequently read that
arises as to whether high amounts of synthetic drugs should be melatonin is an antiinflammatory agent, such a statement is absolutely
preferred over small doses of the extremely well tolerated natural inappropriate, since melatonin can alternately behave not only in an
compound melatonin. This is especially important in the case of antiinflammatory but also a proinflammatory way, in the latter case
agomelatine, which induces hepatotoxicity in a subpopulation of becoming a prooxidant compound (Hardeland et al., 2015), contrary to
patients (Hardeland, 2014b) and, therefore, requires surveillance. As the otherwise prevailing view that melatonin is generally an antiox-
it can be fatal, it is now recommended that it never be administered to idant. The known immune stimulatory actions comprise more upregu-
anyone with pre-existing liver damage, moreover early discontinuation lations of proinflammatory than antiinflammatory cytokines. However,
is recommended in any with suspected damage (Demyttenaere, 2011; many preclinical studies revealed a prevailing antiinflammatory action
Gahr et al., 2013; Gruz et al., 2014; Hardeland, 2014b; Montastruc in relation to aging and in response to strong inflammatory insults.
et al., 2014; Voican et al., 2014). Nevertheless, translation of these findings to the human remains
However, the antidepressant actions of agomelatine differ consider- uncertain. Proinflammatory actions of melatonin in the human have
ably from those of melatonin and several other melatonergic agonists, been observed in arthritis, in which the pineal hormone aggravated the
such as ramelteon. In addition to binding affinities to MT1 and MT2 in disease (Cutolo and Maestroni, 2005; Maestroni et al., 2005). For the
the range of those of melatonin, agomelatine displays the additional same reason, caution is likewise due in all autoimmune diseases.
property of an antagonist at 5-HT2C serotonin receptors. The affinity to Therefore, treatment with melatonin has some limits in the human.
5-HT2C is relatively moderate and may have been a reason for using
higher doses. The 5-HT2C antagonism has been interpreted as a basis of
6. Immunologic alterations and signs of oxidative stress
direct antidepressant actions (Millan et al., 2003). Later, an interaction
between melatonergic agonism and 5-HT2C inhibition has been sug-
Immune dysfunction is strongly associated with numerous psychia-
gested for explaining these effects (Racagni et al., 2011). In contrast to
tric and neurological conditions (Frick et al., 2013; Goldsmith et al.,
melatonin, numerous investigators have tested the antidepressant
2016b). Replicated evidence has demonstrated a strong association
actions of agomelatine in seasonal affective disorder, bipolar disorder
between elevated pro-inflammatory markers and both bipolar and
and MDD. Superiority was reported over several other drugs both as an
unipolar depression (Liu et al., 2012; Modabbernia et al., 2013;
antidepressant and in restoring sleep (Corruble et al., 2013;
Rosenblat et al., 2014; Rosenblat and McIntyre, 2016). This association
Guilleminault, 2005; Ivanov and Samushiya, 2014; Kasper et al.,
has been established by measuring central (i.e. cerebral spinal fluid
2013; Kennedy and Eisfeld, 2007; Martinotti et al., 2012; Owen,
(CSF)) and peripheral (i.e. blood serum levels) cytokine levels, compar-
2009). However, the findings have been multiply reviewed, with
ing subjects with and without mood disorders during periods of
variable judgments (Anonymous, 2013; Bourin and Prica, 2009;
depression, hypomania, mania and euthymia. To date, over fifty studies
Demyttenaere, 2011; Dolder et al., 2008; Eser et al., 2009, 2007;
have been conducted to determine and carefully characterize the
Goodwin, 2009; Guaiana et al., 2013; Howland, 2011a, 2011b;
associations between mood disorders and specific cytokine profiles
Kaminski-Hartenthaler et al., 2015; Kennedy, 2009; Koesters et al.,
(Liu et al., 2012; Modabbernia et al., 2013; Rosenblat et al., 2014;
2013; Langan et al., 2011; MacIsaac et al., 2014; Pandi-Perumal et al.,
Rosenblat and McIntyre, 2016). These studies have provided insights
2009; Singh et al., 2012; Taylor et al., 2014). While positive opinions
into the mechanistic underpinnings of the observed association between
prevailed especially in the earlier publications, an increasing number of
immune dysfunction and mood disorders. Further analysis of these
critical or negative conclusions were present later, especially in the
mechanisms and markers may also lead to the discovery of clinically
larger meta-analyses. The criticism also extended to methodological
relevant biomarkers along with novel treatment targets (Table 3).
problems and to publication bias. However it may be possible that
In a meta-analysis pooling together the results of twenty-nine
larger studies did not reveal overall positive effects, because of
heterogeneity of cohorts. Although such negative collective results
Table 3
may have contributed to the final conclusions in the meta-analyses, no Immunologic alterations and oxidative stress.
specific advantage of agomelatine can be deduced as far as the
correction of circadian dysfunction is concerned, and the contribution • Pro-inflammatory cytokines chronically elevated in both MDD and BD
o They are elevated more in acute states
• Oxidative/nitrosative
of 5-HT2C inhibition to antidepressant effects may be critically valued
stress (O & NS) is associated with mood disorders
as well, at the present state of knowledge. o Effective antidepressants are associated with decreased (O & NS)
As to the conclusion that efficient entrainment requires short
resetting signals and that, in the case of melatonin, rather low doses
• Pro-inflammatory markers are associated with poorer cognitive function
o The association is likely bidirectional
are sufficient, the question remains of whether other beneficial effects • Multiple pathways by which cytokines may influence monoamines
o Possibly via the recently identified brain lymphatic system
of melatonin can be also achieved under these conditions. This concerns
• Chronic migroglial overactivation may damage vital neural circuitry
especially the reduction of neuroinflammation and oxidative stress. A
relationship between oxidative damage and circadian rhythms likely
• Inflammation may up regulate the HPA axis
195
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

studies, a strong association between major depressive disorder (MDD) with current conventional therapies, there has been great interest in
and pro-inflammatory cytokines (in peripheral serum and CSF) was new targets to specifically and effectively alleviate cognitive dysfunc-
identified; interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) and tion in mood disorders. Immune dysfunction has presented as one
soluble IL-2 receptor (sIL-2R) were elevated in MDD subjects compared potential target of interest as pro-inflammatory markers have been
to health controls (Liu et al., 2012). Other inflammatory markers independently associated with poorer cognitive function in mood
associated with MDD include prostaglandin E2 (PGE2), acute phase disorders (Carvalho et al., 2014; Goldsmith et al., 2016a; Rosenblat
reactant C-reactive protein (CRP), IL-1β and IL-2(Felger and Lotrich, et al., 2015).
2013; Goldsmith et al., 2016b; McNamara and Lotrich, 2012). These While the association between pro-inflammatory markers, mood
studies have also suggested that pro-inflammatory cytokines are disorders and cognitive dysfunction has been well established, the
chronically elevated in MDD, even during euthymic periods; however, causative mechanisms underlying this association are still currently
pro-inflammatory markers appear to be more prominently and consis- being investigated (Rosenblat et al., 2014). Evidence from preclinical
tently elevated during major depressive episodes (MDEs). These results and clinical studies have suggested that the association is likely
suggest that depressive disorders are likely associated with a chronic bidirectional in that dysfunction of the innate immune system may
low grade inflammatory state with further perturbation of the innate predispose, precipitate and perpetuate depressive episodes and cogni-
immune system during mood episodes. tive dysfunction as well as vice versa (i.e. depressive episodes may
For bipolar disorder (BD), meta-analytic data pooling together the induce inflammatory changes) (McNamara and Lotrich, 2012; Miller
results of thirty studies revealed a strong association between BD and et al., 2009). Detailed descriptions of potential mechanisms sub-serving
higher serum levels of pro-inflammatory cytokines IL-4, TNF-α, sIL-2R, this bidirectional interaction along with the experimental evidence
IL-1β, IL-6, soluble receptor of TNF-alpha type 1 (STNFR1) and CRP in supporting these proposed mechanisms have been discussed extensively
BD subjects compared to healthy controls (Barbosa et al., 2014a; elsewhere (McNamara and Lotrich, 2012; Miller et al., 2009; Rosenblat
Modabbernia et al., 2013; Munkholm et al., 2013a). While there is et al., 2014). As such, these mechanisms will only be briefly summar-
some variability in cytokine levels during depressive, manic and ized herein.
euthymic periods, accumulating evidence indicates that peripheral Animal and human models have revealed several mechanisms
cytokine abnormalities are persistent, suggesting that BD is also whereby increased activity of the innate immune system may directly
associated with a chronic low-grade inflammatory state with potential impact mood and cognition. With the recent finding of functional
peaks of inflammation during depressive and manic episodes (Barbosa lymphatic vessels in the central nervous system (in an animal model)
et al., 2014a, 2014b, 2013, Brietzke et al., 2009a, 2009b, Munkholm (Louveau et al., 2015), further merit and biologic plausibility is added
et al., 2015, 2013a, 2013b). During periods of euthymia, sTNFR1 and to the potential direct interaction between immune dysfunction and
CRP are the only consistently elevated inflammatory markers (Barbosa neuropsychiatric pathology. The direct effect of cytokines on mono-
et al., 2014a, 2013; Brietzke et al., 2009a; Fernandes et al., 2016). amine levels serves as one key mechanism whereby inflammation may
During manic episodes, serum levels of CRP, IL-6, TNF-α, sTNFR1, IL- affect mood and cognition. Pro-inflammatory cytokines TNF-α, IL-2 and
RA, CXCL10, CXCL11, and IL-4 have been shown to be elevated IL-6 have been shown to directly alter monoamine levels (Capuron
(Barbosa et al., 2014a, 2014b, 2013; Liu et al., 2004). During depressive et al., 2003). IL-2 and interferon (IFN) increase the enzymatic activity
episodes, serum levels of CRP, sTNFR1 and CXCL10 are elevated of indolamine 2,3-dioxygenase (IDO), thus increasing the breakdown of
(Barbosa et al., 2014a, 2014b). tryptophan leading to decreased levels of serotonin and the production
Oxidative/nitrosative stress (O & NS) has also been associated with of depressogenic/anxiogenic tryptophan catabolites (TRYCATs) (Dunn
mood disorders and is intimately connected with immune dysregulation et al., 2005; Rosenblat et al., 2014). Serotonin levels may be further
as inflammation increases O & NS and O & NS increases inflammation modulated through the IL-6 and TNF-α dependent breakdown of 5-HT
(Gill et al., 2010; Lee et al., 2013; Moylan et al., 2014). The O & NS is to 5-hydroxyindoleacetic acid (5-HIAA) (Wang and Dunn, 1998; Zhang
defined as an imbalance between the production of reactive oxygen et al., 2001). Pro-inflammatory cytokines may also induce GTP
species (ROS) and production of antioxidants responsible for neutraliz- cyclohydrolase I (GPT-CH1) leading to alterations in dopamine and
ing ROS. Replicated evidence has demonstrated increased ROS and norepinephrine levels via the GTP pathway (Swardfager et al., 2016).
decreased antioxidants in both MDD and BD, leading to pathologic Over-activation of microglia had been identified as another key
neurodegeneration in key brain regions sub-serving mood and cogni- mechanism of interest (Stertz et al., 2013). Under physiological
tion (Aboul-Fotouh, 2013; Berk et al., 2011; Black et al., 2015; Palta conditions, microglia perform an important role in neuroplasticity,
et al., 2014). More specifically, depressive disorders have been asso- facilitating neural network pruning (Ekdahl, 2012; Harry and Kraft,
ciated with increased levels of pro-oxidant markers, namely, 8-hydroxy- 2012). Pruning of these pathways is essential for maintenance and
2′-deoxyguanosine (8-OHdG), F2-isoprostanes, malondialdehyde growth of more frequently utilized neural pathways (Harry and Kraft,
(MDA) and decreased levels of anti-oxidant molecules, namely, glu- 2012). However, the chronic inflammatory state associated with MDD
tathione (gamma-glutamyl-cysteinyl-glycine; GSH), superoxide dismu- and BD leads to chronic microglia over activation resulting in neuro-
tase (SOD) and glutathione peroxidase (GPx) (Maurya et al., 2016). degeneration by aberrantly destroying important neural pathways
Further, antidepressant response has been associated with decreased (Frick et al., 2013; Stertz et al., 2013). The microglial hypothesis has
O & NS, suggesting a mediational role of O & NS reduction in the been further supported by the previously discussed study by Setiawan
effective treatment of depression (Jimenez-Fernandez et al., 2015). As et al. (2015), which revealed (via PET imaging) increased microglial
such, there has been great interest in further understanding the activation in the ACC, PFC and insula in MDD subjects with a current
mechanisms sub-serving increased O & NS along with the potential MDE, as compared to healthy controls (Setiawan et al., 2015). Post-
novel drug targets these mechanisms may offer. mortem studies have also shown increased markers of inflammation and
Of increasing interest has also been the association between microglial activation in these key brain region (Bezchlibnyk et al.,
immune dysfunction, oxidative stress and specific trans-diagnostic 2001; Rao et al., 2010). The over-activation of microglia also increases
symptoms, such as cognitive dysfunction and anhedonia (Anderson local O & NS, further damaging neural circuitry in key brain regions
et al., 2014; Bauer et al., 2014; Rosenblat et al., 2015; Swardfager et al., sub-serving mood and cognition (Kraft and Harry, 2011; Stertz et al.,
2016). As discussed above, cognitive dysfunction is common and leads 2013).
to significant functional impairment in depressive disorders. Cognitive Another key mechanism whereby inflammation may induce mood
dysfunction often persists throughout euthymic periods as these dysfunction in MDD and BD is HPA axis dysregulation as discussed
symptoms often do not improve, even with the effective treatment of above in Section 4. Increased levels of pro-inflammatory cytokines, IFN,
mood symptoms. Given that cognitive symptoms often do not improve TNF-α and IL-6, up-regulate HPA activity thereby increasing systemic

196
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

cortisol levels leading to hypercortisolemia (Beishuizen and Thijs, adjunctive anti-inflammatories on depressive symptom severity was
2003; Harrison et al., 2009; Wright et al., 2005). Additionally, elevated − 0.40 (p = 0.002), indicative of a moderate and statistically signifi-
levels of inflammatory cytokines decrease glucocorticoid receptor cant antidepressant effect. Heterogeneity of the pooled sample was
synthesis, transport and sensitivity in the hypothalamus and pituitary notably low (I2 = 14%; p = 0.32). Additionally, no manic/hypomanic
(Pace and Miller, 2009; Turnbull and Rivier, 1999). Therefore, the induction or significant treatment emergent adverse events were
negative feedback loop, which usually down-regulates cortisol produc- reported. Of the included agents, adjunctive NAC, an anti-inflammatory
tion, is disabled thereby leading to chronic hypercortisolemia (Pace and and anti-oxidant agent, was shown to have the greatest antidepressant
Miller, 2009; Turnbull and Rivier, 1999). Along with alterations in the effect in BD subjects (Berk et al., 2012, 2008). In a large placebo-
HPA axis, the brain-gut-microbiota axis may also be affected. The brain- controlled, RCT, adjunctive NAC was shown to lower depression scores
gut-microbiota axis has been of increasing interest as a potential throughout the trial with a statistically significant difference compared
bidirectional pathway perpetuating depressive disorders via immune to the placebo group by the primary endpoint of 24 weeks (Berk et al.,
dysfunction (Bercik, 2011; Cryan and Dinan, 2012). 2008). Of note, Soares et al. are currently conducting a phase 2, double-
blind RCT of ASA and NAC as adjunctive treatment for BD
7. Potential treatments based on immunologic changes (NCT01797575).
Interest has recently grown in the use of monoclonal antibody based
With the well-established association between immune dysfunction cytokine inhibitors as these agents may be engineered to specifically
and mood disorders, the immune system serves as a novel target in the target cytokines implicated in the inflammatory-mood pathway.
treatment of depression. Selecting an upstream target (i.e. immune Infliximab (anti-TNF-α) and sirukumab (anti-IL-6) have been of parti-
dysfunction) rather than the downstream effects, such as monoamine cular interest. One key RCT assessed infliximab in treatment resistant
alterations, may allow for potentially improved outcomes and addi- depression (including BD and MDD subjects) (Raison et al., 2013).
tional treatment options in treatment resistant populations. Several Overall, no greater antidepressant effect of infliximab compared to
clinical trials have already been conducted evaluating the antidepres- placebo was observed; however, a significant antidepressant effect was
sant effects of anti-inflammatory agents in a variety of populations, observed for a subgroup of subjects with elevated levels of serum CRP
including but not limited to MDD, bipolar depression and sub-syndro- and TNF-α (Raison et al., 2013). The results of this trial were of
mal depressive symptoms (Kohler et al., 2014; Rosenblat et al., 2016). particular interest as they suggested that stratification using inflamma-
Anti-inflammatory and anti-oxidant agents such as non-steroidal anti- tory biomarkers might help determine which patients may benefit from
inflammatories (NSAIDs), acetylsalicylic acid (ASA), minocycline, N- anti-inflammatory therapies and called into question the validity of
acetylcysteine (NAC) and biologic cytokine inhibitors (e.g. monoclonal previous negative RCTs that did not stratify subjects based on inflam-
antibody based agents such as infliximab) have been investigated for matory status. Currently, a 12-week multisite, double-blind RCT
their antidepressant effects (Rosenblat et al., 2014). Nutraceuticals with evaluating the efficacy, safety, and tolerability of adjunctive infliximab
anti-inflammatory properties, such as omega-3 poly-unsaturated fatty for the treatment of BD subjects with an elevated serum CRP is
acids (omega-3 s) and curcumin, have also been investigated (Bergman underway to a priori further characterize the utility of inflammatory
et al., 2013; Bloch and Hannestad, 2012; Lopresti et al., 2014) stratification (NCT02363738). Sirukumab is also being currently eval-
(Table 4). uated in a similarly designed RCT for MDD with sample stratification
A recent meta-analysis of anti-inflammatory agents, excluding based on CRP levels (NCT02473289). Results from future, stratified
nutraceuticals, in the treatment of depression and depressive symptoms RCTs of cytokine inhibitors may lead to novel treatments and poten-
identified ten randomized controlled trials (RCTs) evaluating the use of tially move the field of psychiatry forward to a more personalized
NSAIDs (n = 4258) and four RCTs investigating cytokine inhibitors medicine approach while simultaneously providing significant insight
(n = 2004) (Kohler et al., 2014). The pooled effect size (standard mean into the pathophysiology of depression.
difference (SMD) of change in depression severity) suggested that anti- The use of naturally occurring anti-inflammatory agents in the
inflammatory treatment reduced depressive symptom severity treatment of BD and MDD has remained of great interest; however, use
(SMD = − 0.34; p = 0.004) compared with placebo. This effect was has been increasingly controversial due to the significant variability of
observed in studies including subjects with depression (SMD = −0.54) RCT results and presumed publication bias (Bloch and Hannestad,
and depressive symptoms (SMD = − 0.27). Sub-analyses suggested 2012; Sarris et al., 2012). Curcumin and omega-3 s have been of
that the antidepressant effect of celecoxib (selective cyclooxygenase 2 greatest interest. Curcumin, an age-old spice, extracted from turmeric
inhibitor) was more reproducible compared to other agents studied. (Curcuma longa) has long been used in complementary and alternative
Notably, meta-analysis of adverse effects suggested good tolerability medicine for its anti-oxidant and anti-inflammatory properties. More
with no evidence of an increased number of infections, gastrointestinal recently, clinical and preclinical studies have shown promising results
or cardiovascular events. for an antidepressant effect with good tolerability associated with daily
In another recent meta-analysis, the use of anti-inflammatory agents curcumin administration (Kaufmann et al., 2015; Sanmukhani et al.,
in the treatment of bipolar depression was assessed (Rosenblat et al., 2014). Most recently, in a RCT (n = 123), MDD subjects were allocated
2016). Eight RCTs (n = 312) assessing adjunctive NSAIDs (n = 53), to one of four treatment conditions, comprising placebo, low-dose
omega-3 s (n = 140), NAC (n = 76) and pioglitazone (n = 44) in the curcumin extract (250 mg twice daily), high-dose curcumin extract
treatment of BD were identified. The overall effect size (SMD) of (500 mg twice daily), or combined low-dose curcumin extract plus
saffron (15 mg twice daily) for 12 weeks (Lopresti and Drummond,
Table 4 2017). The active drug treatments (combined) were associated with
Potential treatments based on immunologic changes. significantly greater improvements in depressive symptoms compared
to placebo (p = 0.03). Active drug treatments also had greater efficacy
• Anti-inflammatory agents, especially celecoxib reduce depression
• Anti-inflammatory
in subjects with atypical depression compared to the remainder of
agents, especially NAC reduce depression in BP
• action
Monoclonal antibody based cytokine inhibitor Infliximab have an antidepressant
in those with elevated CRP and TNF-α
subjects (response rates of 65% versus 35% respectively, p = 0.01). No
differences were found between the differing doses of curcumin or the
• InCurcumin and curcumin/saffron combination has antidepressive action especially
those with atypical depression
curcumin/saffron combination.
Several RCTs have been conducted evaluating the effect of omega-
• remission
An Omega-3 study reported that those with high inflammation have a marked
rate with EPA (eicosapentaenoic acid)
3 s for the acute treatment of depressive symptoms, MDD and BD (Bloch
• Immune modulating agents may only benefit those with immune dysfunction. and Hannestad, 2012; Sarris et al., 2012). Earlier studies suggested a
robust antidepressant effect in both MDD and BD, however, more recent

197
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

studies have been negative (Keck et al., 2006) with recent meta- tory agents show great promise in the treatment of depression in BD and
analyses suggesting a minimal antidepressant effect with significant MDD. What has been discovered is that stratification of patients
publication bias skewing the interpretation of results (Bloch and according to immune status can be important as agents may only
Hannestad, 2012; Sarris et al., 2012). Recently, Rapaport et al. benefit those with immune dysfunction. Further studies will clarify
(Rapaport et al., 2016) successfully predicted antidepressant response these issues.
to omega-3s in MDD by stratifying subjects based on cytokine profiles. We are at a tantalizing point in these investigations. We have much
Based on five inflammatory biomarkers (IL-ra, IL-6, hs-CRP, leptin and information on these processes but insufficient knowledge on key
adiponectin), subjects were separated into two categories, namely, issues. However, there are many, many tools being used to resolve
‘high’ and ‘low’ inflammation. Subjects with high inflammation had a these issues and several very competent groups studying them so that
marked response to eicosapentaenoic acid (EPA) with a 40% remission the answers will surely come.
rate compared to a 25% remission rate in the placebo group. Interest-
ingly, in the low inflammation category, placebo outperformed EPA Funding
with a 44% and 19% remission rate respectively. As such, combining all
subjects, there was no difference in antidepressant effect comparing This work did not receive any specific grant from funding agencies
EPA with placebo. This study highlighted the importance of stratifica- in the public, commercial, or not-for-profit sectors.
tion of samples and provided some insight into a significant potential
cause of heterogeneity in omega-3 RCTs and in anti-inflammatory References
depression studies in general.
Taken together, several anti-inflammatory agents show great pro- Aboul-Fotouh, S., 2013. Coenzyme Q10 displays antidepressant-like activity with
mise in the treatment of depression in BD and MDD. Several RCTs are reduction of hippocampal oxidative/nitrosative DNA damage in chronically stressed
rats. Pharmacol. Biochem. Behav. 104, 105–112. http://dx.doi.org/10.1016/j.pbb.
currently underway as the field of immuno-psychiatry rapidly pro- 2012.12.027.
gresses. The validity and clinical applicability of future studies will be Al-Karawi, D., Jubair, L., 2016. Bright light therapy for nonseasonal depression: meta-
much greater than previous studies if sample stratification (i.e. analysis of clinical trials. J. Affect. Disord. 198, 64–71.
Albrecht, U., 2013. Circadian clocks and mood-related behaviors. [review]. Handb. Exp.
separating sample based on inflammatory status) is implemented. Pharmacol. 217, 227–239.
Previous clinical trials assessing the antidepressant effects of immune American Psychiatric Association, 2013. Diagnostic and Statistical Manual of Mental
modulating agents assumed the traditional approach of administering a Disorders, Fifth Edition (DSM-5), fifth ed. American Psychiatric Association,
Washington, DC.
test drug to subjects stratified solely by DSM diagnosis would be Anderson, G., Berk, M., Dean, O., Moylan, S., Maes, M., 2014. Role of immune-
adequate to discover novel antidepressant agents. However, more inflammatory and oxidative and nitrosative stress pathways in the etiology of
recently, a more personalized medicine approach has been encouraged depression: therapeutic implications. CNS Drugs 28, 1–10. http://dx.doi.org/10.
1007/s40263-013-0119-1.
as it is now recognized that immune dysfunction is an etiological factor
Anonymous, 2013. Agomelatine: a review of adverse effects. Prescrire Int. 22, 70–71.
only in a subset of mood disorder patients. As such, immune modulating Arendt, J., Skene, D.J., 2005. Melatonin as a chronobiotic. Sleep Med. Rev. 9, 25–39.
agents may potentially be only of benefit to patients with immune Arendt, J., Skene, D.J., Middleton, B., Lockley, S.J., Deacon, S., 1997. Efficacy of
dysfunction. Including subjects with normal immune function in melatonin treatment in jet lag, shift work, and blindness. J. Biol. Rhythm. 12,
604–617.
clinical trials may obscure the results and could lead to invalidly Atkinson, M., Kripke, D.F., Wolf, S.R., 1975. Autorhythmometry in manic-depressives.
negative studies, as was demonstrated by the previously discussed Chronobiologia 2, 325–335.
proof-of-concept studies of infliximab (Raison et al., 2013) and EPA Bais, B., Kamperman, A.M., van der Zwaag, M.D., Dieleman, G.C., Harmsen van der Vliet-
Torij, H.W., Bijma, H.H., Lieverse, R., Hoogendijk, W.J., Lambregtse-van den Berg,
(Goldsmith et al., 2016b). M.P., 2016. Bright light therapy in pregnant women with major depressive disorder:
study protocol for a randomized, double-blind, controlled clinical trial. BMC
8. Conclusions Psychiatry 16, 381.
Barbosa, I.G., Bauer, M.E., Machado-Vieira, R., Teixeira, A.L., 2014a. Cytokines in bipolar
disorder: paving the way for neuroprogression. Neural Plast. http://dx.doi.org/10.
The reason for writing this article was to delineate the ways in 1155/2014/360481.
which MDD leads to neurodegeneration and to point the way to Barbosa, I.G., Machado-Vieira, R., Soares, J.C., Teixeira, A.L., 2014b. The immunology of
bipolar disorder. Neuroimmunomodulation 21, 117–122. http://dx.doi.org/10.
practical treatments We have not yet completed that task as there are
1159/000356539.
still many unknowns. However, we have a good idea of the remaining Barbosa, I.G., Rocha, N.P., Bauer, M.E., de Miranda, A.S., Huguet, R.B., Reis, H.J.,
gaps. Zunszain, P.A., Horowitz, M.A., Pariante, C.M., Teixeira, A.L., 2013. Chemokines in
bipolar disorder: trait or state? Eur. Arch. Psychiatry Clin. Neurosci. 263, 159–165.
We know that in depressive disorders as a group and in fact in many
http://dx.doi.org/10.1007/s00406-012-0327-6.
psychiatric disorders there is oftentimes a dysjunction between body Barnard, A.R., Nolan, P.M., 2008. When clocks go bad: neurobehavioural conseuences of
rhythms. It is understandable that a tight coordination of brain, disrupted ccircadian timing. PLoS Genet. 4, 1–8.
let alone body procceses, is essential for normal functioning of such a Baron, K.G., Reid, K.J., 2014. Circadian misalignment and health. Int. Rev. Psychiatry 26,
139–154. http://dx.doi.org/10.3109/09540261.2014.911149.
complicated being as a human. Much more knowledge is needed about Bauer, I.E., Pascoe, M.C., Wollenhaupt-Aguiar, B., Kapczinski, F., Soares, J.C., 2014.
these processes including not only how they go awry but also how to Inflammatory mediators of cognitive impairment in bipolar disorder. J. Psychiatr.
correct them in MDD. MDD patients differ widely from one another in Res. 56, 18–27. http://dx.doi.org/10.1016/j.jpsychires.2014.04.017.
Bechtel, W., 2015. Circadian rhythms and mood disorders: Are the phenomena and
the type of rhythm disorder that they may manifest. Several strategies mechanisms causally related? Front. Psychiatry 6, 118.
for enforcing circadian entrainment are now known but require that the Beck-Friis, J., Kjellman, B.F., Aperia, B., Unden, F., von Rosen, D., Ljunggren, J.G.,
type of rhythm dysfuntion is known. Sleep abnormalities will be only Wetterberg, L., 1985. Serum melatonin in relation to clinical variables in patients
with major depressive disorder and a hypothesis of a low melatonin syndrome. Acta
helpful if they reveal substantial deviations in free-running period Psychiatr. Scand. 71, 319–330.
lengths or poor coupling to external time cues. Of course, if a circadian Beck-Friis, J., Kjellman, B.F., Ljunggren, J.-G., Wetterberg, L., 1984. The pineal gland and
rhythm sleep disorder has been diagnosed, this will suffice for trying a melatonin in affective disorders. In: Brown, G.M., Wainwright, S.D. (Eds.), The Pineal
Gland: Endocrine Aspects, Advances in the Biosciences. Pergamon Press, Oxford, pp.
treatment of enforced circadian entrainment. In the future studies of
313–325.
variants of clock genes may be helpful. Beishuizen, A., Thijs, L.G., 2003. Endotoxin and the hypothalamo-pituitary-adrenal (HPA)
We know that MDD and especially recurrent MDD causes measur- axis. J. Endotoxin Res. 6, 3–24. http://dx.doi.org/10.1179/096805103125001298.
Bellet, M.M., Orozco-Solis, R., Sahar, S., Eckel-Mahan, K., Sassone-Corsi, P., 2011. The
able cognitive problems that are accompanied by immune dysregula-
time of metabolism: NAD +, SIRT1, and the circadian clock. Cold Spring Harb. Symp.
tion and increases in oxidative/nitrosative processes in the brain and Quant. Biol. 76, 31–38.
periphery that could lead to neurodegeneration. Why should that be the Bellivier, F., Geoffroy, P., Etain, B., Scott, J., 2015. Sleep- and circadian
case? What is the underlying mechanism that causes it to happen? What rhythm—associated pathways as therapeutic targets in bipolar disorder. Expert Opin.
Ther. Targets 19, 747–763.
is the optimal method to correct the problems? Several anti-inflamma-

198
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

Benedetti, F., Dallaspezia, S., Colombo, C., Pirovano, A., Marino, E., Smeraldi, E., 2008. A Bunney, B., Li, J., Walsh, D., Stein, R., Vawter, M., Cartagena, P., Barchas, J., Schatzberg,
length polymorphism in the circadian clock gene Per3 influences age at onset of A., Myers, R., Watson, S., Akil, H., Bunney, W., 2015. Circadian dysregulation of
bipolar disorder. Neurosci. Lett. 445, 184–187. http://dx.doi.org/10.1016/j.neulet. clock genes: clues to rapid treatments in major depressive disorder HHS Public
2008.09.002. Access. Mol. Psychiatry 20138, 48–55. http://dx.doi.org/10.1038/mp.2014.138.
Benedetti, F., Riccaboni, R., Locatelli, C., Poletti, S., Dallaspezia, S., Colombo, C., 2014. Bunney, B.G., Bunney, W.E., 2013. Mechanisms of rapid antidepressant effects of sleep
Rapid treatment response of suicidal symptoms to lithium, sleep deprivation, and deprivation therapy: clock genes and circadian rhythms. Biol. Psychiatry 73,
light therapy (chronotherapeutics) in drug-resistant bipolar depression. J. Clin. 1164–1171. http://dx.doi.org/10.1016/j.biopsych.2012.07.020.
Psychiatry 75, 133–140. Burkhart, K., Phelps, J.R., 2009. Amber lenses to block blue light and improve sleep: a
Bengesser, S.A., Reininghaus, E.Z., Lackner, N., Birner, A., Fellendorf, F.T., Platzer, M., randomized trial. Chronobiol. Int. 26, 1602–1612. http://dx.doi.org/10.3109/
Kainzbauer, N., Tropper, B., Hormanseder, C., Queissner, R., Kapfhammer, H.-P., 07420520903523719.
Wallner-Liebmann, S.J., Fuchs, R., Petek, E., Windpassinger, C., Schnalzenberger, M., Buzsáki, G., Watson, B.O., 2012. Brain rhythms and neural syntax: implications for
Reininghaus, B., Evert, B., Waha, A., 2016. Is the molecular clock ticking differently efficient coding of cognitive content and neuropsychiatric disease. Dialogues Clin.
in bipolar disorder? Methylation analysis of the clock gene ARNTL. World J. Biol. Neurosci. 14, 345–367. http://dx.doi.org/10.1097/ALN.0b013e318212ba87.
Psychiatry 14, 1–9. Byrne, E.M., Heath, A.C., Madden, P.A.F., Pergadia, M.L., Hickie, I.B., Montgomery, G.W.,
Bercik, P., 2011. The microbiota-gut-brain axis: learning from intestinal bacteria? Gut 60, Martin, N.G., Wray, N.R., 2014. Testing the role of circadian genes in conferring risk
288–289. http://dx.doi.org/10.1136/gut.2010.226779. for psychiatric disorders. Am. J. Med. Genet. B Neuropsychiatr. Genet. 165, 254–260.
Bergman, J., Miodownik, C., Bersudsky, Y., Sokolik, S., Lerner, P.P., Kreinin, A., http://dx.doi.org/10.1002/ajmg.b.32230.
Polakiewicz, J., Lerner, V., 2013. Curcumin as an add-on to antidepressive treatment: Capuron, L., Neurauter, G., Musselman, D.L., Lawson, D.H., Nemeroff, C.B., Fuchs, D.,
a randomized, double-blind, placebo-controlled, pilot clinical study. Clin. Miller, A.H., 2003. Interferon-alpha-induced changes in tryptophan metabolism.
Neuropharmacol. 36, 73–76. http://dx.doi.org/10.1097/WNF.0b013e31828ef969. Relationship to depression and paroxetine treatment. Biol. Psychiatry 54, 906–914.
Berk, M., Copolov, D.L., Dean, O., Lu, K., Jeavons, S., Schapkaitz, I., Anderson-Hunt, M., Carceller-Sindreu, M., de Diego-Adeliño, J., Serra-Blasco, M., Vives-Gilabert, Y., Martín-
Bush, A.I., 2008. N-acetyl cysteine for depressive symptoms in bipolar disorder—a Blanco, A., Puigdemont, D., Álvarez, E., Pérez, V., Portella, M.J., 2015. Volumetric
double-blind randomized placebo-controlled trial. Biol. Psychiatry 64, 468–475. MRI study of the habenula in first episode, recurrent and chronic major depression.
http://dx.doi.org/10.1016/j.biopsych.2008.04.022. Eur. Neuropsychopharmacol. 25, 2015–2021. http://dx.doi.org/10.1016/j.
Berk, M., Dean, O.M., Cotton, S.M., Gama, C.S., Kapczinski, F., Fernandes, B., Kohlmann, euroneuro.2015.08.009.
K., Jeavons, S., Hewitt, K., Moss, K., Allwang, C., Schapkaitz, I., Cobb, H., Bush, A.I., Cardinali, D.P., Brusco, L.I., Liberczuk, C., Furio, A.M., 2002. The use of melatonin in
Dodd, S., Malhi, G.S., 2012. Maintenance N-acetyl cysteine treatment for bipolar Alzheimer's disease. Neuroendocrinol. Lett. 23, 20–23.
disorder: a double-blind randomized placebo controlled trial. BMC Med. 10, 91. Cardinali, D.P., Pandi-perumal, S.R., Brown, G.M., 2011. Sleep and circadian
http://dx.doi.org/10.1186/1741-7015-10-91. dysregulation in depressive illness pharmacological implications. Clin.
Berk, M., Kapczinski, F., Andreazza, A.C., Dean, O.M., Giorlando, F., Maes, M., Yucel, M., Neuropsychiatry 8, 321–338.
Gama, C.S., Dodd, S., Dean, B., Magalhaes, P.V., Amminger, P., McGorry, P., Malhi, Carroll, B.J., 1982. The dexamethasone suppression test for melancholia. Br. J. Psychiatry
G.S., 2011. Pathways underlying neuroprogression in bipolar disorder: focus on 140, 292–304.
inflammation, oxidative stress and neurotrophic factors. Neurosci. Biobehav. Rev. 35, Carvalho, A.F., Miskowiak, K.K., Hyphantis, T.N., Kohler, C.A., Alves, G.S., Bortolato, B.,
804–817. http://dx.doi.org/10.1016/j.neubiorev.2010.10.001. Sales, P.M., Machado-Vieira, R., Berk, M., McIntyre, R.S., 2014. Cognitive
Berson, D.M., 2007. Phototransduction in ganglion-cell photoreceptors. Pflügers Arch. dysfunction in depression - pathophysiology and novel targets. CNS Neurol. Disord.
454, 849–855. http://dx.doi.org/10.1007/s00424-007-0242-2. Drug Targets 13, 1814–1835.
Bezchlibnyk, Y.B., Wang, J.F., McQueen, G.M., Young, L.T., 2001. Gene expression Chang, H.-C., Guarente, L., 2013. SIRT1 mediates central circadian control in the SCN by
differences in bipolar disorder revealed by cDNA array analysis of post-mortem a mechanism that decays with aging. Cell 153, 1448–1460.
frontal cortex. J. Neurochem. 79, 826–834. Chi, K.F., Korgaonkar, M., Grieve, S.M., 2015. Imaging predictors of remission to anti-
Bilello, J.A., Thurmond, L.M., Smith, K.M., Pi, B., Rubin, R., Wright, S.M., Taub, F., depressant medications in major depressive disorder. J. Affect. Disord. 186, 134–144.
Henry, M.E., Shelton, R.C., Papakostas, G.I., 2015. MDDScore: Confirmation of a http://dx.doi.org/10.1016/j.jad.2015.07.002.
blood test to aid in the diagnosis of major depressive disorder. J. Clin. Psychiatry 76, Choi, S., Han, K.M., Won, E., Yoon, B.J., Lee, M.S., Ham, B.J., 2015. Association of brain-
e199–e206. http://dx.doi.org/10.4088/JCP.14m09029. derived neurotrophic factor DNA methylation and reduced white matter integrity in
Black, C.N., Bot, M., Scheffer, P.G., Cuijpers, P., Penninx, B.W., 2015. Is depression the anterior corona radiata in major depression. J. Affect. Disord. 172, 74–80. http://
associated with increased oxidative stress? A systematic review and meta-analysis. dx.doi.org/10.1016/j.jad.2014.09.042.
Psychoneuroendocrinology 51, 164–175. http://dx.doi.org/10.1016/j.psyneuen. Claustrat, B., Brun, J., Chazot, G., 2005. The basic physiology and pathophysiology of
2014.09.025. melatonin. Sleep Med. Rev. 9, 11–24.
Bloch, M.H., Hannestad, J., 2012. Omega-3 fatty acids for the treatment of depression: Corruble, E., de Bodinat, C., Belaïdi, C., Goodwin, G.M., Belaidi, C., Goodwin, G.M.,
systematic review and meta-analysis. Mol. Psychiatry 17, 1272–1282. http://dx.doi. Agomelatine Study Group, 2013. Efficacy of agomelatine and escitalopram on
org/10.1038/mp.2011.100. depression, subjective sleep and emotional experiences in patients with major
Boivin, D.B., 2000. Influence of sleep-wake and circadian rhythm disturbances in depressive disorder: a 24-wk randomized, controlled, double-blind trial. Int. J.
psychatric disorders. J. Psychiatry Neurosci. 25, 446–458. Neuropsychopharmacol. 16, 2219–2234. http://dx.doi.org/10.1017/
Bonmati-Carrion, M.A., Arguelles-Prieto, R., Martinez-Madrid, M.J., Reiter, R., S1461145713000679.
Hardeland, R., Rol, M.A., Madrid, J.A., 2014. Protecting the melatonin rhythm Crowley, S.K., Youngstedt, S.D., 2012. Efficacy of light therapy for perinatal depression: a
through circadian healthy light exposure. Int. J. Mol. Sci. 15, 23448–23500. http:// review. J. Physiol. Anthropol. 31, 15.
dx.doi.org/10.3390/ijms151223448. Cryan, J.F., Dinan, T.G., 2012. Mind-altering microorganisms: the impact of the gut
Bora, E., Harrison, B.J., Davey, C.G., Yücel, M., Pantelis, C., 2012. Meta-analysis of microbiota on brain and behaviour. Nat. Rev. 13, 701–702. http://dx.doi.org/10.
volumetric abnormalities in cortico-striatal-pallidal-thalamic circuits in major 1038/nrn3346.
depressive disorder. Psychol. Med. 42, 671–681. http://dx.doi.org/10.1017/ Cutolo, M., Maestroni, G.J.M., 2005. The melatonin-cytokine connection in rheumatoid
S0033291711001668. arthritis. Ann. Rheum. Dis. 64, 1109–1111.
Bourin, M., Prica, C., 2009. Melatonin receptor agonist agomelatine: a new drug for Dallaspezia, S., Benedetti, F., 2015. Chronobiology of bipolar disorder: therapeutic
treating unipolar depression. Curr. Pharm. Des. 15, 1675. implication. Curr. Psychiatry Rep. 17, 1–10. http://dx.doi.org/10.1007/s11920-015-
Bouwmans, M.E.J., Bos, E.H., Booij, S.H., van Faassen, M., Oldehinkel, A.J., de Jonge, P., 0606-9.
2015. Intra- and inter-individual variability of longitudinal daytime melatonin Dallaspezia, S., Benedetti, F., 2011. Chronobiological therapy for mood disorders. Expert.
secretion patterns in depressed and non-depressed individuals. Chronobiol. Int. 32, Rev. Neurother. 11, 961–970. http://dx.doi.org/10.1586/ern.11.61.
441–446. http://dx.doi.org/10.3109/07420528.2014.973114. Dallaspezia, S., Locatelli, C., Lorenzi, C., Pirovano, A., Colombo, C., Benedetti, F., 2016.
Brietzke, E., Kauer-Sant'Anna, M., Teixeira, A.L., Kapczinski, F., 2009a. Abnormalities in Sleep homeostatic pressure and PER3 VNTR gene polymorphism influence
serum chemokine levels in euthymic patients with bipolar disorder. Brain Behav. antidepressant response to sleep deprivation in bipolar depression. J. Affect. Disord.
Immun. 23, 1079–1082. http://dx.doi.org/10.1016/j.bbi.2009.04.008. 192, 64–69. http://dx.doi.org/10.1016/j.jad.2015.11.039.
Brietzke, E., Stertz, L., Fernandes, B.S., Kauer-Sant'anna, M., Mascarenhas, M., Escosteguy Dalton, E.J., Rotondi, D., Levitan, R.D., Kennedy, S.H., Brown, G.M., 2000. Use of slow-
Vargas, A., Chies, J.A., Kapczinski, F., 2009b. Comparison of cytokine levels in release melatonin in treatment-resistant depression. J. Psychiatry Neurosci. 25,
depressed, manic and euthymic patients with bipolar disorder. J. Affect. Disord. 116, 48–52.
214–217. http://dx.doi.org/10.1016/j.jad.2008.12.001. Danilenko, K.V., Ivanova, I.A., 2015. Dawn simulation vs. bright light in seasonal
Brown, R., Kocsis, J.H., Caroff, S., Amsterdam, J., Winokur, A., Stokes, P.E., Frazer, A., affective disorder: Treatment effects and subjective preference. J. Affect. Disord. 180,
1985. Differences in nocturnal melatonin secretion between melancholic depressed 87–89.
patients and control subjects. Am. J. Psychiatry 142, 811–816. Danilenko, K.V., Putilov, A.A., 2005. Melatonin treatment of winter depression following
Brusco, L.I., Fainstein, I., Marquez, M., Cardinali, D.P., 1999. Effect of melatonin in total sleep deprivation: waking EEG and mood correlates. Neuropsychopharmacology
selected populations of sleep-disturbed patients. Biol. Signals Recept. 8, 126–131. 30, 1345–1352.
Brzezinski, A., Vangel, M.G., Wurtman, R.J., Norrie, G., Zhdanova, I., Ben-Shushan, A., Delavest, M., Even, C., Benjemaa, N., Poirier, M.-F., Jockers, R., Krebs, M.-O., 2012.
Ford, I., 2005. Effects of exogenous melatonin on sleep: a meta-analysis. Sleep Med. Association of the intronic rs2072621 polymorphism of the X-linked GPR50 gene
Rev. 9, 41–50. with affective disorder with seasonal pattern. Eur. Psychiatry 27, 369–371.
Bumb, J.M., Enning, F., Mueller, J.K., Van Der List, T., Rohleder, C., Findeisen, P., Noelte, Delvecchio, G., Fossati, P., Boyer, P., Brambilla, P., Falkai, P., Gruber, O., Hietala, J.,
I., Schwarz, E., Leweke, F.M., 2016. Differential melatonin alterations in Lawrie, S.M., Martinot, J.L., McIntosh, A.M., Meisenzahl, E., Frangou, S., 2012.
cerebrospinal fluid and serum of patients with major depressive disorder and bipolar Common and distinct neural correlates of emotional processing in bipolar disorder
disorder. Compr. Psychiatry 68, 34–39. http://dx.doi.org/10.1016/j.comppsych. and major depressive disorder: a voxel-based meta-analysis of functional magnetic
2016.03.005. resonance imaging studies. Eur. Neuropsychopharmacol. 22, 100–113. http://dx.doi.

199
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

org/10.1016/j.euroneuro.2011.07.003. bipolar disorder and depression. Mol. Psychiatry 21, 1696–1709. http://dx.doi.org/
Demyttenaere, K., 2011. Agomelatine: a narrative review. Eur. Neuropsychopharmacol. 10.1038/mp.2016.3.
21 (Suppl. 4), S703–S709. http://dx.doi.org/10.1016/j.euroneuro.2011.07.004. Gonzalez, R., Gonzalez, S., Villa, E., Ramirez, M., Zavala, J., Armas, R., Contreras, J.,
Depping, M.S., Wolf, N.D., Vasic, N., Sambataro, F., Thomann, P.A., Wolf, R.C., 2016. Dassori, A., Leach, R.J., Flores, D., Jerez, A., Raventós, H., Ontiveros, A., Nicolini, H.,
Common and distinct structural network abnormalities in major depressive disorder Escamilla, M., 2015. Identification of circadian gene variants in bipolar disorder in
and borderline personality disorder. Prog. Neuropsychopharmacol. Biol. Psychiatry Latino populations. J. Affect. Disord. 186, 367–375. http://dx.doi.org/10.1016/j.jad.
65, 127–133. http://dx.doi.org/10.1016/j.pnpbp.2015.09.007. 2015.07.014.
der Lek, R., Swaab, D.F., Twisk, J., Hol, E.M., Hoogendijk, W.J., Van Someren, E.J., 2008. Goodwin, G.M., 2009. Clinical studies on the efficacy of agomelatine on depressive
Effect of bright light and melatonin on cognitive and noncognitive function in elderly symptoms. CNS Drugs 23 (Suppl. 2), 35–39.
residents of group care facilities: a randomized controlled trial. J. Am. Med. Assoc. Gooley, J.J., Chamberlain, K., Smith, K.a., Khalsa, S.B.S., Rajaratnam, S.M.W., van Reen,
299, 2642–2655. E., Zeitzer, J.M., Czeisler, C.a., Lockley, S.W., 2011. Exposure to room light before
Dibner, C., Schibler, U., Albrecht, U., 2010. The mammalian circadian timing system: bedtime suppresses melatonin onset and shortens melatonin duration in humans. J.
Organization and coordination of central and peripheral clocks. Annu. Rev. Physiol. Clin. Endocrinol. Metab. 96, 463–472. http://dx.doi.org/10.1210/jc.2010-2098.
72, 517–549. http://dx.doi.org/10.1146/annurev-physiol-021909-135821. Gooley, J.J., Lu, J., Fischer, D., Saper, C.B., 2003. A broad role for melanopsin in
Dolberg, O.T., Hirschmann, S., Grunhaus, L., 1998. Melatonin for the treatment of sleep nonvisual photoreception. J. Neurosci. 23, 7093–7106.
disturbances in major depressive disorder. Am. J. Psychiatry 155, 1119–1121. Gordijn, M.C.M., Mannetje, D’t., Meesters, Y., 2012. The effects of blue-enriched light
Dolder, C.R., Nelson, M., Snider, M., 2008. Agomelatine treatment of major depressive treatment compared to standard light treatment in Seasonal Affective Disorder. J.
disorder. Ann. Pharmacother. 42, 1822–1823. Affect. Disord. 136, 72–80.
Dowling, G.A., Burr, R.L., Van Someren, E.J., Hubbard, E.M., Luxenberg, J.S., Mastick, J., Gottlieb, J.F., Terman, M., 2012. Outpatient triple chronotherapy for bipolar depression:
Cooper, B.A., 2008. Melatonin and bright-light treatment for rest-activity disruption case report. J. Psychiatr. Pract. 18, 373–380.
in institutionalized patients with Alzheimer's disease. J. Am. Geriatr. Soc. 56, Gruz, F., Raffa, S., Santucci, C., Papale, R.M., Videla, M.G., Fernandez, M.G., Yantorno, S.,
239–246. Descalzi, V.I., 2014. Agomelatine: fulminant liver failure in a patient with fatty liver.
Drago, A., Monti, B., Ronchi, D. De, Serretti, A., Giovanni, I.R.C.C.S.S., 2015. CRY1 Gastroenterol. Hepatol. 37, 92–94. http://dx.doi.org/10.1016/j.gastrohep.2013.04.
variations impacts on the depressive relapse rate in a sample of bipolar patients. 008.
Psychiatry Investig. 12, 118–124. Guaiana, G., Gupta, S., Chiodo, D., Davies, S.J., Haederle, K., Koesters, M., 2013.
Dridi, D., Zouiten, A., Mansour, H.Ben, 2014. Depression: chronophysiology and Agomelatine versus other antidepressive agents for major depression. Cochrane
chronotherapy. Biol. Rhythm. Res. 45, 77–91. database Syst. Rev. 12. http://dx.doi.org/10.1002/14651858.CD008851.pub2.
Dunn, A.J., Swiergiel, A.H., de Beaurepaire, R., 2005. Cytokines as mediators of Guilleminault, C., 2005. Efficacy of agomelatine versus venlafaxine on subjective sleep of
depression: what can we learn from animal studies? Neurosci. Biobehav. Rev. 29, patients with major depressive diorder. Eur. Neuropsychopharmacol. 15, S419.
891–909. http://dx.doi.org/10.1016/j.neubiorev.2005.03.023. Hack, L.M., Lockley, S.W., Arendt, J., Skene, D.J., 2003. The effects of low-dose 0.5-mg
Dusi, N., Barlati, S., Vita, A., Brambilla, P., 2015. Brain structural effects of antidepressant melatonin on the free-running circadian rhythms of blind subjects. J. Biol. Rhythm.
treatment in major depression. Curr. Neuropharmacol. 13, 458–465. http://dx.doi. 18, 420–429.
org/10.2174/1570159X1304150831121909. Han, K.-M., Kim, D., Sim, Y., Kang, J., Kim, A., Won, E., Tae, W.-S., Ham, B.-J., 2017.
Ekdahl, C.T., 2012. Microglial activation - tuning and pruning adult neurogenesis. Front. Alterations in the brainstem volume of patients with major depressive disorder and
Pharmacol. 3, 41. http://dx.doi.org/10.3389/fphar.2012.00041. their relationship with antidepressant treatment. J. Affect. Disord. 208, 68–75.
Elder, G., Wetherell, M.A., Barclay, N.L., Ellis, J.G., 2014. The cortisol awakening http://dx.doi.org/10.1016/j.jad.2016.08.066.
response—applications and implications for sleep medicine. Sleep Med. Rev. 18, Hardeland, R., 2016. Melatonin and synthetic melatoninergic agonists in psychiatric and
215–224. http://dx.doi.org/10.1016/j.smrv.2013.05.001. age-associated disorders: successful and unsuccessful approaches. Curr. Pharm. Des.
Eniola, K., Bacigalupo, A., Mounsey, A., 2016. PURLs: light therapy for nonseasonal major 22, 1086–1100.
depressive disorder? J. Fam. Pract. 65, 486–488. Hardeland, R., 2014a. Melatonin, noncoding RNAs, messenger RNA stability and
Eser, D., Baghai, T.C., Möller, H.-J., 2007. Evidence of agomelatine's antidepressant epigenetics—evidence, hints, gaps and perspectives. Int. J. Mol. Sci. 15,
efficacy: The key points. Int. Clin. Psychopharmacol. 22 (Suppl. 2), S15–S19. 18221–18252. http://dx.doi.org/10.3390/ijms151018221.
Eser, D., Baghai, T.C., Möller, H.J., 2009. Agomelatine: the evidence for its place in the Hardeland, R., 2014b. Agomelatine and the risk of hepatotoxicity. J. Symptoms Signs 3,
treatment of depression. Core Evid. 4, 171–179. http://dx.doi.org/10.2147/CE. 341–346.
S6005. Hardeland, R., 2012a. Melatonin in aging and disease ― multiple consequences of
Felger, J.C., Lotrich, F.E., 2013. Inflammatory cytokines in depression: neurobiological reduced secretion, options and limits of treatment. Aging Dis. 3 (3), 194–225.
mechanisms and therapeutic implications. Neuroscience 246, 199–229. http://dx. Hardeland, R., 2012b. Rationale and Problems of Melatonergic Treatment. vol. 16. pp.
doi.org/10.1016/j.neuroscience.2013.04.060. 7–14.
Fernandes, B.S., Steiner, J., Molendijk, M.L., Dodd, S., Nardin, P., Goncalves, C.A., Jacka, Hardeland, R., 2009a. Melatonin: signaling mechanisms of a pleiotropic agent. Biofactors
F., Kohler, C.A., Karmakar, C., Carvalho, A.F., Berk, M., 2016. C-reactive protein 35, 183–192. http://dx.doi.org/10.1002/biof.23.
concentrations across the mood spectrum in bipolar disorder: a systematic review and Hardeland, R., 2009b. New approaches in the management of insomnia: weighing the
meta-analysis. Lancet Psychiatry 3, 1147–1156. http://dx.doi.org/10.1016/S2215- advantages of prolonged-release melatonin and synthetic melatoninergic agonists.
0366(16)30370-4. Neuropsychiatr. Dis. Treat. 5, 341–354.
Flynn-Evans, E.E., Tabandeh, H., Skene, D.J., Lockley, S.W., 2014. Circadian rhythm Hardeland, R., Cardinali, D.P., Brown, G.M., Pandi-Perumal, S.R., 2015. Melatonin and
disorders and melatonin production in 127 blind women with and without light brain inflammaging. Prog. Neurobiol. 127–128, 46–63. http://dx.doi.org/10.1016/j.
perception. J. Biol. Rhythm. 29, 215–224. pneurobio.2015.02.001.
Frazer, A., Brown, R., Kocsis, J., Caroff, S., Amsterdam, J., Winokur, A., Sweeney, J., Hardeland, R., Coto-Montes, A., Poeggeler, B., 2003. Circadian rhythms, oxidative stress
Stokes, P., 1985. Patterns of melatonin rhythms in depression. J. Neural Transm. and antioxidative defense mechanisms. Chronobiol. Int. 20, 921–962.
Suppl. 21, 269–290. Harrisberger, F., Smieskova, R., Schmidt, A., Lenz, C., Walter, A., Wittfeld, K., Grabe, H.J.,
Frick, L.R., Williams, K., Pittenger, C., 2013. Microglial dysregulation in psychiatric Lang, U.E., Fusar-Poli, P., Borgwardt, S., 2015. BDNF Val66Met polymorphism and
disease. Clin. Dev. Immunol. http://dx.doi.org/10.1155/2013/608654. hippocampal volume in neuropsychiatric disorders: a systematic review and meta-
Frodl, T., Amico, F., 2014. Is there an association between peripheral immune markers analysis. Neurosci. Biobehav. Rev. 55, 107–118. http://dx.doi.org/10.1016/j.
and structural/functional neuroimaging findings? Prog. Neuropsychopharmacol. neubiorev.2015.04.017.
Biol. Psychiatry 48, 295–303. http://dx.doi.org/10.1016/j.pnpbp.2012.12.013. Harrison, J., Lam, R., Baune, B., McIntyre, R.S., 2016. Selection of cognitive tests for trials
Gagné, A.-M., Lévesque, F., Gagné, P., Hébert, M., 2011. Impact of blue vs red light on of therapeutic agents. Lancet Psychiatry 3, 499.
retinal response of patients with seasonal affective disorder and healthy controls. Harrison, N.A., Brydon, L., Walker, C., Gray, M.A., Steptoe, A., Critchley, H.D., 2009.
Prog. Neuro-Psychopharmacol. Biol. Psychiatry 35, 227–231. Inflammation causes mood changes through alterations in subgenual cingulate
Gahr, M., Freudenmann, R.W., Connemann, B.J., Hiemke, C., Schonfeldt-Lecuona, C., activity and mesolimbic connectivity. Biol. Psychiatry 66, 407–414. http://dx.doi.
2013. Agomelatine and hepatotoxicity: implications of cumulated data derived from org/10.1016/j.biopsych.2009.03.015.
spontaneous reports of adverse drug reactions. Pharmacopsychiatry 46, 214–220. Harry, G.J., Kraft, A.D., 2012. Microglia in the developing brain: a potential target with
http://dx.doi.org/10.1055/s-0033-1353156. lifetime effects. Neurotoxicology 33, 191–206. http://dx.doi.org/10.1016/j.neuro.
Galecki, P., Szemraj, J., Bartosz, G., Bienkiewicz, M., Galecka, E., Florkowski, A., 2012.01.012.
Lewinski, A., Karbownik-Lewinska, M., 2010. Single-nucleotide polymorphisms and Hastings, M.H., Brancaccio, M., Maywood, E.S., 2014. Circadian pacemaking in cells and
mRNA expression for melatonin synthesis rate-limiting enzyme in recurrent circuits of the suprachiasmatic nucleus. J. Neuroendocrinol. 26, 2–10. http://dx.doi.
depressive disorder. J. Pineal Res. 48, 311–317. http://dx.doi.org/10.1111/j.1600- org/10.1111/jne.12125.
079X.2010.00754.x. Hastings, M.H., Goedert, M., 2013. Circadian clocks and neurodegenerative diseases: time
Gill, R., Tsung, A., Billiar, T., 2010. Linking oxidative stress to inflammation: Toll-like to aggregate? Curr. Opin. Neurobiol. 23, 880–887. http://dx.doi.org/10.1016/j.conb.
receptors. Free Radic. Biol. Med. 48, 1121–1132. http://dx.doi.org/10.1016/j. 2013.05.004.
freeradbiomed.2010.01.006. Henriksen, T.E., Skrede, S., Fasmer, O.B., Schoeyen, H., Leskauskaite, I., Bjørke-
Goldsmith, D.R., Haroon, E., Woolwine, B.J., Jung, M.Y., Wommack, E.C., Harvey, P.D., Bertheussen, J., Assmus, J., Hamre, B., Grønli, J., Lund, A., 2016. Blue-blocking
Treadway, M.T., Felger, J.C., Miller, A.H., 2016a. Inflammatory markers are glasses as additive treatment for mania: a randomized placebo-controlled trial.
associated with decreased psychomotor speed in patients with major depressive Bipolar Disord. 18, 221–232. http://dx.doi.org/10.1111/bdi.12390.
disorder. Brain Behav. Immun. 56, 281–288. http://dx.doi.org/10.1016/j.bbi.2016. Herbert, J., 2013. Cortisol and depression: three questions for psychiatry. Psychol. Med.
03.025. 43, 449–469. http://dx.doi.org/10.1017/S0033291712000955.
Goldsmith, D.R., Rapaport, M.H., Miller, B.J., 2016b. A meta-analysis of blood cytokine Howland, R.H., 2011a. A benefit-risk assessment of agomelatine in the treatment of major
network alterations in psychiatric patients: comparisons between schizophrenia, depression. Drug Saf. 34, 709–731. http://dx.doi.org/10.2165/11593960-

200
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

000000000-00000. 167–172. http://dx.doi.org/10.1590/S1516-31802012000300006.


Howland, R.H., 2011b. Publication bias and outcome reporting bias: agomelatine as a Kishi, T., Yoshimura, R., Kitajima, T., Okochi, T., Okumura, T., Tsunoka, T., Yamanouchi,
case example. J. Psychosoc. Nurs. Ment. Health Serv. 49 (9), 11–14. http://dx.doi. Y., Kinoshita, Y., Kawashima, K., Fukuo, Y., Naitoh, H., Umene-Nakano, W., Inada, T.,
org/10.3928/02793695-20110809-01. Nakamura, J., Ozaki, N., Iwata, N., 2010. SIRT1 gene is associated with major
Hua, P., Liu, W., Chen, D., Zhao, Y., Chen, L., Zhang, N., Wang, C., Guo, S., Wang, L., Xiao, depressive disorder in the Japanese population. J. Affect. Disord. 126, 167–173.
H., Kuo, S.H., 2014. Cry1 and Tef gene polymorphisms are associated with major http://dx.doi.org/10.1016/j.jad.2010.04.003.
depressive disorder in the Chinese population. J. Affect. Disord. 157, 100–103. Koesters, M., Guaiana, G., Cipriani, A., Becker, T., Barbui, C., 2013. Agomelatine efficacy
http://dx.doi.org/10.1016/j.jad.2013.11.019. and acceptability revisited: systematic review and meta-analysis of published and
Insel, T., Cuthbert, B., Garvey, M., Heinssen, R., Pine, D.S., Quinn, K., Sanislow, C., Wang, unpublished randomised trials. Br. J. Psychiatry 203, 179–187. http://dx.doi.org/10.
P., 2010. Research Domain Criteria (RDoC): Toward a new classification framework 1192/bjp.bp.112.120196.
for research on mental disorders. Am. J. Psychiatry 167, 748–751. http://dx.doi.org/ Kohler, O., Benros, M.E., Nordentoft, M., Farkouh, M.E., Iyengar, R.L., Mors, O., Krogh, J.,
10.1176/appi.ajp.2010.09091379. 2014. Effect of anti-inflammatory treatment on depression, depressive symptoms, and
Ivanov, S.V., Samushiya, M.A., 2014. Agomelatine in the treatment of depressive adverse effects: a systematic review and meta-analysis of randomized clinical trials.
disorders in clinical practice: multicenter observational CHRONOS study. JAMA Psychiatry 71, 1381–1391. http://dx.doi.org/10.1001/jamapsychiatry.2014.
Neuropsychiatr. Dis. Treat 10, 631–639. http://dx.doi.org/10.2147/NDT.S58994. 1611.
Jaworska, N., Yang, X.-R., Knott, V., Macqueen, G., 2014. A review of fMRI studies during Kraft, A.D., Harry, G.J., 2011. Features of microglia and neuroinflammation relevant to
visual emotive processing in major depressive disorder. World J. Biol. Psychiatry environmental exposure and neurotoxicity. Int. J. Environ. Res. Public Heal. 8,
7321, 1–24. http://dx.doi.org/10.3109/15622975.2014.885659. 2980–3018. http://dx.doi.org/10.3390/ijerph8072980.
Jaworska, N., Yücel, K., Courtright, A., Macmaster, F.P., Sembo, M., Macqueen, G., 2016. Kripke, D., Mullaney, D., Atkinson, M., Wolf, S., 1978. Circadian rhythm disorders in
Subgenual anterior cingulate cortex and hippocampal volumes in depressed youth: manic-depressives. Biol. Psychiatry 13, 335–351.
the role of comorbidity and age. J. Affect. Disord. 190, 726–732. http://dx.doi.org/ Kripke, D.F., Elliott, J.A., Welsh, D.K., Youngstedt, S.D., 2015. Photoperiodic and
10.1016/j.jad.2015.10.064. circadian bifurcation theories of depression and mania. F1000Res. 4, 107. http://dx.
Jimenez-Fernandez, S., Gurpegui, M., Diaz-Atienza, F., Perez-Costillas, L., Gerstenberg, doi.org/10.12688/f1000research.6444.1.
M., Correll, C.U., 2015. Oxidative stress and antioxidant parameters in patients with Kripke, D.F., Nievergelt, C.M., Joo, E., Shekhtman, T., Kelsoe, J.R., 2009. Circadian
major depressive disorder compared to healthy controls before and after polymorphisms associated with affective disorders. J. Circadian Rhythms 7, 2. http://
antidepressant treatment: results from a meta-analysis. J. Clin. Investigation 76, dx.doi.org/10.1186/1740-3391-7-2.
1658–1667. http://dx.doi.org/10.4088/JCP.14r09179. Lam, R.W., Berkowitz, A.L., Berga, S.L., Clark, C.M., Kripke, D.F., Gillin, J.C., 1990.
Jin, X., Shearman, L.P., Weaver, D.R., Zylka, M.J., de Vries, G.J., Reppert, S.M., 1999. A Melatonin suppression in bipolar and unipolar mood disorders. Psychiatry Res. 33,
molecular mechanism regulating rhythmic output from the suprachiasmatic circadian 129–134.
clock. Cell 96, 57–68. Langan, J., Shajahan, P., Martin, D., Carleton, R., 2011. Agomelatine in unipolar
Johansson, C., Willeit, M., Smedh, C., Ekholm, J., Paunio, T., Kieseppä, T., Lichtermann, depression in clinical practice: a retrospective chart review. Ther. Adv.
D., Praschak-Rieder, N., Neumeister, A., Nilsson, L.-G., Kasper, S., Peltonen, L., Psychopharmacol. 1, 175–180.
Adolfsson, R., Schalling, M., Partonen, T., 2003. Circadian clock-related Lavebratt, C., Sjöholm, L.K., Partonen, T., Schalling, M., Forsell, Y., 2010a. PER2
polymorphisms in seasonal affective disorder and their relevance to diurnal variantion is associated with depression vulnerability. Am. J. Med. Genet. B
preference. Neuropsychopharmacology 28, 734–739. http://dx.doi.org/10.1038/sj. Neuropsychiatr. Genet. 153, 570–581. http://dx.doi.org/10.1002/ajmg.b.31021.
npp.1300121. Lavebratt, C., Sjöholm, L.K., Soronen, P., Paunio, T., Vawter, M.P., Bunney, W.E.,
Jones, S.G., Benca, R.M., 2015. Circadian disruption in psychiatric disorders. Sleep Med. Adolfsson, R., Forsell, Y., Wu, J.C., Kelsoe, J.R., Partonen, T., Schalling, M., 2010b.
Clin. 10, 481–493. http://dx.doi.org/10.1016/j.jsmc.2015.07.004. CRY2 is associated with depression. PLoS One 5, 1–8.
Kalenderoglu, A., Çelik, M., Sevgi-Karadag, A., Egilmez, O.B., 2016. Optic coherence Le-Niculescu, H., Patel, S.D., Bhat, M., Kuczenski, R., Faraone, S.V., Tsuang, M.T.,
tomography shows inflammation and degeneration in major depressive disorder McMahon, F.J., Schork, N.J., Nurnberger, J.I., Niculescu, A.B., 2009. Convergent
patients correlated with disease severity. J. Affect. Disord. 204, 159–165. http://dx. functional genomics of genome-wide association data for bipolar disorder:
doi.org/10.1016/j.jad.2016.06.039. Comprehensive identification of candidate genes, pathways and mechanisms. Am. J.
Kaltenboeck, A., Winkler, D., Kasper, S., 2016. Bipolar and related disorders in DSM-5 and Med. Genet. B Neuropsychiatr. Genet. 150, 155–181. http://dx.doi.org/10.1002/
ICD-10. CNS Spectr. 21, 318–323. http://dx.doi.org/10.1017/S1092852916000079. ajmg.b.30887.
Kaminski-Hartenthaler, A., Nussbaumer, B., Forneris, C.A., Morgan, L.C., Gaynes, B.N., Lee, S.Y., Lee, S.J., Han, C., Patkar, A.A., Masand, P.S., Pae, C.U., 2013. Oxidative/
Sonis, J.H., Greenblatt, A., et al., 2015. Melatonin and agomelatine for preventing nitrosative stress and antidepressants: targets for novel antidepressants. Prog. Neuro-
seasonal affective disorder. Cochrane Database Syst. Rev. 11. Psychopharmacol. Biol. Psychiatry 46, 224–235. http://dx.doi.org/10.1016/j.pnpbp.
Karim, A., Tolbert, D., Cao, C., 2006. Disposition kinetics and tolerance of escalating 2012.09.008.
single doses of ramelteon, a high-affinity MT1 and MT2 melatonin receptor agonist Leheste, J.R., Torres, G., 2015. Resveratrol: brain effects on SIRT1, GPR50 and
indicated for treatment of insomnia. J. Clin. Pharmacol. 46, 140–148. photoperiodic signaling. Front. Mol. Neurosci. 8, 61.
Kasper, S., Corruble, E., Hale, A., Lemoine, P., Montgomery, S.A., Quera-Salva, M.A., Leibenluft, E., Feldman-Naim, S., Turner, E.H., Wehr, T.A., Rosenthal, N.E., 1997. Effects
2013. Antidepressant efficacy of agomelatine versus SSRI/SNRI: results from a pooled of exogenous melatonin administration and withdrawal in five patients with rapid-
analysis of head-to-head studies without a placebo control. Int. Clin. cycling bipolar disorder. J. Clin. Psychiatry 58, 383–388.
Psychopharmacol. 28, 12–19. http://dx.doi.org/10.1097/YIC.0b013e328359768e. Lener, M.S., Iosifescu, D.V., 2015. In pursuit of neuroimaging biomarkers to guide
Kaufmann, F.N., Gazal, M., Bastos, C.R., Kaster, M.P., Ghisleni, G., 2015. Curcumin in treatment selection in major depressive disorder: a review of the literature. Ann. N. Y.
depressive disorders: an overview of potential mechanisms, preclinical and clinical Acad. Sci. 1344, 50–65. http://dx.doi.org/10.1111/nyas.12759.
findings. Eur. J. Pharmacol. 76, 1658–1667. http://dx.doi.org/10.1016/j.ejphar. Leppämäki, S., Partonen, T., Vakkuri, O., Lonnqvist, J., Partinen, M., Laudon, M., 2003.
2016.05.026. Effect of controlled-release melatonin on sleep quality, mood, and quality of life in
Kayumov, L., Casper, R.F., Hawa, R.J., Perelman, B., Chung, S.A., Sokalsky, S., Shapiro, subjects with seasonal or weather-associated changes in mood and behaviour. Eur.
C.M., 2005. Blocking low-wavelength light prevents nocturnal melatonin suppression Neuropsychopharmacol. 13, 137–145.
with no adverse effect on performance during simulated shift work. J. Clin. Levoye, A., Dam, J., Ayoub, M.A., Guillaume, J.L., Couturier, C., Delagrange, P., Jockers,
Endocrinol. Metab. 90, 2755–2761. http://dx.doi.org/10.1210/jc.2004-2062. R., 2006. The orphan GPR50 receptor specifically inhibits MT1 melatonin receptor
Kayumov, L., Zhdanova, I.V., Shapiro, C.M., 2000. Melatonin, sleep, and circadian function through heterodimerization. EMBO J. 25, 3012–3023.
rhythm disorders. Semin. Clin. Neuropsychiatry 5, 44–55. Lewy, A.J., 2003. Clinical applications of melatonin in circadian disorders. Dialogues
Keck Jr., P.E., Mintz, J., McElroy, S.L., Freeman, M.P., Suppes, T., Frye, M.A., Altshuler, Clin. Neurosci. 5, 399–413.
L.L., Kupka, R., Nolen, W.A., Leverich, G.S., Denicoff, K.D., Grunze, H., Duan, N., Lewy, A.J., Ahmed, S., Jackson, J.M., Sack, R.L., 1992. Melatonin shifts human circadian
Post, R.M., 2006. Double-blind, randomized, placebo-controlled trials of ethyl- rhythms according to a phase-response curve. Chronobiol. Int. 9, 380–392.
eicosapentanoate in the treatment of bipolar depression and rapid cycling bipolar Lewy, A.J., Bauer, V.K., Cutler, N.L., Sack, R.L., 1998. Melatonin treatment of winter
disorder. Biol. Psychiatry 60, 1020–1022. http://dx.doi.org/10.1016/j.biopsych. depression: a pilot study. Psychiatry Res. 77, 57–61.
2006.03.056. Lewy, A.J., Emens, J., Jackman, A., Yuhas, K., 2006. Circadian uses of melatonin in
Kennedy, S.H., 2009. Agomelatine: Efficacy at each phase of antidepressant treatment. humans. Chronobiol. Int. 23, 403–412.
CNS Drugs 23 (Suppl. 2), 41–47. Lewy, A.J., Emens, J., Songer, J.B., Sims, N., Laurie, A.L., Fiala, S.C., Buti, A.L., 2009.
Kennedy, S.H., Eisfeld, B.S., 2007. Agomelatine and its therapeutic potential in the Winter depression: integrating mood, circadian rhythms, and the sleep/wake and
depressed patient. Neuropsychiatr. Dis. Treat. 3, 423–428. light/dark cycles into a bio-psycho-social-environmental model. Sleep Med. Clin. 4,
Kennedy, S.H., Kutcher, S.P., Ralevski, E., Brown, G.M., 1996. Nocturnal melatonin and 285–299.
24-hour 6-sulphatoxymelatonin levels in various phases of bipolar affective disorder. Lewy, A.J., Emens, J.S., Lefler, B.J., Yuhas, K., Jackman, A.R., 2005. Melatonin entrains
Psychiatry Res. 63, 219–222. free-running blind people according to a physiological dose-response curve.
Kerestes, R., Davey, C.G., Stephanou, K., Whittle, S., Harrison, B.J., 2014. Clinical Chronobiol. Int. 22, 1093–1106.
functional brain imaging studies of youth depression: a systematic review. Lewy, A.J., Emens, J.S., Sack, R.L., Hasler, B.P., Bernert, R.A., 2002. Low, but not high,
NeuroImage Clin. 4, 209–231. http://dx.doi.org/10.1016/j.nicl.2013.11.009. doses of melatonin entrained a free-running blind person with a long circadian
Kessler, R.C., Petukhova, M., Sampson, N.A., Zaslavsky, A.M., Wittchen, H.-U., 2012. period. Chronobiol. Int. 19, 649–658.
Twelve-month and lifetime prevalence and lifetime morbid risk of anxiety and mood Lewy, A.J., Nurnberger Jr., J.I., Wehr, T.A., Pack, D., Becker, L.E., Powell, R.L., Newsome,
disorders in the United States. Int. J. Methods Psychiatr. Res. 21, 169–184. D.A., 1985. Supersensitivity to light: possible trait marker for manic-depressive
Khaleghipour, S., Masjedi, M., Ahade, H., Enayate, M., Pasha, G., Nadery, F., Ahmadzade, illness. Am. J. Psychiatry 142, 725–727.
G., 2012. Morning and nocturnal serum melatonin rhythm levels in patients with Lewy, A.J., Sack, R.L., 1989. The dim light melatonin onset as a marker for circadian
major depressive disorder: an analytical cross-sectional study. Sao Paulo Med. J. 130, phase position. Chronobiol. Int. 6, 93–102.

201
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

Li, J.Z., Bunney, B.G., Meng, F., Hagenauer, M.H., Walsh, D.M., Vawter, M.P., 2013. S.H., 2015a. The impact of cognitive impairment on perceived workforce
Circadian patterns of gene expression in the human brain and disruption in major performance: results from the international mood disorders collaborative project.
depressive disorder. PNAS 110, 9950–9955. http://dx.doi.org/10.1073/pnas. Compr. Psychiatry 56, 279–282. http://dx.doi.org/10.1016/j.comppsych.2014.08.
1305814110/-/DCSupplemental.www.pnas.org/cgi/doi/10.1073/pnas. 051.
1305814110. McIntyre, R.S., Xiao, H.X., Syeda, K., Vinberg, M., Carvalho, A.F., Mansur, R.B.,
Liao, Y., Huang, X., Wu, Q., Yang, C., Kuang, W., Du, M., Lui, S., Yue, Q., Chan, R., Kemp, Maruschak, N., Cha, D.S., 2015b. The prevalence, measurement, and treatment of the
G., Gong, Q., 2013. Is depression a disconnection syndrome? Meta- analysis of cognitive dimension/domain in major depressive disorder. CNS Drugs 29, 577–589.
diffusion tensor imaging studies in patients with MDD. J. Psychiatry Neurosci. 38, http://dx.doi.org/10.1007/s40263-015-0263-x.
49–56. http://dx.doi.org/10.1503/jpn.110180. McNamara, R.K., Lotrich, F.E., 2012. Elevated immune-inflammatory signaling in mood
Lindqvist, D., Mueller, S., Mellon, S.H., Su, Y., Epel, E.S., Reus, V.I., Rosser, R., Mahan, L., disorders: A new therapeutic target? Expert. Rev. Neurother. 12, 1143–1161. http://
Scott Mackin, R., Yang, T.T., Wolkowitz, O.M., 2014. Peripheral antioxidant markers dx.doi.org/10.1586/ern.12.98.
are associated with total hippocampal and CA3/dentate gyrus volume in MDD and Meesters, Y., Dekker, V., Schlangen, L.J.M., Bos, E.H., Ruiter, M.J., 2011. Low-intensity
healthy controls-preliminary findings. Psychiatry Res. Neuroimaging 224, 168–174. blue-enriched white light (750 lux) and standard bright light (10,000 lux) are equally
http://dx.doi.org/10.1016/j.pscychresns.2014.09.002. effective in treating SAD. A randomized controlled study. BMC Psychiatry 11, 17.
Liu, C., Chung, M., 2015. Genetics and epigenetics of circadian rhythms and their Meesters, Y., Winthorst, W.H., Duijzer, W.B., Hommes, V., 2016. The effects of low-
potential roles in neuropsychiatric disorders. Neurosci. Bull. 31, 141–159. http://dx. intensity narrow-band blue-light treatment compared to bright white-light treatment
doi.org/10.1007/s12264-014-1495-3. in sub-syndromal seasonal affective disorder. BMC Psychiatry 16, 27.
Liu, H.C., Yang, Y.Y., Chou, Y.M., Chen, K.P., Shen, W.W., Leu, S.J., 2004. Immunologic Millan, M.J., Gobert, A., Lejeune, F., Dekeyne, A., Newman-Tancredi, A., Pasteau, V.,
variables in acute mania of bipolar disorder. J. Neuroimmunol. 150, 116–122. http:// Rivet, J.M., Cussac, D., 2003. The novel melatonin agonist agomelatine (S20098) is
dx.doi.org/10.1016/j.jneuroim.2004.01.006. an antagonist at 5-hydroxytryptamine2C receptors, blockade of which enhances the
Liu, Y., Ho, R.C., Mak, A., 2012. Interleukin (IL)-6, tumour necrosis factor alpha (TNF- activity of frontocortical dopaminergic and adrenergic pathways. J. Pharmacol. Exp.
alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with Ther. 306, 954–964.
major depressive disorder: A meta-analysis and meta-regression. J. Affect. Disord. Miller, A.H., Maletic, V., Raison, C.L., 2009. Inflammation and its discontents: the role of
139, 230–239. http://dx.doi.org/10.1016/j.jad.2011.08.003. cytokines in the pathophysiology of major depression. Biol. Psychiatry 65, 732–741.
Lockley, S.W., Arendt, J., Skene, D.J., 2007. Visual impairment and circadian rhythm http://dx.doi.org/10.1016/j.biopsych.2008.11.029.
disorders. Dialogues Clin. Neurosci. 9, 301–314. Mishima, K., Tozawa, T., Satoh, K., Matsumoto, Y., Hishikawa, Y., Okawa, M., 1999.
Lockley, S.W., Skene, D.J., James, K., Thapan, K., Wright, J., Arendt, J., 2000. Melatonin Melatonin secretion rhythm disorders in patients with senile dementia of Alzheimer's
administration can entrain the free-running circadian system of blind subjects. J. type with disturbed sleep-waking. Biol. Psychiatry 45, 417–421.
Endocrinol. 164, R1–R6. Modabbernia, A., Taslimi, S., Brietzke, E., Ashrafi, M., 2013. Cytokine alterations in
Lopresti, A.L., Drummond, P.D., 2017. Efficacy of curcumin, and a saffron/curcumin bipolar disorder: a meta-analysis of 30 studies. Biol. Psychiatry 24, 15–25. http://dx.
combination for the treatment of major depression: a randomised, double-blind, doi.org/10.1016/j.biopsych.2013.01.007.
placebo-controlled study. J. Affect. Disord. 207, 188–196. http://dx.doi.org/10. Montastruc, F., Scotto, S., Vaz, I.R., Guerra, L.N., Escudero, A., Sainz, M., Falomir, T.,
1016/j.jad.2016.09.047. Bagheri, H., Herdeiro, M.T., Venegoni, M., Montastruc, J.L., Carvajal, A., Sáinz, M.,
Lopresti, A.L., Maes, M., Maker, G.L., Hood, S.D., Drummond, P.D., 2014. Curcumin for Falomir, T., Bagheri, H., Herdeiro, M.T., Venegoni, M., Montastruc, J.L., Carvajal, A.,
the treatment of major depression: a randomised, double-blind, placebo controlled 2014. Hepatotoxicity related to agomelatine and other new antidepressants: a case/
study. J. Affect. Disord. 167, 368–375. http://dx.doi.org/10.1016/j.jad.2014.06.001. noncase approach with information from the Portuguese, French, Spanish, and Italian
Louveau, A., Smirnov, I., Keyes, T.J., Eccles, J.D., Rouhani, S.J., Peske, J.D., Derecki, pharmacovigilance systems. J. Clin. Psychopharmacol. 34, 327–330. http://dx.doi.
N.C., Castle, D., Mandell, J.W., Lee, K.S., Harris, T.H., Kipnis, J., 2015. Structural and org/10.1097/JCP.0000000000000094.
functional features of central nervous system lymphatic vessels. Nature 523, Moreira, J., Geoffroy, P.A., 2016. Lithium and bipolar disorder: impacts from molecular
337–341. http://dx.doi.org/10.1038/nature14432. to behavioural circadian rhythms. Chronobiol. Int. 33, 35–373.
Lowrey, P.L., Takahashi, J.S., 2004. Mammalian circadian biology: elucidating genome- Moylan, S., Berk, M., Dean, O.M., Samuni, Y., Williams, L.J., O'Neil, A., Hayley, A.C.,
wide levels of temporal organization. Annu. Rev. Genomics Hum. Genet. 5, 407–441. Pasco, J.A., Anderson, G., Jacka, F.N., Maes, M., 2014. Oxidative & nitrosative stress
http://dx.doi.org/10.1146/annurev.genom.5.061903.175925. in depression: why so much stress? Neurosci. Biobehav. Rev. 45, 46–62. http://dx.
Macintyre, D.J., McGhee, K.A., Maclean, A.W., Afzal, M., Briffa, K., Henry, B., Thomson, doi.org/10.1016/j.neubiorev.2014.05.007.
P.A., Muir, W.J., Blackwood, D.H., 2010. Association of GPR50, an X-linked orphan G Munkholm, K., Brauner, J.V., Kessing, L.V., Vinberg, M., 2013a. Cytokines in bipolar
protein-coupled receptor, and affective disorder in an independent sample of the disorder vs. healthy control subjects: a systematic review and meta-analysis. J.
Scottish population. Neurosci. Lett. 475, 169–173. http://dx.doi.org/10.1016/j. Psychiatr. Res. 47, 1119–1133. http://dx.doi.org/10.1016/j.jpsychires.2013.05.018.
neulet.2010.03.072. Munkholm, K., Vinberg, M., Kessing, L.V., 2013b. Cytokines in bipolar disorder: a
MacIsaac, S.E., Carvalho, A.F., Cha, D.S., Mansur, R.B., McIntyre, R.S., 2014. The systematic review and meta-analysis. J. Affect. Disord. 144, 16–27. http://dx.doi.
mechanism, efficacy, and tolerability profile of agomelatine. Expert Opin. org/10.1016/j.jad.2012.06.010.
Pharmacother. 15, 259–274. http://dx.doi.org/10.1517/14656566.2014.862233. Munkholm, K., Weikop, P., Kessing, L.V., Vinberg, M., 2015. Elevated levels of IL-6 and
Maestroni, G.J.M., Cardinali, D.P., Esquifino, A.I., Pandi-Perumal, S.R., 2005. Does IL-18 in manic and hypomanic states in rapid cycling bipolar disorder patients. Brain
melatonin play a disease-promoting role in rheumatoid arthritis? J. Neuroimmunol. Behav. Immun. 43, 205–213. http://dx.doi.org/10.1016/j.bbi.2014.09.021.
158, 106–111. Murphy, M.L., Frodl, T., 2011. Meta-analysis of diffusion tensor imaging studies shows
Malykhin, N.V., Coupland, N.J., 2015. Hippocampal neuroplasticity in major depressive altered fractional anisotropy occurring in distinct brain areas in association with
disorder. Neuroscience 309, 200–213. http://dx.doi.org/10.1016/j.neuroscience. depression. Biol. Mood Anxiety Disord. 1, 3.
2015.04.047. Naismith, S.L., Hermens, D.F., Ip, T.K.C., Bolitho, S., Scott, E., Rogers, N.L., Hickie, I.B.,
Mansour, H.A., Wood, J., Logue, T., Chowdari, K.V., Dayal, M., Kupfer, D.J., Monk, T.H., 2012. Circadian profiles in young people during the early stages of affective disorder.
Devlin, B., Nimgaonkar, V.L., 2006. Association study of eight circadian genes with Transl. Psychiatry 2, e123. http://dx.doi.org/10.1038/tp.2012.47.
bipolar I disorder, schizoaffective disorder and schizophrenia. Genes. Brain. Behav. 5, Nakahata, Y., Kaluzova, M., Grimaldi, B., Sahar, S., Hirayama, J., Chen, D., Guarente,
150–157. http://dx.doi.org/10.1111/j.1601-183X.2005.00147.x. L.P., Sassone-Corsi, P., 2008. The NAD+-dependent deacetylase SIRT1 modulates
Martinotti, G., Sepede, G., Gambi, F., Di Iorio, G., De Berardis, D., Di Nicola, M., Onofrj, CLOCK-mediated chromatin remodeling and circadian control. Cell 134, 329–340.
M., Janiri, L., Di Giannantonio, M., 2012. Agomelatine versus venlafaxine XR in the Nathan, P.J., Burrows, G.D., Norman, T.R., 1999. Melatonin sensitivity to dim white light
treatment of anhedonia in major depressive disorder: a pilot study. J. Clin. in affective disorders. Neuropsychopharmacology 21, 408–413.
Psychopharmacol. 32, 487–491. http://dx.doi.org/10.1097/JCP. Nechita, F., Pirlog, M.-C., Chiriţă, A.L., 2015. Circadian malfunctions in depression -
0b013e31825d6c25. neurobiological and psychosocial approaches. [review]. Romanian J. Morphol.
Martynhak, B.J., Pereira, M., de Souza, C.P., Andreatini, R., 2015. Stretch, shrink, and Embryol. 56, 949–955.
shatter the rhythms: the intrinsic circadian period in mania and depression. CNS Nemeroff, C.B., Weinberger, D., Rutter, M., MacMillan, H.L., Bryant, R.A., Wessely, S.,
Neurol. Disord. Drug Targets 14, 963–969. Stein, D.J., Pariante, C.M., Seemüller, F., Berk, M., Malhi, S., Preisig, M., Brüne, M.,
Maurya, P.K., Noto, C., Rizzo, L.B., Rios, A.C., Nunes, S.O.V., Barbosa, D.S., Sethi, S., Zeni, Lysaker, P., 2013. Forum. Current controversires in psychiatry: DSM-5: a collection of
M., Mansur, R.B., Maes, M., Brietzke, E., 2016. The role of oxidative and nitrosative psychiatrist views on the changes, controversies, and future directions. BMC Med. 11,
stress in accelerated aging and major depressive disorder. Prog. Neuro- 1–19.
Psychopharmacol. Biol. Psychiatry 65, 134–144. http://dx.doi.org/10.1016/j.pnpbp. Nicolaides, N.C., Charmandari, E., Chrousos, G.P., Kino, T., 2014. Circadian endocrine
2015.08.016. rhythms: The hypothalamic-pituitary-adrenal axis and its actions. Ann. N. Y. Acad.
McClung, C.A., 2013. How might circadian rhythms control mood? Let me count the Sci. 1318, 71–80. http://dx.doi.org/10.1111/nyas.12464.
ways. Biol. Psychiatry 74, 242–249. http://dx.doi.org/10.1016/j.biopsych.2013.02. Nievergelt, C.M., Kripke, D.F., Barrett, T.B., Burg, E., Remick, R.A., Sadovnick, A.D.,
019. McElroy, S.L., Keck Jr., P.E., Schork, N.J., Kelsoe, J.R., 2006. Suggestive evidence for
McIntyre, R.S., Cha, D.S., Soczynska, J.K., Woldeyohannes, H.O., Gallaugher, L.A., association of the circadian genes PERIOD3 and ARNTL with bipolar disorder. Am. J.
Kudlow, P., Alsuwaidan, M., Baskaran, A., 2013. Cognitive deficits and functional Med. Genet. B Neuropsychiatr. Genet. 141B, 234–241. http://dx.doi.org/10.1002/
outcomes in major depressive disorder: determinants, substrates, and treatment ajmg.b.30252.
interventions. Depress. Anxiety 30, 515–527. http://dx.doi.org/10.1002/da.22063. Nussbaumer, B., Kaminski-Hartenthaler, A., Forneris, C.A., Morgan, L.C., Sonis, J.H.,
McIntyre, R.S., Lee, Y., 2016. Cognition in major depressive disorder: a “Systemically Gaynes, B.N., Greenblatt, A., Wipplinger, J., Lux, L.J., Winkler, D., Van Noord, M.G.,
Important Functional Index” (SIFI). Curr. Opin. Psychiatry 29, 48–55. http://dx.doi. Hofmann, J., Gartiehner, G., 2015. Light therapy for preventing seasonal affective
org/10.1097/YCO.0000000000000221. disorder. Cochrane Database Syst. Rev. 11.
McIntyre, R.S., Soczynska, J.Z., Woldeyohannes, H.O., Alsuwaidan, M.T., Cha, D.S., O'Connor, S., Agius, M., 2015. A systematic review of structural and functional MRI
Carvalho, A.F., Jerrell, J.M., Dale, R.M., Gallaugher, L.A., Muzina, D.J., Kennedy, differences between psychotic and nonpsychotic depression. Psychiatr. Danub. 27,

202
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

S235–S239. http://dx.doi.org/10.1016/S0924-9338(14)78463-9. N. Am. 39, 125–137. http://dx.doi.org/10.1016/j.psc.2015.09.006.


Oldham, M.A., Ciraulo, D.A., 2014. Bright light therapy for depression: a review of its Rothschild, A.J., 2015. A blood test for depression? J. Clin. Psychiatry 76, e218–e219.
effects on chronobiology and the autonomic nervous system. Chronobiol. Int. 31, http://dx.doi.org/10.4088/JCP.14com09515.
305–319. Rubin, R.T., Heist, E.K., McGeoy, S.S., Hanada, K., Lesser, I.M., 1992. Neuroendocrine
Owen, R.T., 2009. Agomelatine: a novel pharmacological approach to treating aspects of primary endogenous depression. XI. Serum melatonin measures in patients
depression. Drugs Today (Barc) 45, 599–608. and matched control subjects. Arch. Gen. Psychiatry 49, 558–567.
Pace, T.W., Miller, A.H., 2009. Cytokines and glucocorticoid receptor signaling. Sachar, E.J., Hellman, L., Roffwarg, H.P., Halpern, F.S., Fukushima, D.K., Gallagher, T.F.,
Relevance to major depression. Ann. N. Y. Acad. Sci. 1179, 86–105. http://dx.doi. 1973. Disrupted 24-hour patterns of cortisol secretion in psychotic depression. Arch.
org/10.1111/j.1749-6632.2009.04984.x. Gen. Psychiatry 28, 19–24.
Pail, G., Huf, W., Pjrek, E., Winkler, D., Willeit, M., Praschak-Rieder, N., Kasper, S., 2011. Sanmukhani, J., Satodia, V., Trivedi, J., Patel, T., Tiwari, D., Panchal, B., Goel, A.,
Bright-light therapy in the treatment of mood disorders. Neuropsychobiology 64, Tripathi, C.B., 2014. Efficacy and safety of curcumin in major depressive disorder: a
152–162. randomized controlled trial. Phytother. Res. 28, 579–585. http://dx.doi.org/10.
Palta, P., Samuel, L.J., Miller 3rd, E.R., Szanton, S.L., 2014. Depression and oxidative 1002/ptr.5025.
stress: results from a meta-analysis of observational studies. Psychosom. Med. 76, Sarris, J., Mischoulon, D., Schweitzer, I., 2012. Omega-3 for bipolar disorder: meta-
12–19. http://dx.doi.org/10.1097/PSY.0000000000000009. analyses of use in mania and bipolar depression. J. Clin. Psychiatry 73, 81–86. http://
Pandi-Perumal, S.R., Trakht, I., Srinivasan, V., Spence, D.W., Poeggeler, B., Hardeland, R., dx.doi.org/10.4088/JCP.10r06710.
Cardinali, D.P., 2009. The effect of melatonergic and non-melatonergic Sasseville, A., Benhaberou-Brun, D., Fontaine, C., Charon, M.-C., Hebert, M., 2009.
antidepressants on sleep: weighing the alternatives. World J. Biol. Psychiatry 10, Wearing blue-blockers in the morning could improve sleep of workers on a
342–354. permanent night schedule: a pilot study. Chronobiol. Int. 26, 913–925. http://dx.doi.
Papakostas, G.I., Shelton, R.C., Henry, M.E., Bilello, J.A., 2015. Letters to the Editor. J. org/10.1080/07420520903044398.
Clin. Psychiatry 76, e1038. http://dx.doi.org/10.1176/appi.ajp.2015.15060750. Sasseville, A., Hébert, M., 2010. Using blue-green light at night and blue-blockers during
Partonen, T., 2014. Circadian clock proteins in mood regulation. Front. Psychiatry 5, 195. the day to improves adaptation to night work: a pilot study. Prog.
Partonen, T., Treutlein, J., Alpman, A., Frank, J., Johansson, C., Depner, M., Aron, L., Neuropsychopharmacol. Biol. Psychiatry 34, 1236–1242. http://dx.doi.org/10.1016/
Rietschel, M., Wellek, S., Soronen, P., Paunio, T., Koch, A., Chen, P., Lathrop, M., j.pnpbp.2010.06.027.
Adolfsson, R., Persson, M.L., Kasper, S., Schalling, M., Peltonen, L., Schumann, G., Sasseville, A., Paquet, N., Sévigny, J., Hébert, M., 2006. Blue blocker glasses impede the
2007. Three circadian clock genes Per2, Arntl, and Npas2 contribute to winter capacity of bright light to suppress melatonin production. J. Pineal Res. 41, 73–78.
depression. Ann. Med. 39, 229–238. http://dx.doi.org/10.1111/j.1600-079X.2006.00332.x.
Penders, T.M., Stanciu, C.N., Schoemann, A.M., Ninan, P.T., Bloch, R., Saeed, S.A., 2016. Saus, E., Soria, V., Escaramís, G., Vivarelli, F., Crespo, J.M., Kagerbauer, B., Menchòn,
Bright light therapy as augmentation of pharmacotherapy for treatment of J.M., Urretavizcaya, M., Gratacòs, M., Estivill, X., 2010. Genetic variants and
depression: a systematic review and meta-analysis. Prim. Care Companion CNS abnormal processing of pre-miR-182, a circadian clock modulator, in major
Disord. 18. http://dx.doi.org/10.4088/PCC.15r01906. depression patients with late insomnia. Hum. Mol. Genet. 19, 4017–4025. http://dx.
Prasko, J., 2008. Bright light therapy. Neuroendocrinol. Lett. 29, 33–64. doi.org/10.1093/hmg/ddq316.
Price, J.L., Drevets, W.C., 2012. Neural circuits underlying the pathophysiology of mood Saxvig, I.W., Wilhelmsen-Langeland, A., Pallesen, S., Vedaa, Ø., Nordhus, I.-H., Bjorvatn,
disorders. Trends Cogn. Sci. 16, 61–71. http://dx.doi.org/10.1016/j.tics.2011.12. B., 2014. A randomized controlled trial with bright light and melatonin for delayed
011. sleep phase disorder: effects on subjective and objective sleep. Chronobiol. Int. 31,
Price, J.L., Drevets, W.C., 2010. Neurocircuitry of mood disorders. 72–86.
Neuropsychopharmacology 35, 192–216. http://dx.doi.org/10.1038/npp.2009.104. Schnell, A., Albrecht, U., Sandrelli, F., 2014. Rhythm and mood: relationships between
Qin, J., Shen, H., Zeng, L.-L., Jiang, W., Liu, L., Hu, D., 2015. Predicting clinical responses the circadian clock and mood-related behavior. Behav. Neurosci. 128, 326–343.
in major depression using intrinsic functional connectivity. Neuroreport 26, 675–680. http://dx.doi.org/10.1037/a0035883.
http://dx.doi.org/10.1097/WNR.0000000000000407. Schwartz, R.S., Olds, J., 2015. The psychiatry of light. Harv. Rev. Psychiatry 23, 188–194.
Racagni, G., Riva, M.A., Molteni, R., Musazzi, L., Calabrese, F., Popoli, M., Tardito, D., Serfaty, M.A., Osborne, D., Buszewicz, M.J., Blizard, R., Raven, P.W., 2010. A randomized
2011. Mode of action of agomelatine: synergy between melatonergic and 5-HT2C double-blind placebo-controlled trial of treatment as usual plus exogenous slow-
receptors. World J. Biol. Psychiatry 12, 574–587. http://dx.doi.org/10.3109/ release melatonin (6 mg) or placebo for sleep disturbance and depressed mood. Int.
15622975.2011.595823. Clin. Psychopharmacol. 25, 132–142.
Rahman, S.A., Shapiro, C.M., Wang, F., Ainlay, H., Kazmi, S., Brown, T.J., Casper, R.F., Setiawan, E., Wilson, A.A., Mizrahi, R., Rusjan, P.M., Miler, L., Rajkowska, G., Suridjan, I.,
2013. Effects of filtering visual short wavelengths during nocturnal shiftwork on sleep Kennedy, J.L., Rekkas, P.V., Houle, S., Meyer, J.H., 2015. Role of translocator protein
and performance. Chronobiol. Int. 30, 951–962. http://dx.doi.org/10.3109/ density, a marker of neuroinflammation, in the brain during major depressive
07420528.2013.789894. episodes. JAMA Psychiatry 72, 268–275. http://dx.doi.org/10.1001/jamapsychiatry.
Raison, C.L., Rutherford, R.E., Woolwine, B.J., Shuo, C., Schettler, P., Drake, D.F., 2014.2427.
Haroon, E., Miller, A.H., 2013. A randomized controlled trial of the tumor necrosis Sexton, C.E., Mackay, C.E., Ebmeier, K.P., 2009. A systematic review of diffusion tensor
factor antagonist infliximab for treatment-resistant depression: the role of baseline imaging studies in affective disorders. Biol. Psychiatry 66, 814–823. http://dx.doi.
inflammatory biomarkers. JAMA Psychiatry 70, 31–41. http://dx.doi.org/10.1001/ org/10.1016/j.biopsych.2009.05.024.
2013.jamapsychiatry.4. Sexton, C.E., MacKay, C.E., Ebmeier, K.P., 2013. A systematic review and meta-analysis of
Rajendran, B., Janakarajan, V.N., 2016. Circadian clock gene aryl hydrocarbon receptor magnetic resonance imaging studies in late-life depression. Am. J. Geriatr. Psychiatry
nuclear translocator-like polymorphisms are associated with seasonal affective 21, 184–195. http://dx.doi.org/10.1016/j.jagp.2012.10.019.
disorder: an Indian family study. Indian J. Psychiatry 58, 57–60. Shearman, L.P., Sriram, S., Weaver, D.R., Maywood, E.S., Chaves, I., Zheng, B., Kume, K.,
Rao, J.S., Harry, G.J., Rapoport, S.I., Kim, H.W., 2010. Increased excitotoxicity and Lee, C.C., van der Horst, G.T., Hastings, M.H., Reppert, S.M., 2000. Interacting
neuroinflammatory markers in postmortem frontal cortex from bipolar disorder molecular loops in the mammalian circadian clock. Science 288, 1013–1019. http://
patients. Mol. Psychiatry 15, 384–392. http://dx.doi.org/10.1038/mp.2009.47. dx.doi.org/10.1126/science.288.5468.1013.
Rapaport, M.H., Nierenberg, A.A., Schettler, P.J., Kinkead, B., Cardoos, A., Walker, R., Shi, S.Q., White, M.J., Borsetti, H.M., Pendergast, J.S., Hida, A., Ciarleglio, C.M., de
Mischoulon, D., 2016. Inflammation as a predictive biomarker for response to omega- Verteuil, P.A., Cadar, A.G., Cala, C., McMahon, D.G., Shelton, R.C., Williams, S.M.,
3 fatty acids in major depressive disorder: a proof-of-concept study. Mol. Psychiatry Johnson, C.H., 2016. Molecular analyses of circadian gene variants reveal sex-
21, 71–79. http://dx.doi.org/10.1038/mp.2015.22. dependent links between depression and clocks. Transl. Psychiatry 6, e748. http://dx.
Rastad, C., Ulfberg, J., Lindberg, P., 2011. Improvement in fatigue, sleepiness, and health- doi.org/10.1038/tp.2016.9.
related quality of life with bright light treatment in persons with seasonal affective Singer, C., Tractenberg, R.E., Kaye, J., Schafer, K., Gamst, A., Grundman, M., Thomas, R.,
disorder and subsyndromal SAD. Depress. Res. Treat. http://dx.doi.org/10.1155/ Thal, L.J., 2003. A multicenter, placebo-controlled trial of melatonin for sleep
2011/543906. disturbance in Alzheimer's disease. Sleep 26, 893–901.
Robertson, J.M., Tanguay, P.E., 1997. Case study: the use of melatonin in a boy with Singh, S.P., Singh, V., Kar, N., 2012. Efficacy of agomelatine in major depressive disorder:
refractory bipolar disorder. J. Am. Acad. Child Adolesc. Psychiatry 36, 822–825. meta-analysis and appraisal. Int. J. Neuropsychopharmacol. 15, 417–428.
Rock, P.L., Roiser, J.P., Riedel, W.J., Blackwell, A.D., 2014. Cognitive impairment in Sjöholm, L.K., Backlund, L., Cheteh, E.H., Ek, I.R., Frisén, L., Schalling, M., Ösby, U.,
depression: a systematic review and meta-analysis. Psychol. Med. 44, 2029–2040. Lavebratt, C., Nikamo, P., 2010. CRY2 is associated with rapid cycling in bipolar
http://dx.doi.org/10.1017/S0033291713002535. disorderpatients. PLoS One 5, 1–6. http://dx.doi.org/10.1371/journal.pone.
Roiser, J.P., Sahakian, B.J., 2013. Hot and cold cognition in depression. CNS Spectr. 18, 0012632.
139–149. http://dx.doi.org/10.1017/S1092852913000072. Skene, D.J., 2003. Optimization of light and melatonin to phase-shift human circadian
Rosenblat, J.D., Brietzke, E., Mansur, R.B., Maruschak, N.A., Lee, Y., McIntyre, R.S., 2015. rhythms. J. Neuroendocrinol. 15, 438–441.
Inflammation as a neurobiological substrate of cognitive impairment in bipolar Skene, D.J., Deacon, S., Arendt, J., 1996. Use of melatonin in circadian rhythm disorders
disorder: evidence, pathophysiology and treatment implications. J. Affect. Disord. and following phase shifts. Acta Neurobiol. Exp. (Wars) 56, 359–362.
188, 149–159. http://dx.doi.org/10.1016/j.jad.2015.08.058. Smith, M.R., Eastman, C.I., 2009. Phase delaying the human circadian clock with blue-
Rosenblat, J.D., Cha, D.S., Mansur, R.B., McIntyre, R.S., 2014. Inflamed moods: a review enriched polychromatic light. Chronobiol. Int. 26, 709–725. http://dx.doi.org/10.
of the interactions between inflammation and mood disorders. Prog. Neuro- 1080/07420520902927742.
Psychopharmacol. Biol. Psychiatry 53, 23–34. http://dx.doi.org/10.1016/j.pnpbp. Smith, M.R., Revell, V.L., Eastman, C.I., 2009. Phase advancing the human circadian
2014.01.013. clock with blue-enriched polychromatic light. Sleep Med. 10, 287–294.
Rosenblat, J.D., Gregory, J.M., McIntyre, R.S., 2016. Pharmacologic implications of Soria, V., Martinez-Amoros, E., Escaramis, G., Valero, J., Crespo, J.M., Gutierrez-Zotes,
inflammatory comorbidity in bipolar disorder. Curr. Opin. Pharmacol. 29, 63–69. A., Bayes, M., Martorell, L., Vilella, E., Estivill, X., Menchon, J.M., Gratacos, M.,
http://dx.doi.org/10.1016/j.coph.2016.06.007. Urretavizcaya, M., 2010a. Resequencing and association analysis of arylalkylamine
Rosenblat, J.D., McIntyre, R.S., 2016. Bipolar disorder and inflammation. Psychiatr. Clin. N-acetyltransferase (AANAT) gene and its contribution to major depression

203
G.M. Brown et al. Progress in Neuropsychopharmacology & Biological Psychiatry 80 (2018) 189–204

susceptibility. J. Pineal Res. 49, 35–44. http://dx.doi.org/10.1111/j.1600-079X. Cochrane Database Syst. Rev, CD004050. http://dx.doi.org/10.1002/14651858.
2010.00763.x. CD004050.pub2.
Soria, V., Martínez-Amorós, E., Escaramís, G., Valero, J., Pérez-Egea, R., García, C., Tymofiyeva, O., Connolly, C.G., Ho, T.C., Sacchet, M.D., Henje Blom, E., LeWinn, K.Z.,
Gutiérrez-Zotes, A., Puigdemont, D., Bayés, M., Crespo, J.M., Martorell, L., Vilella, E., Xu, D., Yang, T.T., 2017. DTI-based connectome analysis of adolescents with major
Labad, A., Vallejo, J., Pérez, V., Menchón, J.M., Estivill, X., Gratacòs, M., depressive disorder reveals hypoconnectivity of the right caudate. J. Affect. Disord.
Urretavizcaya, M., 2010b. Differential association of circadian genes with mood 207, 18–25. http://dx.doi.org/10.1016/j.jad.2016.09.013.
disorders: CRY1 and NPAS2 are associated with unipolar major depression and Uher, R., Payne, J.L., Pavlova, B., Perlis, R.H., 2014. Major depressive disorder in DSM-5:
CLOCK and VIP with bipolar disorder. Neuropsychopharmacology 35, 1279–1289. implications for clinical practice and research of changes from DSM-IV. Depress.
http://dx.doi.org/10.1038/npp.2009.230. Anxiety 31, 459–471. http://dx.doi.org/10.1002/da.22217.
Soria, V., Urretavizcaya, M., 2009. Circadian rhythms and depression. Actas Españolas van der Lely, S., Frey, S., Garbazza, C., Wirz-Justice, A., Jenni, O.G., Steiner, R., Wolf, S.,
Psiquiatr. 37, 222–232. Cajochen, C., Bromundt, V., Schmidt, C., 2015. Blue blocker glasses as a
Souetre, E., Salvati, E., Belugou, J.L., Pringuey, D., Candito, M., Krebs, B., Ardisson, J.L., countermeasure for alerting effects of evening light-emitting diode screen exposure in
Darcourt, G., Souêtre, E., Salvati, E., Belugou, J.L., Pringuey, D., Candito, M., Krebs, male teenagers. J. Adolesc. Health 56, 113–119. http://dx.doi.org/10.1016/j.
B., Ardisson, J.L., Darcourt, G., 1989. Circadian rhythms in depression and recovery: jadohealth.2014.08.002.
evidence for blunted amplitude as the main chronobiological abnormality. Psychiatry Videnovic, A., Lazar, A.S., Barker, R.A., Overeem, S., 2014. “The clocks that time
Res. 28, 263–278. http://dx.doi.org/10.1016/0165-1781(89)90207-2. us”—circadian rhythms in neurodegenerative disorders. Nat. Rev. Neurol. 10,
Srinivasan, V., Pandi-Perumal, S.R., Cardinali, D.P., Poeggeler, B., Hardeland, R., 2006. 683–693.
Melatonin in Alzheimer's disease and other neurodegenerative disorders. Behav. Voican, C.S., Corruble, E., Naveau, S., Perlemuter, G., 2014. Antidepressant-induced liver
Brain Funct. 2, 15. injury: a review for clinicians. Am. J. Psychiatry 171, 404–415. http://dx.doi.org/10.
Srinivasan, V., Pandi-Perumal, S.R., Trahkt, I., Spence, D.W., Poeggeler, B.H., Hardeland, 1176/appi.ajp.2013.13050709.
R., Cardinali, D.P., 2009a. Melatonin and melatonergic drugs on sleep: possible Vreeburg, S.A., Hoogendijk, W.J., van Pelt, J., Verhagen, J.C., van Dyck, R., Smit, J.H.,
mechanisms of action. Int. J. Neurosci. 119, 821–846. Zitman, F.G., Penninx, B.W., 2009. Major depressive disorder and hypothalamic-
Srinivasan, V., Pandi-Perumal, S.R., Trakht, I., Spence, D.W., Hardeland, R., Poeggeler, B., pituitary-adrenal Axis activity. Arch. Gen. Psychiatry 66, 617–626.
Cardinali, D.P., 2009b. Pathophysiology of depression: role of sleep and the Vrshek-Schallhorn, S., Doane, L.D., Mineka, S., Zinbarg, R.E., Craske, M.G., Adam, E.K.,
melatonergic system. Psychiatry Res. 165, 201–214. 2013. The cortisol awakening response predicts major depression: predictive stability
Stertz, L., Magalhaes, P.V., Kapczinski, F., 2013. Is bipolar disorder an inflammatory over a 4-year follow-up and effect of depression history. Psychol. Med. 43, 483–493.
condition? The relevance of microglial activation. Curr. Opin. Psychiatry 26, 19–26. http://dx.doi.org/10.1017/S0033291712001213.
http://dx.doi.org/10.1097/YCO.0b013e32835aa4b4. Wang, J., Dunn, A.J., 1998. Mouse interleukin-6 stimulates the HPA axis and increases
Stewart, J.W., Halbreich, U., 1989. Plasma melatonin levels in depressed patients before brain tryptophan and serotonin metabolism. Neurochem. Int. 33, 143–154.
and after treatment with antidepressant medication. Biol. Psychiatry 25, 33–38. Wessa, M., Lois, G., 2015. Brain functional effects of psychopharmacological treatment in
Stratmann, M., Konrad, C., Kugel, H., Krug, A., Schöning, S., Ohrmann, P., Uhlmann, C., major depression: a focus on neural circuitry of affective processing. Curr.
Postert, C., Suslow, T., Heindel, W., Arolt, V., Kircher, T., Dannlowski, U., 2014. Neuropharmacol. 13, 466–479. http://dx.doi.org/10.2174/
Insular and hippocampal gray matter volume reductions in patients with major 1570159X13666150416224801.
depressive disorder. PLoS One 9. http://dx.doi.org/10.1371/journal.pone.0102692. Whalley, L.J., Perini, T., Shering, A., Bennie, J., 1991. Melatonin response to bright light
Suzuki, M., Dallaspezia, S., Locatelli, C., Uchiyama, M., Colombo, C., Benedetti, F., 2016. in recovered, drug-free, bipolar patients. Psychiatry Res. 38, 13–19.
Discrepancy between subjective and objective severity as a predictor of response to Wilhelmsen-Langeland, A., Saxvig, I.W., Pallesen, S., Nordhus, I.-H., Vedaa, Ø.,
chronotherapeutics in bipolar depression. J. Affect. Disord. 204, 48–53. Lundervold, A.J., Bjorvatn, B., 2013. A randomized controlled trial with bright light
Swardfager, W., Rosenblat, J.D., Benlamri, M., McIntyre, R.S., 2016. Mapping and melatonin for the treatment of delayed sleep phase disorder: effects on subjective
inflammation onto mood: inflammatory mediators of anhedonia. Neurosci. Biobehav. and objective sleepiness and cognitive function. J. Biol. Rhythm. 28, 306–321.
Rev. 64, 148–166. http://dx.doi.org/10.1016/j.neubiorev.2016.02.017. Wright, C.E., Strike, P.C., Brydon, L., Steptoe, A., 2005. Acute inflammation and negative
Taylor, D., Sparshatt, A., Varma, S., Olofinjana, O., 2014. Antidepressant efficacy of mood: mediation by cytokine activation. Brain Behav. Immun. 19, 345–350. http://
agomelatine: meta-analysis of published and unpublished studies. BMJ 348, g1888. dx.doi.org/10.1016/j.bbi.2004.10.003.
http://dx.doi.org/10.1136/bmj.g1888. Wu, J.C., Kelsoe, J.R., Schachat, C., Bunney, B.G., DeModena, A., Golshan, S., Gillin, J.C.,
Terman, M., Terman, J.S., 2006. Controlled trial of naturalistic dawn simulation and Potkin, S.G., Bunney, W.E., 2009. Rapid and sustained antidepressant response with
negative air ionization for seasonal affective disorder. Am. J. Psychiatry 163, sleep deprivation and chronotherapy in bipolar disorder. Biol. Psychiatry 66,
2126–2133. 298–301.
Thompson, C., Franey, C., Arendt, J., Checkley, S.A., 1988. A comparison of melatonin Young, J.J., Bruno, D., Pomara, N., 2014. A review of the relationship between
secretion in depressed patients and normal subjects. Br. J. Psychiatry 152, 260–265. proinflammatory cytokines and major depressive disorder. J. Affect. Disord. 169,
Thomson, P.A., Wray, N.R., Thomson, A.M., Dunbar, D.R., Grassie, M.A., Condie, A., 15–20. http://dx.doi.org/10.1016/j.jad.2014.07.032.
Walker, M.T., Smith, D.J., Pulford, D.J., Muir, W., Blackwood, D.H., Porteous, D.J., Zawilska, J.B., Skene, D.J., Arendt, J., 2009. Physiology and pharmacology of melatonin
2005. Sex-specific association between bipolar affective disorder in women and in relation to biological rhythms. Pharmacol. Rep. 61, 383–410.
GPR50, an X-linked orphan G protein-coupled receptor. Mol. Psychiatry 10, 470–478. Zhang, J., Terreni, L., De Simoni, M.G., Dunn, A.J., 2001. Peripheral interleukin-6
Turnbull, A.V., Rivier, C.L., 1999. Regulation of the hypothalamic-pituitary-adrenal axis administration increases extracellular concentrations of serotonin and the evoked
by cytokines: actions and mechanisms of action. Physiol. Rev. 79, 1–71. release of serotonin in the rat striatum. Neurochem. Int. 38, 303–308.
Tuunainen, A., Kripke, D.F., Endo, T., 2004. Light therapy for non-seasonal depression.

204

You might also like