In: Serotonergic Systems ISBN: 978-1-62618-287-5 Editor: Reyna O. Villanueva © 2013 Nova Science Publishers, Inc.

No part of this digital document may be reproduced, stored in a retrieval system or transmitted commercially in any form or by any means. The publisher has taken reasonable care in the preparation of this digital document, but makes no expressed or implied warranty of any kind and assumes no responsibility for any errors or omissions. No liability is assumed for incidental or consequential damages in connection with or arising out of information contained herein. This digital document is sold with the clear understanding that the publisher is not engaged in rendering legal, medical or any other professional services.

Chapter I

Interactions of the Serotonergic and Circadian Systems in Health and Disease

Marc Cuesta,1, 2 and Etienne Challet3 1Centre for Study and Treatment of Circadian Rhythms, 2Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada 3Department Neurobiology of Rhythms, Institute of Cellular and Integrative Neurosciences, University of Strasbourg, Strasbourg, France

Abstract

The serotonergic and circadian systems constitute two of the main regulatory signaling networks of the brain. Each of them consists of a highly localized neuronal population that exerts a widespread influence

 Corresponding author: Marc Cuesta, Ph.D. Centre for Study and Treatment of Circadian Rhythms and Laboratory of Molecular Chronobiology Douglas Mental Health University Institute; Department of Psychiatry, McGill University; 6875 LaSalle Boulevard; Montreal (Quebec) H4H 1R3 CANADA; Phone: (514) 761-6131 ext. 2395; Fax: (514) 888-4099; [email protected]. 2 Marc Cuesta and Etienne Challet

on multiple cognitive, behavioral, physiological and molecular functions. Anatomically, these two systems are tightly interconnected and influence each other. In , the circadian system is composed of a central located in the suprachiasmatic nuclei (SCN) of the and peripheral oscillators found in the brain and throughout the body, which are subordinate to the SCN. This circadian organization allows the rhythmic regulation of various physiological functions in a harmonious fashion, including the circadian regulation of serotonin (5-hydroxytryptamine or 5-HT) synthesis and release. The serotonergic system comprises several interconnected nuclei, located in the brainstem, and through its neuronal projections that mainly release 5- HT, regulates numerous functions, such as sleep and wake states, pain, thermoregulation and circadian rhythms. In this chapter, we first define the circadian system and its serotonergic component, as well as the rhythmic regulation of the serotonergic system. Then, we review the serotonergic modulation of the circadian system, including its direct effects as a non-photic factor and its ability to modulate the photic synchronization of the central clock. In the last part, we analyze the potential relationships of the serotonin-circadian interaction with mood disorders.

The Circadian Timing System

From the Earth’s Rotation to Biological Rhythms

Every day, living organisms undergo rhythmic variations of a multitude of biological parameters, such as gene and protein production, physiological functions, behaviors and cognitive processes. These daily variations clearly follow the 24-h light-dark (LD) cycle due to Earth’s rotation. In 1729, the French astronomer de Mairan demonstrated that rhythmic variations are not passive responses to the changing outside world. Indeed, he was the first to report that the daily leaf movements of the sensitive plant (Mimosa pudica) persist in constant darkness (DD), proving their endogenous origin [1].This kind of intrinsic oscillations, ubiquitous in plants and , allows species to anticipate environmental rhythmic variations in a coordinated fashion, and is driven by the circadian system (from the Latin circa and dies, meaning “about”and “one day”, respectively). This system is composed of clocks, found in brain and non-brain tissues. Circadian clocks have been identified in most living organisms from cyanobacteria to humans and fulfill the following properties: they have the ability to generate endogenous rhythms under Interactions of the Serotonergic and Circadian Systems ... 3 constant conditions and to convey them to other structures; they are synchronized or entrained by external and internal cues (called zeitgeber or ZT from the German, meaning “time-giver”), the most potent being the light-dark cycle; they are temperature compensated (i.e. the rhythmicity remains constant even if temperature increases or decreases across a physiological range) [2].

The Central Circadian Clock and its Ability to Generate Rhythmicity

In mammals, the main actor of the circadian system is the central or “master” circadian clock located in the suprachiasmatic nuclei (SCN) of the hypothalamus. This paired structure, containing around 10,000 neurons per nucleus, comprises a ventral “core” region and a dorsal “shell” region [3]. In most SCN neurons, neuropeptides (e.g. VIP in the core; AVP in the shell) are colocalized with GABA and sometimes with glutamate [3]. The neuroanatomy of the SCN and their neuropeptides composition vary considerably across species, as reviewed elsewhere [4]. The first demonstration of the fundamental role of the SCN as the central clock was provided by two independent studies in 1972. Indeed, a bilateral lesion of the SCN resulted in the loss of the cortisol secretion rhythm [5], as well as rest-activity and drinking behavior rhythms [6]. More than 20 years later, Ralph and collaborators [7] showed that a transplantation of fetal SCN was able to restore behavioral rhythmicity in hamsters bearing SCN lesion. This clearly indicates that the primary function of the SCN is to generate internal rhythmicity. Ten more years were necessary to decrypt the underlying molecular mechanisms responsible for the endogenous rhythmicity. In 2000, Shearman and collaborators [8] proposed a mammalian circadian molecular oscillator model (updated since then; see [2, 9]), based on studies using mostly transgenic mice for different genes, called clock genes. In this model, identical in nocturnal and diurnal species, the self-sustained rhythmicity is generated by positive and negative core clock components that form a feedback core loop with a period of about 24 h per cycle (Figure 1; see Table 1 for full names of the different clock components). Two PAS domain helix-loop-helix proteins, CLOCK and BMAL1 form the positive part of the loop. These transcription factors heterodimerize and then activate the transcription from genes containing an E-box (5’-CACGTG-3’) or an E’-box (5’-CACGTT-3’). Among those genes, three Per (1-2-3) and two Cry (1-2) genes constitute the negative 4 Marc Cuesta and Etienne Challet part of the loop. PER and CRY proteins heterodimerize and form a complex with CK1ε/δ that phosphorylates PER and CRY. This facilitates the complex translocation to the nucleus where its quantity increases, leading to the repression of the transcription of several genes by CLOCK/BMAL1, including Per and Cry. This repression is thought to be driven via the recruitment by PER/CRY of a PTB-associated splicing factor/SIN3-histone deacetylase complex (PSF/Sin3-HDAC complex), which deacetylates histones 3 and 4 [10].

Figure 1. Molecular model of the mammalian cell-autonomous oscillator within a SCN cell as described in the text. Genes are represented in italic and proteins in CAPITAL. Nomenclature for each actor is given in Table 1. Abbreviations: CCG, clock-controlled gene; P, phosphate; U, ubiquitin. Interactions of the Serotonergic and Circadian Systems ... 5

Table 1. Non-exhaustive list of the main core clock genes and associated genes mentioned in the text

Gene Alternatename Full name Clock Circadian locomotor output cycle kaput Npas2 Mop4 Neuronal PAS domain-containingprotein 2 Bmal1 Arntl; Mop3 Brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 Per1 Period1 Periodcircadianproteinhomolog 1 Per2 Period2 Periodcircadianproteinhomolog 2 Per3 Period3 Periodcircadianproteinhomolog 3 Cry1 Cryptochrome 1 Cry2 Cryptochrome 2 Rev-erbα Nr1d1 Reverse viral erythroblastis oncogene product alpha Rev-erbβ Nr1d2 Reverse viral erythroblastis oncogene product beta Rorα Rora, Nr1f1 Retinoic acid-related orphan receptor alpha Rorβ Rorb, Nr1f2 Retinoic acid-related orphan receptor beta Rorγ Rorc, Nr1f3 Retinoic acid-related orphan receptor gamma Ck1δ Csnk1d Caseine kinase I delta Ck1ε Csnk1e Caseine kinase I epsilon Ampk AMP-activated protein kinase β-Trcp1 Btrc Beta-transducin repeat containing protein 1 Fbxl3 F-box and leucine-rich repeat protein 3 Fbxl21 F-box and leucine-rich repeat protein 21 Dec1 Bhlhe40 Differentiated embryo chondrocyte protein 1 Dec2 Bhlhe41 Differentiated embryo chondrocyte protein 2 Dbp D-box binding protein Tef Thyrotroph Embryonic Factor Hlf Hepatic leukemia factor E4bp4 E4 promoter-bindingprotein 4 Avp Adh Arginine Timeless Tim Timeless Prokr2 Prokineticin 2

Following this step, PER and CRY are targeted by CK1ε/δ and AMPK, respectively. This leads to their polyubiquitylation by β-TRCP1 and FBXL3 or 6 Marc Cuesta and Etienne Challet

FBXL21, respectively, which are proteins constituting an ubiquitin protein ligase complex, named SKP1-cullin-F-box (SCF). Ultimately, PER and CRY are degraded by the 26S proteasome. Other secondary loops, interconnected with the core loop, allow fine-tune regulations of the circadian clock, by multiple layers of control [2, 11]. For instance, the positive CLOCK/BMAL1 complex also regulates its own production via an activation of the transcription of nuclear orphan receptors genes through E-box located on their promoter (Figure 1). The protein products of these genes, namely REV-ERBα/β and RORα/β/γ, inhibit and activate Bmal1 transcription, respectively, by a competitive binding on ROR- specific response elements (RORE) located on Bmal1 promoter. It appears that the different REV-ERB and ROR proteins are not essential to generate the circadian rhythmicity in the SCN [12]. These proteins would also have a stabilizing role, by maintaining high amplitudes oscillations of clock genes, allowing a better response to synchronizing factors. Several other molecules have been identified as playing a role in the regulation of the circadian clock. For instance, the transcription factors DEC1 and DEC2 are able to reduce E- box mediated transcription in the SCN [13]. Other elements of the proline and acidic amino acid-rich basic leucine zipper (PAR bZIP) transcription factor family (e.g. DBP, TEF, HLF and E4BP4; Table 1) act through a D-box, present on some genes, to form a second accessory feedback loop [2]. Other genes, called clock-controlled genes (ccgs), are not part of any of the different loops but are regulated by them [14]. For instance, some of these genes are directly under the rhythmic control of CLOCK/BMAL1 via E-boxes, such as Avp gene in the SCN [15]. The ccgs are considered as the molecular output of the circadian clock. In other words, they are the link between the basic molecular clock machinery and a multitude of cellular and physiological functions.

The Circadian System as a Hierarchical Interconnected Multi-Oscillatory System

Until recently, the SCN were thought to be the one and only central clock, but we know now that many other brain regions (e.g. hypothalamic nuclei, forebrain) and non-neuronal tissues and cell types (e.g. liver, kidney, muscles, leukocytes) harbor autonomous clocks [16, 17]. These secondary clocks are named peripheral clocks as opposed to the central SCN clock. Virtually, every cell of the body contains circadian clocks (18). Both the SCN cells and Interactions of the Serotonergic and Circadian Systems ... 7 peripheral clock cells can independently self-sustain rhythmicity. However, in vitro, while the SCN neurons form a specialized network that can expresses synchronized rhythmsup to several weeks (i.e. pacemaker activity), the oscillations seen in peripheral clocks dampen out after few days due to desynchronization among the peripheral clock cells [19, 20]. This indicates that peripheral clocks need synchronizing stimuli to exhibit robust circadian at the level of the cellular population. Most of these synchronizing factors originate from the SCN, but feeding-fasting cycles and temperature variations can also directly entrain peripheral clocks [21, 22]. Despite the lack of robust oscillations observed in vitro, there are some peripheral clocks in various brain regions and tissues that can drive local physiological rhythms in the absence of SCN. Two structures have been well characterized, namely the retina [23] and the olfactory bulb [24]. In addition, evidence seems to indicate that the adrenal gland is able to regulate rhythmic parameters, such as steroidogenic acute regulatory protein (StAR) expression, a rate-limiting gene in the regulation of steroidogenesis [25]. Moreover, two SCN-independent oscillators (i.e. food-entrainable oscillator and methamphetamine-sensitive circadian oscillator) can drive circadian behavioral and hormonal rhythms even in SCN-lesioned animals or absence of core clock genes [26-32]. All these major developments in the last decade have started to change the original concept that was shared by chronobiologists for years. The current view of the circadian organization in mammals is that overt rhythms are controlled by a hierarchical system of circadian clocks in the brain and periphery that are subordinate to the central clock in the SCN [33]. This suggests that a system of multiple coupled oscillators is necessary to fine-tune the circadian control of physiology and behavior, including integration of sensory information, metabolic state, social cues, and emotional state. To do so, the SCN convey synchronizing stimuli to the peripheral clocks through a variety of neural and humoral signals. In the central nervous system, the SCN neural projections are very limited and mostly restricted to hypothalamic nuclei (Figure 2A). These nuclei are themselves connected to other brain structures allowing a rhythmic control of peripheral brain clocks. In addition, two neuronal pathways are of particular importance to convey information outside of the brain to peripheral tissues. The first neural pathway connects the SCN to the sympathetic nervous system via the paraventricular nucleus of the hypothalamus (PVN) and the intermediolateral column (IML) to many peripheral organs (Figure 2B). For instance, sympathetic projections 8 Marc Cuesta and Etienne Challet from the IML innervate the adrenal gland, allowing a rhythmic control in its sensitivity to adrenocorticotropic hormone or ACTH [34, 35].

Figure 2. A. Efferent neural projections from the SCN to different brains regions. Abbreviations: AMY, amygdala; ARC, arcuate nucleus; BNST, bed nucleus of the stria terminalis; DMH, dorsomedial nucleus of the hypothalamus; HB, habenula; IGL, intergeniculate leaflet; LS, lateral septum; POA, preoptic area; PVN, paraventricular nucleus of the hypothalamus; PVT, paraventricular nucleus of the thalamus; SCN, suprachiasmatic nuclei; sPVZ, subparaventricular zone; VMH, ventromedial nucleus of the hypothalamus. B. Efferent neural projections from the SCN to one peripheral clock (i.e. adrenal gland), and humoral signals controlled by the SCN. Two outputs (numerated 1 and 2) are represented that link the central clock to the adrenal clock regulating the release of glucocorticoid hormones. A 3rd output (numerated 3) is represented and links the central clock to the pineal gland regulating the release of melatonin hormone. Neuropeptides and hormones are indicated in italic. Abbreviations: ACTH,adrenocorticotropic hormone; AVP, arginine vasopressine; CRH, corticotropin-releasing hormone; IML; intermediolateral columns; Pin, pineal gland; Pit, ; SCG, superior cervical ganglion. Interactions of the Serotonergic and Circadian Systems ... 9

The synthesis of melatonin, a major circadian endocrine factor, is also under the rhythmic control of sympathetic projections. Indeed, from the IML, these projections reach the superior cervical ganglia (SCG), themselves connected to the pineal gland (Figure 2B). Through this pathway, the SCN exert a rhythmic control of the rate-limiting enzyme arylalkylamine N- acetyltransferase (AA-NAT), implicated in melatonin synthesis [36, 37]. The second neural pathway connects the SCN to the parasympathetic nervous system via the PVN and the dorsal motor nucleus of the vagus (DMV) that allows a rhythmic control of a wide range of peripheral tissues functions, such as found in white adipose tissue and liver (for references, see [38, 39]). As we already mentioned, humoral factors represent another link connecting central and peripheral clocks with each other. For instance, the robust rhythm of melatonin is known to facilitate sleep specifically in humans, up-regulate immune parameters and control seasonal functions, such as reproduction [40, 41]. In addition, the synthesis and secretion of glucocorticoids (GCs), another class of hormones, is regulated by the central clock through the hypothalamic-pituitary-adrenal (HPA) axis [25, 42] and thus exhibit a robust circadian rhythm (Figure 2B). This rhythm of GCs (i.e. corticosterone in rats and mice; cortisol in humans) then regulates the synchronization of many peripheral clocks at both molecular and behavioral levels [41, 42]. In peripheral clocks, the circadian rhythmicity relies on the same molecular mechanisms as those found in the SCN clock, although several differences have been identified [43, 44]. For instance, in some brain regions (e.g. striatum, hippocampus), CLOCK is not present and is substituted by one of its analogue, namely NPAS2 (see Table 1). In the liver, both CLOCK and NPAS2 are expressed, but NPAS2 cannot maintain circadian oscillation when CLOCK is missing. RORγ is only expressed in peripheral tissues and would exert the same role than RORα/β. Of note, knowledge of clock gene expression in humans is hampered by the limited possibilities of sampling. Studies must rely on cells that can be easily sampled, or ultimately on post- mortem tissues. Since it is difficult to analyze clock gene expression in the human central clock, several peripheral clocks have been studied instead, such as those present in skin [45], adipose tissue [46, 47], oral mucosa [45], bone marrow [46], colon cells [48, 49], hair follicles [50], and peripheral blood mononuclear cells [51-58]. The results found are relatively similar to those obtained in . In all tissues studied to date, 5 to 10% of the transcriptome displays circadian rhythms, but the subset of rhythmic transcripts is almost entirely 10 Marc Cuesta and Etienne Challet distinct among tissues [14]. This means that the ccgs are not the same in the different clocks found in the SCN and in the different peripheral tissues, implying that the role of these clocks is distinct, and must reflect each structure’s particular functions.

Synchronization of the Central Clock by the Photic Factor

As a relay between the external environment and the internal functions of the body, the SCN regulate the organism’s internal cyclic timing. By this mechanism named “synchronization”, the body can adapt to changes in the environment and thus be in phase with the external world. Among these factors, the 24 h light/dark cycle is the most potent environmental synchronizer of the central clock, but variations in temperature or food availability, for instance, can also affect it [21, 59, 60]. In addition, internal signals, mediating physiological information, can also reach the central clock, which integrates them in conjunction with light cues. These so-called non- photic factors are conveyed in part via serotonergic pathways and will be reviewed later in this chapter. To reach the SCN, the solar light signal or photic factor is mainly detected by the photopigment melanopsin [61], located in a subset of ganglion cells (Figure 3). These melanopsin cells represent 1% to 2.5% of the total number of retinal ganglion cells [61, 62] and project directly to the ventral part of the SCN via the retinohypothalamic tract (RHT) (Figure 3), releasing glutamate and pituitary adenylate cyclase activating peptide (PACAP) [63-65]. In addition, melanopsin cells project to the intergeniculate leaflet (IGL) that is a thalamic structure implicated in the integration of photic and non-photic signals [66-68]. From the IGL, neural projections innervate the ventral part of the SCN through the geniculohypothalamic tract (GHT) (Figure 3) that releases GABA and neuropeptide Y (NPY) [66-68]. From the retina, photic information allows a daily resetting of the central clock. The phase of the SCN can be delayed or advanced up to 2 h per day in most mammalian species, according to the time of day when the retina receives light [69, 70]. By applying discrete light pulses at different time points in animals housed in constant darkness (DD), a phase response curve to light can be defined. Of note, in DD, the period of time when nocturnal animals are awake is considered as the “subjective” night, while it is named “subjective” day when they are asleep. Interactions of the Serotonergic and Circadian Systems ... 11

Figure 3. Main afferent neural projections to the SCN. Grey arrows represent projections that convey photic cues to the SCN, black arrows represent projections that convey non photic cues to the SCN, and the black/grey arrow represent projections that convey photic and/or non-photic cues to the SCN. Neuropeptides and neurotransmitters are indicated in italic. Abbreviations:5-HT, serotonin; DRN, dorsal raphe nucleus; IGL, intergeniculate leaflet; GABA: gamma-aminobutyric acid; GHT, geniculohypothalamic tract; GLU, glutamate; MRN, median raphe nucleus; NPY, neuropeptide Y; PACAP, pituitary adenylatecyclase-activating peptide; RHT, retinohypothalamic tract; SCN, suprachiasmatic nuclei.

The phase response curve to light expresses the magnitude of the phase shift of a central clock marker, such as locomotor activity, as a function of the time of light exposure (Figure 4). In both nocturnal and diurnal species, the phase response curve to light displays a similar pattern with light-induced phase delays or advances occurring mainly during the subjective night (i.e. active and resting period in nocturnal and diurnal species, respectively) [69-72]. This mechanism, for instance, allows the organism to adapt to jetlag: when our body is exposed to a new LD cycle, advanced (i.e westbound travels) or delayed (i.e. eastbound travels) by few hours, the SCN rapidly integrate the timing of the new LD cycle. Then the SCN relay this information, leading to the synchronization of the different rhythmic parameters to the new LD cycle. However, the rate of synchronization differs between the various rhythmic parameters [73], leading to a transient misalignment of the different parts of the circadian system. 12 Marc Cuesta and Etienne Challet

Figure 4. Schematic representation of phase shifts of locomotor activity in response to light pulses in nocturnal rodents and phase response curve to light. At the top of the figure are represented 3 schematic actograms representing the locomotor activity of a nocturnal mouse (A., B. and C). Each row corresponds to 24 h, and 12 days are represented. Animals are kept in constant darkness, as reported by the black and gray bars at the top of each actogram, indicating the subjective night and day, respectively. Hence, mice express their endogenous period (smaller than 24h), explaining why each day, the locomotor activity occurs earlier than the previous day. By convention, in nocturnal species kept in constant darkness, the beginning of the period of activity corresponds to the circadian time 12 or CT12. On the 6th day of recording, a light pulse, represented on the actograms by a grey arrow, is applied at CT6 (A.), CT12 (B.) and CT18 (C.), inducing no effect, a phase delay and a phase advance of the locomotor activity, respectively. The magnitude of the phase shifts is assessed by performing 2 regressions that fit to the onset of activity, represented by gray lines. The first and second regressions are calculated using several days before and after the light pulse, respectively. The phase difference, obtained on the day following the light pulse, gives the value of the phase shift, which is reported on the phase response curve to light, schematized at the bottom of the figure by a gray curve. Each discreet pulse of light, given at a specific time, induces a phase shift with a specific magnitude, and the phase response curve to light represents the integration of all these effects reported on a 24h scale, as represented by the black and gray bars on the x-axis. They are 3 type of response to a light pulse, depending on when it is applied. During the middle of the subjective day (dashed box A), including CT6, no phase shifts are observed. From the end of the subjective day to the middle of the subjective night (dashed box B), including CT12, phase delays are observed. From the middle of the subjective night to the beginning of the subjective day (dashed boxes C), including CT18, phase advances are observed. Abbreviations: CT, circadian time; subj. night, subjective night; subj. day, subjective day. Interactions of the Serotonergic and Circadian Systems ... 13

Retinal illumination leads to glutamate release by RHT terminals, activating N-methyl-D-aspartate (NMDA) receptors present in SCN cells [74]. The subsequent Ca2+ influx induces the synthesis of nitric oxide (NO) [75]. At the beginning of the night, NO activates ryanodine receptors, leading to Ca2+ release from the endoplasmic reticulum [76], while at the end of the night, NO activates guanylate cyclase that produces cGMP [76]. In addition, the cAMP pathway is activated throughout the night in response to photic cues [77]. Together, these 3 pathways induce the phosphorylation of cAMP response element binding (CREB) protein via the activation of the and extracellular signals regulated kinase [78-80]. P-CREB can then translocate to the nucleus and bind to cAMP response element (CRE) present on the promoter of target genes, such as Per1 and Per2 [79-81]. While Per1 transcription is quickly induced throughout the night in response to a light pulse, the induction of Per2 expression is fast at the beginning of the night and slow or even absent at the end of the night [72, 82-84]. In addition, spatial differences have been found. For instance, in response to light, Per1 is induced in the ventral SCN cells, whereas Per2 is expressed in the whole SCN [85, 86]. A causal role of Per1 and Per2 genes in the mediation of light-induced resetting of the central clock has been proposed based on alteration of Per1/2 transcription [87, 88] and studies in Per1/2 mutant mice [89]. In addition, other genes in the SCN are regulated in response to light, such as c-fos [90], Bmal1 [91], Cry2 [72, 92], Rev-erbα and Rorβ [93-95].

The Serotonergic Regulation of the Central Circadian Clock

The Serotonergic Component of the Circadian System

The serotonergic system comprises several nuclei that can be divided in 2 groups, posterior and anterior [96]. Among the anterior group, the median raphe nucleus (MRN) and the dorsal raphe nucleus (DRN) are of particular interest since they are connected with the central clock (Figure 3) [97]. The neural projections from the MRN to the SCN are direct, while the DRN send indirect projections to the SCN via the IGL. Moreover, the MRN and DRN are tightly interconnected [98]. Of note, few studies have described direct projections from the DRN to the SCN in rats and hamsters [99, 100], and retinal fibers connecting raphe nuclei were observed in rats and cats [101, 14 Marc Cuesta and Etienne Challet

102]. These pathways are thought to mediate, at least partially, non-photic information, as reviewed later in this chapter. Hence, the DRM, the MRN, and their input pathways to the SCN can be considered as part of the circadian system. While the raphe nuclei are the primary source of 5-HT in brains, only 50% of the raphe cells that projects to the SCN release 5-HT at their terminals [99, 103, 104]. For instance, in Syrian hamsters, approximately 30% of MRN cells are immunoreactive for vesicular glutamate transporter 3 (VGLUT3) and could then release glutamate [104]. The serotonergic system is with no doubt the neurochemical system that expresses the highest number of 5-HT receptors subtypes. To date, 15 genes coding for those receptors have been described in mammal brains [105]. Among them, 7 different receptors subtypes have been observed in the SCN of the most common species (i.e. rats, mice and hamsters) via immunohistochemistry or autoradiography (Table 2) [106-116]. Less than half of these 5-HT receptors subtypes were described in all 3 species (5-HT1A, 5- HT1B and 5-HT7). The 5-HT2A and 5-HT2Creceptors were only found in rats. To our knowledge, the presence of 5-HT5A receptors in mice was never studied. Importantly, most of these studies did not indicate the pre- or postsynaptic localization of these 5-HT receptors subtypes, as well as the type of neurons on which they are expressed. Of note, the activation of 5-HT3 receptors by different agonists was shown to affect the central clock in rats (see below), but to date, this receptor has only been described in the SCN of Syrian hamsters [116]. It is important to mention that the DRN, MRN and IGL also express 5-HT receptors, such as 5-HT1A (acting as auto-receptors) and 5- HT7 subtypes [108, 117, 118].

Table 2. Localization of 5-HT receptor subtypes in the rat, hamster and mouse SCN

Receptor Rat Hamster Mouse a b c 5-HT1A Yes Yes Yes d e f 5-HT1B Yes Yes Yes g 5-HT2A Yes ? ? g 5-HT2C Yes ? ? k 5-HT3 ? Yes ? h i 5-HT5A Yes Yes ? j a f 5-HT7 Yes Yes Yes ?=Unknown. References as follows: a) [106]; b) [107]; c) [108]; d) [109]; e) [110]; f) [111]; g) [112]; h) [113]; i) [114]; j) [115]; k) [116]. Interactions of the Serotonergic and Circadian Systems ... 15

Circadian Control of the Serotonergic System

Brain 5-HT is synthetized in two steps from the essential amino acid L- tryptophan (L-Trp). During the first step, tryptophan hydroxylase 2 (TPH2), the rate-limiting enzyme of this synthesis pathway, allows L-Trp to be transformed into 5-hydroxyptryptophane (5-HTP) [119]. Then, 5-HTP is quickly decarboxylated into 5-HT by 5-HTP decarboxylase. Interestingly, TPH2 expression is under the control of the circadian system. Indeed, in the DRN and MRN of rats kept in DD, both Tph2 mRNA and TPH2 protein levels expressed a circadian rhythm with a peak at the end of the day and the middle of the night, respectively [120, 121]. In addition, a similar rhythm of TPH2 level was found in the SCN [122] and the IGL [120] with a peak occurring at the end of day. The authors mentioned that the delay observed in the TPH2 peak between these structures suggests that TPH2 is first express in the raphe nuclei and then is transported along the axons to reach terminals located in the SCN and IGL. Moreover, TPH2 enzymatic activity was shown to be rhythmic in rats, with a peak during the middle of the day [123], although this remains contradictory [124]. The underlying mechanisms controlling TPH2 rhythmic synthesis have started to be deciphered. Indeed, in adrenalectomized rats [125] or Syrian hamsters [126], TPH2 rhythm was abolished, but restored when animals received exogenous GCs (i.e. corticosterone in rats and cortisol/corticosterone in Syrian hamsters) in drinking water, mimicking the endogenous rhythm. More recently, it was shown that chronic administration of corticosterone in rats also abolishes Tph2 mRNA rhythm in the DRN [127]. Altogether, these data clearly indicate that the circadian rhythm of plasma GCs is responsible for the rhythm of TPH2. In addition, locomotor activity seems to be able to influence Tph2 expression [125]. Variations of 5-HT content in the whole brain are known for a long time [128] and present elevated values during daytime. More recently, 5-HT content in the SCN region was found to be rhythmic in both nocturnal [95, 129] and diurnal [130] rodents, with maximal values at night and day, respectively. In addition, microdialysis studies indicate a rhythmic release of 5-HT in the SCN of Syrian hamsters [131] and rats [122] with a peak occurring in early night. Similar results were found in the IGL of Syrian hamsters [132]. Interestingly, 5-HT release and content express maximal values few hours later than those of TPH2 levels, suggesting that 5-HT rhythmicity is under the control of TPH2 rhythm. In the mouse midbrain, the 5-HT transporter (5-HTT) that reuptakes extracellular 5-HT from the synaptic 16 Marc Cuesta and Etienne Challet cleft is also under the control of the central clock [133, 134] with higher levels observed at night for both 5-htt mRNA and 5-HTT protein. Hence, the subsequent 5-HT uptake expresses a circadian profile [134]. This circadian regulation is thought to be mediate through the rhythmic control of the activating transcription factor-4 (ATF4) that activates 5-htt transcription. The precise role of the 5-HT rhythmic release into the SCN and IGL is still poorly understood. In the SCN, this rhythm appears to be in phase opposition between nocturnal and diurnal species (i.e. high values during the active period for both behaviors), suggesting that one of its functions is to indicate to the central clock when an is awake and active. As it will be reviewed in the remaining part of this chapter, the serotonergic system exerts multiple other roles as a regulator of the circadian system, but most of them are related to sleep and wake behaviors, such as induced locomotor activity, arousal and sleep deprivation.

Serotonin and the Sleep-Wake Cycle

During the last decade, progress has been made in our understanding of the neural regulation of the human sleep-wake cycle. The current view is that sleep and wake behaviors are generated by complex interactions between endogenous circadian and sleep homeostatic processes, as well as direct effects of light and dark [135]. Interestingly, the sleep-wake cycle is also closely related to the activity of 5-HT neurons. For instance, the activity of 5- HT neurons in the DRN of cats is slow and regular during the wake period [136]. Then, when animals start their sleep period, this activity decreases and loses its rhythmicity, until complete disappearance during the onset of the first REM sleep episode. In addition, as we previously mentioned, 5-HT release in the SCN region occurs during the waking period of animals and is thus correlated to the electrical activity of 5-HT neurons. More recently, it was shown that an induced transient 5-HT deficiency resulted in a transient loss of circadian rhythmicity of locomotor activity and sleep-wake cycle [137]. Taken together, these results suggest that 5-HT (especially from the DRN) not only promotes wake and inhibit REM sleep [138], but also participates in the circadian regulation of sleep and wake behaviors [137]. This indicates that the sleep-wake cycle, the serotonergic system and the circadian central clock are tightly connected.

Interactions of the Serotonergic and Circadian Systems ... 17

Serotonin and Non-Photic Synchronization

In addition to light, many factors can influence the central clock at both molecular and behavioral levels. To study the influence of these non-photic factors, animals are usually kept in DD to prevent not only the synchronizing effect of light, but also its direct effect, called “masking”. Negative masking is a process that diminishes the expression of a circadian pattern [139]. For instance, in nocturnal animals, light exposure at night inhibits locomotor activity [140]. In opposition, positive masking allows a variable to be expressed in addition to its circadian pattern. In diurnal species, light exposure at night induces a bout of locomotor activity at a time when animals are supposed to sleep [141]. In this context, constant conditions such as DD, allow to “unmask” a given variable.

Figure 5. Phase response curve to non-photic factors in nocturnal species. Each discreet application of a non-photic factor, given at a specific time, induces a phase shift of the locomotor activity with a specific magnitude, and the phase response curve represents the integration of all these effects reported on a 24h scale, as represented by the black and gray bars on the x-axis. Phase advances are mainly observed during the subjective day. Abbreviations: CT, circadian time; subj. night, subjective night; subj. day, subjective day.

Examples of non-photic factors and their effects on both rest-activity rhythm and clock gene expression are summarized in Table 3. Hereafter, we will only focus on those that have been linked to the serotonergic system. In nocturnal species, these non-photic factors are mainly effective when applied during the middle of the subjective day. They induce a phase advance of 18 Marc Cuesta and Etienne Challet locomotor activity rhythm (Figure 5) and inhibit expression of clock genes (i.e. Pers). Some studies have reported little behavioral phase delays during the late subjective night, but usually they are barely detectable. In a way somewhat counter-intuitive, thecircadian molecular oscillator model and the photic synchronization are similar in phase according to the astronomical time (e.g. light-dark cycle) in diurnal animals compared to nocturnal ones. By contrast, the responses of the central clock to most non-photic factors are opposite in astronomical phase (but in phase according to the sleep/wake cycle), highlighting the fundamental role of the serotonergic system in the determination and/or the maintenance of diurnal or nocturnal behavior. More importantly, these differences must be taken into serious consideration in both normal and pathological conditions, since humans are genetically diurnal.

Table 3. Main behavioral and molecular effects of different photic and non-photic factors

Nocturnal rodents Diurnal rodents Type of Locomotor Per1/2 Locomotor Per1/2 Factor activity expression activity expression 1. Phase delays: 1. Phase delays: early night early night Increase Increase (wake) (sleep) Light early and late early and late 2. Phase 2. Phase night night advances: advances: late night late night (sleep) (wake) Phase Phase advances Transitory advances Decrease early night ? hyperactivity midday (sleep) (sleep) Phase Phase advances 5-HT advances Decrease No effect night (sleep) midday (sleep) Phase NPY advances Decrease ? ? midday (sleep) Phase Phase delays GABA advances Decrease Decrease Per2 midday (wake) midday (sleep)

Interactions of the Serotonergic and Circadian Systems ... 19

Transitory Hyperactivity and Sleep Deprivation In nocturnal species, a novel running wheel presented during the sleep period (i.e. subjective day) induces a transient hyperactivity that leads to a phase advance of the locomotor activity rhythm [142] and a transient decrease of Per1 and Per2 mRNA levels in the SCN (Table 3) [143, 144]. Hyperactivity seems to be an essential parameter in phase shift induction. Indeed, in immobilized or light runner animals, behavioral phase advances are very small or even undetectable. It has been suggested that the sleep deprivation caused by the hyperactivity could be responsible for these phase- shifts rather than the degree of activity itself [145]. In line with this argument, phase shifts are only observed during the subjective day, when animals are asleep, although there are discrepancies regarding the studied species. For instance, sleep deprivation is ineffective in shifting the SCN of mice [146] and only induces small phase advances in rats [147]. Interestingly, long bouts of activity (minimum of 3 h) are necessary to induce a significant phase shift. This indicates that the circadian system of nocturnal species has a high threshold for wake/activity behavior to alter the circadian system, especially in comparison to light that can induce phase shifts only after few minutes of exposure even at low intensity [70, 148]. This relatively low sensitivity is thought to prevent uncoordinated resetting of the circadian system in response to small amounts of activity (i.e. Zeitgeber noise) that are not necessarily abnormal outside the main active phase. Very few data on hyperactivity or sleep deprivation are available in diurnal species. In diurnal European ground squirrels confined in a running wheel every day at the end of their active period, a synchronization of the locomotor activity can be observed [149]. The authors suggested that this synchronization is the result of daily phase advances induced by hyperactivity. Hence, diurnal and nocturnal species would similarly react to this non-photic factor independently of the phase of the activity cycle. Another hypothesis suggests that this induced activity could lead to sleep deprivation, since it occurred at the far end of the subjective day for a 3 h period. Thus, this non- photic factor could rather act at early night to induce phase shifts, meaning that its action is dependent of the phase of the activity cycle. Data obtained with other non-photic factors in diurnal species [130] are in line with the latest hypothesis and will be discussed later in this chapter. In humans, however, sustained physical exercise performed at the beginning of the night induces small phase delays [150-152] rather than phase advances. Interestingly, at the cerebral level, hyperactivity was shown to induce an endogenous release of 5-HT from the MRN terminals to the SCN of Syrian 20 Marc Cuesta and Etienne Challet hamsters [131]. In addition, lesion of 5-HT fibers, using the 5,7- dihydroxytryptamine (5,7-DHT) neurotoxin, inhibits the behavioral phase advances normally induced by a novel running wheel access [153]. Although it is still unclear which aspect(s) of locomotor activity is(are) responsible for those effects, these results suggests that the serotonergic system acts as an important link between wake/activity behavior and the circadian system. Hence, serotonin can be considered as a crucial candidate to mediate the non- photic synchronization as it will be now detailed.

Serotonin, Serotonergic Agonists and Selective Serotonin Reuptake Inhibitors In nocturnal species, serotonergic influence on the synchronization of the central clock has been extensively studied, both in vitro and in vivo, using different methods of acute administration (systemic or local) and a wide range of agonists, such as serotonin itself, 8-hydroxy-2-di-n-propylaminotetralin (8- OH-DPAT), the most used 5-HT agonist, and fluoxetine, a selective serotonin reuptake inhibitor (SSRI) [95, 154-163]. When administered during the middle of the subjective day, these molecules induce phase-advances of the locomotor activity and transitory inhibition of Per1 and Per2 transcription in the SCN of mice, rats and Syrian hamsters. Interestingly, these effects are similar to those observed in response to transitory hyperactivity. Of note, in rats, it has been shown that mRNA levels of Rev-erbα and Rorβ are increased and decreased, respectively, in response to fluoxetine injection [95]. In addition, in vitro studies indicate that application of 8-OH-DPAT and fluoxetine on SCN slices during the subjective day induce phase advance of the SCN electrical activity [164, 165]. Moreover, by causing a state of serotonin-hypersensitivity (i.e. suppression of the endogenous serotonergic tone), it is possible to induce very large phase shifts in response to 5-HT agonists in vitro [165] and hyperactivity in vivo [166, 167]. Interestingly, in nocturnal species, recreational drugs, such as cocaine or 3,4-methylenedioxymethamphetamine (MDMA or 'Ecstasy'), known to act on the serotonergic system by different mechanisms, can mimic [168] or alter [169] the non-photic synchronization of the central clock, respectively. To date, there is only one study that examined the serotonergic modulation on the central clock of Arvicanthisansorgei, also known as Sudanian grass rats [130], a diurnal rodent model [170]. Strikingly, the temporal window of circadian sensitivity of this rodent to (+)8-OH-DPAT (i.e. positive enantiomer of 8-OH-DPAT) and fluoxetine is opposed to that observed in nocturnal rodents. In other words, this agonist induces phase advances of the locomotor Interactions of the Serotonergic and Circadian Systems ... 21 activity essentially during the subjective night, meaning that such phase-shifts are dependent on the sleep-wake cycle. Indeed, for both nocturnal and diurnal rodents, phase advances are observed during their rest period. This result highlights one more time the pivotal role of the serotonergic system in the transmission of wake/activity information to the central clock. Surprisingly, systemic injections of (+)8-OH-DPAT and fluoxetine at subjective dawn and dusk failed to modify Per1 and Per2 mRNA expression in the SCN of Sudanian grass rats, as it was observed in nocturnal rodents. This suggests that other clock genes could be modified in response to acute serotonergic input in diurnal species, since Per1 and Per2 mRNA levels are relatively low at dawn and dusk. Another hypothesis suggests that Per1 and Per2 expression could be influenced on a different time scale in diurnal rodents. Indeed, a change in the expression of these genes was measured one hour after the treatment, while in some nocturnal rodents (i.e. Syrian hamster), the effects on Per expression were only observed 2 hours after a serotonergic injection [160]. Several studies have explored the different 5-HT receptor subtypes that could be responsible for the non-photic effects observed in response to a serotonergic stimulation. Since 8-OH-DPAT binds to 5-HT1A and 5-HT7 receptors, two subtypes of receptors present in the SCN (Table 2), extensive research has been performed to delineate their respective role. Apparently, both receptors subtypes are needed to observed non-photic effects. Indeed, both 5-HT1A antagonists (e.g. pindolol) [171] and 5-HT7 antagonists (e.g. 2a- [4-(4-phenyl-1,2,3,6-tetrahydropyridyl)butyl]-2a,3,4,5-tetrahydro- bezo[cd]indol-2(1H)-one or DR404 and (2R)-1-[(3-hydroxyphenyl)sulfonyl]- 2[2-(4-methyl-1-piperidinyl)ethyl]pyrrolidine or SB269970) [159, 172] block phase shift induced by 8-OH-DPAT treatment and mutant mice lacking either 5-HT1A [173] or 5-HT7 [174] receptors subtype do not express behavioral phase shifts in response to 8-OH-DPAT. Of note, 5-HT5A receptors could also be implicated, at least in rats [175], but this in vitro result needs to be confirmed with in vivo studies. No data are yet available in diurnal species, but it is reasonable to suggest that both type of receptors are implicated, since (+)8-OH-DPAT induces behavioral phase shifts [130]. Besides their presence in the SCN, 5-HT1A and 5-HT7 receptors are also expressed in the DRM, NRM and IGL [108, 117, 118]. Electrical stimulation of the DRN and MRN, and local injections of 8-OH-DPAT into the DRN and IGL induce phase advances of the locomotor activity rhythm [155, 172, 176]. However, these structures and their 5-HT receptors are not essential to elicit these effects, since a local injection of 8-OH-DPAT into the SCN leads to behavioral phase advances [155, 159]. 22 Marc Cuesta and Etienne Challet

While 5-HT1A and 5-HT7 receptors seem to act in synergy, the intracellular pathways they activate are opposed: 5-HT1A and 5-HT7 receptors activation leads to inhibition [177] and activation [178] of the adenylatecyclase (AC) pathway, respectively. It is possible that within the SCN, these receptors are expressed in different neuronal subtypes. According to this hypothesis, 5-HT7 receptors activation inhibits GABA release and GABA-activated current into the SCN of rats [179] and the DRN of - pigs [180], and 5-HT1A receptors activation leads to a decrease of glutamate release into the SCN of Syrian hamsters [181]. In addition, in vivo local applications of 5-HT and 8-OH-DPAT inhibit the SCN electrical activity [182, 183]. Based on these results, 5-HT1A and 5-HT7 receptors could be localized in glutamatergic and GABAergic neurons in the SCN, respectively, but further investigations are required to confirm this hypothesis and delineate the respective functions of these neuronal subtypes in the mediation of non-photic information to the central clock.

Neuropeptide Y, GABA and the IGL As an integrator of photic and serotonergic non-photic cues that are then conveyed to the SCN via NPY and GABA release, the IGL represents an additional modulator in the synchronization of the central clock. In nocturnal species, local NPY injections into the SCN [184], electrical stimulations of the geniculo-hypothalamic projections [185], and in vitro NPY application on SCN slices [186] induce phase shifts similar to those observed in response to hyperactivity and 5-HT agonist treatments. In addition, intra- cerebroventricular injections of anti-NPY antibodies [187], as well as IGL lesions [188] lead to an inhibitory effect on behavioral phase shifts induced by an access to a novel wheel. More recently, it was shown that increased locomotor activity and local injections of 8-OH-DPAT in the IGL induce an increased NPY release in the SCN [189]. At the cellular level, NPY is thought to bind to Y2 receptors [190, 191], which activate the protein kinase C [192], leading to a transitory inhibition of Per1 and Per2 mRNA level in the SCN [193, 194], similar to that observed for the other non-photic factors. In addition to NPY, IGL neurons are known to release GABA. Interestingly, local injections into the SCN of GABAergic agonists for

GABAA receptors (e.g. muscimol) induce non-photic phase-shifting responses and an inhibition of Per1 and Per2 mRNA level in the SCN of Syrian hamsters [195-197], similar to those observed in response to hyperactivity, and 5-HT and NPY agonist treatments in nocturnal species. In contrast, data obtained in response to muscimol in the diurnal Nile grass rat [197, 198] are Interactions of the Serotonergic and Circadian Systems ... 23 different than those obtained in response to serotonergic drugs in the diurnal Sudanian grass rat [130]. Indeed, when injected during the subjective day, muscimol induces behavioral phase delays and a transitory inhibition of Per2 expression. This suggests that GABAergic input from the IGL and/or GABAergic neurons into the SCN could have a different role in the synchronization of the central clock between nocturnal and diurnal species. Collectively, all the above results indicate that the serotonergic system, directly from the DRN and MRN, and indirectly through the IGL releasing NPY and GABA, act in mediating locomotor activity-induced phase-shifting effects on the central clock. The challenge in the future years will be to delineate the exact pathway linking the locomotor activity to the serotonergic system, and the cellular and molecular mechanisms regulating the non-photic synchronization in diurnal species. This will represent valuable informationthat could lead to innovative therapies in “diurnal humans” suffering from jet-lag, insomnia or shift-work related alterations.

Serotonin as a Photic Factor

As we mentioned, serotonin is clearly implicated in the non-photic synchronization of the central clock. Surprisingly, only 2 in vivo studies showed this type of effect on the circadian system of rats [95, 158], while there are plenty of in vitro data available in this rodent [158, 162, 165, 171, 199, 200]. The lack of in vivo observations might be due to a specificity found in rats that has not yet been described in mice and Syrian hamsters. Indeed, during the subjective night, systemic and local injections of non-specific serotonergic agonists, such as quipazine (5-HT3> 5-HT1A> 5-HT1B), (±)-2,5- dimethoxy-4-iodoamphetamine or DOI (5-HT2A> 5-HT2C> 5-HT2B), 1-(3- chlorophenyl)piperazine or mCPP (5-HT2C> 5-HT2B> 5-HT2A> 5-HT1), and specific agonists for 5-HT3 (1-(m-chlorophenyl)biguanide or mCPBG) and 5- HT2C (8,9-dichloro-2,3,4,4a-tetrahydro-1H-pyrazino[1,2-a]quin oxalin-5(6H)- one or WAY-161503) receptors, produce phase shifts of the locomotor activity that are similar to those observed in response to light [95, 201-205]. In addition, most of these compounds induce an increase of Per1 and Per2 mRNA, and FOS protein levels in the SCN when applied at night [202, 206, 207]. The serotonergic modulation of the central clock in rats is thus paradoxical, since both photic-like and non-photic effects can be observed. The different 5-HT receptors subtypes present in the SCN could partially explain why such opposite responses can be observed after a serotonergic 24 Marc Cuesta and Etienne Challet stimulation. First, a global activation of all 5-HT receptors present in the SCN, through an increase of endogenous 5-HT in response to fluoxetine, leads to non-photic rather than photic-like response [95]. This suggests that the serotonergic system mediates mainly non-photic information in rats. Second, as it was previously described, 5-HT1A and 5-HT7 receptors are thought to mediate non-photic effects observed during daytime in nocturnal rodents, including rats. However, these receptors are not implicated in the photic-like effects, since both (+)8-OH-DPAT and (2S)-(+)-5-(1,3,5-trimethylpyrazol-

4yl)-2-(dimethylamino)tetralin (AS19), a potent 5-HT7 agonist, failed to mimic the effect of light when applied at the end of the subjective night [95]. Third, 5-HT3 and 5-HT2C receptors were described as responsible for the induction of photic-like responses. Indeed, injection of specific 5-HT2C (WAY-161503) [95] and 5-HT3 (mCPBG) [95, 202] agonists mimic the effects of light on the central clock.

While 5-HT2C receptors were described in the SCN of rats [112] at the post-synaptic level [207], 5-HT3 receptors were localized only in the SCN of Syrian hamsters [116]. However, it was shown in rats that enucleation, leading to RHT degeneration, prevents the behavioral phase advances induced by quipazine [201]. This indirect evidence suggests that 5-HT3 receptors would be localized at the presynaptic level on RHT terminals. Their activation would lead to a release of glutamate from RHT terminals inducing photic phase shifts. Of note, since 5-HT2A receptor subtype is present in the SCN of rats [112] and has an affinity for DOI, a serotonergic agonist that induces photic- like effects, we cannot exclude its possible involvement in these effects. While there is extensive literature on the photic-like effects observed in rats, there are almost no data available in mice or Syrian hamsters.The localization of 5-HT3 and 5-HT2C receptors in the SCN of these species has not yet been fully described and to our knowledge, only one study used a 5-HT2C receptor agonist (Ro-600175) in Syrian hamsters, but in order to study the modulation of the photic synchronization rather than to test possible photic- like effects [208]. To date, the only study showing possible photic-like effects in Syrian hamsters used an agonist with particular properties, named 8-[2-[4- (2-methoxyphenyl)-1-piperazinyl]ethyl]-8-azaspiro[4.5]decane-7,9-dione or

BMY 7378 [209]. This mixed compound act on 5-HT1A receptors, but as an antagonist when they are located at the postsynaptic level, and as an agonist in presynaptic receptors. BMY 7378 induces phase shifts only when administered at the end of the subjective night. These effects would be a consequence of the pharmacological properties of this agonist rather than a real photic-like effect observable in rats. Interactions of the Serotonergic and Circadian Systems ... 25

Serotonin as a Modulator of the Photic Synchronization

In this chapter, we previously described the well-characterized effects of light, as well as those related to non-photic factors, on the synchronization of the central clock in rodents kept in laboratory conditions. These conditions allow the study of one specific stimulus by a rigorous control of different variables that could influence the circadian system. In the wild, however, it seems obvious that different synchronizing factors simultaneously reach the SCN. Responses of the circadian system to the combination of these factors are mediated by the central clock and can sometimes be complex. Hereafter, the different effects observed in response to the interaction of photic and serotonergic non-photic factors will be reviewed.

Negative Modulationof the Photic Synchronization in Nocturnal Species As for others non-photic factors, such as novel wheel access [210], NPY [211, 212] and GABA agonists [213, 214], the serotonergic modulation of the photic synchronization of the central clock has been extensively studied in mice and Syrian hamsters, and more recently in rats, providing similar results. In these 3 nocturnal species, systemic or local injections of 8-OH-DPAT and fluoxetine performed during the subjective night and preceding light pulses, inhibits light-induced phase shifts of the locomotor activity (Figure 6) [95, 146, 215-218]. Surprisingly, 8-OH-DPAT was described as less effective in mice compared to Syrian hamster [218], and in rats, its positive enantiomer was used to test only the modulation of light-induced phase advances [95]. Other serotonergic related compounds were shown to affect the photic synchronization. For instance, in Syrian hamsters, light-induced electrical activity of SCN neurons is inhibited by pre-treatment with 8-OH-DPAT, but also with 5-carboxamidotryptamine (5-CT) and 5-HT itself [182, 183]. Of note, light, when applied during the subjective day, is able to reduce behavioral phase advance induced by the most common non-photic factors [154, 155, 194, 195, 219-221]. At the molecular level, it has been shown that 8-OH-DPAT and quipazine inhibit light-induced FOS expression in the SCN [216, 222-224], with a stronger inhibition observed in Syrian hamsters compared to rats. To date, there is only one study that described the modulation of clock gene expression in response to serotonergic stimulation associated with light pulse [95]. 26 Marc Cuesta and Etienne Challet

Figure 6. Schematic representation of the serotonergic modulation of the photic synchronization at the level of the locomotor activity of nocturnal animals. Schematic actograms representing the locomotor activity of a nocturnal mouse in response to a light pulse (A.), an injection of a serotonergic agonist (i.e. 8-OH-DPAT) followed by a light pulse (B.) and an injection of 8-OH-DPAT (C), performed at CT12 (left panel in A., B., and C.) and CT18 (right panel in A., B., and C.). Each row corresponds to 24 h, and 12 days are represented. Animals are kept in constant darkness, as reported by the black and gray bars at the top of each actogram, indicating the subjective night and day, respectively. Hence, mice express their endogenous period (smaller than 24h), explaining why each day, the locomotor activity occurs earlier than the previous day. By convention, in nocturnal species kept in constant darkness, the beginning of the period of activity corresponds to the circadian time 12 or CT12. On the 6th day of recording, the treatment, represented on the actograms by a grey arrow, is applied at CT12 or CT18, to test the effects of 8-OH-DPAT on light-induced phase delays and advances, respectively. The magnitude of the phase shifts is assessed by performing 2 regressions that fit to the onset of activity, represented by gray lines. The first and second regressions are calculated using several days before and after the light pulse, respectively. The phase difference, obtained on the day following the light pulse, gives the value of the phase shift. The light-induced phase advance and delay (A.) are both reduced when animals receive an injection of 8-OH-DPAT prior to the light pulse (B.), while 8-OH-DPAT alone does not induce phase shift at CT12 and CT18 (C.) Abbreviations: CT, circadian time; subj. night, subjective night; subj. day, subjective day. Interactions of the Serotonergic and Circadian Systems ... 27

When applied at the end of the subjective night, systemic injections of fluoxetine enhance the light-induced increase of clock gene expression in the SCN, strongly for Per1, and slightly for Per2 and Rorβ. These results challenge the unifying theory established to explain the molecular responses to the interactions between photic and non-photic factors [144]. In this theory, non-photic factors are supposed to inhibit the light-induced increase of clock gene expression. Although this theory was confirmed for some non-photic factors, such as NPY and GABA [225-227], contradictory results were found in response to novel wheel access, behavioral arousal and fluoxetine injection [95, 228, 229]. Thus, despite similar behavioral responses, the molecular mechanisms implicated in the interaction between photic and non-photic factors are variables, complex, and probably not limited to modifications of core clock gene expression in the SCN. As for the non-photic synchronization, 8-OH-DPAT is the most effective agonist used in nocturnal rodents to study the serotonergic modulation of the photic synchronization [95, 216, 217]. Both 5-HT1A and 5-HT7 receptors subtypes have been described as responsible for the responses observed. Indeed, 8-OH-DPAT application is able to inhibit light-induced electrical activity of SCN slices [182, 183], but this inhibition is more or less counteracted by pre-treatment using 5-HT1A [182] receptors antagonists, and strongly reduced with 5-HT7 receptors antagonists [183]. In addition, glutamate release from RHT in response to optic nerve stimulation was shown to be reduced after an activation of 5-HT7 receptors [230]. Another study showed that in Syrian hamsters, 5-HT1A and 5-HT2C antagonist injections are able to increase the inhibition of light-induced behavioral phase shifts elicited by the SSRI citalopram, while 5-HT7 antagonist is ineffective [231]. Mutant mice lacking either 5-HT1A [173] or 5-HT7 [174] receptors subtype also provide us some insights on the role of these receptors. While phase advances are normally observed in response to a light pulse given at the end of the subjective night, phase delays were observed in mutant mice for 5-HT7 receptors. In mutant mice for 5-HT1A receptors, light-induced phase advances are larger than those observed in wild-type animals. Of note, injection of the potent 5-HT7 agonist, AS19, failed to inhibit light-induced phase advance, suggesting a more important role for 5-HT1A receptors in rats [95]. This result, however, has to be interpreted carefully, since AS19 is a relatively new compound and its specificity to 5-HT7receptors is still controversial [232]. Recently, one study showed that the blockade of 5-HT1B and 5-HT5A receptors subtypes using specific antagonists lead to an inhibition of light-induced behavioral phase advances in Syrian hamsters [231]. While this is the first 28 Marc Cuesta and Etienne Challet

demonstration of the implication of 5-HT5A receptors in these effects, data showing the role of 5-HT1B receptors on the negative modulation of the photic synchronization were already available [110, 181, 230, 233, 234]. Briefly, once activated, 5-HT1B receptors induces a decrease in the release of glutamate from the RHT, normally induced in response to light. While all these receptors subtypes have been described in the SCN (Table 2), most of them are also present in the DRN, MRN and IGL and their activation could participate in the behavioral response observed. For instance,

5-HT5A receptors are thought to be localized presynaptically in the raphe nuclei, and their blockade would lead to an increase of 5-HT release into the SCN, explaining why a negative modulation of the photic synchronization was observed in the study of Gannon and collaborators [231]. There are at least five 5-HT receptors subtypes (5-HT1A, 5-HT1B, 5-HT2C, 5-HT5A and 5-HT7) that are involved in the negative modulation of the photic synchronization of the central clock. This demonstrates how important is the role of the serotonergic system on the mechanisms regulating the circadian system. Nevertheless, since local injections of 8-OH-DPAT in the SCN inhibit light-induced phase shifts of the locomotor activity rhythm [217], 5-HT1A and 5-HT7 receptors localized in the SCN are thought to be the predominant actors. More studies are needed to delineate the respective role of these different subtypes of receptors in function of their localization in one or several parts of the serotonergic component of the circadian system. Indeed, association of the right pharmacological compounds with bright-light therapy could prevent circadian rhythms from becoming pathologically modified, as it can be observed in mood disorders (see below).

Positive Modulation of the Photic Synchronization in Nocturnal Species In nocturnal rodents, as we just described, the serotonergic system mainly exerts negative modulation on the photic synchronization of the central clock.

In rats, however, it was shown that an activation of 5-HT2C receptors at the end of the subjective night, using the specific agonist WAY-161503, leads to the potentiation of light-induced behavioral phase advances [95]. This receptor subtype is also responsible for the photic-like effects observed in that species [95, 202] and it is thus not surprising to observe such an additional effect in response to the association of this photic-like factor with the photic factor. Strikingly, 5-HT3 receptors that are also implicated in the photic-like effects do not participate in the positive modulation of the photic synchronization [95]. This lack of effect could be due to the localization of this subtype of Interactions of the Serotonergic and Circadian Systems ... 29 receptor. Indeed, they are supposed to be localized on RHT terminals and to mimic the effect of light, their activation induces a release of glutamate from RHT terminals. It is possible that this release is negligible in comparison to the massive release induced by a light pulse, explaining why an additional effect is not observed. In the case of the 5-HT2C receptors activation, since they are thought to be localized postsynaptically in the SCN and the intracellular pathway they activate is independent of glutamate release, they could potentiate light-induced phase shifts of the locomotor activity rhythm. In mice and Syrian hamsters they are almost no data available on a possible positive modulation of the photic synchronization through the activation of 5-HT receptors. One study described that 5-HT2C receptors activation does not modify light-induced phase advance in Syrian hamsters [208]. In contrast, 5-HT2C antagonist injections were shown to increase the inhibition of light-induced phase advances elicited by the SSRI citalopram [231]. Taken together, these results suggest a different role for 5- HT2C receptors activation in Syrian hamster compared to the effects seen in rats. It has been proposed that in Syrian hamsters, the blockade of 5-HT2C receptors at the level of the raphe nuclei would lead to an increase of 5-HT release into the SCN [231], thus reinforcing the elevation of extracellular 5-HT in the SCN, elicited by citalopram. More data are needed to confirm this hypothesis, but it is clear that 5-HT2C receptors, as a function of their localization, regulate the synchronization of the circadian system in very different ways between rats and Syrian hamsters. It is possible, however, to observe a positive modulation of the photic synchronization in Syrian hamsters. Indeed injections of mixed agonists, such as 1-(2-methoxyphenyl)- 4(4-(2-phtalimido)butyl)piperazine or NAN-190, were shown to potentiate light-induced phase shifts [208, 209, 235-240]. The critical involvement of 5-

HT1A receptors in this effect was described [240], meaning that the effects observed are due to the specific pharmacological properties of these mixed agonists. In addition, Smith and collaborators [240] found that the potentiation effects of NAN-190 were due, at least in part, to a down-regulation of some genes and biochemical pathways involved in the molecular responses of the central clock to light.

Positive Modulation of the Photic Synchronization in Diurnal Species Interactions between photic and non-photic factors have not been extensively studied in diurnal rodents. To date, there are few publications on the inhibitory effects of GABAA and GABAB agonists on the light-induced 30 Marc Cuesta and Etienne Challet behavioral phase advances obtained in Nile grass rats [198, 226, 241]. These results are similar to those obtained in the nocturnal Syrian hamster [213, 214]. This suggests that, in contrast to what has been observed for GABA agonists as a non-photic factor, these compounds act on the photic synchronization in a similar way between nocturnal and diurnal species, despite some differences at the molecular level [197, 226] Interestingly, in the diurnal Sudanian grass rat, systemic injections of (+)8-OH-DPAT and fluoxetine were shown to potentiate both light-induced behavioral phase advances and delays [130]. This result highlights an opposed serotonergic modulation of the photic synchronization between nocturnal and diurnal species, as it was described for the serotonergic non-photic modulation of the central clock. Moreover, it appears that the non-photic regulation of the photic synchronization varies in function of a given non-photic factor (GABA vs. 5-HT agonists) between nocturnal and diurnal species, as it was also described for the non-photic modulation of the central clock. Taken together, it is thus tempting to hypothesize that the differential effects observed in response to these non-photic factors are linked to the mechanisms underlying diurnal and nocturnal behaviors. We already mentioned that the serotonergic system conveys information on wake/arousal behavior to the central clock. Hence, it is not surprising that the responses of the SCN to serotonergic cues differ between nocturnal and diurnal species, since their respective sleep-wake patterns are in phase opposition. The differences observed for GABA agonists suggest that the IGL (that release GABA) or the SCN GABAergic neurons could provide differential information to the SCN in function of the diurnal or nocturnal behavior. At the beginning of the subjective night, systemic injections of (+)8-OH- DPAT and fluoxetine coupled with light pulses induce an increase of Per1 and Rev-erbα mRNA levels in the SCN, while light pulse alone does not modify the expression of these genes [130]. At the end of the subjective night, these injections potentiate and inhibit the light-induced expression of Per1 and Rorβ mRNA in the SCN, respectively [130]. Interestingly, while these results are similar for Per1 at the end of the subjective night in diurnal and nocturnal species, they are in opposition for Rorβ [95]. This suggests an important differential role for Rorβ in response to the serotonergic modulation of the photic synchronization between diurnal and nocturnal species. Of note, the molecular effects observed in response to a serotonergic stimulation are different than those seen after a GABAergic injection, confirming the differential role of these 2 non-photic factors in diurnal species. Once again, these results have to be interpreted carefully, since translational and post- Interactions of the Serotonergic and Circadian Systems ... 31 translational modification could occur in response to the association of photic and non-photic factors. As for the serotonergic non-photic synchronization of the central clock, the serotonergic modulation of the photic synchronization differs greatly between nocturnal and diurnal species, thus reinforcing the need to obtain more data on diurnal species that could then be used in humans for the treatment of different pathologies affecting the circadian system.

Interactions of the Serotonergic and Circadian Systems in Pathological Conditions

As a fundamental coordinator of various biological functions, the endogenous circadian system strongly participates in overall health and longevity. For optimal function, circadian rhythms should be aligned with respect to the sleep-wake cycle. Accordingly, a temporal misalignment between the circadian system and the sleep-wake cycle could have deleterious effects as seen in many disorders, including mental disorders. In addition, several therapeutic approaches based on manipulation of circadian synchronizers have been used successfullyto treat symptoms of several of these disorders. Interestingly, the symptomatic presence of irregular or disrupted sleep-wake cycleinmood disorders, associated with perturbation in the level of different neurotransmitters, including serotonin, raises the question whether these alterations are a consequence of a dysfunctional circadian system. In the last part of this chapter, we first present the impact of a deficiency of the serotonergic neurotransmission on the circadian system and then we review the sleep, circadian rhythms and serotonergic disturbances observed in mood disorders.

Effects of Chronic Serotonergic Deficiency or Input on the Central Clock

While there is an extensive literature on the effects of discrete serotonergic cues on the central clock, studies describing the impact of a deficiency of the serotonergic system or the chronic effects of serotonergic drugs used in the 32 Marc Cuesta and Etienne Challet treatment of mood disorders on the circadian system are more limited. In Syrian hamsters, destruction of 5-HT fibers, using 5,7-DHT, a toxin that selectively kills serotonergic neurons, leads to a lengthening of the active period in LD and DD [242, 243]. Interestingly, in constant light conditions (LL), the lengthening of the endogenous period observed in response to this regimen, disappears in 5,7-DHT treated animals [242]. In mice, 5-HT fiber lesion associated with enucleation lead to a lengthening of the active phase and the endogenous period, as wells as increased total activity and peculiar patterns of activity during the active phase [244]. In addition, destruction of 5- HT fibers results in the loss of observable behavioral phase advances in response to a novel wheel access given during the middle of the subjective day [153]. More recently, the serotonin transporter knockout (5-HTT KO) mouse model was used to study the effects of lifelong elevated 5-HT levels on circadian rhythms [245]. Unexpectedly, while some alterations of sleep architecture present in patients suffering from mood disorders were previously described in 5-HTT KO mice [246, 247], no differences in the set of locomotor activity parameters (i.e. amplitude, period, acrophase and total activity) were found in those mice compared to wild-type mice, when kept in LD or DD. Interestingly, 5-HTT KO mice presented attenuated light-induced behavioral phase delay compared to wild-type mice. The authors suggested that the constant elevated level of extracellular 5-HT, which is supposed to induce a downregulation of the different 5-HT receptors, could have been inefficient to downregulate 5-HT1B receptors located in the RHT terminals, resulting in elevated 5-HT1B activation that would be responsible of the inhibition of the light-induced phase shifts. Despite this result, the lack of circadian perturbations in 5-HTT KO mice might be explained with compensatory changes. Several studies explored the effects of a chronic treatment of fluoxetine, an antidepressant that increases levels of extracellular 5-HT, as seen in 5-HHT KO mice. While some studies showed an absence of effect on the locomotor activity rhythm of rats [248], others reported a shortening of the endogenous period in mice in response to fluoxetine [249]. Chronic fluoxetine treatment would also lead to a downregulation of 5-HT7 receptors [250], as well as 5-HT1B receptors but only at night [251]. The latest result is then in opposition with the hypothesis proposed to explain the effect seen in 5-HTT KO mice. Another recent study investigated the effect of a developmental disruption of the serotonin system [252]. This disruption was obtained by knocking down PC12 ETS domain-containing transcription factor 1 or Pet-1, a transcription Interactions of the Serotonergic and Circadian Systems ... 33 factor that colocalizes with TPH neurons from embryonic day 12.5. Pet-1 was shown to be necessary for the maintenance of a serotonergic phenotype in adults [253] and mice lacking this gene express increased aggression and anxiety-like behavior [254]. Pet-1 KO mice show a significant disruption of the 5-HT with a loss of around 70% of 5-HT cells and a very little expression of the key molecules of the 5-HT system (e.g. TPH, 5-HTT) in the remaining 30%. In addition, Pet-1 KO mice show virtually no-5-HT labeling in the SCN and a very small number of 5-HT labeling in the MRN and DRN. At the circadian level, Pet-1 KO mice express a lengthening of the endogenous period of locomotor activity in DD, as seen in mice treated with 5,7-DHT. This suggests a possible slowing down of the central clock in absence of serotonin. If this was true, this effect would not be mediated by 5-HT1A, 5- HT1B nor 5-HT7 receptors, since mice lacking one of these receptors do not show a lengthening of their endogenous period [173, 174, 255]. Pet-1 KO mice also express a redistribution of their active period in different light dark cycles, with a significant shift of activity from early to late night, leading to a bimodal pattern. Taken together, the results indicate a clear role for 5-HT in the regulation of the timing of locomotor activity, and 5-HT deficiency results in alteration of the circadian system.

Interaction of the Serotonergic and Circadian Systems in Mood Disorders

Mood disorders are a consequence of genetic, environmental, physiological, and psychological factors. This group of complex and multifactorial disorders essentially comprises recurrent unipolar depressive disorder (also known as major depressive disorders or MDD), seasonal affective disorders (SAD) and bipolar affective disorders (BPD).

Major Depressive Disorders In most countries, the number of people who will suffer from MDD during their life is estimated between 8 and 12%. Alteration of the serotonergic system, such as its under-activation, has long been implicated in the pathophysiology of depression [256, 257]. In addition pharmacological treatments using SSRIs have been shown to be efficient in the treatment of depression. Interestingly, another overarching feature of depression is the systematic presence of sleep and circadian disruptions. Indeed, up to 90% of depressed patients suffer from insomnia, which additionally represents a risk 34 Marc Cuesta and Etienne Challet factor for developing and relapsing into MDD. Depressed patients also often present advanced rhythms, as evidenced by reduced rapid eye movement (REM) sleep latency, earlier distribution of REM sleep during the night, and early morning awakenings [258, 259]. Other circadian disturbances have been observed such as greater nocturnal core body temperature, higher overall mean temperature, greater plasma cortisol and lower plasma melatonin levels, although this remains controversial [258, 260-262]. Abnormal phase advances of the circadian system compared to the sleep-wake cycle were also reported. Based on the systematic presence of circadian disturbances in many psychiatric diseases, including mood disorders, many scientific publications over the past years started to focus on the possible association of various clock gene polymorphisms (e.g. single-nucleotide polymorphisms or SNPs) with mental illnesses in humans. However, the results of most of these studies have to be interpreted keeping in mind the limitations commonly related to candidate gene association studies, such as population stratification, sample size and multiple testing issues. Some of these studies revealed that variants of several core clock genes, namely CRY1 and NPAS2 [263], as well as other circadian genes, such as TIM [264] are associated with MDD. Moreover, variants of PROKR2, which is implicated in circadian and homeostatic regulation of sleep in rodents, were found to be moderately associated with MDD [265]. In addition to association studies, a higher expression of CLOCK, PER1, and BMAL1 mRNA in peripheral blood leukocytes among individuals with a history of depression compared to healthy subjects was described [266]. Interestingly, manipulation of the circadian clock can be effective as a treatment for mood disorders [267, 268]. Indeed, chronotherapies, such as bright light exposure can reset circadian phase (depending on the time of application) and be useful for correcting the state of misalignment between the endogenous circadian system and the sleep schedule.Another chronotherapy, named sleep phase advance therapy, could act in the same way by reducing this temporal misalignment. Other non-pharmacological interventions such as sleep deprivation (partial, total, or selective for REM sleep) have been successfully used for MDD on a short-term scale, but would rely on homeostatic rather than circadian processes.As reviewed earlier in this chapter, the serotonergic and circadian systems are extensively intertwined and both systems are altered in MDD. The linkage of these two systems is also apparent clinically, since bright light therapy was described to accelerate and enhance therapeutic effects of SSRIs in MDD [269-271]. In addition, agomelatine, a new promising drug to treat mood disorders, act on both systems at the same time [272]. As a melatonin agonist, it influences the circadian system by a Interactions of the Serotonergic and Circadian Systems ... 35

possible stabilization of the sleep-wake cycle and as a 5-HT2C receptor antagonist, it acts on different parts of the serotonergic system.

Seasonal Affective Disorder SAD is another common mood disorder in which the linkage between serotonergic and circadian system and its clinical relevance might be best illustrated. SAD affects around 10% of the population living in non-tropical zones. The winter form of SAD is characterized by depressive symptoms occurring during the fall and winter months. This might be linked to altered secretion of melatonin, although this remains controversial [258, 273]. According to another model, the phase shift hypothesis, SAD patients become depressed because a misalignment occurs between their circadian system and their sleep schedule [258]. Most of the time, this misalignment is caused by a circadian phase delay associated with the later dawn in winter. In addition, SAD patients complaints of disturbed sleep (e.g. hypersomnia), and present increased non-REM sleep duration and greater slow-wave activity per minute of non-REM sleep [259, 274]. Association studies indicate that variants of PER2, NPAS2 and BMAL1 are significantly associated with SAD [259, 275, 276]. Moreover, certain allelic combinations of SNPs of these 3 genes have an additive effect, leading to an increased risk of developing SAD by 4.43 over other genotypes and 10.67 over the most protective haplotypes [275]. In addition, prior studies such as that of Johansson and colleagues [276] showed a relationship between the PER3 p.Val647Gly genotypeand morningness /eveningness, reinforcing the association between clock genes polymorphism and chronotype. While SAD is considered as a circadian disorder, there is evidence involving the serotonergic system. Polymorphism of the short-repeat allele of 5-HTT gene (5-HTTLPR) is associated with increased seasonality in mood and with atypical depressive symptoms present in SAD [277, 278]. In addition, variants of the gene coding for 5-HT2A receptors are directly associated with increased risk of developing SAD [279, 280]. Interestingly, tph2 gene expression in the raphe nuclei of Syrian hamsters is variable according to the season, with winter-like light cycles suppressing the circadian rhythm of its mRNA expression [126, 281]. This suggests an effect of the photoperiod on a key component of the serotonergic system that could be seen in SAD patients. In accordance with this hypothesis, 5-HT turnover, obtained by measuring 5-HT and its metabolites in blood, is lowest in winter and the rate of its production is increased in response to the duration of bright light [96]. Moreover, in vivo measures of serotonin transporter binding in the human brain are higher at the fall and winter and decreases as the day length 36 Marc Cuesta and Etienne Challet increases [282-284]. Bright light exposure and/or exogenous melatonin administration have been successfully used as attempts to correct abnormal phase position present in SAD [285, 286]. In addition, pharmacotherapy with SSRIs and agomelatine is also effective to alleviate SAD [272, 285, 286]. Overall, seasonal light cues, mediated by the circadian system, influence the serotonergic system in the brain, and both systems contribute to seasonal regulation of mood at the physiological level.

Conclusion

Etiological determinants of mood disorders are still poorly understood although several pieces of evidence point toward fundamental alterations of circadian rhythmicity in addition to a deficiency of the serotonergic system. A model has been proposed for bipolar disorders [287] that could also be used for MDD and SAD. In this model, variants of certain genes, especially clock genes, would affect the ability of the circadian system to adapt to environmental changes and would predispose to sleep-wake disturbances. Considering the interconnections between the circadian and serotonergic systems, it is reasonable to suggest that sleep disturbances and circadian deficits could alter the serotonergic system (as wells as other system such as dopaminergic system), in turn affecting mood regulation. A vicious cycle would then begin, where mood deregulation would affect sleep quality and quantity, which would reinforce the temporal misalignment between the sleep- wake cycle and the circadian system. The convergence of these two systems in mood regulation is well illustrated, especially in SAD, in which there is evidence for perturbation of both systems, and manipulation of each of these systems is amenable to therapeutic strategies. The challenge in the near future will be to better understand the pathophysiology of this serotonergic/circadian cycle that could help to find most appropriate treatments.

References

[1] de Mairan J. Observation botanique. Hist. Acad. Roy. Sci. 1729. [2] Lowrey PL, Takahashi JS. Genetics of circadian rhythms in Mammalian model organisms. Advances in genetics. 2011;74:175-230. eng. Interactions of the Serotonergic and Circadian Systems ... 37

[3] Welsh DK, Takahashi JS, Kay SA. : cell autonomy and network properties. Annual review of physiology. 2010;72:551-77. eng. [4] Morin LP, Shivers KY, Blanchard JH, Muscat L. Complex organization of mouse and rat suprachiasmatic nucleus. Neuroscience. 2006;137(4):1285-97. eng. [5] Moore RY, Eichler VB. Loss of a circadian adrenal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain research. 1972 Jul 13;42(1):201-6. eng. [6] Stephan FK, Zucker I. Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proceedings of the National Academy of Sciences of the United States of America. 1972 Jun;69(6):1583-6. eng. [7] Ralph MR, Foster RG, Davis FC, Menaker M. Transplanted suprachiasmatic nucleus determines circadian period. Science. 1990 Feb 23;247(4945):975-8. eng. [8] Shearman LP, Sriram S, Weaver DR, Maywood ES, Chaves I, Zheng B, et al. Interacting molecular loops in the mammalian circadian clock. Science. 2000 May 12;288(5468):1013-9. eng. [9] Ko CH, Takahashi JS. Molecular components of the mammalian circadian clock. Human molecular genetics. 2006 Oct 15;15 Spec No 2:R271-7. eng. [10] Duong HA, Robles MS, Knutti D, Weitz CJ. A molecular mechanism for circadian clock negative feedback. Science. 2011 Jun 17;332(6036):1436-9. eng. [11] Mazzoccoli G, Pazienza V, Vinciguerra M. Clock genes and clock- controlled genes in the regulation of metabolic rhythms. Chronobiology international. 2012 Apr;29(3):227-51. eng. [12] Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA. Redundant function of REV-ERBalpha and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS genetics. 2008 Feb;4(2):e1000023. eng. [13] Rossner MJ, Oster H, Wichert SP, Reinecke L, Wehr MC, Reinecke J, et al. Disturbed clockwork resetting in Sharp-1 and Sharp-2 single and double mutant mice. PloS one. 2008;3(7):e2762. eng. [14] Duffield GE. DNA microarray analyses of circadian timing: the genomic basis of biological time. Journal of neuroendocrinology. 2003 Oct;15(10):991-1002. eng. 38 Marc Cuesta and Etienne Challet

[15] Jin X, Shearman LP, Weaver DR, Zylka MJ, de Vries GJ, Reppert SM. A molecular mechanism regulating rhythmic output from the suprachiasmatic circadian clock. Cell. 1999 Jan 8;96(1):57-68. eng. [16] Stratmann M, Schibler U. Properties, entrainment, and physiological functions of mammalian peripheral oscillators. Journal of biological rhythms. 2006 Dec;21(6):494-506. [17] Cermakian N, Boivin DB. The regulation of central and peripheral circadian clocks in humans. Obesity reviews : an official journal of the International Association for the Study of Obesity. 2009 Nov;10 Suppl 2:25-36. [18] Balsalobre A, Damiola F, Schibler U. A serum induces circadian gene expression in mammalian tissue culture cells. Cell. 1998 Jun 12;93(6):929-37. eng. [19] Guo H, Brewer JM, Lehman MN, Bittman EL. Suprachiasmatic regulation of circadian rhythms of gene expression in hamster peripheral organs: effects of transplanting the pacemaker. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006 Jun 14;26(24):6406-12. eng. [20] Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U. Circadian gene expression in individual fibroblasts: cell-autonomous and self- sustained oscillators pass time to daughter cells. Cell. 2004 Nov 24;119(5):693-705. eng. [21] Buhr ED, Yoo SH, Takahashi JS. Temperature as a universal resetting cue for mammalian circadian oscillators. Science. 2010 Oct 15;330(6002):379-85. eng. [22] Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes and development. 2000 Dec 1;14(23):2950-61. eng. [23] Tosini G, Menaker M. Circadian rhythms in cultured mammalian retina. Science. 1996 Apr 19;272(5260):419-21. eng. [24] Granados-Fuentes D, Tseng A, Herzog ED. A circadian clock in the olfactory bulb controls olfactory responsivity. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006 Nov 22;26(47):12219-25. eng. [25] Son GH, Chung S, Kim K. The adrenal peripheral clock: glucocorticoid and the circadian timing system. Frontiers in neuroendocrinology. 2011 Oct;32(4):451-65. eng. Interactions of the Serotonergic and Circadian Systems ... 39

[26] Tataroglu O, Davidson AJ, Benvenuto LJ, Menaker M. The methamphetamine-sensitive circadian oscillator (MASCO) in mice. Journal of biological rhythms. 2006 Jun;21(3):185-94. eng. [27] Honma S, Yasuda T, Yasui A, van der Horst GT, Honma K. Circadian behavioral rhythms in Cry1/Cry2 double-deficient mice induced by methamphetamine. Journal of biological rhythms. 2008 Feb;23(1):91-4. eng. [28] Mohawk JA, Baer ML, Menaker M. The methamphetamine-sensitive circadian oscillator does not employ canonical clock genes. Proceedings of the National Academy of Sciences of the United States of America. 2009 Mar 3;106(9):3519-24. eng. [29] Cuesta M, Aungier J, Morton AJ. The methamphetamine-sensitive circadian oscillator is dysfunctional in a transgenic mouse model of Huntington's disease. Neurobiology of disease. 2012 Jan;45(1):145-55. eng. [30] Challet E, Mendoza J, Dardente H, Pevet P. Neurogenetics of food anticipation. The European journal of neuroscience. 2009 Nov;30(9):1676-87. eng. [31] Mistlberger RE. Food-anticipatory circadian rhythms: concepts and methods. The European journal of neuroscience. 2009 Nov;30(9):1718- 29. eng. [32] Verwey M, Amir S. Food-entrainable circadian oscillators in the brain. The European journal of neuroscience. 2009 Nov;30(9):1650-7. eng. [33] Balsalobre A. Clock genes in mammalian peripheral tissues. Cell and tissue research. 2002 Jul;309(1):193-9. eng. [34] Kalsbeek A, van Heerikhuize JJ, Wortel J, Buijs RM. A diurnal rhythm of stimulatory input to the hypothalamo-pituitary-adrenal system as revealed by timed intrahypothalamic administration of the vasopressin V1 antagonist. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1996 Sep 1;16(17):5555-65. eng. [35] Buijs RM, Wortel J, Van Heerikhuize JJ, Feenstra MG, Ter Horst GJ, Romijn HJ, et al. Anatomical and functional demonstration of a multisynaptic suprachiasmatic nucleus adrenal (cortex) pathway. The European journal of neuroscience. 1999 May;11(5):1535-44. eng. [36] Garidou ML, Bartol I, Calgari C, Pevet P, Simonneaux V. In vivo observation of a non-noradrenergic regulation of arylalkylamine N- acetyltransferase gene expression in the rat pineal complex. Neuroscience. 2001;105(3):721-9. eng. 40 Marc Cuesta and Etienne Challet

[37] Kalsbeek A, Garidou ML, Palm IF, Van Der Vliet J, Simonneaux V, Pevet P, et al. Melatonin sees the light: blocking GABA-ergic transmission in the paraventricular nucleus induces daytime secretion of melatonin. The European journal of neuroscience. 2000 Sep;12(9):3146- 54. eng. [38] Kalsbeek A, Yi CX, la Fleur SE, Buijs RM, Fliers E. Suprachiasmatic nucleus and autonomic nervous system influences on awakening from sleep. International review of neurobiology. 2010;93:91-107. eng. [39] Dibner C, Schibler U, Albrecht U. The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annual review of physiology. 2010;72:517-49. eng. [40] Altun A, Ugur-Altun B. Melatonin: therapeutic and clinical utilization. International journal of clinical practice. 2007 May;61(5):835-45. eng. [41] Revel FG, Masson-Pevet M, Pevet P, Mikkelsen JD, Simonneaux V. Melatonin controls seasonal breeding by a network of hypothalamic targets. Neuroendocrinology. 2009;90(1):1-14. eng. [42] Kalsbeek A, van der Spek R, Lei J, Endert E, Buijs RM, Fliers E. Circadian rhythms in the hypothalamo-pituitary-adrenal (HPA) axis. Molecular and cellular endocrinology. 2012 Feb 5;349(1):20-9. eng. [43] Mendoza J, Challet E. Brain clocks: from the suprachiasmatic nuclei to a cerebral network. The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry. 2009 Oct;15(5):477-88. eng. [44] Dardente H, Cermakian N. Molecular circadian rhythms in central and peripheral clocks in mammals. Chronobiology international. 2007;24(2):195-213. eng. [45] Bjarnason GA, Jordan RC, Wood PA, Li Q, Lincoln DW, Sothern RB, et al. Circadian expression of clock genes in human oral mucosa and skin: association with specific cell-cycle phases. The American journal of pathology. 2001 May;158(5):1793-801. eng. [46] Otway DT, Mantele S, Bretschneider S, Wright J, Trayhurn P, Skene DJ, et al. Rhythmic diurnal gene expression in human adipose tissue from individuals who are lean, overweight, and type 2 diabetic. Diabetes. 2011 May;60(5):1577-81. eng. [47] Garaulet M, Ordovas JM, Gomez-Abellan P, Martinez JA, Madrid JA. An approximation to the temporal order in endogenous circadian rhythms of genes implicated in human adipose tissue metabolism. Journal of cellular physiology. 2010 Dec 6. Eng. [48] Mostafaie N, Kallay E, Sauerzapf E, Bonner E, Kriwanek S, Cross HS, et al. Correlated downregulation of estrogen receptor beta and the Interactions of the Serotonergic and Circadian Systems ... 41

circadian clock gene Per1 in human colorectal cancer. Molecular carcinogenesis. 2009 Jul;48(7):642-7. eng. [49] Pardini L, Kaeffer B, Trubuil A, Bourreille A, Galmiche JP. Human intestinal circadian clock: expression of clock genes in colonocytes lining the crypt. Chronobiology international. 2005;22(6):951-61. eng. [50] Akashi M, Soma H, Yamamoto T, Tsugitomi A, Yamashita S, Nishida E, et al. Noninvasive method for assessing the human circadian clock using hair follicle cells. Proceedings of the National Academy of Sciences of the United States of America. 2010 Aug 31;107(35):15643-8. eng. [51] Boivin DB, James FO, Wu A, Cho-Park PF, Xiong H, Sun ZS. Circadian clock genes oscillate in human peripheral blood mononuclear cells. Blood. 2003 Dec 1;102(12):4143-5. eng. [52] Teboul M, Barrat-Petit MA, Li XM, Claustrat B, Formento JL, Delaunay F, et al. Atypical patterns of circadian clock gene expression in human peripheral blood mononuclear cells. Journal of Molecular Medicine (Berl). 2005 Sep;83(9):693-9. eng. [53] James FO, Boivin DB, Charbonneau S, Belanger V, Cermakian N. Expression of clock genes in human peripheral blood mononuclear cells throughout the sleep/wake and circadian cycles. Chronobiology international. 2007;24(6):1009-34. eng. [54] James FO, Cermakian N, Boivin DB. Circadian rhythms of melatonin, cortisol, and clock gene expression during simulated night shift work. Sleep. 2007 Nov;30(11):1427-36. eng. [55] Fukuya H, Emoto N, Nonaka H, Yagita K, Okamura H, Yokoyama M. Circadian expression of clock genes in human peripheral leukocytes. Biochemical and biophysical research communications. 2007 Mar 23;354(4):924-8. eng. [56] Archer SN, Viola AU, Kyriakopoulou V, von Schantz M, Dijk DJ. Inter- individual differences in habitual sleep timing and entrained phase of endogenous circadian rhythms of BMAL1, PER2 and PER3 mRNA in human leukocytes. Sleep. 2008 May;31(5):608-17. eng. [57] Kusanagi H, Hida A, Satoh K, Echizenya M, Shimizu T, Pendergast JS, et al. Expression profiles of 10 circadian clock genes in human peripheral blood mononuclear cells. Neuroscience research. 2008 Jun;61(2):136-42. eng. [58] Kavcic P, Rojc B, Dolenc-Groselj L, Claustrat B, Fujs K, Poljak M. The impact of sleep deprivation and nighttime light exposure on clock gene 42 Marc Cuesta and Etienne Challet

expression in humans. Croatian medical journal. 2011 Oct 15;52(5):594-603. eng. [59] Challet E, Malan A, Pevet P. Daily hypocaloric feeding entrains circadian rhythms of wheel-running and body temperature in rats kept in constant darkness. Neuroscience letters. 1996 Jun 14;211(1):1-4. eng. [60] Challet E, Jacob N, Vuillez P, Pevet P, Malan A. Fos-like immunoreactivity in the circadian timing system of calorie-restricted rats fed at dawn: daily rhythms and light pulse-induced changes. Brain research. 1997 Oct 3;770(1-2):228-36. eng. [61] Hattar S, Liao HW, Takao M, Berson DM, Yau KW. Melanopsin- containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science. 2002 Feb 8;295(5557):1065-70. eng. [62] Morin LP, Blanchard JH, Provencio I. Retinal ganglion cell projections to the hamster suprachiasmatic nucleus, intergeniculate leaflet, and visual midbrain: bifurcation and melanopsin immunoreactivity. The Journal of comparative neurology. 2003 Oct 20;465(3):401-16. eng. [63] Morin LP, Goodless-Sanchez N, Smale L, Moore RY. Projections of the suprachiasmatic nuclei, subparaventricular zone and retrochiasmatic area in the golden hamster. Neuroscience. 1994 Jul;61(2):391-410. eng. [64] Miller AM, Chappell R, Obermeyer WH, Benca RM. Analysis of the retinohypothalamic tract in congenic albino and pigmented rats. Brain research. 1996 Nov 25;741(1-2):348-51. eng. [65] Smale L, Boverhof J. The suprachiasmatic nucleus and intergeniculate leaflet of niloticus, a diurnal murid rodent from East Africa. The Journal of comparative neurology. 1999 Jan 11;403(2):190-208. eng. [66] Goel N, Governale MM, Jechura TJ, Lee TM. Effects of intergeniculate leaflet lesions on circadian rhythms in Octodon degus. Brain research. 2000 Sep 22;877(2):306-13. eng. [67] Morin LP. Serotonin and the regulation of mammalian circadian rhythmicity. Annals of medicine. 1999 Feb;31(1):12-33. eng. [68] Morin LP, Allen CN. The circadian visual system, 2005. Brain research reviews. 2006 Jun;51(1):1-60. eng. [69] Daan S, Pittendrigh, S.C. . A functional analysis of circadian pacemakers in nocturnal rodents. II. The variability of phase response curves. J. Comp. Physiol. 1976;106:253-66. [70] Takahashi JS, DeCoursey PJ, Bauman L, Menaker M. Spectral sensitivity of a novel photoreceptive system mediating entrainment of Interactions of the Serotonergic and Circadian Systems ... 43

mammalian circadian rhythms. Nature. 1984 Mar 8-14;308(5955):186-8. eng. [71] Mahoney M, Bult A, Smale L. Phase response curve and light-induced fos expression in the suprachiasmatic nucleus and adjacent hypothalamus of Arvicanthis niloticus. Journal of biological rhythms. 2001 Apr;16(2):149-62. eng. [72] Caldelas I, Poirel VJ, Sicard B, Pevet P, Challet E. Circadian profile and photic regulation of clock genes in the suprachiasmatic nucleus of a diurnal mammal Arvicanthis ansorgei. Neuroscience. 2003;116(2):583- 91. eng. [73] Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, et al. Resetting central and peripheral circadian oscillators in transgenic rats. Science. 2000 Apr 28;288(5466):682-5. eng. [74] Vuillez P, Jacob N, Teclemariam-Mesbah R, Van Rossum A, Vivien- Roels B, Pevet P. Effect of NMDA receptor antagonist MK-801 on light-induced Fos expression in the suprachiasmatic nuclei and on melatonin production in the Syrian hamster. Journal of neuroendocrinology. 1998 Sep;10(9):671-7. eng. [75] Ding JM, Chen D, Weber ET, Faiman LE, Rea MA, Gillette MU. Resetting the biological clock: mediation of nocturnal circadian shifts by glutamate and NO. Science. 1994 Dec 9;266(5191):1713-7. eng. [76] Ding JM, Buchanan GF, Tischkau SA, Chen D, Kuriashkina L, Faiman LE, et al. A neuronal ryanodine receptor mediates light-induced phase delays of the circadian clock. Nature. 1998 Jul 23;394(6691):381-4. eng. [77] Takahashi H, Umeda N, Tsutsumi Y, Fukumura R, Ohkaze H, Sujino M, et al. Mouse dexamethasone-induced RAS protein 1 gene is expressed in a circadian rhythmic manner in the suprachiasmatic nucleus. Brain research Molecular brain research. 2003 Jan 31;110(1):1-6. eng. [78] Ginty DD, Kornhauser JM, Thompson MA, Bading H, Mayo KE, Takahashi JS, et al. Regulation of CREB phosphorylation in the suprachiasmatic nucleus by light and a circadian clock. Science. 1993 Apr 9;260(5105):238-41. eng. [79] Obrietan K, Impey S, Smith D, Athos J, Storm DR. Circadian regulation of cAMP response element-mediated gene expression in the suprachiasmatic nuclei. The Journal of biological chemistry. 1999 Jun 18;274(25):17748-56. eng. [80] von Gall C, Duffield GE, Hastings MH, Kopp MD, Dehghani F, Korf HW, et al. CREB in the mouse SCN: a molecular interface coding the phase-adjusting stimuli light, glutamate, PACAP, and melatonin for 44 Marc Cuesta and Etienne Challet

clockwork access. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1998 Dec 15;18(24):10389-97. eng. [81] Ding JM, Faiman LE, Hurst WJ, Kuriashkina LR, Gillette MU. Resetting the biological clock: mediation of nocturnal CREB phosphorylation via light, glutamate, and nitric oxide. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1997 Jan 15;17(2):667-75. eng. [82] Albrecht U, Sun ZS, Eichele G, Lee CC. A differential response of two putative mammalian circadian regulators, mper1 and mper2, to light. Cell. 1997 Dec 26;91(7):1055-64. eng. [83] Miyake S, Sumi Y, Yan L, Takekida S, Fukuyama T, Ishida Y, et al. Phase-dependent responses of Per1 and Per2 genes to a light-stimulus in the suprachiasmatic nucleus of the rat. Neuroscience letters. 2000 Nov 10;294(1):41-4. eng. [84] Shearman LP, Zylka MJ, Weaver DR, Kolakowski LF, Jr., Reppert SM. Two period homologs: circadian expression and photic regulation in the suprachiasmatic nuclei. Neuron. 1997 Dec;19(6):1261-9. eng. [85] Dardente H, Klosen P, Caldelas I, Pevet P, Masson-Pevet M. Phenotype of Per1- and Per2-expressing neurons in the suprachiasmatic nucleus of a diurnal rodent (Arvicanthis ansorgei): comparison with a nocturnal species, the rat. Cell and tissue research. 2002 Oct;310(1):85-92. eng. [86] Moriya T, Horikawa K, Akiyama M, Shibata S. Correlative association between N-methyl-D-aspartate receptor-mediated expression of period genes in the suprachiasmatic nucleus and phase shifts in behavior with photic entrainment of clock in hamsters. Molecular pharmacology. 2000 Dec;58(6):1554-62. eng. [87] Akiyama M, Kouzu Y, Takahashi S, Wakamatsu H, Moriya T, Maetani M, et al. Inhibition of light- or glutamate-induced mPer1 expression represses the phase shifts into the mouse circadian locomotor and suprachiasmatic firing rhythms. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1999 Feb 1;19(3):1115- 21. eng. [88] Wakamatsu H, Takahashi S, Moriya T, Inouye ST, Okamura H, Akiyama M, et al. Additive effect of mPer1 and mPer2 antisense oligonucleotides on light-induced phase shift. Neuroreport. 2001 Jan 22;12(1):127-31. eng. [89] Albrecht U, Zheng B, Larkin D, Sun ZS, Lee CC. MPer1 and mper2 are essential for normal resetting of the circadian clock. Journal of biological rhythms. 2001 Apr;16(2):100-4. eng. Interactions of the Serotonergic and Circadian Systems ... 45

[90] de Arriba Zerpa GA, Guido ME, Bussolino DF, Pasquare SJ, Castagnet PI, Giusto NM, et al. Light exposure activates retina ganglion cell lysophosphatidic acid acyl transferase and phosphatidic acid phosphatase by a c-fos-dependent mechanism. Journal of neurochemistry. 1999 Sep;73(3):1228-35. [91] Abe H, Honma S, Namihira M, Tanahashi Y, Ikeda M, Honma K. Circadian rhythm and light responsiveness of BMAL1 expression, a partner of mammalian clock gene Clock, in the suprachiasmatic nucleus of rats. Neuroscience letters. 1998 Dec 18;258(2):93-6. eng. [92] Okamura H, Miyake S, Sumi Y, Yamaguchi S, Yasui A, Muijtjens M, et al. Photic induction of mPer1 and mPer2 in cry-deficient mice lacking a biological clock. Science. 1999 Dec 24;286(5449):2531-4. eng. [93] Masana MI, Sumaya IC, Becker-Andre M, Dubocovich ML. Behavioral characterization and modulation of circadian rhythms by light and melatonin in C3H/HeN mice homozygous for the RORbeta knockout. American journal of physiology Regulatory, integrative and comparative physiology. 2007 Jun;292(6):R2357-67. eng. [94] Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, et al. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell. 2002 Jul 26;110(2):251-60. eng. [95] Cuesta M, Clesse D, Pevet P, Challet E. New light on the serotonergic paradox in the rat circadian system. Journal of neurochemistry. 2009 Jul;110(1):231-43. eng. [96] Jacobs BL, Azmitia EC. Structure and function of the brain serotonin system. Physiological Reviews. 1992 Jan 1;72:165-229. eng. [97] Meyer-Bernstein EL, Morin LP. Differential serotonergic innervation of the suprachiasmatic nucleus and the intergeniculate leaflet and its role in circadian rhythm modulation. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1996 Mar 15;16(6):2097-111. eng. [98] Mosko SS, Haubrich D, Jacobs BL. Serotonergic afferents to the dorsal raphe nucleus: evdience from HRP and synaptosomal uptake studies. Brain research. 1977 Jan 7;119(2):269-90. eng. [99] Leander P, Vrang N, Moller M. Neuronal projections from the mesencephalic raphe nuclear complex to the suprachiasmatic nucleus and the deep pineal gland of the golden hamster (Mesocricetus auratus). The Journal of comparative neurology. 1998 Sep 14;399(1):73-93. eng. 46 Marc Cuesta and Etienne Challet

[100] Kawano H, Decker K, Reuss S. Is there a direct retina-raphe- suprachiasmatic nucleus pathway in the rat? Neuroscience letters. 1996 Jul 12;212(2):143-6. eng. [101] Foote WE, Taber-Pierce E, Edwards L. Evidence for a retinal projection to the midbrain raphe of the cat. Brain research. 1978 Nov 3;156(1):135-40. eng. [102] Shen H, Semba K. A direct retinal projection to the dorsal raphe nucleus in the rat. Brain research. 1994 Jan 28;635(1-2):159-68. eng. [103] Hay-Schmidt A, Vrang N, Larsen PJ, Mikkelsen JD. Projections from the raphe nuclei to the suprachiasmatic nucleus of the rat. Journal of chemical neuroanatomy. 2003 Jul;25(4):293-310. eng. [104] Yamakawa GR, Antle MC. Phenotype and function of raphe projections to the suprachiasmatic nucleus. The European journal of neuroscience. 2010 Jun;31(11):1974-83. eng. [105] Hoyer D, Martin G. 5-HT receptor classification and nomenclature: towards a harmonization with the . Neuropharmacology. 1997 Apr-May;36(4-5):419-28. eng. [106] Wright DE, Seroogy KB, Lundgren KH, Davis BM, Jennes L. Comparative localization of serotonin1A, 1C, and 2 receptor subtype mRNAs in rat brain. The Journal of comparative neurology. 1995 Jan 16;351(3):357-73. [107] Duncan MJ, Short J, Wheeler DL. Comparison of the effects of aging on 5-HT7 and 5-HT1A receptors in discrete regions of the circadian timing system in hamsters. Brain research. 1999 May 22;829(1-2):39-45. [108] Bonaventure P, Nepomuceno D, Kwok A, Chai W, Langlois X, Hen R, et al. Reconsideration of 5-hydroxytryptamine (5-HT)(7) receptor distribution using [(3)H]5-carboxamidotryptamine and [(3)H]8-hydroxy- 2-(di-n-propylamino)tetraline: analysis in brain of 5-HT(1A) knockout and 5-HT(1A/1B) double-knockout mice. The Journal of pharmacology and experimental therapeutics. 2002 Jul;302(1):240-8. eng. [109] Manrique C, Francois-Bellan AM, Segu L, Becquet D, Hery M, Faudon M, et al. Impairment of serotoninergic transmission is followed by adaptive changes in 5HT1B binding sites in the rat suprachiasmatic nucleus. Brain research. 1994 Nov 7;663(1):93-100. [110] Pickard GE, Smith BN, Belenky M, Rea MA, Dudek FE, Sollars PJ. 5- HT1B receptor-mediated presynaptic inhibition of retinal input to the suprachiasmatic nucleus. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1999 May 15;19(10):4034-45. Interactions of the Serotonergic and Circadian Systems ... 47

[111] Belenky MA, Pickard GE. Subcellular distribution of 5-HT(1B) and 5- HT(7) receptors in the mouse suprachiasmatic nucleus. The Journal of comparative neurology. 2001 Apr 9;432(3):371-88. [112] Moyer RW, Kennaway DJ. Immunohistochemical localization of serotonin receptors in the rat suprachiasmatic nucleus. Neuroscience letters. 1999 Aug 27;271(3):147-50. [113] Oliver KR, Kinsey AM, Wainwright A, Sirinathsinghji DJ. Localization of 5-ht(5A) receptor-like immunoreactivity in the rat brain. Brain research. 2000 Jun 9;867(1-2):131-42. [114] Duncan MJ, Jennes L, Jefferson JB, Brownfield MS. Localization of serotonin(5A) receptors in discrete regions of the circadian timing system in the Syrian hamster. Brain research. 2000 Jun 30;869(1- 2):178-85. [115] Neumaier JF, Sexton TJ, Yracheta J, Diaz AM, Brownfield M. Localization of 5-HT(7) receptors in rat brain by immunocytochemistry, in situ hybridization, and agonist stimulated cFos expression. Journal of chemical neuroanatomy. 2001 Jan;21(1):63-73. [116] Carrillo M, Ricci LA, Schwartzer JJ, Melloni RH. Immunohistochemical characterization of 5-HT(3A) receptors in the Syrian hamster forebrain. Brain research. 2010 May 6;1329:67-81. [117] Chalmers DT, Watson SJ. Comparative anatomical distribution of 5- HT1A receptor mRNA and 5-HT1A binding in rat brain--a combined in situ hybridisation/in vitro receptor autoradiographic study. Brain research. 1991 Oct 4;561(1):51-60. eng. [118] Duncan MJ, Hensler JG. Aging alters in a region-specific manner serotonin transporter sites and 5-HT(1A) receptor-G protein interactions in hamster brain. Neuropharmacology. 2002 Jul;43(1):36-44. eng. [119] Lovenberg W, Jequier E, Sjoerdsma A. Tryptophan hydroxylation: measurement in pineal gland, brainstem, and carcinoid tumor. Science. 1967 Jan 13;155(3759):217-9. eng. [120] Malek ZS, Pevet P, Raison S. Circadian change in tryptophan hydroxylase protein levels within the rat intergeniculate leaflets and raphe nuclei. Neuroscience. 2004;125(3):749-58. eng. [121] Malek ZS, Dardente H, Pevet P, Raison S. Tissue-specific expression of tryptophan hydroxylase mRNAs in the rat midbrain: anatomical evidence and daily profiles. The European journal of neuroscience. 2005 Aug;22(4):895-901. eng. [122] Barassin S, Raison S, Saboureau M, Bienvenu C, Maitre M, Malan A, et al. Circadian tryptophan hydroxylase levels and serotonin release in the 48 Marc Cuesta and Etienne Challet

suprachiasmatic nucleus of the rat. The European journal of neuroscience. 2002 Mar;15(5):833-40. eng. [123] Kan JP, Chouvet G, Hery F, Debilly G, Mermet A, Glowinski J, et al. Daily variations of various parameters of serotonin metabolism in the rat brain. I. Circadian variations of tryptophan-5-hydroxylase in the raphe nuclei and the striatum. Brain research. 1977 Mar 4;123(1):125-36. eng. [124] McLennan IS, Lees GJ. Diurnal changes in the kinetic properties of tryptophan hydroxylase from rat brain. Journal of neurochemistry. 1978 Aug;31(2):557-9. eng. [125] Malek ZS, Sage D, Pevet P, Raison S. Daily rhythm of tryptophan hydroxylase-2 messenger ribonucleic acid within raphe neurons is induced by corticoid daily surge and modulated by enhanced locomotor activity. Endocrinology. 2007 Nov;148(11):5165-72. eng. [126] Nexon L, Sage D, Pevet P, Raison S. Glucocorticoid-mediated nycthemeral and photoperiodic regulation of tph2 expression. The European journal of neuroscience. 2011 Apr;33(7):1308-17. eng. [127] Donner NC, Montoya CD, Lukkes JL, Lowry CA. Chronic non-invasive corticosterone administration abolishes the diurnal pattern of tph2 expression. Psychoneuroendocrinology. 2012 May;37(5):645-61. eng. [128] Quay WB. Differences in circadian rhythms in 5-hydroxytryptamine according to brain region. The American journal of physiology. 1968 Dec;215(6):1448-53. eng. [129] Poncet L, Denoroy L, Jouvet M. Daily variations in in vivo tryptophan hydroxylation and in the contents of serotonin and 5- hydroxyindoleacetic acid in discrete brain areas of the rat. Journal of neural transmission General section. 1993;92(2-3):137-50. eng. [130] Cuesta M, Mendoza J, Clesse D, Pevet P, Challet E. Serotonergic activation potentiates light resetting of the main circadian clock and alters clock gene expression in a diurnal rodent. Experimental neurology. 2008 Apr;210(2):501-13. eng. [131] Dudley TE, DiNardo LA, Glass JD. Endogenous regulation of serotonin release in the hamster suprachiasmatic nucleus. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1998 Jul 1;18(13):5045-52. eng. [132] Grossman GH, Farnbauch L, Glass JD. Regulation of serotonin release in the Syrian hamster intergeniculate leaflet region. Neuroreport. 2004 Jan 19;15(1):103-6. eng. [133] Ushijima K, Sakaguchi H, Sato Y, To H, Koyanagi S, Higuchi S, et al. Chronopharmacological study of antidepressants in forced swimming Interactions of the Serotonergic and Circadian Systems ... 49

test of mice. The Journal of pharmacology and experimental therapeutics. 2005 Nov;315(2):764-70. eng. [134] Ushijima K, Koyanagi S, Sato Y, Ogata T, Matsunaga N, Fujimura A, et al. Role of activating transcription factor-4 in 24-hour rhythm of serotonin transporter expression in the mouse midbrain. Molecular pharmacology. 2012 Aug;82(2):264-70. eng. [135] Tsai JW, Hannibal J, Hagiwara G, Colas D, Ruppert E, Ruby NF, et al. Melanopsin as a sleep modulator: circadian gating of the direct effects of light on sleep and altered sleep homeostasis in Opn4(-/-) mice. PLoS biology. 2009 Jun 9;7(6):e1000125. [136] Trulson ME, Jacobs BL. Raphe unit activity in freely moving cats: correlation with level of behavioral arousal. Brain research. 1979 Mar 9;163(1):135-50. eng. [137] Nakamaru-Ogiso E, Miyamoto H, Hamada K, Tsukada K, Takai K. Novel biochemical manipulation of brain serotonin reveals a role of serotonin in the circadian rhythm of sleep-wake cycles. The European journal of neuroscience. 2012 Jun;35(11):1762-70. eng. [138] Monti JM. The role of dorsal raphe nucleus serotonergic and non- serotonergic neurons, and of their receptors, in regulating waking and rapid eye movement (REM) sleep. Sleep medicine reviews. 2010 Oct;14(5):319-27. eng. [139] Aschoff J, Daan, S, Honma KI. Zeitgebers, entrainment, and masking: some unsettled questions. 1st ed. Aschoff J, Daan, S et al., editor. Berlin: Springer-Verlag; 1982. [140] Redlin U. Neural basis and biological function of masking by light in mammals: suppression of melatonin and locomotor activity. Chronobiology international. 2001 Sep;18(5):737-58. eng. [141] Redlin U, Mrosovsky N. Nocturnal activity in a diurnal rodent (Arvicanthis niloticus): the importance of masking. Journal of biological rhythms. 2004 Feb;19(1):58-67. eng. [142] Reebs SG, Mrosovsky N. Effects of induced wheel running on the circadian activity rhythms of Syrian hamsters: entrainment and phase response curve. Journal of biological rhythms. 1989 Spring;4(1):39-48. eng. [143] Maywood ES, Mrosovsky N, Field MD, Hastings MH. Rapid down- regulation of mammalian period genes during behavioral resetting of the circadian clock. Proceedings of the National Academy of Sciences of the United States of America. 1999 Dec 21;96(26):15211-6. eng. 50 Marc Cuesta and Etienne Challet

[144] Maywood ES, Mrosovsky N. A molecular explanation of interactions between photic and non-photic circadian clock-resetting stimuli. Brain research Gene expression patterns. 2001 Aug;1(1):27-31. eng. [145] Antle MC, Mistlberger RE. Circadian clock resetting by sleep deprivation without exercise in the Syrian hamster. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2000 Dec 15;20(24):9326-32. eng. [146] Challet E, Turek FW, Laute M, Van Reeth O. Sleep deprivation decreases phase-shift responses of circadian rhythms to light in the mouse: role of serotonergic and metabolic signals. Brain research. 2001 Aug 3;909(1-2):81-91. eng. [147] Mistlberger RE. Effects of daily schedules of forced activity on free- running rhythms in the rat. Journal of biological rhythms. 1991 Spring;6(1):71-80. eng. [148] Sharma VK, Chandrashekaran MK. Rapid phase resetting of a mammalian circadian rhythm by brief light pulses. Chronobiology international. 1997 Nov;14(6):537-48. [149] Hut RA, Mrosovsky N, Daan S. Nonphotic entrainment in a diurnal mammal, the European ground squirrel (Spermophilus citellus). Journal of biological rhythms. 1999 Oct;14(5):409-19. eng. [150] Buxton OM, Frank SA, L'Hermite-Baleriaux M, Leproult R, Turek FW, Van Cauter E. Roles of intensity and duration of nocturnal exercise in causing phase delays of human circadian rhythms. The American journal of physiology. 1997 Sep;273(3 Pt 1):E536-42. eng. [151] Buxton OM, Lee CW, L'Hermite-Baleriaux M, Turek FW, Van Cauter E. Exercise elicits phase shifts and acute alterations of melatonin that vary with circadian phase. American journal of physiology Regulatory, integrative and comparative physiology. 2003 Mar;284(3):R714-24. eng. [152] Van Reeth O, Sturis J, Byrne MM, Blackman JD, L'Hermite-Baleriaux M, Leproult R, et al. Nocturnal exercise phase delays circadian rhythms of melatonin and thyrotropin secretion in normal men. The American journal of physiology. 1994 Jun;266(6 Pt 1):E964-74. eng. [153] Edgar DM, Reid MS, Dement WC. Serotonergic afferents mediate activity-dependent entrainment of the mouse circadian clock. The American journal of physiology. 1997 Jul;273(1 Pt 2):R265-9. eng. [154] Caldelas I, Challet E, Saboureau M, Pevet P. Light and melatonin inhibit in vivo serotonergic phase advances without altering serotonergic- induced decrease of per expression in the hamster suprachiasmatic Interactions of the Serotonergic and Circadian Systems ... 51

nucleus. Journal of molecular neuroscience : MN. 2005;25(1):53-63. eng. [155] Challet E, Scarbrough K, Penev PD, Turek FW. Roles of suprachiasmatic nuclei and intergeniculate leaflets in mediating the phase-shifting effects of a serotonergic agonist and their photic modulation during subjective day. Journal of biological rhythms. 1998 Oct;13(5):410-21. eng. [156] Cutrera RA, Ouarour A, Pevet P. Effects of the 5-HT1a receptor agonist 8-OH-DPAT and other non-photic stimuli on the circadian rhythm of wheel-running activity in hamsters under different constant conditions. Neuroscience letters. 1994 May 19;172(1-2):27-30. eng. [157] Cutrera RA, Saboureau M, Pevet P. Phase-shifting effect of 8-OH- DPAT, a 5-HT1A/5-HT7 receptor agonist, on locomotor activity in golden hamster in constant darkness. Neuroscience letters. 1996 May 24;210(1):1-4. eng. [158] Edgar DM, Miller JD, Prosser RA, Dean RR, Dement WC. Serotonin and the mammalian circadian system: II. Phase-shifting rat behavioral rhythms with serotonergic agonists. Journal of biological rhythms. 1993 Spring;8(1):17-31. eng. [159] Ehlen JC, Grossman GH, Glass JD. In vivo resetting of the hamster circadian clock by 5-HT7 receptors in the suprachiasmatic nucleus. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2001 Jul 15;21(14):5351-7. eng. [160] Horikawa K, Yokota S, Fuji K, Akiyama M, Moriya T, Okamura H, et al. Nonphotic entrainment by 5-HT1A/7 receptor agonists accompanied by reduced Per1 and Per2 mRNA levels in the suprachiasmatic nuclei. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2000 Aug 1;20(15):5867-73. eng. [161] Horikawa K, Shibata S. Phase-resetting response to (+)8-OH-DPAT, a serotonin 1A/7 receptor agonist, in the mouse in vivo. Neuroscience letters. 2004 Sep 23;368(2):130-4. eng. [162] Prosser RA, Miller JD, Heller HC. A serotonin agonist phase-shifts the circadian clock in the suprachiasmatic nuclei in vitro. Brain research. 1990 Nov 26;534(1-2):336-9. eng. [163] Mendoza J, Clesse D, Pevet P, Challet E. Serotonergic potentiation of dark pulse-induced phase-shifting effects at midday in hamsters. Journal of neurochemistry. 2008 Aug;106(3):1404-14. eng. [164] Sprouse J, Li X, Stock J, McNeish J, Reynolds L. Circadian rhythm phenotype of 5-HT7 receptor knockout mice: 5-HT and 8-OH-DPAT- 52 Marc Cuesta and Etienne Challet

induced phase advances of SCN neuronal firing. Journal of biological rhythms. 2005 Apr;20(2):122-31. [165] Prosser RA, Lee HM, Wehner A. Serotonergic pre-treatments block in vitro serotonergic phase shifts of the mouse suprachiasmatic nucleus circadian clock. Neuroscience. 2006 Oct 13;142(2):547-55. [166] Knoch ME, Gobes SM, Pavlovska I, Su C, Mistlberger RE, Glass JD. Short-term exposure to constant light promotes strong circadian phase- resetting responses to nonphotic stimuli in Syrian hamsters. The European journal of neuroscience. 2004 May;19(10):2779-90. [167] Mistlberger RE, Belcourt J, Antle MC. Circadian clock resetting by sleep deprivation without exercise in Syrian hamsters: dark pulses revisited. Journal of biological rhythms. 2002 Jun;17(3):227-37. [168] Glass JD, Brager AJ, Stowie AC, Prosser RA. Cocaine modulates pathways for photic and nonphotic entrainment of the mammalian SCN circadian clock. American journal of physiology Regulatory, integrative and comparative physiology. 2012 Mar 15;302(6):R740-50. [169] Colbron S, Jones M, Biello SM. MDMA alters the response of the circadian clock to a photic and non-photic stimulus. Brain research. 2002 Nov 22;956(1):45-52. [170] Cuesta M, Clesse D, Pevet P, Challet E. From daily behavior to hormonal and neurotransmitters rhythms: comparison between diurnal and nocturnal rat species. Hormones and behavior. 2009 Feb;55(2):338- 47. eng. [171] Tominaga K, Shibata S, Ueki S, Watanabe S. Effects of 5-HT1A receptor agonists on the circadian rhythm of wheel-running activity in hamsters. European journal of pharmacology. 1992 Apr 7;214(1):79-84. [172] Duncan MJ, Grear KE, Hoskins MA. Aging and SB-269970-A, a selective 5-HT7 receptor antagonist, attenuate circadian phase advances induced by microinjections of serotonergic drugs in the hamster dorsal raphe nucleus. Brain research. 2004 May 15;1008(1):40-8. [173] Smith VM, Sterniczuk R, Phillips CI, Antle MC. Altered photic and non-photic phase shifts in 5-HT(1A) receptor knockout mice. Neuroscience. 2008 Dec 2;157(3):513-23. [174] Gardani M, Biello SM. The effects of photic and nonphotic stimuli in the 5-HT7 receptor knockout mouse. Neuroscience. 2008 Mar 3;152(1):245- 53. [175] Sprouse J, Reynolds L, Braselton J, Schmidt A. Serotonin-induced phase advances of SCN neuronal firing in vitro: a possible role for 5-HT5A receptors? Synapse. 2004 Nov;54(2):111-8. Interactions of the Serotonergic and Circadian Systems ... 53

[176] Meyer-Bernstein EL, Morin LP. Electrical stimulation of the median or dorsal raphe nuclei reduces light-induced FOS protein in the suprachiasmatic nucleus and causes circadian activity rhythm phase shifts. Neuroscience. 1999;92(1):267-79. [177] De Vivo M, Maayani S. Characterization of the 5-hydroxytryptamine1a receptor-mediated inhibition of forskolin-stimulated adenylate cyclase activity in guinea pig and rat hippocampal membranes. The Journal of pharmacology and experimental therapeutics. 1986 Jul;238(1):248-53. [178] Jasper JR, Kosaka A, To ZP, Chang DJ, Eglen RM. Cloning, expression and pharmacology of a truncated splice variant of the human 5-HT7 receptor (h5-HT7b). British journal of pharmacology. 1997 Sep;122(1):126-32. [179] Kawahara F, Saito H, Katsuki H. Inhibition by 5-HT7 receptor stimulation of GABAA receptor-activated current in cultured rat suprachiasmatic neurones. The Journal of physiology. 1994 Jul 1;478 ( Pt 1):67-73. [180] Roberts C, Thomas DR, Bate ST, Kew JN. GABAergic modulation of 5- HT7 receptor-mediated effects on 5-HT efflux in the guinea-pig dorsal raphe nucleus. Neuropharmacology. 2004 Jun;46(7):935-41. [181] Srkalovic G, Selim M, Rea MA, Glass JD. Serotonergic inhibition of extracellular glutamate in the suprachiasmatic nuclear region assessed using in vivo brain microdialysis. Brain research. 1994 Sep 12;656(2):302-8. [182] Ying SW, Rusak B. Effects of serotonergic agonists on firing rates of photically responsive cells in the hamster suprachiasmatic nucleus. Brain research. 1994 Jul 18;651(1-2):37-46. [183] Ying SW, Rusak B. 5-HT7 receptors mediate serotonergic effects on light-sensitive suprachiasmatic nucleus neurons. Brain research. 1997 May 2;755(2):246-54. [184] Huhman KL, Albers HE. Neuropeptide Y microinjected into the suprachiasmatic region phase shifts circadian rhythms in constant darkness. Peptides. 1994;15(8):1475-8. [185] Harrington ME. The ventral lateral geniculate nucleus and the intergeniculate leaflet: interrelated structures in the visual and circadian systems. Neuroscience and biobehavioral reviews. 1997 Sep;21(5): 705-27. [186] Biello SM, Golombek DA, Harrington ME. Neuropeptide Y and glutamate block each other's phase shifts in the suprachiasmatic nucleus in vitro. Neuroscience. 1997 Apr;77(4):1049-57. 54 Marc Cuesta and Etienne Challet

[187] Biello SM, Janik D, Mrosovsky N. Neuropeptide Y and behaviorally induced phase shifts. Neuroscience. 1994 Sep;62(1):273-9. [188] Schuhler S, Pitrosky B, Saboureau M, Lakhdar-Ghazal N, Pevet P. Role of the thalamic intergeniculate leaflet and its 5-HT afferences in the chronobiological properties of 8-OH-DPAT and triazolam in syrian hamster. Brain research. 1999 Dec 4;849(1-2):16-24. [189] Glass JD, Guinn J, Kaur G, Francl JM. On the intrinsic regulation of neuropeptide Y release in the mammalian suprachiasmatic nucleus circadian clock. The European journal of neuroscience. 2010 Mar;31(6):1117-26. [190] Golombek DA, Biello SM, Rendon RA, Harrington ME. Neuropeptide Y phase shifts the circadian clock in vitro via a Y2 receptor. Neuroreport. 1996 May 17;7(7):1315-9. [191] Huhman KL, Gillespie CF, Marvel CL, Albers HE. Neuropeptide Y phase shifts circadian rhythms in vivo via a Y2 receptor. Neuroreport. 1996 May 17;7(7):1249-52. [192] Biello SM, Golombek DA, Schak KM, Harrington ME. Circadian phase shifts to neuropeptide Y In vitro: cellular communication and signal transduction. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1997 Nov 1;17(21):8468-75. [193] Fukuhara C, Brewer JM, Dirden JC, Bittman EL, Tosini G, Harrington ME. Neuropeptide Y rapidly reduces Period 1 and Period 2 mRNA levels in the hamster suprachiasmatic nucleus. Neuroscience letters. 2001 Nov 16;314(3):119-22. [194] Maywood ES, Okamura H, Hastings MH. Opposing actions of neuropeptide Y and light on the expression of circadian clock genes in the mouse suprachiasmatic nuclei. The European journal of neuroscience. 2002 Jan;15(1):216-20. [195] Mintz EM, Jasnow AM, Gillespie CF, Huhman KL, Albers HE. GABA interacts with photic signaling in the suprachiasmatic nucleus to regulate circadian phase shifts. Neuroscience. 2002;109(4):773-8. [196] Smith RD, Inouye S, Turek FW. Central administration of muscimol phase-shifts the mammalian circadian clock. Journal of comparative physiology A, Sensory, neural, and behavioral physiology. 1989 Feb;164(6):805-14. [197] Novak CM, Ehlen JC, Paul KN, Fukuhara C, Albers HE. Light and GABA)(A) receptor activation alter period mRNA levels in the SCN of diurnal Nile grass rats. The European journal of neuroscience. 2006 Nov;24(10):2843-52. Interactions of the Serotonergic and Circadian Systems ... 55

[198] Novak CM, Albers HE. Circadian phase alteration by GABA and light differs in diurnal and nocturnal rodents during the day. Behavioral neuroscience. 2004 Jun;118(3):498-504. [199] Medanic M, Gillette MU. Serotonin regulates the phase of the rat suprachiasmatic circadian pacemaker in vitro only during the subjective day. The Journal of physiology. 1992 May;450:629-42. [200] Sprouse J, Braselton J, Reynolds L. Fluoxetine modulates the circadian biological clock via phase advances of suprachiasmatic nucleus neuronal firing. Biological psychiatry. 2006 Oct 15;60(8):896-9. [201] Graff C, Kohler M, Pevet P, Wollnik F. Involvement of the retinohypothalamic tract in the photic-like effects of the serotonin agonist quipazine in the rat. Neuroscience. 2005;135(1):273-83. [202] Graff C, Challet E, Pevet P, Wollnik F. 5-HT3 receptor-mediated photic- like responses of the circadian clock in the rat. Neuropharmacology. 2007 Feb;52(2):662-71. [203] Kohler M, Kalkowski A, Wollnik F. Serotonin agonist quipazine induces photic-like phase shifts of the circadian activity rhythm and c-Fos expression in the rat suprachiasmatic nucleus. Journal of biological rhythms. 1999 Apr;14(2):131-40. [204] Kennaway DJ, Rowe SA, Ferguson SA. Serotonin agonists mimic the phase shifting effects of light on the melatonin rhythm in rats. Brain research. 1996 Oct 21;737(1-2):301-7. [205] Kennaway DJ, Moyer RW. Serotonin 5-HT2c agonists mimic the effect of light pulses on circadian rhythms. Brain research. 1998 Sep 28;806(2):257-70. [206] Varcoe TJ, Kennaway DJ, Voultsios A. Activation of 5-HT2C receptors acutely induces Per gene expression in the rat suprachiasmatic nucleus at night. Brain research Molecular brain research. 2003 Nov 26;119(2):192-200. [207] Varcoe TJ, Kennaway DJ. Activation of 5-HT2C receptors acutely induces Per1 gene expression in the rat SCN in vitro. Brain research. 2008 May 13;1209:19-28. [208] Gannon RL, Millan MJ. The corticotropin-releasing factor (CRF)(1) receptor antagonists CP154,526 and DMP695 inhibit light-induced phase advances of hamster circadian activity rhythms. Brain research. 2006 Apr 14;1083(1):96-102. [209] Gannon RL. Serotonergic serotonin (1A) mixed agonists/antagonists elicit large-magnitude phase shifts in hamster circadian wheel-running rhythms. Neuroscience. 2003;119(2):567-76. 56 Marc Cuesta and Etienne Challet

[210] Ralph MR, Mrosovsky N. Behavioral inhibition of circadian responses to light. Journal of biological rhythms. 1992 Winter;7(4):353-9. [211] Weber ET, Rea MA. Neuropeptide Y blocks light-induced phase advances but not delays of the circadian activity rhythm in hamsters. Neuroscience letters. 1997 Aug 15;231(3):159-62. [212] Lall GS, Biello SM. Attenuation of circadian light induced phase advances and delays by neuropeptide Y and a neuropeptide Y Y1/Y5 receptor agonist. Neuroscience. 2003;119(2):611-8. [213] Gillespie CF, Huhman KL, Babagbemi TO, Albers HE. Bicuculline increases and muscimol reduces the phase-delaying effects of light and VIP/PHI/GRP in the suprachiasmatic region. Journal of biological rhythms. 1996 Jun;11(2):137-44. [214] Gillespie CF, Mintz EM, Marvel CL, Huhman KL, Albers HE. GABA(A) and GABA(B) agonists and antagonists alter the phase- shifting effects of light when microinjected into the suprachiasmatic region. Brain research. 1997 Jun 13;759(2):181-9. [215] Gannon RL, Millan MJ. Evaluation of serotonin, noradrenaline and dopamine reuptake inhibitors on light-induced phase advances in hamster circadian activity rhythms. Psychopharmacology. 2007 Dec;195(3):325-32. [216] Rea MA, Glass JD, Colwell CS. Serotonin modulates photic responses in the hamster suprachiasmatic nuclei. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1994 Jun;14(6):3635-42. [217] Weber ET, Gannon RL, Rea MA. Local administration of serotonin agonists blocks light-induced phase advances of the circadian activity rhythm in the hamster. Journal of biological rhythms. 1998 Jun;13(3):209-18. [218] Antle MC, Ogilvie MD, Pickard GE, Mistlberger RE. Response of the mouse circadian system to serotonin 1A/2/7 agonists in vivo: surprisingly little. Journal of biological rhythms. 2003 Apr;18(2):145- 58. [219] Mrosovsky N. Double-pulse experiments with nonphotic and photic phase-shifting stimuli. Journal of biological rhythms. 1991 Summer;6(2):167-79. [220] Biello SM, Mrosovsky N. Blocking the phase-shifting effect of neuropeptide Y with light. Proceedings Biological sciences / The Royal Society. 1995 Feb 22;259(1355):179-87. Interactions of the Serotonergic and Circadian Systems ... 57

[221] Penev PD, Turek FW, Wallen EP, Zee PC. Aging alters the serotonergic modulation of light-induced phase advances in golden hamsters. The American journal of physiology. 1997 Feb;272(2 Pt 2):R509-13. [222] Recio J, Pevet P, Masson-Pevet M. Serotonergic modulation of photically induced increase in melatonin receptor density and Fos immunoreactivity in the suprachiasmatic nuclei of the rat. Journal of neuroendocrinology. 1996 Nov;8(11):839-45. [223] Glass JD, Selim M, Rea MA. Modulation of light-induced C-Fos expression in the suprachiasmatic nuclei by 5-HT1A receptor agonists. Brain research. 1994 Feb 28;638(1-2):235-42. [224] Selim M, Glass JD, Hauser UE, Rea MA. Serotonergic inhibition of light-induced fos protein expression and extracellular glutamate in the suprachiasmatic nuclei. Brain research. 1993 Sep 10;621(2):181-8. [225] Brewer JM, Yannielli PC, Harrington ME. Neuropeptide Y differentially suppresses per1 and mRNA induced by light in the suprachiasmatic nuclei of the golden hamster. Journal of biological rhythms. 2002 Feb;17(1):28-39. eng. [226] Ehlen JC, Novak CM, Karom MC, Gamble KL, Albers HE. Interactions of GABA A receptor activation and light on period mRNA expression in the suprachiasmatic nucleus. Journal of biological rhythms. 2008 Feb;23(1):16-25. eng. [227] Yokota SI, Horikawa K, Akiyama M, Moriya T, Ebihara S, Komuro G, et al. Inhibitory action of brotizolam on circadian and light-induced per1 and per2 expression in the hamster suprachiasmatic nucleus. British journal of pharmacology. 2000 Dec;131(8):1739-47. eng. [228] Christian CA, Harrington ME. Three days of novel wheel access diminishes light-induced phase delays in vivo with no effect on per1 induction by light. Chronobiology international. 2002 Jul;19(4):671-82. eng. [229] Edelstein K, de la Iglesia HO, Schwartz WJ, Mrosovsky N. Behavioral arousal blocks light-induced phase advances in locomotor rhythmicity but not light-induced Per1 and Fos expression in the hamster suprachiasmatic nucleus. Neuroscience. 2003;118(1):253-61. eng. [230] Smith BN, Sollars PJ, Dudek FE, Pickard GE. Serotonergic modulation of retinal input to the mouse suprachiasmatic nucleus mediated by 5- HT1B and 5-HT7 receptors. Journal of biological rhythms. 2001 Feb;16(1):25-38. [231] Gannon RL, Peglion JL, Millan MJ. Differential influence of selective 5- HT5A vs 5-HT1A, 5-HT1B, or 5-HT2C receptor blockade upon light- 58 Marc Cuesta and Etienne Challet

induced phase shifts in circadian activity rhythms: interaction studies with citalopram. European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology. 2009 Dec;19(12):887-97. [232] Eriksson TM, Holst S, Stan TL, Hager T, Sjogren B, Ogren SO, et al. 5- HT1A and 5-HT7 receptor crosstalk in the regulation of emotional memory: implications for effects of selective serotonin reuptake inhibitors. Neuropharmacology. 2012 Nov;63(6):1150-60. [233] Pickard GE, Weber ET, Scott PA, Riberdy AF, Rea MA. 5HT1B receptor agonists inhibit light-induced phase shifts of behavioral circadian rhythms and expression of the immediate-early gene c-fos in the suprachiasmatic nucleus. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1996 Dec 15;16(24):8208-20. [234] Pickard GE, Rea MA. TFMPP, a 5HT1B receptor agonist, inhibits light- induced phase shifts of the circadian activity rhythm and c-Fos expression in the mouse suprachiasmatic nucleus. Neuroscience letters. 1997 Aug 8;231(2):95-8. [235] Byku M, Gannon RL. Effects of the 5HT1A agonist/antagonist BMY 7378 on light-induced phase advances in hamster circadian activity rhythms during aging. Journal of biological rhythms. 2000 Aug;15(4):300-5. [236] Matsuda T, Yoshikawa T, Suzuki M, Asano S, Somboonthum P, Takuma K, et al. Novel benzodioxan derivative, 5-(3-[((2S)-1,4- benzodioxan-2- ylmethyl)amino]propoxy)-1,3-benzodioxole HCl (MKC-242), with a highly potent and selective agonist activity at rat central serotonin1A receptors. Japanese journal of pharmacology. 1995 Dec;69(4):357-66. [237] Rea MA, Barrera J, Glass JD, Gannon RL. Serotonergic potentiation of photic phase shifts of the circadian activity rhythm. Neuroreport. 1995 Jul 10;6(10):1417-20. [238] Sterniczuk R, Stepkowski A, Jones M, Antle MC. Enhancement of photic shifts with the 5-HT1A mixed agonist/antagonist NAN-190: intra- suprachiasmatic nucleus pathway. Neuroscience. 2008 May 15;153(3):571-80. [239] Takahashi S, Yoshinobu Y, Aida R, Shimomura H, Akiyama M, Moriya T, et al. Extended action of MKC-242, a selective 5-HT(1A) receptor agonist, on light-induced Per gene expression in the suprachiasmatic nucleus in mice. Journal of neuroscience research. 2002 May 15;68(4):470-8. Interactions of the Serotonergic and Circadian Systems ... 59

[240] Smith VM, Hagel K, Antle MC. Serotonergic potentiation of photic phase shifts: examination of receptor contributions and early biochemical/molecular events. Neuroscience. 2010 Jan 13;165(1):16-27. [241] Novak CM, Ehlen JC, Huhman KL, Albers HE. GABA(B) receptor activation in the suprachiasmatic nucleus of diurnal and nocturnal rodents. Brain research bulletin. 2004 Jul 15;63(6):531-5. [242] Meyer-Bernstein EL, Blanchard JH, Morin LP. The serotonergic projection from the median raphe nucleus to the suprachiasmatic nucleus modulates activity phase onset, but not other circadian rhythm parameters. Brain research. 1997 Apr 25;755(1):112-20. [243] Smale L, Michels KM, Moore RY, Morin LP. Destruction of the hamster serotonergic system by 5,7-DHT: effects on circadian rhythm phase, entrainment and response to triazolam. Brain research. 1990 May 7;515(1-2):9-19. [244] Mistlberger RE, Bossert JM, Holmes MM, Marchant EG. Serotonin and feedback effects of behavioral activity on circadian rhythms in mice. Behavioural brain research. 1998 Nov;96(1-2):93-9. [245] Pang RD, Holschneider DP, Miller JD. Circadian rhythmicity in serotonin transporter knockout mice. Life sciences. 2012 Oct 5;91(11- 12):365-8. [246] Alexandre C, Popa D, Fabre V, Bouali S, Venault P, Lesch KP, et al. Early life blockade of 5-hydroxytryptamine 1A receptors normalizes sleep and depression-like behavior in adult knock-out mice lacking the serotonin transporter. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006 May 17;26(20):5554-64. [247] Wisor JP, Wurts SW, Hall FS, Lesch KP, Murphy DL, Uhl GR, et al. Altered rapid eye movement sleep timing in serotonin transporter knockout mice. Neuroreport. 2003 Feb 10;14(2):233-8. [248] Wollnik F. Effects of chronic administration and withdrawal of antidepressant agents on circadian activity rhythms in rats. Pharmacology, biochemistry, and behavior. 1992 Oct;43(2):549-61. [249] Possidente B, Lumia AR, McEldowney S, Rapp M. Fluoxetine shortens circadian period for wheel running activity in mice. Brain research bulletin. 1992 Apr;28(4):629-31. [250] Mullins UL, Gianutsos G, Eison AS. Effects of antidepressants on 5- HT7 receptor regulation in the rat hypothalamus. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 1999 Sep;21(3):352-67. 60 Marc Cuesta and Etienne Challet

[251] Duncan MJ, Hester JM, Hopper JA, Franklin KM. The effects of aging and chronic fluoxetine treatment on circadian rhythms and suprachiasmatic nucleus expression of neuropeptide genes and 5-HT1B receptors. The European journal of neuroscience. 2010 May;31(9): 1646-54. [252] Paulus EV, Mintz EM. Developmental disruption of the serotonin system alters circadian rhythms. Physiology and behavior. 2012 Jan 18;105(2):257-63. [253] Liu C, Maejima T, Wyler SC, Casadesus G, Herlitze S, Deneris ES. Pet- 1 is required across different stages of life to regulate serotonergic function. Nature neuroscience. 2010 Oct;13(10):1190-8. [254] Hendricks TJ, Fyodorov DV, Wegman LJ, Lelutiu NB, Pehek EA, Yamamoto B, et al. Pet-1 ETS gene plays a critical role in 5-HT neuron development and is required for normal anxiety-like and aggressive behavior. Neuron. 2003 Jan 23;37(2):233-47. [255] Sollars PJ, Ogilvie MD, Rea MA, Pickard GE. 5-HT1B receptor knockout mice exhibit an enhanced response to constant light. Journal of biological rhythms. 2002 Oct;17(5):428-37. [256] Ressler KJ, Nemeroff CB. Role of serotonergic and noradrenergic systems in the pathophysiology of depression and anxiety disorders. Depression and anxiety. 2000;12 Suppl 1:2-19. [257] Meltzer H. Serotonergic dysfunction in depression. The British journal of psychiatry Supplement. 1989 Dec(8):25-31. [258] Boivin DB. Influence of sleep-wake and circadian rhythm disturbances in psychiatric disorders. Journal of psychiatry and neuroscience : JPN. 2000 Nov;25(5):446-58. eng. [259] Lamont EW, Legault-Coutu D, Cermakian N, Boivin DB. The role of circadian clock genes in mental disorders. Dialogues in clinical neuroscience. 2007;9(3):333-42. eng. [260] Persaud R. Nocturnal sweating and temperature in depression'. Acta psychiatrica Scandinavica. 2000 Mar;101(3):251. eng. [261] Keller J, Flores B, Gomez RG, Solvason HB, Kenna H, Williams GH, et al. Cortisol circadian rhythm alterations in psychotic major depression. Biological psychiatry. 2006 Aug 1;60(3):275-81. eng. [262] Pacchierotti C, Iapichino S, Bossini L, Pieraccini F, Castrogiovanni P. Melatonin in psychiatric disorders: a review on the melatonin involvement in psychiatry. Frontiers in neuroendocrinology. 2001 Jan;22(1):18-32. eng. Interactions of the Serotonergic and Circadian Systems ... 61

[263] Soria V, Martinez-Amoros E, Escaramis G, Valero J, Perez-Egea R, Garcia C, et al. Differential association of circadian genes with mood disorders: CRY1 and NPAS2 are associated with unipolar major depression and CLOCK and VIP with bipolar disorder. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2010 May;35(6):1279-89. eng. [264] Utge SJ, Soronen P, Loukola A, Kronholm E, Ollila HM, Pirkola S, et al. Systematic analysis of circadian genes in a population-based sample reveals association of TIMELESS with depression and sleep disturbance. PloS one. 2010;5(2):e9259. eng. [265] Kishi T, Kitajima T, Tsunoka T, Okumura T, Ikeda M, Okochi T, et al. Possible association of prokineticin 2 receptor gene (PROKR2) with mood disorders in the Japanese population. Neuromolecular medicine. 2009;11(2):114-22. eng. [266] Gouin JP, Connors J, Kiecolt-Glaser JK, Glaser R, Malarkey WB, Atkinson C, et al. Altered expression of circadian rhythm genes among individuals with a history of depression. Journal of affective disorders. 2010 Oct;126(1-2):161-6. eng. [267] Pail G, Huf W, Pjrek E, Winkler D, Willeit M, Praschak-Rieder N, et al. Bright-light therapy in the treatment of mood disorders. Neuropsychobiology. 2011;64(3):152-62. eng. [268] Wirz-Justice A. Chronobiology and mood disorders. Dialogues in clinical neuroscience. 2003 Dec;5(4):315-25. eng. [269] Kripke DF. Light treatment for nonseasonal depression: speed, efficacy, and combined treatment. Journal of affective disorders. 1998 May;49(2):109-17. [270] Loving RT, Kripke DF, Shuchter SR. Bright light augments antidepressant effects of medication and wake therapy. Depression and anxiety. 2002;16(1):1-3. [271] Benedetti F, Colombo C, Serretti A, Lorenzi C, Pontiggia A, Barbini B, et al. Antidepressant effects of light therapy combined with sleep deprivation are influenced by a functional polymorphism within the promoter of the serotonin transporter gene. Biological psychiatry. 2003 Oct 1;54(7):687-92. [272] Gorwood P. Restoring circadian rhythms: a new way to successfully manage depression. Journal of psychopharmacology. 2010 Aug;24(2 Suppl):15-9. [273] Magnusson A, Boivin D. Seasonal affective disorder: an overview. Chronobiology international. 2003 Mar;20(2):189-207. eng. 62 Marc Cuesta and Etienne Challet

[274] Schwartz PJ, Rosenthal NE, Kajimura N, Han L, Turner EH, Bender C, et al. Ultradian oscillations in cranial thermoregulation and electroencephalographic slow-wave activity during sleep are abnormal in humans with annual winter depression. Brain research. 2000 Jun 2;866(1-2):152-67. eng. [275] Partonen T, Treutlein J, Alpman A, Frank J, Johansson C, Depner M, et al. Three circadian clock genes Per2, Arntl, and Npas2 contribute to winter depression. Annals of medicine. 2007;39(3):229-38. eng. [276] Johansson C, Willeit M, Smedh C, Ekholm J, Paunio T, Kieseppa T, et al. Circadian clock-related polymorphisms in seasonal affective disorder and their relevance to diurnal preference. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2003 Apr;28(4):734-9. eng. [277] Rosenthal NE, Mazzanti CM, Barnett RL, Hardin TA, Turner EH, Lam GK, et al. Role of serotonin transporter promoter repeat length polymorphism (5-HTTLPR) in seasonality and seasonal affective disorder. Molecular psychiatry. 1998 Mar;3(2):175-7. [278] Johansson C, Willeit M, Levitan R, Partonen T, Smedh C, Del Favero J, et al. The serotonin transporter promoter repeat length polymorphism, seasonal affective disorder and seasonality. Psychological medicine. 2003 Jul;33(5):785-92. [279] Lee HJ, Sung SM, Lim SW, Paik JW, Leen K. Seasonality associated with the serotonin 2A receptor -1438 A/G polymorphism. Journal of affective disorders. 2006 Oct;95(1-3):145-8. [280] Molnar E, Lazary J, Benko A, Gonda X, Pap D, Mekli K, et al. Seasonality and winter-type seasonal depression are associated with the rs731779 polymorphism of the serotonin-2A receptor gene. European neuropsychopharmacology: the journal of the European College of Neuropsychopharmacology. 2010 Sep;20(9):655-62. [281] Nexon L, Poirel VJ, Clesse D, Pevet P, Raison S. Complex regional influence of photoperiod on the nycthemeral functioning of the dorsal and median raphe serotoninergic system in the Syrian hamster. The European journal of neuroscience. 2009 Nov;30(9):1790-801. [282] Praschak-Rieder N, Willeit M, Wilson AA, Houle S, Meyer JH. Seasonal variation in human brain serotonin transporter binding. Archives of general psychiatry. 2008 Sep;65(9):1072-8. [283] Willeit M, Sitte HH, Thierry N, Michalek K, Praschak-Rieder N, Zill P, et al. Enhanced serotonin transporter function during depression in seasonal affective disorder. Neuropsychopharmacology : official Interactions of the Serotonergic and Circadian Systems ... 63

publication of the American College of Neuropsychopharmacology. 2008 Jun;33(7):1503-13. [284] Kalbitzer J, Erritzoe D, Holst KK, Nielsen FA, Marner L, Lehel S, et al. Seasonal changes in brain serotonin transporter binding in short serotonin transporter linked polymorphic region-allele carriers but not in long-allele homozygotes. Biological psychiatry. 2010 Jun 1;67(11):1033-9. [285] Ruhrmann S, Kasper S, Hawellek B, Martinez B, Hoflich G, Nickelsen T, et al. Effects of fluoxetine versus bright light in the treatment of seasonal affective disorder. Psychological medicine. 1998 Jul;28(4):923- 33. [286] Lam RW, Levitt AJ, Levitan RD, Enns MW, Morehouse R, Michalak EE, et al. The Can-SAD study: a randomized controlled trial of the effectiveness of light therapy and fluoxetine in patients with winter seasonal affective disorder. The American journal of psychiatry. 2006 May;163(5):805-12. [287] Harvey AG. Sleep and circadian rhythms in bipolar disorder: seeking synchrony, harmony, and regulation. The American journal of psychiatry. 2008 Jul;165(7):820-9. eng.