1-s2.0-s0306452215008076-main

Upload: astrimentari

Post on 07-Jul-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/19/2019 1-s2.0-S0306452215008076-main

    1/40

    IDENTIFICATION OF INTELLECTUAL DISABILITY GENES SHOWINGCIRCADIAN CLOCK-DEPENDENT EXPRESSION IN THEMOUSE HIPPOCAMPUS

    J. RENAUD, a,b,c F. DUMONT, a,c M. KHELFAOUI, e

    S. R. FOISSET, e F. LETOURNEUR, a,c T. BIENVENU, a,b,c

    O. KHWAJA, d O. DORSEUIL a,b,cy AND P. BILLUART a,b,c*†

    a Institut National de la Santé  et de la Recherche Mé dicale

    (INSERM), U1016, Institut Cochin, Paris, France

    b Université  Paris Descartes Paris V, Institut Cochin, Paris, France

    c Centre National de la Recherche Scientifique (CNRS),

    UMR8104, Institut Cochin, Paris, France

    d Roche Pharma Research and Early Development,

    Roche Innovation Center Basel, Basel, Switzerland e Centre National de la Recherche Scientifique (CNRS), UMR,

    INCI, Strasbourg, France

    Abstract—Sleep is strongly implicated in learning,

    especially in the reprocessing of recently acquired memory.

    Children with intellectual disability (ID) tend to have sleep-

    wake disturbances, which may contribute to the pathophys-

    iology of the disease. Given that sleep is partly controlled by

    the circadian clock, we decided to study the rhythmic

    expression of genes in the hippocampus, a brain structure

    which plays a key role in memory in humans and

    rodents. By investigating the hippocampal transcriptome

    of adult mice, we identified 663 circadian clock controlled

    (CCC) genes, which we divided into four categories basedon their temporal pattern of expression. In addition to the

    standard core clock genes, enrichment analysis identified

    several transcription factors among these hippocampal

    CCC genes, and our findings suggest that genes from one

    cluster regulate the expression of those in another. Interest-

    ingly, these hippocampal CCC genes were highly enriched

    in sleep/wakefulness-related genes. We show here that

    several genes in the glucocorticoid signaling pathway,

    which is involved in memory, show a CCC pattern of expres-

    sion. However, ID genes were not enriched among these

    CCC genes, suggesting that sleep or learning and memory

    disturbances observed in patients with ID are probably not

    related to the circadian clock in the hippocampus.

     2015 IBRO. Published by Elsevier Ltd. All rights reserved.

    Key words: circadian clock, hippocampus, sleep, memory,

    intellectual disability.

    INTRODUCTION

    Intellectual disability (ID) is defined by an overall

    intelligence quotient (IQ) lower than 70 and deficits in

    cognitive abilities with an onset before 18 years of 

    age. This disorder affects 1–3% of the population.

    The underlying causes of ID are heterogeneous andinclude genetic and/or environmental factors that

    influence the development and function of the central

    nervous system. Genetic factors comprise mutations in

    genes encoding proteins involved in neurogenesis,

    neuronal migration, synaptic function, transcription or 

    translation. Many children with ID have problems with

    memory and learning. Indeed, several memorization

    processes and associated structures may be impaired in

    ID, which limits the ability to learn broader cognitive

    skills. For example, patients with Down’s or Williams

    syndrome show working memory deficits (Jarrold et al.,

    1999). Numerous studies report that many children with

    ID show delayed settling or frequent night waking(Didde and Sigafoos, 2001; Robinson and Richdale,

    2004; Sajith and Clarke, 2007).

    Sleep is implicated in long-term memory, especially in

    the reprocessing of recently acquired memory (Gais and

    Born, 2004; Ji and Wilson, 2007). Indeed, sleep plays a

    fundamental role in the neurocognitive performance of 

    infants and children (Born and Wilhelm, 2012). More

    precisely, slow-wave sleep activity helps to consolidate

    hippocampal-dependent episodic memory. Sleep is con-

    trolled by two main regulatory processes: a homeostatic

    and a circadian one. The homeostatic process tracks

    the need for sleep whereas the distribution of sleep

    throughout the day is strongly regulated by circadian

    rhythm (CR). Circadian clock genes may also be directlyinvolved in sleep homeostasis (Naylor et al., 2000).

    Sleep/wake-related genes have been reported to be

    expressed in the cortex, cerebellum, forebrain and

    hypothalamus in mice and rats (Terao et al., 2003,

    2006; Cirelli et al., 2004).

    The circadian clock is a fundamental system

    regulating many aspects of behavior and physiology,

    including sleep–wake cycles but also blood pressure,

    body temperature, and metabolism. The

    suprachiasmatic nucleus (SCN) of the hypothalamus

    contains the master circadian clock, which is reset daily

    http://dx.doi.org/10.1016/j.neuroscience.2015.08.066

    0306-4522/ 2015 IBRO. Published by Elsevier Ltd. All rights reserved.

    *Correspondence to: P. Billuart, Institut National de la Sante ´  et de laRecherche Me ´ dicale (INSERM), U1016, Institut Cochin, Paris,France

    E-mail address: [email protected]   (P. Billuart).y These authors contributed equally to this work.

     Abbreviations: AA, ascorbic acid; ASD, Autism Spectrum Disorders;CCC, circadian clock controlled; CR, circadian rhythm; CT, CircadianTime; HSPs, heat shock proteins; ID, intellectual disability; IPA,Ingenuity Pathway Analysis; IQ, intelligence quotient; REM, rapid eyemovement; SCN, suprachiasmatic nucleus; SWG, sleep-wakefulness-related genes; SWS, slow wave sleep; TF, transcription factor; TFBSs,transcription factor binding sites.

    Neuroscience  308 (2015) 11–50

    11

    http://dx.doi.org/10.1016/j.neuroscience.2015.08.066mailto:[email protected]://dx.doi.org/10.1016/j.neuroscience.2015.08.066http://dx.doi.org/10.1016/j.neuroscience.2015.08.066http://dx.doi.org/10.1016/j.neuroscience.2015.08.066mailto:[email protected]://dx.doi.org/10.1016/j.neuroscience.2015.08.066http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    2/40

    by light that is detected by retinal ganglion cells and

    relayed via signaling through the retinohypothalamic

    tract. The circadian clock consists of a group of highly

    conserved core clock proteins (e.g. Rev-erba, Clock,

    Bmal1, Per1, Per2, Per3), the expression of which is

    regulated through transcriptional, translational and post-

    translational feedback loops. These regulatory mechanismsgenerate and maintain the cyclic expression of clock

    genes as well as those they regulate (called output

    genes) over a 24-h period (Bell-Pedersen et al., 2005;

    Ko and Takahashi, 2006; Dardente and Cermakian,

    2007). The SCN coordinates peripheral oscillators like

    the liver, kidney and cortex, which can be self-

    sustained, although their correct functioning depends on

    the other oscillators. Clock mechanisms in the SCN and

    peripheral oscillators are identical, and involve the 24-h

    cyclic expression of core clock genes (Reppert and

    Weaver, 2002; Yoo et al., 2004).

    CR has been studied in many non-neuronal tissues,

    especially in the liver because it regulates metabolism.Several studies report that about 10% of transcripts

    show circadian clock controlled (CCC) expression in the

    liver, most of which are involved in metabolic functions

    (Panda et al., 2002; Storch et al., 2002; Cho et al.,

    2012). Microarrays and RNA-seq have been used to

    examine the transcriptome at different Circadian Time

    (CT) points in several regions of the mouse brain, includ-

    ing the SCN (Panda et al., 2002), prefrontal cortex (Yang

    et al., 2007), cerebellum, hypothalamus and brain stem

    (Zhang et al., 2014). The core clock genes are expressed

    in various brain structures, such as the hippocampus and

    cortex (Namihira et al., 1999; Reick et al., 2001;

    Wakamatsu et al., 2001; Shieh, 2003; Yang et al., 2007;

    Jilg et al., 2010) and studies on mutant mice suggest thatthe disruption of clock genes alters hippocampal-

    dependent memory formation (Garcia, 2000; Wang

    et al., 2009; Jilg et al., 2010). The hippocampus controls

    memory and learning, processes spatial information and

    episodic memory, and consolidates information from

    short-term to long-term memory. These processes are

    regulated by CR (Chaudhury and Colwell, 2002;

    Chaudhury et al., 2005; Ruby et al., 2008; Gerstner 

    et al., 2009). For instance, circadian oscillations in hip-

    pocampal MAPK activity are implicated in the formation

    and persistence of memory (Eckel-Mahan et al., 2008;

    Phan et al., 2011). In addition, adult neurogenesis in the

    dentate gyrus is also controlled by the circadian molecular 

    clock (Bouchard-Cannon et al., 2013; Schnell et al.,

    2014).

    Although the link between cognitive impairment,

    memory and sleep as well as that between CR and

    sleep are well established, little is known about the

    relationship between the circadian clock and ID. The

    enrichment of CCC genes among the known ID genes

    would favor the hypothesis that the circadian clock-

    mediated deregulation of memory processes is involved

    in the pathogenesis of ID.

    In this context, we analyzed genome-wide rhythmic

    gene expression in the hippocampus, focusing on

    circadian regulation. Using the JTK cycle algorithm, we

    demonstrate here that about 3.5% of transcripts show

    circadian-regulated expression in the mouse hippo-

    campus. We divided these genes into four categories

    based on their temporal expression, and show that each

    category is characterized by specific molecular and

    cellular signatures. The category containing genes

    that were strongly expressed at the transition phase

    between light and dark was highly enriched in sleep/wake-related genes.

    Furthermore, from a list of 673 ID-associated genes,

    we identified 30 genes (4.4%) showing circadian-

    dependent expression in the hippocampus. This

    proportion was not significantly higher than that expected

    by chance (3.5%), showing that the pathogenesis of ID

    is probably not related to disturbances in the circadian

    clock in the hippocampus.

    EXPERIMENTAL PROCEDURES

    Animals

    Animal experiments were performed in accordance withthe European Communities Council Directive (86/809/

    EEC) on the care and use of animals and were

    approved by the local ethics committee. All animals

    were housed singly in cages in a room maintained at

    2 3 ± 1 C under a 12/12 light–dark cycle (bright light,

    200 lx). Nine-week-old male C57/BL6 mice were killed in

    the dark and hippocampi were quickly excised under a

    red-light lamp and snap-frozen in liquid nitrogen. The

    dark/dark period started at 7 p.m. Four CT points were

    sampled. The CT18 samples were taken after 30 h of 

    continuous darkness, after which the other CT samples

    were taken at 6-h intervals: CT0, CT6 and CT12 after 

    36, 42 and 48 h of continuous darkness, respectively.

    Microarray analysis

    The SurePrint G3 Mouse GE 8 60 K microarray (Agilent

    Technologies, Santa Clara, CA, USA) was used for 

    expression profiling experiments.

    Total RNA was extracted using RNA-PLUS (MP

    Biomedicals, Santa Ana, CA, USA), treated with rDNase

    (Macherey-Nagel EURL, Hoerdt, France) and cleaned

    with an RNA clean-up kit (Macherey-Nagel EURL,

    Hoerdt, France). RNA transcripts were quantified using

    a Nanodrop spectrophotometer (Thermo Fischer 

    Scientific, Waltham, MA, USA) and quality was

    monitored with the Agilent 2100 Bioanalyzer (AgilentTechnologies, Santa Clara, CA, USA). Each biological

    replicate, i.e. total RNA from the hippocampus of one

    mouse, was hybridized to one chip.

    Real-time RT-PCR

    Microarray results were validated by performing RT-PCR

    on the same samples used for hybridization. DNase

    I–treated RNA was reverse transcribed using the

    QuantiTect reverse transcription kit (Qiagen, Hilden,

    Germany) according to the manufacturer’s instructions.

    The expression of two clock genes was evaluated

    (Rev-erba   and   Arntl ). Reactions were performed in

    triplicate in an LC480 LightCycler (Roche Applied

    12 J. Renaud et al. /Neuroscience 308 (2015) 11–50

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    3/40

    Science, Indianapolis, IN, USA). The   Gapdh   and

    Cyclophilin A   housekeeping genes were used as internal

    controls. Average normalized delta-Ct values were used

    to calculate the fold difference in expression between

    samples. The following primers were used: mouse

    Rev-erba   (ID #Mm00520708_m1), mouse Arntl (ID

    #Mm00500226_m1) and Gapdh (ID #Mm99999915_g1)from Applied Biosystems, and cyclophilin A forward:

    50-GTG ACT TTA CAC GCC ATA ATG-30 and reverse:

    50-ACA AGA TGC CAG GAC CTG TAT-30, gene ID for 

    cyclophilin A (or Ppia) (uc007hyn.1). A one-way ANOVA

    was performed to assess the significance of expression

    differences between samples.

    Western blotting

    Total protein extracted from the hippocampus was

    separated by SDS–PAGE, followed by Western blotting

    with a rabbit antibody against Clock (1:1000, Merck

    Millipore, Fontenay sous Bois, France, AB2203) and a

    mouse antibody against Gapdh (1:4000, AmbionThermo Fischer Scientific, Waltham, MA, USA,

    AM4300). Data were expressed as the mean ± SEM.

    The significance of differences in protein levels between

    samples was tested by an unpaired Student t  test.

    PCA and JTK analysis

    Raw data from the Agilent extraction file were first

    normalized using g-processed signal values and

    quantile normalization. Expression data were then

    summarized to obtain the gene-level expression value.

    First, variations in gene expression were analyzed

    using Principal Component Analysis to eliminate

    technical bias and outliers. The JTK cycle algorithm(Hughes et al., 2010) was then applied to identify rhythmic

    genes. An adjusted p-value < 0.05 was considered to be

    significant (JTK-positive). These two analyses were

    performed using R statistical software.

    Identification and clustering of genes

    To identify clusters of genes among JTK-positive genes,

    we applied the  k -means clustering algorithm (Pearson

    dissimilarity and centroid linkage) as implemented in the

    open source clustering software Cluster 3.0 (Eisen

    et al., 1998). To view the clustering results generated by

    Cluster 3.0, we used Alok Saldanha’s Java Treeview,

    which displays hierarchical as well as k -means clusteringresults.

    Functional analysis of genes controlled by the

    circadian clock

    The 663 JTK-positive genes and the genes of each

    cluster were uploaded to Ingenuity Pathway Analysis

    (IPA, Ingenuity Systems Inc., Redwood City, CA, USA)

    to analyze molecular and cellular functions and

    canonical pathways associated with the dataset.

    Analysis of transcription factor (TF) binding sites

    Promoter sequences were analyzed using the‘‘Overrepresented transcription factor binding sites” and

    Matinspector tools in Genomatix software (GmbH,

    Munich, Germany). The software calculates  z -scores

    that correspond to the distance from the population

    mean expressed in units of the standard deviation of the

    population. The z -scores are calculated with a continuity

    correction using the formula  z  = (x E 0.5)/S , where   x 

    is the number of found matches in the input data,  E   isthe expected value and S   is the standard deviation.

    To determine the degree of inter-regulation among the

    genes in each cluster, for each TF, we calculated the

    mean of the   z -scores of all the clusters. For each

    cluster and each TF, the z -score was normalized by the

    mean value of the z -scores. The sum of the normalized

    z -scores was then averaged.

    RESULTS

    CCC genes in the hippocampus

    To identify CCC genes in the mouse hippocampus, we

    conducted our study in dark–dark conditions to

    overcome synchronizing effect of the SCN in response

    to light. We previously reported that the expression of 

    some clock genes (Rev-erba   and Arntl ) oscillates in the

    hippocampus in such conditions (Valnegri et al., 2011).

    To identify systematically genes with a CCC pattern of 

    expression, we used Agilent microarray technology to

    examine the transcriptome of the hippocampus of three

    mice at four CT points. We used the JTK cycle algorithm

    (Hughes et al., 2010) to identify and characterize genes

    with rhythmic expression. This algorithm accurately mea-

    sures the period, phase, acrophase and amplitude of 

    cycling transcripts. Among the 948 JTK-positive genes

    (adjusted p-value < 0.05), 663 showed cyclic expressionof a 24-h period and could thus be defined as CCC genes

    (Table 1).

    To assess the circadian rhythmicity of the

    hippocampus, we first examined oscillations in the

    expression of core clock genes (Fig. 1 and Table 1). We

    found that   Clock    (1a) and   Rev-erba   mRNAs (1b)

    oscillate with a period of 24 h. Furthermore, we

    previously reported that levels of Rev-erba   protein show

    a rhythmic pattern (Valnegri et al., 2011). We thus

    assessed the abundance of Clock protein in the hip-

    pocampus over 24 h in dark/dark conditions (1c). Protein

    levels peaked at CT0 (the ratio of Clock to Gapdh was

    1.539 ± 0.084 at CT0 vs. 0.4174 ± 0.007 at CT18,

    0.5473 ± 0.082 at CT12 and 0.4745 ± 0.06515 at CT6, p-values = 0.0056, 0.004 and 0.002, respectively).

    These results suggest that the expression of clock genes

    in the hippocampus shows circadian oscillations at both

    the mRNA and protein level. The analysis of microarray

    data revealed that   Arntl , also known as  Bmal1   (1d),

    Per1   (1e), and Per2  (1f) also showed CCC expression.

    Real-time PCR analysis of two major clock genes, Rev-

    erba   and   Arntl , the expression of which are temporally

    opposed, confirmed the findings of our microarray exper-

    iments (Fig. 1c, d, g , h). Thus, these results show that the

    abundance of clock genes mRNA oscillates in a typical

    cyclic manner in the hippocampus.

    We next performed an Ingenuity Pathway Analysis(IPA) on our dataset of 663 genes to identify the

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   13

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    4/40

    Table 1.   JTK analysis of the 948 genes with an adjusted   p-value < 0.05 (JTK-positive). The 663 JTK-positive genes with a period of 24 h are

    highlighted in red

    14 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    5/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   15

  • 8/19/2019 1-s2.0-S0306452215008076-main

    6/40

    Table 1  (continued )

    16 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    7/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   17

  • 8/19/2019 1-s2.0-S0306452215008076-main

    8/40

    Table 1  (continued )

    18 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    9/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   19

  • 8/19/2019 1-s2.0-S0306452215008076-main

    10/40

    Table 1  (continued )

    20 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    11/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   21

  • 8/19/2019 1-s2.0-S0306452215008076-main

    12/40

    Table 1  (continued )

    22 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    13/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   23

  • 8/19/2019 1-s2.0-S0306452215008076-main

    14/40

  • 8/19/2019 1-s2.0-S0306452215008076-main

    15/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   25

  • 8/19/2019 1-s2.0-S0306452215008076-main

    16/40

    Table 1  (continued )

    26 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    17/40

    Table 1  (continued )

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   27

  • 8/19/2019 1-s2.0-S0306452215008076-main

    18/40

    Table 1  (continued )

    28 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    19/40

    pathways and molecular/cellular functions associatedwith these CCC genes. The core-clock genes (Per1,Per2 ,   Arntl ,   Rev-erba   and   Clock ) were significantlyenriched among the top canonical pathways ( p-value =2.34 103)   (Fig. 2). We also identified other criticalcomponents of the clock, Cry1 and Cry2, in a list of 

    upstream regulators ( p-value of overlap = 8.26 105and 8.26 105, respectively). Cry proteins interact with

    Per proteins and associate with Clock and Arntl torepress transcription.

    Co-regulation of CCC genes

    We next defined clusters of genes based on their 

    expression levels at the four CT points analyzed to

    identify co-regulated CCC genes using cluster and java

    tree view software. The 663 JTK-positive genes were

    grouped into four clusters showing a distinct temporal

    expression pattern and containing between 48 and 308

    genes (Fig. 3a). Clock genes mapped to different

    clusters depending on their peak of expression and weassumed that genes in the same cluster were

    co-regulated during circadian activity (Fig. 3b–e).

    Given that sleep is a circadian process and implicated

    in learning and memory, we wondered whether our 663

    CCC genes were enriched in sleep-wakefulness-related

    genes (SWG). Approximately 5% of the transcripts

    expressed in the rat cortex and cerebellum are

    regulated by sleep/wakefulness (Cirelli et al., 2004). By

    comparing our list of 663 hippocampal CCC genes with

    the mouse orthologs of the genes reported by Cirelli

    et al., we found 52 genes potentially regulated by SWG

    (i.e. 7.85% of our entire list) (Cirelli et al., 2004). A

    Chi-square test with Yates continuity correction, which is

    used for large sample sizes, showed that CCC geneswere significantly enriched in SWG ( p < 0.0001). We also

    examined for the presence of SWG in each cluster.

    Clusters 1 and 4 were significantly enriched in SWG

    (11% and 8.3%, respectively) whereas cluster 2 and

    cluster 3 did not contain a significant proportion of these

    genes (1.06% and 4.25%, respectively). Genes in cluster 

    4 showed a peak of expression at CT12, which is relevant

    because CT12 is a transition phase between light and

    dark. Per2  was classified as a cluster 1 gene, the expres-

    sion of which peaked at CT18. Individuals presenting a

    mutation in the phosphorylation site of PER2 suffer from

    familial advanced sleep phase syndrome (FAPS), which

    is characterized by early sleep onset and offset (Toh,2001). Our findings suggest that sleep/wakefulness and

    circadian activity are two inter-connected processes in

    the hippocampus.

    Genes involved in post-translational modifications and

    protein ubiquitination pathways were highly enriched incluster 1 ( p-value = 3.17 107 and 3.91 104

    respectively). These two pathways are two well-known

    circadian processes (Mehra et al., 2009). Indeed, post-translational modifications such as phosphorylation,acetylation and ubiquitination of clock proteins contributeto the entrainment of the clock. Ubiquitination, in particu-lar, regulates the half-life of proteins and is clearly essen-

    tial to control daily fluctuations in protein abundance. Inour dataset, heat shock proteins (HSPs) were over-represented among the proteins involved in post-translational modifications. Many HSPs show circadianregulation and variations in their expression levels during

    sleep and have been suggested to control the clock inperipheral organs (Terao et al., 2003; Reinke et al.,2008; Yan et al., 2008). Furthermore, HSPs interact with

    the glucocorticoid receptor in a manner dependent on

    the circadian oscillations (Furay et al., 2006). Interest-ingly, glucocorticoid signaling was also in the topcanonical pathway ( p-value = 1.07 103) and wasover-represented in cluster 2 ( p-value = 2.11 104).

    Glucocorticoids are strongly implicated in the hippocampal-dependent consolidation of memory (de Quervain et al.,2009), and Arntl-deficient mice present deficits in gluco-corticoid production and a poor behavioral response tostress (Leliavski et al., 2014). Moreover, circadian oscilla-tion of glucocorticoid levels promotes the formation of dendritic spines in the cortex (Liston et al., 2013). Another CCC gene identified in this analysis was the glucocorti-

    coid receptor, NR3C1 (nuclear receptor subfamily 3,group C, member 1) identified by IPA as an upstream reg-

    ulator of transcription ( p-value of overlap = 1.4 103).This receptor functions both as a transcriptional activator that binds to glucocorticoid response elements in thepromoters of glucocorticoid responsive genes and as a

    regulator of other TFs. This result suggests that thecircadian clock influences the hippocampus and memorythrough the regulation of glucocorticoid signaling.

    In addition, the antioxidant action of vitamin C was an

    enriched pathway ( p-value = 3.68 104) that was over-

    represented in cluster 3 ( p-value = 1.09 102).

    Ascorbic acid (AA), also known as vitamin C,

    accumulates in the brain, especially in the hippocampus

    and its antioxidant function is neuroprotective

    (MacGregor et al., 1996). Furthermore, the amount of AA oscillates with CR in the SCN (Wang et al., 2012).

    Table 1  (continued )

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   29

    http://-/?-http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    20/40

  • 8/19/2019 1-s2.0-S0306452215008076-main

    21/40

    Regarding metabolism, cluster 1, which contains genes

    highly expressed during an active phase (dark phase),

    was enriched in genes involved in lipid metabolism. Lipids

    are important sources of energy and are provided in large

    quantities in the diet, with mice feeding preferentially dur-

    ing the arousal phase (Pan and Hussain, 2009). Overall,

    metabolism is a circadian-regulated process and is clo-

    sely related to sleep–wake and feeding–fasting cycles.

    Moreover, metabolic processes influence the functionand timing of the clock (Peek et al., 2012). Indeed, cellular 

    redox state influences DNA binding of the Clock-Arntl het-

    erodimer (Rutter et al., 2001). Therefore, it is not surpris-

    ing to find metabolism pathways highly represented in our 

    list of hippocampal CCC genes.

    Transcriptional regulation of CCC genes

    Gene expression is regulated by the binding of TFs to

    promoter regions, which is temporally and spatially

    controlled. We thus wondered whether the coordinated

    expression of genes in each cluster may be explained

    by the presence of specific transcription factor bindingsites (TFBSs) in the promoters of the hippocampal CR

    genes. We screened the genes in each cluster for 

    enrichment in TFBSs using the search tool ‘‘Over-

    represented TFBSs” in Genomatix software. The

    software analyzes the promoter regions of genes

    (500 bp upstream and 100 bp downstream from the

    transcription start site) and assigns a statistical

    enrichment value, named a   z -score, to each TFBS.

    First, we noticed that the matrix family v$HIFF (E box-

    element target sequence: CACGTG) was highlyrepresented (z -score = 19.3) in cluster 3 (peak of 

    expression at CT6) (data not shown). This matrix family

    is strongly associated with CR and with the expression

    of genes regulated by Clock and Arntl. Moreover, this

    analysis showed substantial overlap in the TFBSs

    identified in each cluster (Fig. 4a). We categorized

    TFBSs into four groups based on their enrichment in

    each cluster, with group 1, 2, 3 and 4 containing

    TFBSs enriched in one, two, three or four clusters,

    respectively. Group 4 included 74.3% of the TFBSs

    whereas groups 1, 2 and 3 contained only 12.4%,

    3.8% and 9.5% of the TFBSs, respectively (Fig. 4b).

    We conclude that the transcriptional regulation of hippocampal CCC genes appears to be similar among

    Fig. 2.  Network of circadian core clock genes. Network of the core clock genes identified as circadian oscillating genes in our microarray analysis

    (highlighted in pink). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   31

  • 8/19/2019 1-s2.0-S0306452215008076-main

    22/40

    Fig. 3.  Clustering of the 663 JTK-positive genes according to their temporal expression. (a) Heat map showing the expression profiles of the 663

    JTK-positive genes. Four clusters are identified. (b–e) Plots showing the distinct temporal expression patterns of JTK-positive genes in the four 

    clusters. Clock genes associated with each cluster are shown in red. Cluster 1 (b) contains genes showing an expression peak at CT18, including

    Per2 and Clock. Cluster 2 (c) contains genes showing an expression peak at CT0, including Arnt1. Cluster 3 (d) gene contains genes showing an

    expression peak at CT6, including Rev-erba. Cluster 4 (e) contains genes showing an expression peak at CT12, including Cry1 and Per1. In panel

    (b,c,d,e), expression peak is shown by a blue star. (For interpretation of the references to color in this figure legend, the reader is referred to the

    web version of this article.)

    Fig. 4.   Identification of TFBSs in the 663 CCC genes. (a) Table showing the number of TFBSs shared between clusters. (b) Clustering of TFBSs

    based on their enrichment in the various clusters. Four groups can be identified (G1–G4). G4 contains TFBSs present in all the clusters whereas G1

    contains TFBSs present specifically in one cluster.

    32 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    23/40

    clusters. Therefore, circadian oscillations in the

    expression of these genes may be generated by a

    restricted number of specific TFs.

    Given that the clock is regulated transcriptionally and

    that clock genes are TFs, we determined the proportion

    of TFs among the CCC genes in the hippocampus.

    Among our four clusters, cluster 2 was the most

    enriched in TFs (24.21%) whereas cluster 3 included

    very few TFs (7.55%). We then identified the TFs in

    each cluster that may regulate the expression of genes

    in the other clusters. We quantified the potential inter-

    regulation of gene expression between clusters bycalculating a normalized   z -score value (Fig. 5). We

    found that TFs from cluster 1 may strongly regulate the

    expression of genes from clusters 3 and 4. Cluster 2

    contained TFs that may control the temporal expression

    of genes from clusters 1, 3 and 4. This analysis thus

    highlights transcriptional regulation between our clusters

    that may underlie their different temporal patterns of 

    expression, thereby illustrating the typical functioning of 

    a clock.

    Identification of CCC ID genes in the hippocampus

    Next, we wanted to explore the link between ID and the

    circadian clock. From the literature and from publicdatabases, we identified a list of 673 genes involved in

    ID and Autism Spectrum Disorders (ASD) (Table 2), 30

    (4.45%) of which showed CCC expression in the

    hippocampus according to our analysis (Table 3). This

    proportion is not significantly different from that

    expected by chance alone ( p-value = 0.1), suggesting

    that clock output genes in the hippocampus are not

    associated with ID. However,  Fgfr3   and   Aff2 , both of 

    which are included in this list of 30 genes, have already

    been implicated in sleep-related respiratory disturbances

    and sleep enhanced multifocal spiking, respectively

    (Musumeci et al., 2000; Schlu ¨ ter et al., 2011).

    Furthermore,  Per1   was also included in this list of 30CCC regulated ID/ASD genes (Nicholas et al., 2007).

    Polymorphisms in Per1, which encodes a critical compo-

    nent of the circadian clock, are associated with extreme

    diurnal preference (Carpen et al., 2006).

    IPA also showed that lipid metabolism was an over-

    represented biological function ( p-value = 5.05 105

    to 3.89 102). ID and lipid metabolism disorders have

    already been described as two potentially related

    diseases (Thiel et al., 2003; Voronov et al., 2008;

    Matsumata et al., 2014). Lipid metabolism is known to

    be a clock-controlled process (Sancar and Brunner,

    2014).

    Finally, we divided the 30 CCC ID genes in eachcluster according to their temporal pattern of expression

    (Fig. 6). These results are of significant interest for 

    clinicians. Indeed, children with ID may have different

    clinical profiles of sleep/wake defects depending on the

    expression pattern of the ID gene involved.

    DISCUSSION

    Circadian clocks drive the CR of biological processes in

    various tissues. This system is organized into a master 

    circadian clock, the SCN that synchronizes the

    peripheral oscillators. Instead of a single cerebral clock,

    the brain contains a system of multiple local oscillators

    exhibiting CR in different structures (Guilding andPiggins, 2007). Indeed, the expression of  Per1  and  Per2 

    clock genes only partially overlaps in various brain

    regions and their promoters are activated in a cell type-

    specific manner (neurons for   Per1   vs. glial cells for 

    Per2 ) (Feillet et al., 2008; Cheng et al., 2009).

    The hippocampus is entrained by the SCN suggesting

    that these regions are connected physically or in an

    indirect manner. Pathways connecting the two

    structures have been identified in rats (Moga et al.,

    1995). In addition, the release of melatonin from the

    pineal gland (Perreau-Lenz et al., 2003) or hypocretin-

    expressing neurons of the lateral hypothalamus (Yang

    et al., 2013) may constitute an indirect link between themaster clock and the hippocampus.

    Fig. 5.   Quantification of potential inter-regulation of gene expression between clusters. (a) A normalized mean z -score value was assigned to each

    cluster as an enrichment factor. (b) Model of inter-regulation between clusters.

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   33

    http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    24/40

    Table 2. List of the 673 ID-ASD genes. ID genes have been compiled from literature and from public databases ( http://www.genes2cognition.org; http://

    www.g2conline.org; http://www.ggc.org/research/molecular-studies/xlid.html; http://www.gencodys.eu; https://gene.sfari.org/autdb/Welcome.do)

    Gene symbol Gene name

    A2bp1 ataxin 2-binding protein 1

    Aass aminoadipate-semialdehyde synthase

    Abat 4-aminobutyrate aminotransferaseAbcc8 ATP-binding cassette, sub-family C (CFTR/MRP), member 8

    Abcd1 ATP-binding cassette, sub-family D (ALD), member 1

    Abhd5 abhydrolase domain containing 5

    Acbd6 acyl-Coenzyme A binding domain containing 6

    Acox1 acyl-Coenzyme A oxidase 1, palmitoyl

    Acsl4 acyl-CoA synthetase long-chain family member 4

    Acy1 aminoacylase 1

    Ada adenosine deaminase

    Adk adenosine kinase

    Adora2a adenosine A2a receptor 

    Adra2b adrenergic receptor, alpha 2b

    Adrb2 adrenergic receptor, beta 2

    Adsl adenylosuccinate lyase

    Aff2 AF4/FMR2 family, member 2

    Aga aspartylglucosaminidaseAgtr2 angiotensin II receptor, type 2

    Ahi1 Abelson helper integration site 1

    Ak1 adenylate kinase 1

    Aldh18a1 aldehyde dehydrogenase 18 family, member A1

    Aldh3a2 aldehyde dehydrogenase family 3, subfamily A2

    Aldh4a1 aldehyde dehydrogenase 4 family, member A1

    Aldh5a1 aldehyde dehydrogenase family 5, subfamily A1

    Aldoa aldolase A, fructose-bisphosphate

    Alg12 asparagine-linked glycosylation 12 (alpha-1,6-mannosyltransferase)

    Alg2 asparagine-linked glycosylation 2 (alpha-1,3-mannosyltransferase)

    Alg3 asparagine-linked glycosylation 3 (alpha-1,3-mannosyltransferase)

    Alg6 asparagine-linked glycosylation 6 (alpha-1,3,-glucosyltransferase)

    Alox5ap arachidonate 5-lipoxygenase activating protein

    Alx1 ALX homeobox 1

    Alx4 aristaless-like homeobox 4Amt aminomethyltransferase

    Ankrd11 ankyrin repeat domain 11

    Ap1s2 adaptor-related protein complex 1, sigma 2 subunit

    Ap4e1 adaptor-related protein complex AP-4, epsilon 1

    Ap4m1 adaptor-related protein complex AP-4, mu 1

    Apc adenomatosis polyposis coli

    Ar androgen receptor  

    Arg1 arginase, liver  

    Arhgef6 Rac/Cdc42 guanine nucleotide exchange factor (GEF) 6

    Arhgef9 CDC42 guanine nucleotide exchange factor (GEF) 9

    Arnt2 aryl hydrocarbon receptor nuclear translocator 2

    Arsa arylsulfatase A

    Arx aristaless-related homeobox

    Asah1 N-acylsphingosine amidohydrolase 1

    Ascc3 activating signal cointegrator 1 complex subunit 3Ascl1 achaete-scute complex homolog 1 (Drosophila)

    Asl argininosuccinate lyase

    Asmt acetylserotonin O-methyltransferase

    Aspa aspartoacylase

    Aspm asp (abnormal spindle)-like, microcephaly associated (Drosophila)

    Ass1 argininosuccinate synthetase 1

    Astn2 astrotactin 2

    Atic 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase

    Atn1 atrophin 1

    Atp10a ATPase, class V, type 10A

    Atp2a2 ATPase, Ca++ transporting, cardiac muscle, slow twitch 2

    Atp5e ATP synthase, H+ transporting, mitochondrial F1 complex, epsilon subunit

    Atp6ap2 ATPase, H+ transporting, lysosomal accessory protein 2

    Atp7a ATPase, Cu++ transporting, alpha polypeptide

    34 J. Renaud et al. /Neuroscience 308 (2015) 11–50

    http://www.genes2cognition.org/http://www.g2conline.org/http://www.g2conline.org/http://www.ggc.org/research/molecular-studies/xlid.htmlhttp://www.gencodys.eu/https://gene.sfari.org/autdb/Welcome.dohttps://gene.sfari.org/autdb/Welcome.dohttp://www.gencodys.eu/http://www.ggc.org/research/molecular-studies/xlid.htmlhttp://www.g2conline.org/http://www.g2conline.org/http://www.genes2cognition.org/

  • 8/19/2019 1-s2.0-S0306452215008076-main

    25/40

  • 8/19/2019 1-s2.0-S0306452215008076-main

    26/40

  • 8/19/2019 1-s2.0-S0306452215008076-main

    27/40

    Table 2  (continued )

    Gene symbol Gene name

    Ehmt1 euchromatic histone methyltransferase 1

    Ehmt2 euchromatic histone lysine N-methyltransferase 2

    Eif2ak3 eukaryotic translation initiation factor 2 alpha kinase 3

    Eif2s3x eukaryotic translation initiation factor 2, subunit 3, structural gene X-linkedEif4e eukaryotic translation initiation factor 4E

    Elp2 elongator acetyltransferase complex subunit 2

    Emx2 empty spiracles homeobox 2

    En2 engrailed 2

    Entpd1 ectonucleoside triphosphate diphosphohydrolase 1

    Ercc2 excision repair cross-complementing rodent repair deficiency, complementation group 2

    Ercc3 excision repair cross-complementing rodent repair deficiency, complementation group 3

    Ercc5 excision repair cross-complementing rodent repair deficiency, complementation group 5

    Ercc6 excision repair cross-complementing rodent repair deficiency, complementation group 6

    Ercc8 excision repaiross-complementing rodent repair deficiency, complementation group 8

    Erlin2 ER lipid raft-associated 2

    Esr1 estrogen receptor 1 (alpha)

    Esr2 estrogen receptor 2 (beta)

    Esrrb estrogen-related receptor, beta

    F13a1 coagulation factor XIII, A1 subunitFabp5 fatty acid binding protein 5

    Fabp7 fatty acid binding protein 7, brain

    Fam126a family with sequence similarity 126, member A

    Fanca Fanconi anemia, complementation group A

    Fancb Fanconi anemia, complementation group B

    Fancc Fanconi anemia, complementation group C

    Fasn fatty acid synthase

    Fbn1 fibrillin 1

    Fbxo33 F-box protein 33

    Fbxo40 F-box protein 40

    Fezf2 Fez family zinc finger 2

    Fgd1 FYVE, RhoGEF and PH domain containing 1

    Fgf14 fibroblast growth factor 14

    Fgfr1 fibroblast growth factor receptor 1

    Fgfr2 fibroblast growth factor receptor 2Fgfr3 fibroblast growth factor receptor 3

    Fh1 fumarate hydratase 1

    Fhit fragile histidine triad gene

    Fkrp fukutin-related protein

    Fktn fukutin

    Flna filamin, alpha

    Flt1 FMS-like tyrosine kinase 1

    Fmr1 fragile X mental retardation syndrome 1

    Folr1 folate receptor 1 (adult)

    Foxe1 forkhead box E1

    Foxg1 forkhead box G1

    Foxp1 forkhead box P1

    Foxp2 forkhead box P2

    Fras1 Fraser syndrome 1 homolog (human)

    Frk fyn-related kinaseFrmpd4 FERM and PDZ domain containing 4

    Fry furry homolog (Drosophila)

    Ftsj1 FtsJ homolog 1 (E. coli)

    Fuca1 fucosidase, alpha-L- 1, tissue

    G6pdx glucose-6-phosphate dehydrogenase X-linked

    Gabra4 gamma-aminobutyric acid (GABA) A receptor, subunit alpha 4

    Gabrb1 gamma-aminobutyric acid (GABA) A receptor, subunit beta 1

    Gabrb3 gamma-aminobutyric acid (GABA) A receptor, subunit beta 3

    Gale UDP-galactose-4-epimerase

    Galnt13 UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 13

    Galt galactose-1-phosphate uridyl transferase

    Gamt guanidinoacetate methyltransferase

    Gatm glycine amidinotransferase (L-arginine:glycine amidinotransferase)

    Gcdh glutaryl-Coenzyme A dehydrogenase

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   37

  • 8/19/2019 1-s2.0-S0306452215008076-main

    28/40

    Table 2  (continued )

    Gene symbol Gene name

    Gch1 GTP cyclohydrolase 1

    Gck glucokinase

    Gcsh glycine cleavage system protein H (aminomethyl carrier)

    Gdi1 guanosine diphosphate (GDP) dissociation inhibitor 1Gfap glial fibril lary acidic protein

    Glb1 galactosidase, beta 1

    Gldc glycine decarboxylase

    Gli3 GLI-Kruppel family member GLI3

    Glo1 glyoxalase 1

    Glra2 glycine receptor, alpha 2 subunit

    Gm2a GM2 ganglioside activator protein

    Gnas GNAS (guanine nucleotide binding protein, alpha stimulating) complex locus

    Gnpat glyceronephosphate O-acyltransferase

    Gns glucosamine (N-acetyl)-6-sulfatase

    Gon4l gon-4-like (C.elegans)

    Gpc3 glypican 3

    Gphn gephyrin

    Gpi1 glucose phosphate isomerase 1

    Gpx1 glutathione peroxidase 1Grcc10 gene rich cluster, C10 gene

    Gria3 glutamate receptor, ionotropic, AMPA3 (alpha 3)

    Grik2 glutamate receptor, ionotropic, kainate 2 (beta 2)

    Grin2a glutamate receptor, ionotropic, NMDA2A (epsilon 1)

    Grin2b glutamate receptor, ionotropic, NMDA2B (epsilon 2)

    Grm8 glutamate receptor, metabotropic 8

    Grpr gastrin releasing peptide receptor 

    Gss glutathione synthetase

    Gstm1 glutathione S-transferase, mu 1

    Gucy2f guanylate cyclase 2f 

    Gusb glucuronidase, beta

    Gyk glycerol kinase

    Hadh hydroxyacyl-Coenzyme A dehydrogenase

    Hccs holocytochrome c synthetase

    Hdac4 histone deacetylase 4Hesx1 homeobox gene expressed in ES cells

    Hexa hexosaminidase A

    Hexb hexosaminidase B

    Hla-a major histocompatibility complex, class I, A

    Hla-drb1 major histocompatibility complex, class II, DR beta 1

    Hlcs holocarboxylase synthetase (biotin- [propriony-Coenzyme A carboxylase (ATP-hydrolyzing)] ligase)

    Hmgcl 3-hydroxy-3-methylglutaryl-Coenzyme A lyase

    Hnrnph2 heterogeneous nuclear ribonucleoprotein H2

    Hoxa1 homeobox A1

    Hoxb1 homeobox B1

    Hpd 4-hydroxyphenylpyruvic acid dioxygenase

    Hprt hypoxanthine guanine phosphoribosyl transferase

    Hras Harvey rat sarcoma virus oncogene

    Hs3st5 heparan sulfate (glucosamine) 3-O-sulfotransferase 5

    Hsd11b1 hydroxysteroid 11-beta dehydrogenase 1Hsd17b10 hydroxysteroid (17-beta) dehydrogenase 10

    Hspd1 heat shock protein 1 (chaperonin)

    Htr1b 5-hydroxytryptamine (serotonin) receptor 1B

    Htr2a 5-hydroxytryptamine (serotonin) receptor 2A

    Htr3a 5-hydroxytryptamine (serotonin) receptor 3A

    Htr3c 5-hydroxytryptamine (serotonin) receptor 3, family member C

    Htr7 5-hydroxytryptamine (serotonin) receptor 7

    Huwe1 HECT, UBA and WWE domain containing 1

    Ids iduronate 2-sulfatase

    Idua iduronidase, alpha-L-

    Igbp1 immunoglobulin (CD79A) binding protein 1

    Igbp1b immunoglobulin (CD79A) binding protein 1b

    Igf1 insulin-like growth factor 1

    Ikbkg inhibitor of kappaB kinase gamma

    38 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    29/40

    Table 2  (continued )

    Gene symbol Gene name

    Il1rapl1 interleukin 1 receptor accessory protein-like 1

    Immp2l IMP2 inner mitochondrial membrane peptidase-like (S. cerevisiae)

    Inpp1 inositol polyphosphate-1-phosphatase

    Inpp4a inositol polyphosphate-4-phosphatase, type IInpp5e inositol polyphosphate-5-phosphatase E

    Iqsec2 IQ motif and Sec7 domain 2

    Itga4 integrin alpha 4

    Itga7 integrin alpha 7

    Itgb3 integrin beta 3

    Itgb7 integrin beta 7

    Jag1 jagged 1

    Jmjd1c jumonji domain containing 1C

    Kank1 KN motif and ankyrin repeat domains 1

    Kcnj1 potassium inwardly-rectifying channel, subfamily J, member 1

    Kcnj10 potassium inwardly rectifying channel, subfamily J, member 10

    Kcnj11 potassium inwardly rectifying channel, subfamily J, member 11

    Kcnk9 potassium channel, subfamily K, member 9

    Kcnma1 potassium large conductance calcium-activated channel, subfamily M, alpha member 1

    Kctd7 potassium channel tetramerization domain containing 7Kdm5a lysine (K)-specific demethylase 5A

    Kdm5c lysine (K)-specific demethylase 5C

    Kdm6b KDM1 lysine (K)-specific demethylase 6B

    Kiaa1586 Kiaa1586

    Kif7 kinesin family member 7

    Kirrel3 kin of IRRE like 3 (Drosophila)

    Klf8 Kruppel-like factor 8

    Klhl15 kelch-like 15

    Kmt2c lysine (K)-specific methyltransferase 2C

    Kras v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog

    L1cam L1 cell adhesion molecule

    L2hgdh L-2-hydroxyglutarate dehydrogenase

    Lama1 laminin, alpha 1

    Lama2 laminin, alpha 2

    Lamb1 laminin B1Lamp2 lysosomal-associated membrane protein 2

    Large l ike-glycosyltransferase

    Larp7 La ribonucleoprotein domain family, member 7

    Las1l LAS1-like (S. cerevisiae)

    Lrfn5 leucine rich repeat and fibronectin type III domain containing 5

    Lrpprc leucine-rich PPR-motif containing

    Lrrc1 leucine rich repeat containing 1

    Lzts2 leucine zipper, putative tumor suppressor 2

    Macrod2 MACRO domain containing 2

    Magt1 magnesium transporter 1

    Man1b1 mannosidase, alpha, class 1B, member 1

    Man2b1 mannosidase 2, alpha B1

    Manba mannosidase, beta A, lysosomal

    Maoa monoamine oxidase A

    Map2 microtubule-associated protein 2Map2k1 mitogen-activated protein kinase kinase 1

    Map2k2 mitogen-activated protein kinase kinase 2

    Mark1 MAP/microtubule affinity-regulating kinase 1

    Mat1a methionine adenosyltransferase I, alpha

    Mbd1 methyl-CpG binding domain protein 1

    Mbd3 methyl-CpG binding domain protein 3

    Mbd4 methyl-CpG binding domain protein 4

    Mbd5 methyl-CpG binding domain protein 5

    Mbtps2 membrane-bound transcription factor peptidase, site 2

    Mccc1 methylcrotonoyl-Coenzyme A carboxylase 1 (alpha)

    Mccc2 methylcrotonoyl-Coenzyme A carboxylase 2 (beta)

    Mcoln1 mucolipin 1

    Mcph1 microcephaly, primary autosomal recessive 1

    Mdga2 MAM domain containing glycosylphosphatidylinositol anchor 2

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   39

  • 8/19/2019 1-s2.0-S0306452215008076-main

    30/40

    Table 2  (continued )

    Gene symbol Gene name

    Mecp2 methyl CpG binding protein 2

    Med12 mediator complex subunit 12

    Med13l mediator complex subunit 13-like

    Med17 mediator complex subunit 17Med23 mediator complex subunit 23

    Med25 mediator complex subunit 25

    Mef2c myocyte enhancer factor 2C

    Met met proto-oncogene

    Mgat2 mannoside acetylglucosaminyltransferase 2

    Mid1 midline 1

    Mkks McKusick–Kaufman syndrome

    Mll2 myeloid/lymphoid or mixed-lineage leukemia 2

    Mlycd malonyl-CoA decarboxylase

    Mmachc methylmalonic aciduria cblC type, with homocystinuria

    Mocs1 molybdenum cofactor synthesis 1

    Mocs2 molybdenum cofactor synthesis 2

    Mogs mannosyl-oligosaccharide glucosidase

    Mpdu1 mannose-P-dolichol utilization defect 1

    Mtf1 metal response element binding transcription factor 1Mthfr 5,10-methylenetetrahydrofolate reductase

    Mtm1 X-linked myotubular myopathy gene 1

    Mtr 5-methyltetrahydrofolate-homocysteine methyltransferase

    Mtrr 5-methyltetrahydrofolate-homocysteine methyltransferase reductase

    Myo16 myosin XVI

    Myo5a myosin VA

    Naga N-acetyl galactosaminidase, alpha

    Naglu alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB)

    Nbea neurobeachin

    Nbn nibrin

    Nd5 NADH dehydrogenase subunit 5

    Ndnl2 necdin-like 2

    Ndp Norrie disease (pseudoglioma) (human)

    Ndst1 N-deacetylase/N-sulfotransferase (heparan glucosaminyl) 1

    Ndufa1 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 1Ndufs1 NADH dehydrogenase (ubiquinone) Fe-S protein 1

    Ndufs2 NADH dehydrogenase (ubiquinone) Fe-S protein 2

    Ndufs4 NADH dehydrogenase (ubiquinone) Fe-S protein 4

    Ndufs7 NADH dehydrogenase (ubiquinone) Fe-S protein 7

    Ndufs8 NADH dehydrogenase (ubiquinone) Fe-S protein 8

    Ndufv1 NADH dehydrogenase (ubiquinone) flavoprotein 1

    Neu1 neuraminidase 1

    Nf1 neurofibromatosis 1

    Nhs Nance-Horan syndrome (human)

    Nkx2-1 NK2 homeobox 1

    Nlgn1 neuroligin 1

    Nlgn3 neuroligin 3

    Nlgn4-x neuroligin 4, X-linked

    Nlgn4-y neuroligin 4, Y-linked

    Nlrp3 NLR family, pyrin domain containing 3Nos1ap nitric oxide synthase 1 (neuronal) adaptor protein

    Nos2a nitric oxide synthase 2A, inducible

    Npas2 neuronal PAS domain protein 2

    Npc1 Niemann-Pick type C1

    Npc2 Niemann-Pick type C2

    Nras neuroblastoma ras oncogene

    Nrcam neuronal cell adhesion molecule

    Nrp2 neuropilin 2

    Nrxn1 neurexin I

    Nsd1 nuclear receptor-binding SET-domain protein 1

    Nsdhl NAD(P)-dependent steroid dehydrogenase-like

    Ntng1 netrin G1

    Ntrk1 neurotrophic tyrosine kinase, receptor, type 1

    Ntrk3 neurotrophic tyrosine kinase, receptor, type 3

    40 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    31/40

    Table 2  (continued )

    Gene symbol Gene name

    Oat ornithine aminotransferase

    Ocrl oculocerebrorenal syndrome of Lowe

    Odf3l2 outer dense fiber of sperm tails 3-like 2

    Ofd1 oral-facial-digital syndrome 1 gene homolog (human)Ophn1 oligophrenin 1

    Oprm1 opioid receptor, mu 1

    Or1c1 olfactory receptor, family 1, subfamily C, member 1

    Otc ornithine transcarbamylase

    Oxtr oxytocin receptor  

    Pafah1b1 platelet-activating factor acetylhydrolase, isoform 1b, subunit 1

    Pah phenylalanine hydroxylase

    Pak3 p21 protein (Cdc42/Rac)-activated kinase 3

    Pank2 pantothenate kinase 2

    Park2 Parkinson disease (autosomal recessive, juvenile) 2, parkin

    Parp1 poly (ADP-ribose) polymerase family, member 1

    Pax8 paired box 8

    Pcca propionyl-Coenzyme A carboxylase, alpha polypeptide

    Pccb propionyl Coenzyme A carboxylase, beta polypeptide

    Pcdh10 protocadherin 10Pcdh19 protocadherin 19

    Pcdh9 protocadherin 9

    Pcp4 Purkinje cell protein 4

    Pcx pyruvate carboxylase

    Pde9a phosphodiesterase 9A

    Pdha1 pyruvate dehydrogenase E1 alpha 1

    Pdhx pyruvate dehydrogenase complex, component X

    Pdp1 pyruvate dehydrogenase phosphatase catalytic subunit 1

    Pdzd4 PDZ domain containing 4

    Pecr peroxisomal trans-2-enoyl-CoA reductase

    Pepd peptidase D

    Per1 period circadian clock 1

    Pex1 peroxisomal biogenesis factor 1

    Pex10 peroxisomal biogenesis factor 10

    Pex12 peroxisomal biogenesis factor 12Pex13 peroxisomal biogenesis factor 13

    Pex16 peroxisomal biogenesis factor 16

    Pex19 peroxisomal biogenesis factor 19

    Pex3 peroxisomal biogenesis factor 3

    Pex5 peroxisomal biogenesis factor 5

    Pex5l peroxisomal biogenesis factor 5-like

    Pex6 peroxisomal biogenesis factor 6

    Pex7 peroxisomal biogenesis factor 7

    Pgk1 phosphoglycerate kinase 1

    Phf6 PHD finger protein 6

    Phf8 PHD finger protein 8

    Pik3cg phosphoinositide-3-kinase, catalytic, gamma polypeptide

    Pitx1 paired-like homeodomain transcription factor 1

    Pla2g6 phospholipase A2, group VI

    Pln phospholambanPlp1 proteolipid protein (myelin) 1

    Pmm2 phosphomannomutase 2

    Pnp purine-nucleoside phosphorylase

    Polr3b polymerase (RNA) III (DNA directed) polypeptide B

    Pomgnt1 protein O-linked mannose beta 1,2-N-acetylglucosaminyltransferase

    Pomt1 protein-O-mannosyltransferase 1

    Pon1 paraoxonase 1

    Porcn porcupine homolog (Drosophila)

    Pou1f1 POU domain, class 1, transcription factor 1

    Ppox protoporphyrinogen oxidase

    Ppp1r3f protein phosphatase 1, regulatory (inhibitor) subunit 3F

    Ppt1 palmitoyl-protein thioesterase 1

    Pqbp1 polyglutamine binding protein 1

    Prkcb protein kinase C, beta

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   41

  • 8/19/2019 1-s2.0-S0306452215008076-main

    32/40

    Table 2  (continued )

    Gene symbol Gene name

    Prkcg protein kinase C, gamma

    Prkra protein kinase, interferon inducible double stranded RNA-dependent activator 

    Prmt10 protein arginine methyltransferase 10 (putative)

    Prps1 phosphoribosyl pyrophosphate synthetase 1Prrt2 proline-rich transmembrane protein 2

    Prss12 protease, serine 12 neurotrypsin (motopsin)

    Psap prosaposin

    Psmd10 proteasome (prosome, macropain) 26S subunit, non-ATPase, 10

    Ptch1 patched homolog 1

    Ptch2 patched homolog 2

    Ptchd1 patched domain containing 1

    Pten phosphatase and tensin homolog

    Ptgs2 prostaglandin-endoperoxide synthase 2

    Ptpn11 protein tyrosine phosphatase, non-receptor type 11

    Pts 6-pyruvoyl-tetrahydropterin synthase

    Pvrl1 poliovirus receptor-related 1

    Pycr1 pyrroline-5-carboxylate reductase 1

    Qdpr quinoid dihydropteridine reductase

    Rab39b RAB39B, member RAS oncogene familyRaf1 v-raf-leukemia viral oncogene 1

    Rai1 retinoic acid induced 1

    Ralgds ral guanine nucleotide dissociation stimulator 

    Rapgef4 Rap guanine nucleotide exchange factor (GEF) 4

    Rb1cc1 RB1-inducible coiled-coil 1

    Rbm10 RNA binding motif protein 10

    Rbm28 RNA binding motif protein 28

    Reep3 receptor accessory protein 3

    Reln reelin

    Rfwd2 ring finger and WD repeat domain 2

    Rgs7 regulator of G protein signaling 7

    Rhoxf1 Rhox homeobox family, member 1

    Rims3 regulating synaptic membrane exocytosis 3

    Rlim ring finger protein, LIM domain interacting

    Robo1 roundabout homolog 1 (Drosophila)Rpgrip1l Rpgrip1-like

    Rpl10 ribosomal protein L10

    Rps6ka2 ribosomal protein S6 kinase, polypeptide 2

    Rps6ka3 ribosomal protein S6 kinase polypeptide 3

    Sar1b SAR1 gene homolog B (S. cerevisiae)

    Satb2 special AT-rich sequence binding protein 2

    Sc5d sterol-C5-desaturase (fungal ERG3, delta-5-desaturase) homolog (S. cerevisiae)

    Scaper S phase cyclin A-associated protein in the ER

    Scn1a sodium channel, voltage-gated, type I, alpha

    Scn2a1 sodium channel, voltage-gated, type II, alpha 1

    Sdc2 syndecan 2

    Sdha succinate dehydrogenase complex, subunit A, flavoprotein (Fp)

    Sema5a sema domain, seven thrombospondin repeats (type 1 and type 1-like), transmembrane domain (TM) and short cytoplasmic

    domain, (semaphorin) 5A

    Setbp1 SET binding protein 1Sez6l2 seizure-related 6 homolog like 2

    Sgsh N-sulfoglucosamine sulfohydrolase (sulfamidase)

    Sh3kbp1 SH3-domain kinase binding protein 1

    Shank2 SH3/ankyrin domain gene 2

    Shank3 SH3/ankyrin domain gene 3

    Shh sonic hedgehog

    Shoc2 soc-2 (suppressor of clear) homolog (C. elegans)

    Shroom4 shroom family member 4

    Six3 sine oculis-related homeobox 3

    Slc12a1 solute carrier family 12, member 1

    Slc12a6 solute carrier family 12, member 6

    Slc16a2 solute carrier family 16 (monocarboxylic acid transporters), member 2

    Slc17a5 solute carrier family 17 (anion/sugar transporter), member 5

    Slc1a1 solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, system Xag), member 1

    42 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    33/40

    Table 2  (continued )

    Gene symbol Gene name

    Slc25a12 solute carrier family 25 (mitochondrial carrier, Aralar), member 12

    Slc25a15 solute carrier family 25 (mitochondrial carrier ornithine transporter), member 15

    Slc2a1 solute carrier family 2 (facilitated glucose transporter), member 1

    Slc31a1 solute carrier family 31, member 1Slc35c1 solute carrier family 35, member C1

    Slc4a10 solute carrier family 4, sodium bicarbonate cotransporter-like, member 10

    Slc4a4 solute carrier family 4 (anion exchanger), member 4

    Slc5a5 solute carrier family 5 (sodium iodide symporter), member 5

    Slc6a4 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4

    Slc6a8 solute carrier family 6 (neurotransmitter transporter, creatine), member 8

    Slc7a7 solute carrier family 7 (cationic amino acid transporter, y+ system), member 7

    Slc9a6 solute carrier family 9 (sodium/hydrogen exchanger), member 6

    Slc9a9 solute carrier family 9 (sodium/hydrogen exchanger), member 9

    Smc1a structural maintenance of chromosomes 1A

    Smc3 structural maintenance of chromosomes 3

    Smpd1 sphingomyelin phosphodiesterase 1, acid lysosomal

    Sms spermine synthase

    Snap29 synaptosomal-associated protein 29

    Sos1 son of sevenless homolog 1 (Drosophila)Sox10 SRY (sex determining region Y)-box 10

    Sox3 SRY (sex determining region Y)-box 3

    Spg11 spastic paraplegia 11

    Srd5a3 steroid 5 alpha-reductase 3

    Srpx2 sushi-repeat-containing protein, X-linked 2

    St7 suppression of tumorigenicity 7

    Stag2 stromal antigen 2

    Stard8 START domain containing 8

    Stk39 serine/threonine kinase 39

    Stxbp1 syntaxin binding protein 1

    Sucla2 succinate-Coenzyme A ligase, ADP-forming, beta subunit

    Suclg2 succinate-Coenzyme A ligase, GDP-forming, beta subunit

    Suox sulfite oxidase

    Surf1 surfeit gene 1

    Syn1 synapsin ISyngap1 synaptic Ras GTPase activating protein 1 homolog (rat)

    Syp synaptophysin

    Syt17 synaptotagmin XVII

    Taf1 TAF1 RNA polymerase II, TATA box binding protein (TBP)-associated factor 

    Taf2 TAF2 RNA polymerase II, TATA box binding protein (TBP)-associated factor 

    Taf7 TAF7 RNA polymerase II, TATA box binding protein (TBP)-associated factor 

    Taf7l TAF7-like RNA polymerase II, TATA box binding protein (TBP)-associated factor 

    Taf9b TAF9B RNA polymerase II, TATA box binding protein (TBP)-associated factor 

    Tat tyrosine aminotransferase

    Tbc1d24 TBC1 domain family, member 24

    Tbce tubulin-specific chaperone E

    Tcf4 transcription factor 4

    Tcn2 transcobalamin 2

    Tdgf1 teratocarcinoma-derived growth factor 1

    Tdo2 tryptophan 2,3-dioxygenaseTg thyroglobulin

    Tgfbr1 transforming growth factor, beta receptor I

    Tgfbr2 transforming growth factor, beta receptor II

    Tgif1 TGFB-induced factor homeobox 1

    Th tyrosine hydroxylase

    Thoc2 THO complex 2

    Thrb thyroid hormone receptor beta

    Timm8a1 translocase of inner mitochondrial membrane 8A1

    Tmem135 transmembrane protein 135

    Agmo alkylglycerol monooxygenase

    Tmem67 transmembrane protein 67

    Tph2 tryptophan hydroxylase 2

    Tpi1 triosephosphate isomerase 1

    Tpo thyroid peroxidase

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   43

  • 8/19/2019 1-s2.0-S0306452215008076-main

    34/40

    We present here data showing clock genes in thehippocampus are expressed in a circadian-dependent

    manner in dark/dark conditions. Consistently, the

    abundance of Rev-erba   protein and mRNA was

    inversely correlated, which was expected given that

    Rev-erba   represses its own transcription (Valnegri et al.,

    2011). The abundance of Clock protein peaked at CT0,

    about 3 h after the peak in clock transcription. This analy-

    sis of mRNA and protein abundance suggests that the

    hippocampus presents some characteristics of an

    endogenous clock. However, previous experiments

    examining the expression of clock genes at the mRNA

    and protein level in light/dark conditions showed that the

    expression of most clock genes peaks simultaneously,

    suggesting the absence of an endogenous oscillator in

    the hippocampus (Jilg et al., 2010). Therefore, it is impor-tant to conduct parallel studies in both dark/dark and

    light/dark conditions to determine how the SCN synchro-

    nization by light influences gene expression in the

    hippocampus.

    The expression of circadian oscillating genes is very

    tissue specific. Indeed, Panda et al. found little overlap

    in CCC genes expressed in the SCN and those

    expressed in the liver (about 8%) (Panda et al., 2002).

    Similarly, among the 663 hippocampal circadian genes

    identified in this study, less than 3% correspond to

    circadian-regulated genes in the SCN. This tissue speci-

    ficity suggests that most of these genes have specific

    functions in the hippocampus. Indeed, we identified

    pathways related to memory in our list of hippocampal

    Table 2  (continued )

    Gene symbol Gene name

    Tpp1 tripeptidyl peptidase I

    Trappc9 trafficking protein particle complex 9

    Trmt1 tRNA methyltransferase 1

    Tsc1 tuberous sclerosis 1Tsc2 tuberous sclerosis 2

    Tshb thyroid stimulating hormone, beta subunit

    Tshr thyroid stimulating hormone receptor 

    Tsn translin

    Tspan7 tetraspanin 7

    Tuba1a tubulin, alpha 1A

    Tuba8 tubulin, alpha 8

    Tubb2b tubulin, beta 2B class IIB

    Tubb3 tubulin, beta 3 class III

    Tusc3 tumor suppressor candidate 3

    Ube2a ubiquitin-conjugating enzyme E2A

    Ube2h ubiquitin-conjugating enzyme E2H

    Ube3a ubiquitin protein ligase E3A

    Ubr7 ubiquitin protein ligase E3 component n-recognin 7 (putative)

    Upb1 ureidopropionase, betaUpf3b UPF3 regulator of nonsense transcripts homolog B (yeast)

    Usp27x ubiquitin specific peptidase 27, X chromosome

    Vash1 vasohibin 1

    Vldlr very low density lipoprotein receptor 

    Vps13b vacuolar protein sorting 13B (yeast)

    Vrk1 vaccinia-related kinase 1

    Wdr45b WD repeat domain 45B

    Wdr62 WD repeat domain 62

    Wnk3 WNK lysine deficient protein kinase 3

    Wnt2 wingless-type MMTV integration site family, member 2

    Xpa xeroderma pigmentosum, complementation group A

    Xpc xeroderma pigmentosum, complementation group C

    Xpnpep3 X-prolyl aminopeptidase (aminopeptidase P) 3, putative

    Ywhae tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, epsilon polypeptide

    Yy1 YY1 transcription factor  Zbtb16 zinc finger and BTB domain containing 16

    Zbtb40 zinc finger and BTB domain containing 40

    Zcchc8 zinc finger, CCHC domain containing 8

    Zdhhc15 zinc finger, DHHC domain containing 15

    Zdhhc9 zinc finger, DHHC domain containing 9

    Zeb2 zinc finger E-box binding homeobox 2

    Zfp526 zinc finger protein 526

    Zfp592 zinc finger protein 592

    Zfp711 zinc finger protein 711

    Zic2 zinc finger protein of the cerebellum 2

    Zic3 zinc finger protein of the cerebellum 3

    44 J. Renaud et al. /Neuroscience 308 (2015) 11–50

    http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    35/40

    Table 3.  Circadian clock controlled genes involved in ID. List of the 30 ID genes showing a CCC pattern of expression in the mouse hippocampus

    among the 673 ID genes

    Gene

    symbol

    Gene name Chromosome

    (H/M)

    Function Disease(s) also

    associated to ID

    GO annotations

    Human Mus

    musculus

     AFF2 Aff2    AF4/FMR2 family,

    member 2

    X RNA-binding protein Fragile XE syndrome G-quadruplex RNA

    binding

     ALG2 Alg2    asparagine-linked

    glycosylation 2

    (alpha-1,3-

    mannosyltransferase)

    9/4 alpha 1,3

    mannosyltransferase

    congenital disorder of 

    glycosylation and

    congenital disorder of 

    glycosylation type ii

    calcium-dependent

    protein binding and

    protein

    heterodimerization

    activity.

     ASS1 Ass1   argininosuccinate

    synthase 1

    9/2 control of blood pressure citrullinemia and

    citrullinemia type i

    protein binding and

    ATP binding

    BBS7 Bbs7     Bardet-Biedl

    syndrome 7

    4/3 ciliogenesis bbs7-related Bardet-

    Biedl syndrome and

    Bardet-Biedl

    syndrome 7

    protein binding

    BRAF Braf     Braf transforming

    gene

    7/6 transduction of mitogenic

    signals from the cell

    membrane to the nucleus

    colorectal cancer and

    hairy cell leukemia

    protein kinase

    activity and

    calcium ion binding

    CDH22 Cdh22     cadherin 22 20/2 calcium-dependent cell

    adhesion proteins

    colorectal cancer calcium ion binding

    CLCNKB Clcnkb   chloride channel,

    voltage-sensitive Kb

    1/4 voltage-gated chloride

    channel

    bartter syndrome type

    3 and bartter 

    syndrome type 4

    voltage-gated

    chloride channel

    activity

    CLN6 Cln6     ceroid-lipofuscinosis,

    neuronal 6

    15/9 degradation of post-

    translationally modified

    proteins in lysosomes

    ceroid-lipofuscinosis,

    neuronal 6, variant

    late infantile and

    ceroid lipofuscinosis

    neuronal 6.

    protein binding and

    protein

    homodimerization

    activity

    DAPK1 Dapk1   death-associated

    protein kinase 1

    9/13 calcium/calmodulin-

    dependent serine/threonine

    kinase

    pediatric lymphoma

    and central nervous

    system lymphoma

    protein kinase

    activity and

    identical protein

    binding

    DBT Dbt     dihydrolipoamide

    branched chain

    transacylase E2

    1/3 the branched-chain alpha-

    keto dehydrogenase complex

    catalyzes the overall

    conversion of alpha-keto

    acids to acyl-CoA and CO(2)

    maple syrup urine

    disease and maple

    syrup urine disease

    type 2

    dihydrolipolylysine-

    residue (2-

    methylpropanoyl)

    transferase activity

    and cofactor 

    binding

    FABP5 Fabp5     fatty acid binding

    protein 5

    8/3 high specificity for fatty acids bladder squamous

    cell carcinoma and

    psoriasis

    transporter activity

    and fatty acid

    binding

    FGFR3 Fgfr 3     fibroblast growth

    factor receptor 3

    4/5 tyrosine-protein kinase acting

    as cell-surface receptor for 

    fibroblast growth factors and

    playing an essential role in the

    regulation of cell proliferation,

    differentiation and apoptosis

    achondroplasia and

    hypochondroplasia

    protein tyrosine

    kinase activity and

    fibroblast growth

    factor-activated

    receptor activity

    FKRP Fkrp   fukutin-related protein 19/7 post-translational modification

    of dystroglycan

    limb-girdle muscular 

    dystrophy type 2i and

    muscular dystrophy-

    dystroglycanopathy

    transferase activity

    GALE Gale   UDP-galactose-4-

    epimerase

    1/4 epimerization of UDP-glucose

    to UDP-galactose and the

    epimerization of UDP-N-

    acetylglucosamine to UDP-N-

    acetylgalactosamine

    epimerase deficiency

    galactosemia and

    erythrocyte galactose

    epimerase deficiency

    coenzyme binding

    and protein

    homodimerization

    activity

    GLDC Gldc     glycine

    decarboxylase

    9/19 degradation of glycine gldc-related glycine

    encephalopathy and

    glycine

    encephalopathy

    electron carrier 

    activity and lyase

    activity

    (continued on next page)

    J. Renaud et al. / Neuroscience 308 (2015) 11–50   45

  • 8/19/2019 1-s2.0-S0306452215008076-main

    36/40

    Table 3  (continued )

    Gene

    symbol

    Gene name Chromosome

    (H/M)

    Function Disease(s) also

    associated to ID

    GO annotations

    Human Mus

    musculus

    GUCY2F Gucy2f    guanylate cyclase 2F X resynthesis of cGMP after 

    light activation of the visual

    signal transduction cascade

    retinitis and retinal

    disease

    protein

    heterodimerization

    activity and protein

    homodimerization

    activity

    HEXB Hexb   hexosaminidase B 5/13 degradation of GM2

    gangliosides and of other 

    molecules containing terminal

    N-acetyl hexosamines

    sandhoff disease and

    cervical cancer,

    cation binding and

    protein

    homodimerization

    activity

    KMT2C Kmt 2c    lysine (K)-specific

    methyltransferase 2C

    7/5 histone methyltransferase transcriptional

    coactivation

    MAGT1 Magt1   magnesium

    transporter 1

    X N-glycosylation and Mg(2+)

    transport

    x-linked magnesium

    deficiency with

    epstein-barr virus

    infection and

    neoplasia and t

    lymphocyte

    deficiency

    magnesium ion

    transmembrane

    transporter activity

    MKKS Mkks   McKusick–Kaufman

    syndrome

    20/2 folding of proteins upon ATP

    hydrolysis

    Bardet-Biedl

    syndrome and

    McKusick–Kaufman

    syndrome

    unfolded protein

    binding and ATP

    binding

    PARK2 Park2     Parkinson disease

    (autosomal

    recessive, juvenile) 2,

    parkin

    6/17 part of a multiprotein E3

    ubiquitin ligase complex,

    catalyzing the covalent

    attachment of ubiquitin

    moieties onto substrate

    proteins

    Parkinson’s disease

    and parkin type of 

     juvenile Parkinson

    disease

    protein kinase

    binding and

    ubiquitin protein

    ligase binding.

    PCDH10 Pcdh10    protocadherin 10 4/3 potential calcium-dependent

    cell-adhesion protein

    dravet syndrome and

    nasopharyngitis

    calcium ion binding

    PECR Pecr     peroxisomal trans-2-

    enoyl-CoA reductase

    2/1 chain elongation of fatty acids alcoholism and

    hirschsprung’s

    disease

    2,4-dienoyl-CoA

    reductase

    (NADPH) activity

    and nucleotide

    binding

    PER1 Per1   period circadian clock

    1

    17/11 transcription factor acting as a

    repressor in the circadian

    transcriptional loop

    advanced sleep

    phase syndrome and

    delayed sleep phase

    syndrome

    E-box binding and

    signal transducer 

    activity

    PEX3 Pex3     peroxisomal

    biogenesis factor 3

    6/10 peroxisome biosynthesis and

    integrity

    zellweger syndrome,

    complementation

    group g and

    peroxisome

    biogenesis disorder 

    10a

    lipid binding and

    protein

    dimerization

    activity

    PSMD10 PSMD10    proteasome

    (prosome,

    macropain) 26S

    subunit, non-ATPase,

    10

    X chaperone during the

    assembly of the 26S

    proteasome

    hepatocellular 

    carcinoma and

    autism spectrum

    disorder 

    protein binding and

    transcription factor 

    binding

    RPL10 RPL10     ribosomal protein L10 X protein synthesis, component

    of the 60S subunit of the

    ribosome

    autism spectrum

    disorder and wilms

    tumor 

    structural

    constituent of 

    ribosome

    SLC7A7 SLC7A7    solute carrier family 7

    (cationic amino acid

    transporter, y+

    system), member 7

    14 sodium-independent uptake

    of dibasic amino acids and

    sodium-dependent uptake of 

    some neutral amino acids

    lysinuric protein

    intolerance and

    cystinuria

    amino acid

    transmembrane

    transporter activity

    ZBTB40 ZBTB40    zinc finger and BTB

    domain containing 40

    1/4 transcriptional regulation DNA binding

    46 J. Renaud et al. /Neuroscience 308 (2015) 11–50

  • 8/19/2019 1-s2.0-S0306452215008076-main

    37/40

  • 8/19/2019 1-s2.0-S0306452215008076-main

    38/40

    context. Learning entrains hippocampal memory-specific

    neuronal networks, through the cAMP and MAPK path-

    ways. The amplification or modulation of oscillations in

    these signaling pathways may encode memory before

    it is transferred from the hippocampus to the cortex

    for long-term storage (Eckel-Mahan and Storm, 2009).

    It also appears that this transfer of information betweenthese two structures depends on sleep (Ji and Wilson,

    2007).

    The role of core clock genes in memory formation,

    synaptic plasticity and learning is further strengthened

    by the recent derivation of a  Per1   KO mouse. These

    mice present daytime deficits in working memory

    performance together with the loss of CR-regulated

    CREB phosphorylation in the hippocampus (Rawashdeh

    et al., 2014). Although synaptic plasticity is closely related

    to memory and learning (Whitlock et al., 2006), genes

    encoding excitatory synaptic proteins were not enriched

    in our dataset of CCC genes. The multicellular environ-

    ment of the hippocampus may explain this lack of enrichment. Alternatively, although we focused on tran-

    scriptional regulation in our study, post-transcriptional

    and post-translational modifications may also drive the

    circadian expression of synaptic proteins. Indeed, pro-

    cesses such as RNA methylation (Fustin et al., 2013)

    and protein phosphorylation (Gallego and Virshup,

    2007) are key regulators of the circadian clock.

    Sleep is partly controlled by CR. Specific sequences

    of neuronal activity are replayed during sleep, and sleep

    and consolidation of memory are related processes

    (Shank and Margoliash, 2009; Cairney et al., 2014).

    Furthermore, sleep promotes the formation of dendritic

    spines in the cortex after learning (Yang et al., 2014).

    We show here that hippocampal CCC genes are enrichedin SWG. This overlap supports the idea that sleep, the

    circadian clock and memory are related processes.

    Slow wave sleep (SWS), occurring during non REM

    sleep (non-rapid eye movement sleep), is the deepest

    form of sleep. It is related to the consolidation and

    strengthening of memory and reflects communication

    between the hippocampus and cortex (Huber and Born,

    2014). In humans, slow EEG frequencies are implicated

    in long-term memory performance and consolidation

    (Marshall et al., 2006; Moroni et al., 2014). The highest

    levels of SWS occur during childhood, which supports

    the cognitive role of this type of sleep (Fogel et al.,

    2012). Children with ID, in particular those with Fragile

    X or Down syndrome, show lower than normal levels of 

    A1 EEG slow oscillations, a subtype that reflects the

    deepening of sleep and is associated with higher cogni-

    tive function (Miano et al., 2007, 2008). Aff2,  also known

    as   Fmr2 , has also been implicated in the sleep/wake

    processes in patients with FRAXE-linked ID (Musumeci

    et al., 2000). Interestingly, we also identified  Aff2   as a

    CCC ID gene, suggesting that its rhythmic expression is

    important for memory consolidation.

    Overall, our list of CCC ID genes will be very useful for 

    retrospective and prospective clinical studies. Indeed, it

    would be interesting to search for sleep defects in

    children with ID and mutations in one of these 30 CCC

    genes. In addition these genes are good candidates for 

    mutation screening in patients with ID and sleep

    abnormalities. Our findings may also help clinicians to

    decide at what time of day patients should receive

    education. Indeed, the benefit of dedicated care may

    depend on the rhythmicity of the expression of CCC ID

    genes. Further studies examining this link may lead to

    new therapeutic strategies.

    CONCLUSIONS

    Our analysis demonstrates that about 3.5% of genes

    expressed in the mouse hippocampus show a CCC

    pattern of expression and that ID genes are not

    enriched among these CCC genes. In addition, we

    identify a list of 30 ID genes showing CCC expression.

    Mutations in these genes may lead to defects in the

    circadian regulation of hippocampal-dependent memory

    consolidation and/or in sleep/wake disturbances

    observed in patients with ID.

     Acknowledgments—We thank Cochin Institute genomic facility 

    for technical assistance. We thank Dr. Etienne Challet (from

    INCI, Strasbourg, France) for his expertise on CR. This work 

    was supported by grants from Inserm, Agence Nationale de la

    Recherche (ANR-2010-BLANC-1434-03), Europe (Euro FP7,

    Gencodys, 241995) and Roche (Agreement, N 121531A10).

    J.R. received a postdoctoral fellowship from Roche.

    REFERENCES

    Aguilar-Arnal L, Sassone-Corsi P (2015) Chromatin landscape and

    circadian dynamics: spatial and temporal organization of clock

    transcription. Proc Natl Acad Sci U S A 112:6863–6870.

    Bell-Pedersen D, Cassone VM, Earnest DJ, Golden SS, Hardin PE,

    Thomas TL, Zoran MJ (2005) Circadian rhythms from multiple

    oscillators: lessons from diverse organisms. Nat Rev Genet

    6:544–556.

    Born J, Wilhelm I (2012) System consolidation of memory during

    sleep. Psychol Res 76:192–203.

    Bouchard-Cannon P, Mendoza-Viveros L, Yuen A, Kærn M, Cheng

    H-YM (2013) The circadian molecular clock regulates adult

    hippocampal neurogenesis by controlling the timing of cell-cycle

    entry and exit. Cell Rep 5:961–973.

    Cairney SA, Durrant SJ, Hulleman J, Lewis PA (2014) Targeted

    memory reactivation during slow wave sleep facilitates emotional

    memory consolidation. Sleep 37(701–707):707A.

    Carpen JD, von Schantz M, Smits M, Skene DJ, Archer SN (2006) A

    silent polymorphism in the PER1 gene associates with extremediurnal preference in humans. J Hum Genet 51:1122–1125.

    Chaudhury D, Colwell CS (2002) Circadian modulation of learning

    and memory in fear-conditioned mice. Behav Brain Res

    133:95–108.

    Chaudhury D, Wang LM, Colwell CS (2005) Circadian regulation of 

    hippocampal long-term potentiation. J Biol Rhythms 20:225–236.

    Cheng H-YM, Alvarez-Saavedra M, Dziema H, Choi YS, Li A,

    Obrietan K (2009) Segregation of expression of mPeriod gene

    homologs in neurons and glia: possible divergent roles of 

    mPeriod1 and mPeriod2 in the brain. Hum Mol Genet

    18:3110–3124.

    Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong L-W,

    DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J, Downes

    M, Panda S, Evans RM (2012) Regulation of circadian behaviour 

    and metabolism by REV-ERB-a   and REV-ERB-b. Nature

    485:123–127.

    48 J. Renaud et al. /Neuroscience 308 (2015) 11–50

    http://refhub.elsevier.com/S0306-4522(15)00807-6/h0005http://refhub.elsevier.com/S0306-4522(15)00807-6/h0005http://refhub.elsevier.com/S0306-4522(15)00807-6/h0005http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0015http://refhub.elsevier.com/S0306-4522(15)00807-6/h0015http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0025http://refhub.elsevier.com/S0306-4522(15)00807-6/h0025http://refhub.elsevier.com/S0306-4522(15)00807-6/h0025http://refhub.elsevier.com/S0306-4522(15)00807-6/h0030http://refhub.elsevier.com/S0306-4522(15)00807-6/h0030http://refhub.elsevier.com/S0306-4522(15)00807-6/h0030http://refhub.elsevier.com/S0306-4522(15)00807-6/h0035http://refhub.elsevier.com/S0306-4522(15)00807-6/h0035http://refhub.elsevier.com/S0306-4522(15)00807-6/h0035http://refhub.elsevier.com/S0306-4522(15)00807-6/h0040http://refhub.elsevier.com/S0306-4522(15)00807-6/h0040http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://refhub.elsevier.com/S0306-4522(15)00807-6/h0050http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://refhub.elsevier.com/S0306-4522(15)00807-6/h0045http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0040http://refhub.elsevier.com/S0306-4522(15)00807-6/h0040http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0035http://refhub.elsevier.com/S0306-4522(15)00807-6/h0035http://refhub.elsevier.com/S0306-4522(15)00807-6/h0035http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0030http://refhub.elsevier.com/S0306-4522(15)00807-6/h0030http://refhub.elsevier.com/S0306-4522(15)00807-6/h0030http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0025http://refhub.elsevier.com/S0306-4522(15)00807-6/h0025http://refhub.elsevier.com/S0306-4522(15)00807-6/h0025http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://refhub.elsevier.com/S0306-4522(15)00807-6/h0020http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0015http://refhub.elsevier.com/S0306-4522(15)00807-6/h0015http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://refhub.elsevier.com/S0306-4522(15)00807-6/h0010http://-/?-http://refhub.elsevier.com/S0306-4522(15)00807-6/h0005http://refhub.elsevier.com/S0306-4522(15)00807-6/h0005http://refhub.elsevier.com/S0306-4522(15)00807-6/h0005http://-/?-http://-/?-http://-/?-

  • 8/19/2019 1-s2.0-S0306452215008076-main

    39/40

    Cirelli C, Gutierrez CM, Tononi G (2004) Extensive and divergent

    effects of sleep and wakefulness on brain gene expression.

    Neuron 41:35–43.

    Dardente H, Cermakian N (2007) Molecular circadian rhythms in

    central and peripheral clocks in mammals. Chronobiol Int

    24:195–213.

    De Quervain DJ-F, Aerni A, Schelling G, Roozendaal B (2009)

    Glucocorticoids and the regulation of memory in health anddisease. Front Neuroendocrinol 30:358–370.

    Didde R, Sigafoos J (2001) A review of the nature and treatment of 

    sleep disorders in individuals with developmental disabilities. Res

    Dev Disabil 22:255–272.

    Eckel-Mahan KL, Phan T, Han S, Wang H, Chan GCK, Scheiner ZS,

    Storm DR (2008) Circadian oscillation of hippocampal MAPK

    activity and cAmp: implications for memory persistence. Nat

    Neurosci 11:1074–1082.

    Eckel-Mahan KL, Storm DR (2009) Circadian rhythms and memory:

    not so simple as cogs and gears. EMBO Rep 10:584–591.

    Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster 

    analysis and display of genome-wide expression patterns. Proc

    Natl Acad Sci U S A 95:14863–14868.

    Feillet CA, Mendoza J, Albrecht U, Pe ´ vet P, Challet