You are on page 1of 56

Accepted Manuscript

Evolution of circadian rhythms: from bacteria to human

Utpal Bhadra, Nirav Thakkar, Paromita Das, Manika Pal-Bhadra

PII: S1389-9457(17)30190-9
DOI: 10.1016/j.sleep.2017.04.008
Reference: SLEEP 3383

To appear in: Sleep Medicine

Received Date: 12 January 2017


Revised Date: 7 April 2017
Accepted Date: 18 April 2017

Please cite this article as: Bhadra U, Thakkar N, Das P, Pal-Bhadra M, Evolution of circadian rhythms:
from bacteria to human, Sleep Medicine (2017), doi: 10.1016/j.sleep.2017.04.008.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

Review

Evolution of circadian rhythms: from bacteria to human

PT
Utpal Bhadra a,*, Nirav Thakkar a, Paromita Das b, Manika Pal-Bhadra b
a
Functional Genomics and Gene Silencing Group, Centre for Cellular and Molecular
Biology, Hyderabad-500007, India

RI
b
Centre for Chemical Biology, Indian Institute of Chemical Technology, Hyderabad-500007,
India

SC
Running Title: Evolutionary shift of Circadian rhythm

U
AN
* Corresponding author. Functional Genomics and Gene Silencing Group, Centre for Cellular

and Molecular Biology, Uppal Road, Hyderabad, India-50007. Tel.: +91 40 2719 2513; fax:
M

+91 40 2716 0591.


D

E-mail address: utpal@ccmb.res.in (U. Bhadra).


TE
C EP
AC
ACCEPTED MANUSCRIPT

ABSTRACT

The human body persists in its rhythm as per its initial time zone, and transition always
occurs as per solar movement around the earth over 24 h. But during transition, while

PT
traveling, exceeding that of the earth the movements supercedes, which is far beyond the
toleration of the body’s biological clock. This poses an alteration in our physiological
activities of sleep–wake pattern, mental alertness, organ movement, and eating habits,

RI
causing them to temporarily lose the track of time. This is further re-synchronized with the
physiological cues of the destination over time. The mechanism of resetting of the clocks

SC
with varying time zones and cues occur in organisms from bacteria to humans. It is the result
of the evolution of different pathways and molecular mechanisms over the time. There has
been evolution of numerous comprehensive mechanisms using various research tools to get

U
a deeper insight into the rapid turnover of molecular mechanisms in various species. This
AN
review reports insights into the evolution of the circadian mechanism and its evolutionary
shift which is vital and plays a major role in assisting different organisms to adapt in different
zones and controls their internal biological clocks with changing external cues.
M

Keywords:
D

Feedback loop
Bacteria
TE

Human evolutionary divergence


Transcriptional and post-transcriptional regulation
C EP
AC

Introduction

The last universal common ancestor (LUCA) and all the subsequent life forms that evolved

from it, including humans, have eternally experienced the diurnal changes in the form of day

and night [1,2]. Evolutionary pressures and natural selection have favored the formation of
ACCEPTED MANUSCRIPT
genetic circuits that keep track of time and alter the physiology and behavior of life forms so

as to adapt to the daily changes in light and temperature [3]. The day–night cycle resulting

from the earth’s rotation provides cues to these genetic circuits, which act as internal

timekeepers. This allows the life forms to adapt not only to the diurnal changes but also to

the annual changes reflected in the duration of the day versus night [4,5]. This timekeeper

PT
genetic circuit or the organism’s internal clock inform it about when to venture out for

hunting, when to hide, when to store food for the adverse season, when to prepare for

RI
hibernation/estivation, and even when to reproduce. These behavioral responses are often

critical to the survival of an organism. Experiments in cyanobacteria, show that in

SC
competition experiments, strains that exhibit an internal clock periodicity, which is similar to

external environmental periodicity, have a better Darwinian fitness compared to mutant

U
strains whose internal periodicity is not synchronous with external environmental periodicity.
AN
Drosophila lines with a mutation in genes involved in the maintenance of internal periodicity

exhibit different rates of development as well as differences in their lifespan. In higher


M

animals such as humans, the periodicity of circadian rhythms is most prominently expressed
D

as the sleep–wake cycle. Continued long-term disturbances in a sleep–wake cycle of higher


TE

animals have been linked to several disorders ranging from reduced cognitive ability to a

higher incidence of cancers.


EP

The genetic circuits that regulate the internal clock or pacemaker are conserved from

bacteria to plants and humans, though the individual components or the cogs of these
C

genetic circuits differ [6]. These circadian pacemakers can make an approximate
AC

measurement of time and so their phase needs to be set daily in order to keep the internal

clock in synchrony with the environmental clock. Thus, it is not surprising to know that light is

the leading entrainment cue for all circadian circuits encompassing organisms from

unicellular ones to mammals. In animals, like Zebrafish and Drosophila, light–dark cycles

can entrain peripheral oscillators also, which obviously is only possible if at least some

photons reach all internal organs. The evolution of larger and opaque organisms
ACCEPTED MANUSCRIPT
necessitated the development of different non-photic entrainment pathways, comprising a

highly specialized master pacemaker in the suprachiasmatic nucleus (SCN) in mammals [7–

9]. In this review, we discuss the development of circadian clocks in different organisms from

prokaryotes to plants and from plants to animals, its molecular pathways and the similarities

and variation during the evolutionary time.

PT
Circadian rhythms in cyanobacteria

RI
Circadian rhythms are not restricted to eukaryotes, but also exist in the most primitive

organisms like cyanobacteria. The organism Synechococcus elongates PCC 7942 is a

SC
preferable genetic model for understanding the molecular details pertaining to clock gene

regulation. Pioneer workers like Susan Golden, Carl Johnson, Masahiro Ishiura, and Takao

U
Kondo have incorporated luciferase to monitor rhythmic expression of the gene causing first
AN
clock mutants in cyanobacteria to evolve. Cyanobacteria are the simplest known organisms

known to exhibit circadian rhythms (Fig. 1). Several species of Cyanobacteria are capable of
M

photosynthesis and nitrogen fixation. Oxygen is a by-product of photosynthesis. However,


D

the enzymes involved in nitrogen fixation are inhibited in the presence of oxygen. Unlike

heterocystous filamentous cyanobacteria, non-heterocystous and unicellular cyanobacteria


TE

cannot spatially separate photosynthesis and nitrogen fixation; hence, they resort to
EP

temporal separation of these two processes. Photosynthesis takes place during the daylight,

while nitrogen fixation peaks during night/dark. Interestingly, this rhythmicity is maintained
C

even when they are shifted to constant light (LL), albeit with temperature rhythmicity.
AC

Reporter gene (luciferase) insertions throughout the chromosome of unicellular

cyanobacteria, Synechococcus elongates revealed that all the 800 insertion lines, which

displayed quantifiable fluorescence, exhibited circadian rhythmicity. Ethylmethane sulfonate

(EMS) mutagenesis and rescue experiments showed that the kai gene cluster, composed of

kaiA, kaiB and kaiC genes, is crucial for the maintenance of circadian rhythmicity. kaiA is

transcribed as a monocistronic mRNA from the kaiA promoter while kaiB and kaiC are

transcribed as a di-cistronic mRNA from the kaiBC promoter, which is then translated into
ACCEPTED MANUSCRIPT
two distinct proteins, such as KaiB and KaiC. The process of phosphorylation of the KaiC is

regulated by two proteins, namely KaiA and KaiB. KaiA protein positively regulates the KaiC

phosphorylation and activates accumulation of phosphorylated KaiC, while the KaiB protein

is the negative regulator and promotes the accumulation of non-phosphorylated KaiC protein

[10–12]. Phosphorylated KaiC protein is suggested to stop expression of the kaiBC gene,

PT
thus forming the negative feedback regulation of the kaiBC gene expression [13]. It has been

discovered [14] that rhythmic phosphorylation of KaiC protein persists in cyanobacteria in

RI
constant dark conditions (DD). In DD conditions, transcription, translation, as well as

degradation processes, are inhibited [15]. It was further studied [16] that the KaiC

SC
phosphorylation rhythm in an in vitro system occurs only if proper amounts of KaiA, KaiB and

KaiC proteins as well as ATP exist [16–18]. Research until now reveals the biochemical

U
interactions among KaiA, KaiB, and KaiC proteins and their role in driving circadian
AN
oscillations, but still a regulatory mechanism of phosphorylation of KaiC protein needs to be

elucidated.
M

The loci, crm (circadian rhythm modulator) and rpaB (regulator of phycobilisome association
D

B) comprise the principal clock. crm comprises the gene encoding a 62 peptide is situated
TE

upstream of rpaA presumed earlier to affect the expression of RpaA [19]. Earlier, the genes

kai or rpaA were only thought to be responsible for causing arrhythmia, which was later
EP

disproved [19].

The Crm peptide acts as an allosteric modulator of RpaA, acting as codon initiating
C

translation complement to a crm1 mutant. It may be that the altered peptide of transposon
AC

insertion recessively exerts an effect adverse to the clock. The RpaB response regulator

also plays a part in regulating transcription of the circadian genes. Being highly conserved

[20] it conjugates with the histidine kinase NblS, to regulate genes related to cold, light, and

osmotic and oxidative stress [21–23]. When phosphorylated, RpaB binds to HLR1 (highlight

regulatory 1) regulating the gene expression [21,23]. RpaB activates gene expression, but

usually it inhibits expression of the promoters downstream [21,23,24].


ACCEPTED MANUSCRIPT
An HLR1 element in the PkaiBC promoter shows peak binding at dawn when the light is

constant [24], which is 12 h apart from the peak stage of RpaA getting bound to PkaiBC.

Thus, RpaA and RpaB cooperatively perform regulation of the expression of the circadian

target, by displacing RpaB. Genes like kaiBC (class 1) and purF (class 2), that regulate

RpaA have been found to undergo negative regulation by RpaB (87). A phosphorylated

PT
RpaA shows circadian oscillation, while RpaB phosphorylation occurs when cells are grown

under 12 h of light followed by 12 h of dark [25]. The degree of phosphorylation decreased

RI
with an overexpressed RpaB receiver domain, which suggests that besides RpaA, PraB too

competes for target binding and function as RpaA modulator via regulating RpaB

SC
phosphorylation [25]. This clearly unfolds the cross-talk occurring between the circadian

pathway and the environmentally governed pathway and the synchronizations occurring to

U
generate rhythmic expression of the genes. To regulate a number of peroxide levels [26], an
AN
antioxidant named peroxiredoxin is found to be beneficial; circadian rhythms are therefore

better modified using a conserved marker of peroxiredoxin oxidation [2,27,28]. The 24-h
M

redox cycles are entrainable and also temperature compensated when checked for the
D

oxidized active state. In the eukaryotic cells, however, the rhythms generated via the
TE

peroxiredoxin oxidation function independently of the feedback loops regulating transcription

and translation [28]. Organisms like flies, mice, Neurospora, S. elongates and the archaeon
EP

Halobacterium salinarum sp. NRC-1 show rhythms of peroxiredoxin oxidation [2], which

suggest that there may be circadian rhythm in archaeal organisms too. S. elongate rhythms
C

of peroxiredoxin oxidation show that besides the canonical clock mechanisms, rhythms are
AC

driven via an oscillator still unknown to researchers, which might use more light on the

ancient clock driven by oxidative cycles. In this way, a model previously thought of as linear

with input, oscillator, and output, has emerged to be an intricately integrated network,

comprising elements with multiple functions. Hence, it has made huge leaps in expanding

our knowledge of the cyanobacterial circadian clock with future prospects of unfolding

several more interesting aspects in the system.


ACCEPTED MANUSCRIPT
Internal ‘clock’ of Neurospora

Over several decades, Neurospora crassa, the filamentous fungus, has served as a brilliant

model for the study of biological, regulatory and circadian rhythms. It typically thrives on

woody biomass in the laboratory on agar-containing medium. Recent studies have

demonstrated the disruption of the natural circadian clock in the dark with a consistent

PT
metabolism, extending the possibility of removal of the negative feedback loop operating in

case of the circadian oscillator. The core clock component, frq[29] gets activated by the

RI
White Collar Complex (WCC), which is made of two PAS domains having White Collar-1

SC
(WC-1) and White Collar-2(WC-2) [30–32]. WC-1 is the photoreceptor for blue light while

WC-2 helps in the light signaling pathway. The circadian late night and early morning cause

U
promotion of the transcription of frq in the presence of WCC. FRQ shows a peak during the
AN
circadian midday 4, where FRQ operates through a negative feedback loop, which causes

inhibition of transcription of frq gene. Once inhibition occurs, frq levels show a decline in the
M

circadian in the afternoon and in the early evening. FRQ also upregulates the transcription of

WC-1 and wc-2. The gene, vivid (vvd) is found to be controlled by WCC and shows photo-
D

adaptation [33]. VVD shows a competitive interaction with WC-1 to inhibit the formation of
TE

WCC and transcription of vvd [34].


EP

Thus, the circadian rhythms get stabilized by the interlocking of the negative and positive

feedback loops [35,36]. FRQ undergoes progressive phosphorylation, making it susceptible


C

to ubiquitination and degradation that change its conformation [34,35,37]. Once degraded,
AC

FRQ generates a fresh cycle of activation of transcription by WCC.

Most studies on Neurospora crassa pertain to circadian rhythms, since it reproduces both

asexually as well as sexually during the day/light, while at night/dark, aerial hyphae are

produced perpendicularly to form the asexual spores, macroconidia. The conidia formation

process is regulated in a daily manner and peaks can be seen just before the dawn. The

rhythm of growth and conidia formation is monitored using ‘race tubes’. At 25°C, the growth
ACCEPTED MANUSCRIPT
front advances with a periodicity of about 21.6 h [38]. Many mutations that resulted in

changes in this periodicity were mapped to the frq gene. FRQ protein regulates its own

transcription in a classical negative feedback loop. As the FRQ protein accumulates it is

progressively phosphorylated, by calcium/calmodulin-dependent kinase (CaMK1) and casein

kinase II (CKII), which marks it for degradation. Inhibition of FRQ phosphorylation reduces

PT
FRQ turnover and interestingly also increases the frq mRNA levels, suggesting that

phosphorylation is not only important for FRQ turnover but also its ability to negatively

RI
regulate its own transcription. Two PAS domain containing transcription factor proteins –

WC1 and WC2 – form a complex and induce the transcription of frq. WC1 protein acts as a

SC
photoreceptor and its transcription is induced by light, while WC2 is constitutively expressed.

Phosphorylated FRQ inhibits the transcription-promoting activity of theWC1–WC2 complex,

U
thus inhibiting its own transcription. Turnover of phosphorylated FRQ at night permits the
AN
WC1–WC2 complex to activate transcription of freq. Another PAS domain containing protein,

VVD controls the light response of internal clock to definitive periods of time, such that even
M

in constant light conditions, the light-induced processes occur only transiently. VVD also
D

regulates its own transcription in a negative feedback loop [39–42]. Rhythmicity of the
TE

internal clock is also responsive to temperature. At higher temperatures, the FRQ oscillates

at a higher level, such that the trough level of FRQ at 28°C is higher than the peak level at
EP

21°C. Thus, shifting of Neurospora culture growing at 21–28°C would result in phase s hifting

of the internal clock, which would now correspond to the time of night when the FRQ is
C

lowest. Response to temperature may be an intrinsic property of FRQ protein, as some


AC

mutants of FRQ are not responsive to temperature change. Normal environmental conditions

assume a higher temperature during the day/light phase and a lower temperature

corresponding to the dark/night phase. Interestingly, when Neurospora is placed in

conditions where it is competing for temperature–light, entrainment happens with

temperature cycle, such that conidiation is observed during the light/daytime (lower

temperature). This suggests that light may not be the dominant signal for entrainment of

Neurospora clock [43–46].


ACCEPTED MANUSCRIPT
There is an intricate communication between the cell cycle and circadian clock that regulates

cell division cycles, whose alterations cause diseases like cancer. A highly conserved

coupling has been found between the circadian clock and cell cycle via the serine/threonine

protein kinase-29, which currently proposes that circadian oscillations of cell cycle partners

help to synchronize nuclear divisions. It experimentally demonstrates G1 and G2 cyclins,

PT
CLN-1 and CLB-1 to show oscillation with bioluminescence reporters. Such oscillations are

found to be abolished in the frq knockout mutant. Thus, it shows that the molecular dynamics

RI
of cell cycle that modulate the circadian clock synchronize nuclear divisions in Neurospora.

SC
Circadian Rhythm in Plants

U
AN
Plants were found to show a variety of movement that was driven by rhythms, like the leaf

movement, growth, germination, stomatal movement, enzyme activity, gas exchange, flower
M

opening, photosynthetic activity, and fragrance emission [47]. Kloppstech [48] had elucidated

circadian rhythm in the pea in the abundance of three nuclear transcripts that code for the
D

light-harvesting chlorophyll a/b binding protein (LHCB), the small subunit of ribulose-1,5-
TE

bisphosphate carboxylase/oxygenase and an early light-induced protein. In wheat, the rate

of Cab-1 transcription was found to be controlled by circadian rhythms [49]. Amongst, pea
EP

and wheat, Arabidopsis thaliana was preferable to combine forward genetic analysis with

molecular gene cloning [50]. Later, the rate of transcription and accumulation of LHCB
C

transcript in Arabidopsis [51] were found to regulated by circadian factors.


AC

Arabidopsis show a rhythmic movement of the cotyledon and leaf, which is mostly based on

differential growth. The elongation rate of the abaxial and adaxial cells of the petiole has a

circadian rhythm that helps in oscillating the positions of cotyledons and leaves. Besides,

there is a change in the rate of elongation of hypocotyl [52] and stem inflorescence [53].The

Arabidopsis genome sequencing has [54] identified novel genes having interlocked feedback

loops that are conserved. The Arabidopsis genome sequencing has led to the identification
ACCEPTED MANUSCRIPT
of different orthologs of the recognized clock proteins. The Arabidopsis clock is unique

bearing conserved feedback loops consisting of three interlocked feedback loops, with two

single Myb motif transcription factors, LATE ELONGATED HYPOCOTYL (LHY) and

CIRCADIAN AND CLOCK ASSOCIATED1 (CCA1). CCA1 and LHY are responsible for

repression of the expression of TOC1, the evening-phased protein; this is carried out by the

PT
evening elements binding in its promoter region [55, 56].The family of PSEUDO-RESPONSE

REGULATOR (PRR) protein has a PSEUDO-RECEIVER (PR) domain and a CONSTANS

RI
(CO) domain, CO-like domain, TOC1 (CCT) domain and these proteins act as a general

repressor of transcription with DNA binding activity. TOC1 is a member of this protein family

SC
[57]. TOC1 mediates DNA binding and transcriptional activities through the C-terminus CCT

motif and the N-terminus PR motif, respectively [57–59]. Other experiments revealed that in

U
the evening, the expression of TOC1 inhibits the accumulation of CCA1/LHY. These factors
AN
control the positive regulation of three TOC1 relatives, PRR5, PRR7, andPRR9. TOC1 is a

clock component that controls the oscillation of the mRNA and the protein produced thereof.
M

CCA1/LHY is negatively regulated by PRR5/7/9. CCA1 and LHY are transcriptionally


D

repressed post-degradation by ZTL to regulate the titer of TOC1, which primarily determines
TE

the circadian period [60]. Other experiments revealed that in the evening, the expression of

TOC1 inhibits the accumulation of CCA1/LHY. In the morning, mRNA abundance of


EP

PSEUDORESPONSE REGULATOR 7 (PRR7) and PRR9 is caused by CCA1/LHY

[4,61,62]. In the evening, the circadian loop comprises GIGANTEA (GI), TOC1, EARLY
C

FLOWERING 3 and 4(ELF3 and ELF4), and LUX ARRHYTHMO (LUX) [63,64] (Fig. 2).
AC

There is a formation of complex like evening complex (EC). It is made of ELF3, ELF4 and

LUX suppress the LUX expression, ELF4, GI, TOC1 and PRR9 (Fig. 1) [65–67]. Evening

loop and morning loop are involved in up the fundamental model of the circadian clock in

Arabidopsis [55,68–71].

A novel family of proteins comprising ZEITLUPE (ZTL), LOV KELCH PROTEIN2 (LKP2),

and FLAVIN binding KELCH REPEAT F-BOX (FKF), with PAS/LOV domains, Kelch repeats,
ACCEPTED MANUSCRIPT
and F-boxes [72–74], has revealed interesting aspects of the role of protein degradation in

the circadian clock of Arabidopsis. The LOV domains are photoactive, while FKF is solely

concerned with photoperiodism, while ZTL and LKP2 alter the clock. Mutations in ztl-1 and

ztl-2 cause alterations in the length of the period of various rhythm [73,75,76]. The

abundance of ZTL mRNA is not regulated by the clock, the protein is in its peak at dusk, and

PT
troughs at dawn. The proteasomal degradation pathways are modulated by F-box proteins

that specifically undergo poly-ubiquitination of targets for degradation. Thus, the activity of

RI
TOC1 is regulated through repression of transcription by CCA1 and LHY, which determines

the circadian period.

SC
In Arabidopsis, a positive correlation exists between the period length of a set of natural

factors and the length of the day encountered at latitude of origin of the factors, which

U
determine whether the altered clock function gets selected under differing environmental
AN
conditions. The length of the period depends upon carbon fixation, biomass, and survival too

[77] by altering the timing of flowering [78]. The lengthened period delay of CO mRNA
M

accumulation, which increases the critical day length essential for CO protein accumulation.
D

This late flowering is, therefore, advantageous in the high altitudes before the freezing
TE

weather sets in.

Circadian rhythms in Drosophila


EP

Drosophila melanogaster is a holometabolous insect having defined stages of development


C

from egg, to larva, then pupa, and finally to an adult. Larva, which emerges from the egg,
AC

passes through three instar stages and then forms an immobile pupa. Inside the pupa,

metamorphosis transforms it into a fly, which on development, emerges from the pupa in a

process called eclosion [79]. It was observed that eclosion is ‘gated’ by the circadian clock,

such that most flies eclose 1–2 h before dawn [80–82]. Further, the locomotor behavior

assays show that wild-type flies are active during the day/light and inactive during the

night/dark in an LD (light–dark) cycle. Even in constant darkness, flies continue to maintain

the rhythm showing a high locomotor active phase and an inactive (resting) phase
ACCEPTED MANUSCRIPT
corresponding to subjective day and night [83]. In addition to locomotion and eclosion, other

behaviors controlled by the clock are egg laying, courtship, gustatory sensitivity, olfactory

sensitivity and learning and memory. Caffeine suppresses the resting phase in flies, which is

similar to its effect on sleep in humans. Similarly, hydroxyzine, a sedative drug used for the

treatment of anxiety and tension in humans, enhances the resting phase in Drosophila [84–

PT
89] (Fig. 3).

The Drosophila clock

RI
In a mutagenesis screen, the mutants that displayed short (pers), long (perl) and arrhythmic

SC
(per0) periodicities mapped to the gene ‘period’ (per) on X chromosome of Drosophila

melanogaster, encoding a PAS domain protein. Different periodicities were observed in

U
eclosion as well as locomotor activity [90]. Timeless (tim) was identified in a screen for
AN
recessive mutations that affect the Drosophila clock. As with per gene, short (tims), long

(timl) and arrhythmic (tim0) mutations of tim have also been identified. dClock (dClk) and
M

cycle (cyc; also known as dBmal1) encode bHLH/PAS domain transcription factors that form
D

an integral part of the internal clock in flies. Drosophila doubletime (dbt) gene encodes

casein kinase 1, which is directly or indirectly involved in phosphorylation of PER and its
TE

clearance. SHAGGY (SGG) phosphorylates TIM and controls its entry into the nucleus and
EP

thereby role in a feedback loop. Clearance of TIM is regulated by light, although light directly

does not affect TIM protein in vitro. TIM degradation is important in resetting the internal
C

clock. Mutant flies that lack eyes or the visual transduction pathways can be entrained to
AC

light and exhibit degradation of TIM in response to light, indicating the presence of a

photoreceptor, which functions independently of vision. CRYPTOCHROME (CRY) was

found to perform the function of the circadian photoreceptor. In cryb mutants, TIM is not

degraded in response to light and both per and time expressed constitutively at most places,

where they are expressed with the exception of lateral neurons. Their expression in lateral

neurons continues to be rhythmic in cryb mutant flies and locomotor rhythms are maintained

[91–99] (Table 1).


ACCEPTED MANUSCRIPT
Transcriptional and post-transcriptional feedback models

Biological timekeeping is carried out by the circadian clock. The periodical oscillation of the

circadian clock is known as circadian rhythm. In the case of Drosophila melanogaster, the

period (per) and timeless (tim) core genes are responsible for the circadian rhythm. Gene

expression of per is regulated at transcriptional and post-transcriptional levels. Per genes

PT
undergo changes in the circadian rhythm at the transcriptional level. The rise of per and tim

mRNA in the evening/dark phase leads to accumulation of PER and TIM in the cytoplasm.

RI
But Tim protein is light sensitive and accumulates only after dark and stabilizes when it

SC
forms a dimer along with PERIOD. Again here, PER alone is unstable and dimerization with

TIM stabilizes it. In the absence of TIM, PER is phosphorylated by Double time (Dbt), which

U
has kinase activity, and afterwards it is ubiquitinated by Slimb the F-Box protein and
AN
degraded in the proteasome. Later, the TIM and PER heterodimers enter the nucleus and

accumulate. Further, this dimer binds to Clk/Cyc dimers via a Per-Clk interaction. This
M

interaction causes hyperphosphorylation of the clock and thus prevents Clk/Cyc dimers from

binding to the DNA. This negatively regulates transcription of per and tim genes. This
D

creates a first negative feedback loop where Period inhibits its own transcription. Further Clk
TE

mRNA is transcribed in an opposite way to tim/per mRNA, achieving a peak in the early

morning. Clk/Cyc dimers are formed, which activate transcription of two proteins, namely
EP

Vrille and Pdp1e. Both proteins bind to the V/P boxes in the promoter region of Clk. Vrille

inhibits the transcription of Clk while Pdp1e activates Clk transcription. This forms the
C

second negative feedback loop integrated within the first [3,82,100–105] (Fig. 4).
AC

Evolutionary divergence in clock principles

Ever since life dawned on this planet, there has been a sea change in the environment to

produce the amiable and pleasant conditions that exist at present. With very insignificant

changes in the revolution of the earth around its orbit being approximately 365 days, there

have been appreciable changes in the period of rotation of the earth on its axis, and also in
ACCEPTED MANUSCRIPT
the period of revolution of the moon around the earth. This has brought about significant

variations in the circadian clock over the evolutionary time scales. Over the past 4 billion

years, the distance between the earth and the moon has changed greatly, owing to the

effects of the tidal drag, and this has revealed that initially, the earth completed one rotation

cycle in a period of 4 h which extended to approx. 20 h during the Cambrian period [106].

PT
Organisms evolved and persisted in the ever-changing environment, by virtue of their

Darwinian fitness which resonated between the endogenous periodicity of the organism and

RI
the period changes of the organism’s environment, which was very beautifully depicted by

the cyanobacteria, Synechococcus [107]. This organism simultaneously evolved a circadian

SC
mechanism that caused the organism to delay its mechanism over a millennium about 1.9

billion years ago, using KaiB for 0.25–0.5 billion years ago followed by KaiA until the

U
geophysical period extended to 24 h as in the present. The cryptochromes present now
AN
perform functions in both animals as well as plant circadian clocks. The Cry genes have

undergone extensive duplication, as a result of which, there is a modulation of functions. In


M

mammals, the CRYs play a role of negative regulation of the circadian clock [108], with no
D

significant role in light signaling [109–111].


TE

The six Cry genes in zebrafish negatively regulate light signaling while in Drosophila, the

single cry gene helps in blue-light signaling the central pacemaker neurons [112].The two
EP

Cry genes in Arabidopsis help in transmitting blue-light signaling to the clock [113]. In plants,

the photolyase domain may have undergone fusion with the C-terminal DAS domain
C

400 million years ago since this domain concurrently occurs in both mosses and angiosperm
AC

[114]. The C-terminal domains have undergone extensive divergence in animals, such as

Arabidopsis [115], but in Drosophila [97] and Xenopus [116], they play a role in regulation.

Cryptochromes, therefore, act as molecules that exist as homologs/paralogs to the clock,

playing similar roles in plants and animals. The divergence between the two phyla occurred

about 1.6 million years ago, in the pre-Cambrian age. The cryptochromes not only act as

part of the cellular response to ultraviolet (UV) damage but also evolve for executing

functions related to the clock. p53 undergoes phosphorylation by casein kinase 2 when
ACCEPTED MANUSCRIPT
subjected to UV damage, thus they activate the tumor suppressor [117]. Besides the yeast

casein kinase 1, HRR25 function in transcriptional response to UV damage. These proteins

are important for the phosphorylation of main plant and mammalian [118] clock proteins [70],

Neurospora [115], flies [93,119]. But these proteins are also crucial for the Wnt signaling,

being important players. Post-Cambrian, the evolution of clock genes is mainly due to gene

PT
duplication and functional divergence. Per genes show maximum divergence and are

believed to have duplicated first in the vertebrate lineages.

RI
Duplication in higher eukaryotes exists, where a copy sustains the original functions while

the other undergoes changes in their amino acids to become functionally divergent [120].The

SC
same genetic trend is set by the circadian system, where the gene related to the clock such

as the per, Cry, cycle, and Clock exists in single copies in insects but as duplicate copies in

U
vertebrates. The duplication of a clock gene in a lineage causes it to shuffle to another
AN
chromosome, with the creation of variation with respect to the ancestral copy. The two

copies show divergence with overlapping occurring very frequently, as in the case of the
M

three mPer genes in the murine model that have rhythmic responses in the SCN but have
D

variable responses to light [121].


TE

The mper2 presumptively looks like the positive regulator of the gene, Bmal1 [108] but does

not modulate the rhythm although it regulates the circadian rhythm. The very same per gene
EP

in quails shows a functional divergence only with qPer2, but not with qPer1 mRNA when

induced by light [122]. The Drosophila timeless (tim) genes have two paralogs, tim and tim2
C

(also called timeout), but a single copy of Tim exists in mammals [123–125]. The Tim in
AC

mammals apparently is orthologous to tim2 in the fly, which means that tim2/mTim is the

ancestor and has undergone a recent duplication in the insects, which is evident in Dipterans

and Lepidopterans only, but is missing from the genome of the honeybee. Thus, we can

conclude that the duplication of tim may have preceded the above two orders more than

200 million years ago. mTim has been reported to regulate the clock in mammals [126] but

Drosophila does not provide any such evidence for tim2.


ACCEPTED MANUSCRIPT
Gene duplication is exemplified by another example of the silkmoth, Antheraea pernyi per

gene has two paralogs on different sex chromosomes perZand perW [127]. Several other

sequences of the per encode an antisense transcript in the female sex chromosome W but

are missing in the homogametic males (chromosomes ZZ), who are rhythmic, which proves

that the perW is not essential for clock function. The genes perW and perZ show a great

PT
degree of identical sequence, which suggests that perW may evolve by translocating very

recently from the Z chromosome [127].

RI
There are certain situations when paralogs show divergence and in doing so lose their

complete function, turning out to be pseudogenes, as in the case of the hPer4 in human and

SC
rmPer4 in Rhesus monkey [124,128], in that neither gets transcribed nor codes for

contiguous segments that produce the PER-like protein. Per4 may have its origin from Per3

U
via retrotransposition, which perhaps might have to move the gene to its current position
AN
[128]. After such findings shedding light on the evolution of the clock gene per, there is much

waiting to be discovered about the evolution of the other genes such as cry.
M

Protein regulation of circadian rhythms


D
TE

CLOCK is Circadian Locomotor output cycles kaput. The clock is responsible for the

generation of the circadian rhythms. In the case of most of the clock genes Per-Arnt-Sim
EP

(PAS) domain mediates protein interaction and helps in the regulation of the circadian

rhythm. PAS is also related to the transcription factor and also works as a heterodimer.
C

There are numerous numbers of Rof clock genes being cloned in different genera of
AC

organisms. In the case of neuro POR white collar 1 and white collar 2 and frequency are

present, in the case of humans period (per) and timeless (tim) genes are present. In

Drosophila period (per) double time, timeless (tim), and clock genes are present. In the case

of mammals period genes per 1, per 2, per3, bmal1, clock and timeless (tim) genes are

present. In the case of mammals clock genes or bmal 1genes through the help of

transcription binding factor, E-Box, which is located near the period gene, activate per genes

namely per 1, per 2, per 3. After the transcription of per 1, per 2, per 3, mRNAs are moved
ACCEPTED MANUSCRIPT
and are translated to PER 1 PER 2 and PER 3 proteins. CRY 1 and CRY 2 are

cryptochrome proteins and they act as a negative feedback loop. PER after interaction with

cryptochrome proteins, translocated per proteins from the cytoplasm to the nucleus.

Heterodimers of per and cry translocate back to the nucleus repressing their own

transcription by the complex formation of CLOCK and BMAL1. There is another regulatory

PT
loop by a heterodimer of CLOCK and BMAL1. The retinoic acid-related orphan nuclear

receptor is activated for transcription. There is competition between REV-ERBα and RORα

RI
to bind to the retinoic acid related orphan nuclear receptor. Bmal 1 is activated for

transcription by RORs whereas transcription process is repressed by REV-ERBα. ROR and

SC
REV-ERBα are both responsible for positive and negative regulation of the circadian

oscillation [92,93,119,129–134].

U
AN
The mammalian clock environment

Physiological parameters such as temperature, blood pressure, metabolism, retinal


M

electroretinogram responses and the circulating titers of hormones in mammals are


D

subjected to a 24-h rhythm that persists even in the absence of the cycles of sleep/wake or
TE

light/dark [56,135–137].The circadian clock in mammals has three main components which

include the input signaling pathways, the main pacemaker or the central oscillator and output
EP

signaling pathways. The signals or cues received from the environment are known as

zeitgebers and get communicated to the main pacemaker through the input pathway where
C

they synchronize the oscillations in the pacemaker with respect to the changes in the solar
AC

day–night cycle. Zeitgebers includes factors such as temperature, light, noise, food, and

melatonin [138].The pacemaker for the circadian clock includes two clusters of neurons

called the SCN, comprising about ~100,000 neurons in each, present in the hypothalamus

just above the optic chiasma [139]. Light cues are perceived by the retinal cells and get

transmitted to this region via the retinohypothalamic tract where signal transduction occurs

via the influx of calcium after an interaction with the NMDA receptors to activate the IP3 and

ryanodine receptors. The central oscillator then generates and maintains the rhythmicity
ACCEPTED MANUSCRIPT
upon the necessary input perception. These signals are then diffused consecutively to the

peripheral organs via the output pathway such as the glucocorticoid pathway [140].The core

genes of the mammalian circadian clock comprise 14 genes, including members of the Per,

Cry, Bmal, Clock, Ror and Rev-Erb families, that show an interaction via the positive and

negative feedback loops of the transcription and translation [141].

PT
The circadian day ensues with the CLOCK/BMAL1 activator heteromeric complex formation,

which binds the E-box sequences at the promoter of the genes, Per, Cry, Ror and Rev-Erb

RI
[118,138,142]. The two major loops, REV-ERB/Bmal/ROR and PER/CRY loops,

interconnect [142]. The transcription of the Per 1, 2, 3 and Cry 1, 2 genes activates in the

SC
PER/CRY loop and, in turn, inhibits the transcription of the clock genes via the

CLOCK/BMAL1.The REV-ERB/Bmal/ROR loop helps in driving the oscillations which are

U
self-sustained [142]. Both of the loops actually operate independently but need to
AN
interconnect concomitantly to produce oscillations within a period of 24 h, which is why they

comprise two transcription/translation feedback loops (TTFLs) [143,144].


M

As an exception to the transcriptional rhythms, in some cases there also exists a non-
D

transcriptional/translational rhythm as in the case of the human red blood cells. These cells
TE

are non-nucleated where peroxiredoxins undergo oxidization in a circadian rhythm with the

temperature-compensation ability of the oscillators [15,16,145,146]. On the contrary, the


EP

nucleated cells are influenced by the cytoplasmic peroxiredoxin clock, which takes place via

the transcriptional clock factors [146].


C

Resetting the human clock


AC

The biological clock is an innate mechanism of one’s body. This clock regulates the normal

physiological functioning of the body. There are several genes responsible for the biological

clock. The circadian clock gets reset by an external signal such that these rhythms

generated get synchronized with variations occurring daily in the environment [147]. The

SCN present in the anterior position in the hypothalamus is responsible for the physiological
ACCEPTED MANUSCRIPT
functioning of our body. The SCN works as a pacemaker of the body. Several genes, namely

per, cry, clock, are responsible for the functioning of the biological clock (Table 2). Although

in today’s world of globalization and fast-moving life, external sources such as mobile

phones laptops in some ways regulate a human biological clock. But among several factors,

light is the major factor in the regulation of circadian rhythm in the mammalian body. The

PT
mper1 is a gene sequence present in mammals which is homologous to the per gene in

Drosophila and for genes in Neurospora. The per and freq are clock genes in Drosophila and

RI
Neurospora. For the determination expression profile of mper1 and its potential role, light

treatment was used. The light treatment helped to reset the circadian clock. The reaction of

SC
circadian rhythm towards light is used as a mechanistic process to determine the circadian

pacemaker and the organization of the circadian system. The expression profile of mper1

U
peaks during daytime and lowers during the night. This expression profile suggests
AN
dependency of mper1 on the daytime light [29,118,148–155] (Fig. 5).
M

The clock gene expression is thought to be a culmination of molecular mechanisms

operating under the influence of light that translate mRNA to a functional regulatory protein.
D

Light acts as the chief modulator and cue for the entrainment of the biological clock. The
TE

ambient light/dark cycle intricately regulates the phases of the SCN. There is a very fast

entrainment in the presence of transient light exposure at night [156]. Light stimulates the
EP

retinal ganglion cells that express melanopsin and release the neurotransmitter at the

retinohypothalamic tract terminals [156,157], which binds to the postsynaptic receptor. Then
C

they activate the second messenger-based signal transduction pathway in the SCN. Light
AC

cues, when perceived by the central oscillator, cause the stimulation of the mitogen and

stress-activated kinases that activate CREB. CREB is responsible for the expression of

genes with CREB in the promoters such as c-Fos and Per1 [158]. The mRNA translation

occurs mainly at the initiation step [159] with the CREB co-activator, CRTC [160,161].

Translation initiates when the m7GpppN structure present at 5′-end gets recognized by the

eukaryotic translation initiation factor 4E [162], where eIF4E binds the complex of eIF4F
ACCEPTED MANUSCRIPT
having eIF4A and eIF4G [159,163]. eIF4E acts as the rate-limiting translation factor whose

presence in abundance occurs due to the presence of mTOR protein. The mTOR obstructs

binding of eIF4E to eIF4G, hence the mRNA translation is obstructed [159,163].The activity

of the eIF4E gets modified by the p38 and p42/44 MAP kinases through phosphorylation at a

single amino acid, Ser209 [164]. The MAPK interacts with the MAPK-interacting

PT
serine/threonine kinases such as MNK1 and MNK2 [165].

A recent study has exposed that MAPK/MNK phosphorylation of the cap-binding protein,

RI
eIF4E, that in turn regulates the translational initiation of the mRNA of the clock proteins

PER1 and PER2. This phosphorylation is intricately regulated in the SCN by light and shows

SC
a constant circadian rhythm. In situations demanding an upregulated PER, eIF4E gets

phosphorylated to trigger the Per protein; but under constant environmental light conditions,

U
eIF4E just timely modulates the PER synthesis.
AN
An unprecedented increase in the PER leads to the disruption of the dynamic balance of the

feedback loops and causes the resetting of the phase of the SCN clock [157]. In this way,
M

the activity of eIF4E is highly regulated to ensure a well-timed protein synthesis and normal
D

functioning of the circadian clock.


TE

A common link in the clock control process between plant and animals
EP

The circadian clock generates certain kinds of oscillations known as circadian rhythm.

Circadian rhythm is found in all kinds of organisms starting from Cyanobacteria, Neurospora,
C

plants, mammals, and other animals. In plants, there are three transcriptional loops present
AC

in the plant circadian clock. PSEUDO-RESPONSE REGULATOR 1 (PRR1) or TOC 1 is the

clock regulatory gene, Circadian Clock associated 1 (CCA1) and LATE ELONGATED

HYPOCOTYL (LHL). CCA1 and LHL bind to TOC 1 and block its transcription. TOC 1 is

hypothesized to activate an unknown compound X. It has been considered that there may be

other evening-faced genes. In the case of animals, SCN is an important component of the

circadian clock. It is considered to be a pacemaker of the circadian clock. The mammalian

circadian oscillation requires both negative and positive elements. The negative elements
ACCEPTED MANUSCRIPT
are per 1, per 2 genes and cry 1 and cry 2. The positive-acting proteins are Bmal 1 and clock

genes. In plants, circadian rhythm is essential for maintaining the balance of the whole

system. Circadian rhythm in plants helps plants to get accustomed to seasonal changes,

enzymatic activity, stomatal changes, gas exchange, germination, pollination, leaf

movement, photosynthetic activity, fragrance emission, and growth. It also helps in

PT
synchronization to the light/dark cycle. In the case of animal circadian genes, per 1, per 2,

cry 1, and cry 2 are homologous to the plant circadian rhythm. The function of both genes is

RI
to maintain circadian oscillation and thus help in the correct running of the biological clock

[55,77,166–171].

SC
From bacteria to humans

U
In the case of cyanobacteria, kaiA, kaiB, and kaiC are important in the regulation of circadian
AN
oscillation. These three genes are very important for circadian oscillation. Deletion of any of

the genes will result in arrhythmicity of the system. Kai BC complex and Kai A transcript are
M

numerous. In the case of negative feedback regulation, Kai BC is repressed by


D

overexpression of KaiC. This results in the elimination of the loss of rhythmicity. On the other

hand, positive feedback regulation is found when Kai A overexpression activates Kai B C
TE

complex. In the case of mammals, period genes per 1, per 2, per3, bmal1, clock, and
EP

timeless (tim) genes are present. In the case of mammals, clock genes or bmal 1 genes

through the help of transcription binding factor, E-Box, which is located near the period gene,
C

activates per genes namely per 1, per 2, per 3. After the transcription of per 1, per 2, and per
AC

3, mRNAs are moved and are translated to PER 1, PER 2, and PER 3 proteins. CRY 1 and

CRY 2 are cryptochrome proteins and they act as a negative feedback loop. PER after

interaction with cryptochrome proteins translocate per proteins from the cytoplasm to the

nucleus. Then heterodimers of per and cry translocate back to the nucleus repressing their

own transcription by the complex formation of CLOCK and BMAL1. There is another

regulatory loop by a heterodimer of CLOCK and BMAL1. The retinoic acid-related orphan

nuclear receptor is activated for transcription. There is a competition between REV-ERBα


ACCEPTED MANUSCRIPT
and RORα to bind to the retinoic-acid-related orphan nuclear receptor. Bmal 1 is activated

for transcription by RORs whereas the transcription process is repressed by REV-ERBα.

ROR and REV-ERBα are both responsible for positive and negative regulation of the

circadian oscillation. Bacterial circadian rhythm is very important; circadian rhythm plays a

role in cell division and adaption to the environment. Similarly, in the case of humans,

PT
circadian rhythm carries out physiological work. It helps in creating a balance in the all the

physiological behavior, starting from the sleeping pattern, hormonal balance, feeding pattern,

RI
cell regeneration and other biological activities. In the case of both cyanobacteria and

humans, genes responsible for the circadian clock are homologous. Both in cyanobacteria

SC
and humans, circadian rhythm is responsible for maintaining balance in physiological

activities. There are differences in the structural organization of the genes, but they perform

U
the same task of maintaining circadian rhythm. The regulation of genes responsible for
AN
circadian rhythm in cyanobacteria and humans is also similar [12,172–178].
M

Bioinformatics outlook in the circadian evolutionary processes


D

Bioinformatics studies pertaining to circadian rhythm involve the use of an appropriate

experimental model that helps to organize information and helps to understand the
TE

hypothesis to be investigated. Such models might include a diagram correlating different


EP

genes and proteins or may be some mathematical equations. Models can be implemented

as contextual or conceptual; they may be represented as diagrams or in the form of


C

computations. Quantitatively they may be discrete, continuous, stochastic or deterministic


AC

[179]. A model to determine circadian attributes needs to have essentially the characteristics

of rhythmicity with a circadian period and should get compensated with temperature and

should be able to be entrained by external cues with amplitudes that can produce a

functional output and never get dampened under constant conditions. To date, various

models have been created to mimic the circadian clock in diverse species through

mathematical differential equations as in the case of mammals, such as mus musculus,

[142,180,181], for insects such as D. melanogaster [182–185], plants particularly, A. thaliana


ACCEPTED MANUSCRIPT
[186–188] Ostreococcus tauri [189,190], fungi (Neurospora crassa) [191–193], and

cyanobacteria [194,195]. The core components of the clock mechanism need to be set to

find out its properties such as the endogenous self-sustained oscillations, robustness,

cellular synchronization and ability to be entrained by external cues. This can be achieved by

setting up models whose other functions may be to understand the alterations in the

PT
properties of the clock and to correlate the situation with a de-regulated pathological

condition [142,196]. Furthermore, there are certain models that try to simulate the network of

RI
the circadian components at the level of the system using clock-controlled genes which react

to a circadian rhythm in a tissue-specific manner [197–199], hence they can be an answer to

SC
studying circadian effects in a specific tissue. It is important to understand the structure of

the gene regulatory network via the identification of various enhancer or repressor binding

U
sites at the promoter of the gene using ChIP-seq.
AN
A simple sequence or position weight matrix (PWM) can help in identifying motifs such as
M

the E-, D-boxes, and RREs, for the principal clock genes. Then this matrix can be matched

to the genomic positions and analyzed using statistics of occurrence of the specific gene
D

targeting the clock [200].The Goodwin oscillator represents the first biochemical negative-
TE

feedback oscillator model that interconnects three components with rates of production and

degradation [201]. Coupled oscillators represent the focus on different influences on the
EP

different clock components [202]. A mathematical description of the circadian system may be

obtained using the conventional ordinary differential equations (ODEs), gene expression
C

changes over time [180,181,203], or the delayed differential equations (DDEs) to explicitly
AC

express time [183,204]. The latter has been found to be useful in overcoming the missing

factor and in simplifying the model [205]. Besides, Boolean models sometimes mimic the

basic properties of the core components of the clock in A. thaliana [206]. For the preparation

of a mathematical model from data, one needs to overlook the experimental inaccuracies via

statistics [207].
ACCEPTED MANUSCRIPT
The changes in gene expression should be unfolded via luciferase assays [208,209],

producing data having direct or indirect information about the gene’s circadian phenotype.

Trigonometric functions can be commonly used, such as the one below, which can fit the

expression into a linear regression [210] corresponding to the phase shifts in each of its

peaks.

PT
RI
The circadian model is primarily the consolidation of the network structure with an additional

SC
data on time-series. The network structure designs a set of the equation and the time series

determines the values of the model such as the rate of inhibition of the proteins or RNAs or

U
any other experimental parameters. Large and complex mathematical models are usually
AN
optimized using a global optimization algorithm like the Evolutionary Algorithms [211] or

approaches based on the Markov Chain Monte Carlo (MCMC) sampling, like the simulated
M

annealing [212]. COSOPT is an algorithm that helps to incorporate the dampening factor and
D

provides necessary information about the periods or the amplitude [213]. A database called
TE

CircaDB [214] has been found to be instrumental in annotating the expression of several

circadian genes with time. Alternately, one can use the maSigPro package meant for
EP

Bioconductor to understand the differential expression of the genes with time. The online

service provider BioDare is useful for recognizing the period of the circadian time [215].
C

Analysis of the bifurcation diagrams identifies key parameters pertaining to a particular


AC

circadian phenotype that mimics a pathological condition, and hence identifies the genes or

proteins that disrupt the biological clock and hence find out its role in the disease

progression.

Systems biology methods involving analyzing and integrating the different types of large-

scale data such as those of gene co-expression, ChIP-seq data, protein–protein interactions,

GO terms and analysis of the KEGG pathways [216,217] are preferred for understanding the

relation of the clock-controlled genes to a larger network of the similarly functioning genes
ACCEPTED MANUSCRIPT
[218–220]. Hence, an integration of such interdisciplinary approaches may furnish newer

strategies in the field of chemotherapeutic treatment and may shed abundant light in

unleashing factors that will restore the circadian rhythm timing in various diseases due to

perturbed clock factors.

PT
Conclusion

RI
Taken together, the diversity of hierarchical mechanisms of an individual organism is

impressive, tightly organized, but limited to their normal sleep/wake distribution. The

SC
machinery can be compared to an orchestra’s conductor. The circadian rhythms combat and

overcome many gestures to set the timing of the sleep/wake cycle of individual musicians

U
playing their own instruments. However, to overcome sleeping disorders, the success of the
AN
circadian symphony not only depends on the role of the conductor but also relies on the

individual organisms as musicians to play their roles.


M

Remarkable progress in the field of circadian rhythms or sleep/wake cycles has been made
D

in recent decades. The identification of many key genes and their roles in sleep/wake
TE

balance and the discovery of the roles in peripheral oscillators have advanced our

understanding, but a clear picture of the circadian timing in various organisms is far from
EP

complete. We speculate that the hierarchical evolution and inventory of clock factors have

just begun. The present report is in support of this, but numerous clock components still
C

remain to be identified in the future.


AC

The mechanism of circadian rhythm in the fly provides an excellent opportunity as a model

organism, and can promote the understanding of a complex mechanism such as the

sleep/wake rhythm in human. In many organisms, sleep disorders should focus on

interconnecting the central oscillation to sleep behaviors and physiology. The cyclic rhythmic

transcripts and their integration with diversified related protein molecule and small molecule
ACCEPTED MANUSCRIPT
rhythm might provide cutting edge technological emerge. These complete pictures in the

different organism might show an overall picture of sleep behavior and physiology.

In addition, we pointed out that the relationship or coupling of clock protein and specific

signals cascade illustrated the different molecular mechanisms of circadian rhythm. The

endeavors between physiology and circadian behaviors will require the applicability of many

PT
genetic tools, genomics, proteomics, physiology, and behavior. In fact, in different

organisms, major attraction lies in multidisciplinary events, and it is speculated that future

RI
experiments for illustrating circadian rhythm in different organisms will not only bring us

SC
enormous exciting surprises, but will also reward us with immense adventures.

Conflict of interest

U
The authors declare that they have no competing interests.
AN
Acknowledgements
M

We greatly acknowledge the discussion and technical assistance provided by all lab
D

members. The work was supported by an HFSP Young Investigator Grant (RGY020),

Wellcome trust fellowship (GP0145) awarded to UB. Wellcome Trust International


TE

Fellowship was awarded to MPB (GAP0065). We dedicate this manuscript to the memory of
EP

a renowned physician in Calcutta, Dr Malaya Kr. Pal, who inspired us in Science.


C

References
AC

[1] Glansdorff N, Xu Y, Labedan B. The last universal common ancestor: emergence, constitution and

genetic legacy of an elusive forerunner. Biol Direct 2008;3:1.

[2] Edgar RS, Green EW, Zhao Y, van Ooijen G, Olmedo M, Qin X, et al. Peroxiredoxins are conserved

markers of circadian rhythms. Nature 2012;485:459-64.

[3] Panda S, Hogenesch JB, Kay SA. Circadian rhythms from flies to human. Nature 2002;417:329-35.

[4] Harmer SL. The circadian system in higher plants. Annu Rev Plant Biol 2009;60:357-77.
ACCEPTED MANUSCRIPT
[5] Brunner M, Schafmeier T. Transcriptional and post-transcriptional regulation of the circadian

clock of cyanobacteria and Neurospora. Genes Dev 2006;20:1061-74.

[6] Bell-Pedersen D, Cassone VM, Earnest DJ, Golden SS, Hardin PE, Thomas TL, et al. Circadian

rhythms from multiple oscillators: lessons from diverse organisms. Nat Rev Genet 2005;6:544-56.

[7] Schibler U, Sassone-Corsi P. A web of circadian pacemakers. Cell 2002;111:919-22.

PT
[8] Whitmore D, Foulkes NS, Sassone-Corsi P. Light acts directly on organs and cells in culture to set

the vertebrate circadian clock. Nature 2000;404:87-91.

RI
[9] Giebultowicz JM. Peripheral clocks and their role in circadian timing: insights from insects. Phil

SC
Trans R Soc Lond B Biol Sci 2001;356:1791-9.

[10] Sehgal A. Molecular Biology of Circadian Rhythms. Hoboken, N.J.: Wiley–Liss; 2004.

U
[11] Kurosawa G, Aihara K, Iwasa Y. A model for the circadian rhythm of cyanobacteria that
AN
maintains oscillation without gene expression. Biophys J 2006;91:2015-23.

[12] Ishiura M, Kutsuna S, Aoki S, Iwasaki H, Andersson CR, Tanabe A, et al. Expression of a gene
M

cluster kaiABC as a circadian feedback process in cyanobacteria. Science 1998;281:1519-23.


D

[13] Nishiwaki T, Satomi Y, Nakajima M, Lee C, Kiyohara R, Kageyama H, et al. Role of KaiC
TE

phosphorylation in the circadian clock system of Synechococcus elongatus PCC 7942. Proc Natl Acad

Sci U S A 2004;101:13927-32.
EP

[14] Tomita JT, Feigen GA. Serological identification and physical-chemical properties of the non-

spasmogenic principle (NSP) in tetanus toxin. Immunochemistry 1969;6:421-35.


C

[15] Tomita J, Nakajima M, Kondo T, Iwasaki H. No transcription-translation feedback in circadian


AC

rhythm of KaiC phosphorylation. Science 2005;307:251-4.

[16] Nakajima M, Imai K, Ito H, Nishiwaki T, Murayama Y, Iwasaki H, et al. Reconstitution of circadian

oscillation of cyanobacterial KaiC phosphorylation in vitro. Science 2005;308:414-5.

[17] Golden SS, Ishiura M, Johnson CH, Kondo T. Cyanobacterial circadian rhythms. Annu Rev Plant

Biol 1997;48:327-54.
ACCEPTED MANUSCRIPT
[18] Rust MJ, Golden SS, O'Shea EK. Light-driven changes in energy metabolism directly entrain the

cyanobacterial circadian oscillator. Science. 2011;331:220-3.

[19] Boyd JS, Bordowitz JR, Bree AC, Golden SS. An allele of the crm gene blocks cyanobacterial

circadian rhythms. Proc Natl Acad Sci U S A 2013;110:13950-5.

[20] Ashby MK, Mullineaux CW. Cyanobacterial ycf27 gene products regulate energy transfer from

PT
phycobilisomes to photosystems I and II. FEMS Microbiol Lett 1999;181:253-60.

[21] Kappell AD, van Waasbergen LG. The response regulator RpaB binds the high light regulatory 1

RI
sequence upstream of the high-light-inducible hliB gene from the cyanobacterium Synechocystis PCC

SC
6803. Arch Microbiol 2007;187:337-42.

[22] López-Redondo ML, Moronta F, Salinas P, Espinosa J, Cantos R, Dixon R, et al. Environmental

U
control of phosphorylation pathways in a branched two-component system. Molec Microbiol
AN
2010;78:475-89.

[23] van Waasbergen LG, Dolganov N, Grossman AR. nblS, a gene involved in controlling
M

photosynthesis-related gene expression during high light and nutrient stress in Synechococcus
D

elongatus PCC 7942. J Bacteriol 2002;184:2481-90.


TE

[24] Hanaoka M, Tanaka K. Dynamics of RpaB–promoter interaction during high light stress, revealed

by chromatin immunoprecipitation (ChIP) analysis in Synechococcus elongatus PCC 7942. Plant J


EP

2008;56:327-35.

[25] Espinosa J, Boyd JS, Cantos R, Salinas P, Golden SS, Contreras A. Cross-talk and regulatory
C

interactions between the essential response regulator RpaB and cyanobacterial circadian clock
AC

output. Proc Natl Acad Sci U S A 2015;112:2198-203.

[26] Hall A, Karplus PA, Poole LB. Typical 2-Cys peroxiredoxins – structures, mechanisms and

functions. FEBS J 2009;276:2469-77.

[27] Schreiber W, Stuehlinger HG, Brunner M, Hollenstein U. Preclinical management of

thromboembolic disorders. Semin Thromb Hemost 1996;22:3-13.


ACCEPTED MANUSCRIPT
[28] O'Neill JS, Van Ooijen G, Dixon LE, Troein C, Corellou F, Bouget F-Y, et al. Circadian rhythms

persist without transcription in a eukaryote. Nature 2011;469:554-8.

[29] Dunlap JC. Molecular bases for circadian clocks. Cell 1999;96:271-90.

[30] Cheng P, He Q, He Q, Wang L, Liu Y. Regulation of the Neurospora circadian clock by an RNA

helicase. Genes Dev 2005;19:234-41.

PT
[31] Liu Y, Bell-Pedersen D. Circadian rhythms in Neurospora crassa and other filamentous fungi.

Eukaryot Cell 2006;5:1184-93.

RI
[32] Aronson BD, Johnson KA, Loros JJ, Dunlap JC. Negative feedback defining a circadian clock:

SC
autoregulation of the clock gene frequency. Science 1994;263:1578-84.

[33] Schafmeier T, Haase A, Kaldi K, Scholz J, Fuchs M, Brunner M. Transcriptional feedback of

U
Neurospora circadian clock gene by phosphorylation-dependent inactivation of its transcription
AN
factor. Cell 2005;122:235-46.

[34] Baker CL, Kettenbach AN, Loros JJ, Gerber SA, Dunlap JC. Quantitative proteomics reveals a
M

dynamic interactome and phase-specific phosphorylation in the Neurospora circadian clock. Molec
D

Cell 2009;34:354-63.
TE

[35] Querfurth C, Diernfellner A, Heise F, Lauinger L, Neiss A, Tataroglu O, et al. Posttranslational

regulation of Neurospora circadian clock by CK1a-dependent phosphorylation. Cold Spring Harb


EP

Symp Quant Biol 2007;72:177-83.

[36] Querfurth C, Diernfellner AC, Gin E, Malzahn E, Höfer T, Brunner M. Circadian conformational
C

change of the Neurospora clock protein FREQUENCY triggered by clustered hyperphosphorylation of


AC

a basic domain. Molec Cell 2011;43:713-22.

[37] Tang C-T, Li S, Long C, Cha J, Huang G, Li L, et al. Setting the pace of the Neurospora circadian

clock by multiple independent FRQ phosphorylation events. Proc Natl Acad Sci U S A

2009;106:10722-7.

[38] Bell-Pedersen D. Circadian rhythms in Neurospora crassa. Mycology Series 2002;15:187-214.


ACCEPTED MANUSCRIPT
[39] Shrode LB, Lewis ZA, White LD, Bell-Pedersen D, Ebbole DJ. vvd is required for light adaptation

of conidiation-specific genes of Neurospora crassa, but not circadian conidiation. Fungal Genet Biol

2001;32:169-81.

[40] Schneider K, Perrino S, Oelhafen K, Li S, Zatsepin A, Lakin-Thomas P, et al. Rhythmic conidiation

in constant light in vivid mutants of Neurospora crassa. Genetics 2009;181:917-31.

PT
[41] Lakin-Thomas PL, Coté GG, Brody S. Circadian rhythms in Neurospora crassa: biochemistry and

genetics. Crit Rev Microbiol 1990;17:365-416.

RI
[42] Feldman JF, Dunlap JC. Neurospora crassa: a unique system for studying circadian rhythms.

SC
Photochemical and Photobiological Reviews: Springer; 1983. pp. 319-68.

[43] Heintzen C, Liu Y. The Neurospora crassa circadian clock. Advances in genetics. 2007;58:25-66.

U
[44] Bell-Pedersen D. Understanding circadian rhythmicity in Neurospora crassa: from behavior to
AN
genes and back again. Fungal Genet Biol 2000;29:1-18.

[45] Gardner GF, Feldman JF. The frq locus in Neurospora crassa: a key element in circadian clock
M

organization. Genetics 1980;96:877-86.


D

[46] Lewis Z, Correa A, Schwerdtfeger C, Link K, Xie X, Gomer R, et al. Overexpression of White
TE

Collar-1 (WC-1) activates circadian clock-associated genes, but is not sufficient to induce most

light-regulated gene expression in Neurospora crassa. Molec Microbiol 2002;45:917-31.


EP

[47] Cumming BG, Wagner E. Rhythmic processes in plants. Annu Rev Plant Physiol 1968;19:381-416.

[48] Kloppstech K. Diurnal and circadian rhythmicity in the expression of light-induced plant nuclear
C

messenger RNAs. Planta 1985;165:502-6.


AC

[49] Nagy F, Kay SA, Chua N-H. A circadian clock regulates transcription of the wheat Cab-1 gene.

Genes Dev 1988;2:376-82.

[50] Somerville C, Koornneef M. A fortunate choice: the history of Arabidopsis as a model plant. Nat

Rev Genet 2002;3:883-9.

[51] Millar AJ, Kay SA. Circadian control of cab gene transcription and mRNA accumulation in

Arabidopsis. Plant Cell 1991;3:541-50.


ACCEPTED MANUSCRIPT
[52] Dowson-Day MJ, Millar AJ. Circadian dysfunction causes aberrant hypocotyl elongation patterns

in Arabidopsis. Plant J 1999;17:63-71.

[53] Jouve L, Greppin H, Degli Agosti R. Arabidopsis thaliana floral stem elongation: evidence for an

endogenous circadian rhythm. Plant Physiol Biochem 1998;36:469-72.

[54] Initiative AG. Analysis of the genome sequence of the flowering plant Arabidopsis thaliana.

PT
Nature 2000;408:796.

[55] Alabadı ́ D, Oyama T, Yanovsky MJ, Harmon FG, Más P, Kay SA. Reciprocal regulation between

RI
TOC1 and LHY/CCA1 within the Arabidopsis circadian clock. Science 2001;293:880-3.

SC
[56] Green RM, Tobin EM. The role of CCA1 and LHY in the plant circadian clock. Dev Cell 2002;2:516-

8.

U
[57] Gendron JM, Pruneda-Paz JL, Doherty CJ, Gross AM, Kang SE, Kay SA. Arabidopsis circadian clock
AN
protein, TOC1, is a DNA-binding transcription factor. Proc Natl Acad Sci U S A 2012;109:3167-72.

[58] Tiwari SB, Shen Y, Chang HC, Hou Y, Harris A, Ma SF, et al. The flowering time regulator
M

CONSTANS is recruited to the FLOWERING LOCUS T promoter via a unique cis-element. New
D

Phytologist 2010;187:57-66.
TE

[59] Pokhilko A, Fernández AP, Edwards KD, Southern MM, Halliday KJ, Millar AJ. The clock gene

circuit in Arabidopsis includes a repressilator with additional feedback loops. Mol Syst Biol
EP

2012;8:574.

[60] Mehta N, Cheng H-YM. Micro-managing the circadian clock: The role of microRNAs in biological
C

timekeeping. J Molec Biol 2013;425:3609-24.


AC

[61] Farré EM, Harmer SL, Harmon FG, Yanovsky MJ, Kay SA. Overlapping and distinct roles of PRR7

and PRR9 in the Arabidopsis circadian clock. Curr Biol 2005;15:47-54.

[62] Nakamichi N, Kiba T, Henriques R, Mizuno T, Chua N-H, Sakakibara H. PSEUDO-RESPONSE

REGULATORS 9, 7, and 5 are transcriptional repressors in the Arabidopsis circadian clock. Plant Cell

2010;22:594-605.
ACCEPTED MANUSCRIPT
[63] Mas P, Yanovsky MJ. Time for circadian rhythms: plants get synchronized. Curr Opin Plant Biol

2009;12:574-9.

[64] Pruneda-Paz JL, Kay SA. An expanding universe of circadian networks in higher plants. Trends

Plant Sci 2010;15:259-65.

[65] Helfer A, Nusinow DA, Chow BY, Gehrke AR, Bulyk ML, Kay SA. LUX ARRHYTHMO encodes a

PT
nighttime repressor of circadian gene expression in the Arabidopsis core clock. Curr Biol

2011;21:126-33.

RI
[66] Nusinow DA, Helfer A, Hamilton EE, King JJ, Imaizumi T, Schultz TF, et al. The ELF4-ELF3-LUX

SC
complex links the circadian clock to diurnal control of hypocotyl growth. Nature 2011;475:398-402.

[67] Herrero E, Kolmos E, Bujdoso N, Yuan Y, Wang M, Berns MC, et al. EARLY FLOWERING4

U
recruitment of EARLY FLOWERING3 in the nucleus sustains the Arabidopsis circadian clock. Plant Cell
AN
2012;24:428-43.

[68] Hicks KA, Millar AJ, Carré IA, Somers DE. Conditional circadian dysfunction of the Arabidopsis
M

early-flowering 3 mutant. Science 1996;274:790.


D

[69] Harmer SL, Hogenesch JB, Straume M, Chang H-S, Han B, Zhu T, et al. Orchestrated transcription
TE

of key pathways in Arabidopsis by the circadian clock. Science 2000;290:2110-3.

[70] Sugano S, Andronis C, Ong MS, Green RM, Tobin EM. The protein kinase CK2 is involved in
EP

regulation of circadian rhythms in Arabidopsis. Proc Natl Acad Sci U S A 1999;96:12362-6.

[71] Somers DE, Devlin PF, Kay SA. Phytochromes and cryptochromes in the entrainment of the
C

Arabidopsis circadian clock. Science 1998;282:1488-90.


AC

[72] Nelson DC, Lasswell J, Rogg LE, Cohen MA, Bartel B. FKF1, a clock-controlled gene that regulates

the transition to flowering in Arabidopsis. Cell 2000;101:331-40.

[73] Somers DE, Schultz TF, Milnamow M, Kay SA. ZEITLUPE encodes a novel clock-associated PAS

protein from Arabidopsis. Cell 2000;101:319-29.

[74] Schultz TF, Kiyosue T, Yanovsky M, Wada M, Kay SA. A role for LKP2 in the circadian clock of

Arabidopsis. Plant Cell 2001;13:2659-70.


ACCEPTED MANUSCRIPT
[75] Somers DE, Kim W-Y, Geng R. The F-box protein ZEITLUPE confers dosage-dependent control on

the circadian clock, photomorphogenesis, and flowering time. Plant Cell 2004;16:769-82.

[76] Dodd AN, Parkinson K, Webb AA. Independent circadian regulation of assimilation and stomatal

conductance in the ztl-1 mutant of Arabidopsis. New Phytologist 2004;162:63-70.

[77] Dodd AN, Salathia N, Hall A, Kévei E, Tóth R, Nagy F, et al. Plant circadian clocks increase

PT
photosynthesis, growth, survival, and competitive advantage. Science 2005;309:630-3.

[78] Yanovsky MJ, Kay SA. Molecular basis of seasonal time measurement in Arabidopsis. Nature

RI
2002;419:308-12.

SC
[79] Sang JH. Drosophila melanogaster: the fruit fly. Encyclopedia of Genetics 2001:157-62.

[80] Konopka RJ, Benzer S. Clock mutants of Drosophila melanogaster. Proc Natl Acad Sci U S A

U
1971;68:2112-6.
AN
[81] Pittendrigh CS, Daan S. A functional analysis of circadian pacemakers in nocturnal rodents. J

Comp Physiol 1976;106:223-52.


M

[82] Peschel N, Helfrich-Förster C. Setting the clock–by nature: circadian rhythm in the fruitfly
D

Drosophila melanogaster. FEBS Lett 2011;585:1435-42.


TE

[83] Aschoff J. Circadian activity pattern with two peaks. Ecology 1966;47:657-62.

[84] Shaw PJ, Cirelli C, Greenspan RJ, Tononi G. Correlates of sleep and waking in Drosophila
EP

melanogaster. Science 2000;287:1834-7.

[85] Tononi G, Cirelli C, Shaw PJ, Greenspan RJ. Methods for identifying compounds that modulate
C

vigilance states. Google Patents; 2004.


AC

[86] Gilestro GF. Video tracking and analysis of sleep in Drosophila melanogaster. Nature Protocols

2012;7:995-1007.

[87] Wu MN, Ho K, Crocker A, Yue Z, Koh K, Sehgal A. The effects of caffeine on sleep in Drosophila

require PKA activity, but not the adenosine receptor. J Neurosci 2009;29:11029-37.

[88] Chatterjee A, Tanoue S, Houl JH, Hardin PE. Regulation of gustatory physiology and appetitive

behavior by the Drosophila circadian clock. Curr Biol 2010;20:300-9.


ACCEPTED MANUSCRIPT
[89] Hendricks JC, Finn SM, Panckeri KA, Chavkin J, Williams JA, Sehgal A, et al. Rest in Drosophila is a

sleep-like state. Neuron 2000;25:129-38.

[90] Wager-Smith K, Kay SA. Circadian rhythm genetics: from flies to mice to humans. Nature Genet

2000;26:23-7.

[91] Emery P, So WV, Kaneko M, Hall JC, Rosbash M. CRY, a Drosophila clock and light-regulated

PT
cryptochrome, is a major contributor to circadian rhythm resetting and photosensitivity. Cell

1998;95:669-79.

RI
[92] Price JL, Blau J, Rothenfluh A, Abodeely M, Kloss B, Young MW. double-time is a novel

SC
Drosophila clock gene that regulates PERIOD protein accumulation. Cell 1998;94:83-95.

[93] Kloss B, Price JL, Saez L, Blau J, Rothenfluh A, Wesley CS, et al. The Drosophila clock gene

U
double-time encodes a protein closely related to human casein kinase Iε. Cell 1998;94:97-107.
AN
[94] Lee C, Parikh V, Itsukaichi T, Bae K, Edery I. Resetting the Drosophila clock by photic regulation

of PER and a PER-TIM complex. Science 1996;271:1740.


M

[95] Saez L, Young MW. Regulation of nuclear entry of the Drosophila clock proteins period and
D

timeless. Neuron 1996;17:911-20.


TE

[96] Lee C, Bae K, Edery I. The Drosophila CLOCK protein undergoes daily rhythms in abundance,

phosphorylation, and interactions with the PER–TIM complex. Neuron 1998;21:857-67.


EP

[97] Rosato E, Codd V, Mazzotta G, Piccin A, Zordan M, Costa R, et al. Light-dependent interaction

between Drosophila CRY and the clock protein PER mediated by the carboxy terminus of CRY. Curr
C

Biol 2001;11:909-17.
AC

[98] Baylies MK, Vosshall LB, Sehgal A, Young MW. New short period mutations of the Drosophila

clock gene per. Neuron 1992;9:575-81.

[99] Konopka R, Wells S, Lee T. Mosaic analysis of a Drosophila clock mutant. Molec Gen Genet

1983;190:284-8.

[100] Glossop NR, Lyons LC, Hardin PE. Interlocked feedback loops within the Drosophila circadian

oscillator. Science 1999;286:766-8.


ACCEPTED MANUSCRIPT
[101] Allada R, White NE, So WV, Hall JC, Rosbash M. A mutant Drosophila homolog of mammalian

Clock disrupts circadian rhythms and transcription of period and timeless. Cell 1998;93:791-804.

[102] Darlington TK, Wager-Smith K, Ceriani MF, Staknis D, Gekakis N, Steeves TD, et al. Closing the

circadian loop: CLOCK-induced transcription of its own inhibitors per and tim. Science

1998;280:1599-603.

PT
[103] Rutila JE, Suri V, Le M, So WV, Rosbash M, Hall JC. CYCLE is a second bHLH-PAS clock protein

essential for circadian rhythmicity and transcription of Drosophila period and timeless. Cell

RI
1998;93:805-14.

SC
[104] Hamblen MJ, White NE, Emery PT, Kaiser K, Hall JC. Molecular and behavioral analysis of four

period mutants in Drosophila melanogaster encompassing extreme short, novel long, and

U
unorthodox arrhythmic types. Genetics 1998;149:165-78.
AN
[105] Kim EY, Bae K, Ng FS, Glossop NR, Hardin PE, Edery I. Drosophila CLOCK protein is under

posttranscriptional control and influences light-induced activity. Neuron 2002;34:69-81.


M

[106] Krasinsky GA, Brumberg VA. Secular increase of astronomical unit from analysis of the major
D

planet motions, and its interpretation. Celest Mech Dynam Astron 2004;90:267-88.
TE

[107] Ouyang Y, Andersson CR, Kondo T, Golden SS, Johnson CH. Resonating circadian clocks

enhance fitness in cyanobacteria. Proc Natl Acad Sci U S A 1998;95:8660-4.


EP

[108] Shearman LP, Sriram S, Weaver DR, Maywood ES, Chaves I, Zheng B, et al. Interacting

molecular loops in the mammalian circadian clock. Science 2000;288:1013-9.


C

[109] Trebing D, Brunner M, Kroning Y, Goring HD. [Young man with verrucous heel tumor ]. J Dtsch
AC

Dermatol Ges 2003;1:739-41.

[110] Panda S, Provencio I, Tu DC, Pires SS, Rollag MD, Castrucci AM, et al. Melanopsin is required for

non-image-forming photic responses in blind mice. Science 2003;301:525-7.

[111] Hattar S, Lucas RJ, Mrosovsky N, Thompson S, Douglas R, Hankins MW, et al. Melanopsin and

rod–cone photoreceptive systems account for all major accessory visual functions in mice. Nature

2003;424:75-81.
ACCEPTED MANUSCRIPT
[112] Working PK, Newman MS, Sullivan T, Brunner M, Podell M, Sahenk Z, et al. Comparative

intravenous toxicity of cisplatin solution and cisplatin encapsulated in long-circulating, pegylated

liposomes in cynomolgus monkeys. Toxicol Sci 1998;46:155-65.

[113] Cashmore AR. Cryptochromes: enabling plants and animals to determine circadian time. Cell

2003;114:537-43.

PT
[114] Lin C, Shalitin D. Cryptochrome structure and signal transduction. Annu Rev Plant Biol

2003;54:469-96.

RI
[115] Yang B, Kallio H. Composition and physiological effects of sea buckthorn (Hippophae) lipids.

SC
Trends Food Sci Technol 2002;13:160-7.

[116] Zhu X, Lafferty J, Ghahramani Z. Combining active learning and semi-supervised learning using

U
gaussian fields and harmonic functions. ICML 2003 workshop on the continuum from labeled to
AN
unlabeled data in machine learning and data mining 2003.

[117] Kapoor M, Lozano G. Functional activation of p53 via phosphorylation following DNA damage
M

by UV but not γ radiation. Proc Natl Acad Sci U S A 1998;95:2834-7.


D

[118] Lowrey PL, Takahashi JS. Mammalian circadian biology: elucidating genome-wide levels of
TE

temporal organization. Annu Rev Genomics Hum Genet 2004;5:407.

[119] Akten B, Jauch E, Genova GK, Kim EY, Edery I, Raabe T, et al. A role for CK2 in the Drosophila
EP

circadian oscillator. Nature Neurosci 2003;6:251-7.

[120] Ohno S. Gene duplication and the uniqueness of vertebrate genomes circa 1970–1999.
C

Seminars in cell & developmental biology: Elsevier; 1999. p. 517-22.


AC

[121] Zylka MJ, Shearman LP, Weaver DR, Reppert SM. Three period homologs in mammals:

differential light responses in the suprachiasmatic circadian clock and oscillating transcripts outside

of brain. Neuron 1998;20:1103-10.

[122] Yoshimura T, Suzuki Y, Makino E, Suzuki T, Kuroiwa A, Matsuda Y, et al. Molecular analysis of

avian circadian clock genes. Molec Brain Res 2000;78:207-15.


ACCEPTED MANUSCRIPT
[123] Benna C, Scannapieco P, Piccin A, Sandrelli F, Zordan M, Rosato E, et al. A second timeless gene

in Drosophila shares greater sequence similarity with mammalian tim. Curr Biol 2000;10:R512-R3.

[124] Clayton JD, Kyriacou CP, Reppert SM. Keeping time with the human genome. Nature

2001;409:829-31.

[125] Gotter AL, Manganaro T, Weaver DR, Kolakowski LF, Possidente B, Sriram S, et al. A time-less

PT
function for mouse timeless. Nature Neurosci 2000;3:755.

[126] Barnes JW, Tischkau SA, Barnes JA, Mitchell JW, Burgoon PW, Hickok JR, et al. Requirement of

RI
mammalian Timeless for circadian rhythmicity. Science 2003;302:439-42.

SC
[127] Gotter AL, Levine JD, Reppert SM. Sex-linked period genes in the silkmoth, Antheraea pernyi:

implications for circadian clock regulation and the evolution of sex chromosomes. Neuron

U
1999;24:953-65.
AN
[128] Gotter AL, Reppert SM. Analysis of human Per4. Molec Brain Res 2001;92:19-26.

[129] Hardin PE. Transcription regulation within the circadian clock: the E-box and beyond. J Biol
M

Rhythms 2004;19:348-60.
D

[130] Grima B, Lamouroux A, Chelot E, Papin C, Limbourg-Bouchon B, Rouyer F. The F-box protein
TE

slimb controls the levels of clock proteins period and timeless. Nature 2002;420:178-82.

[131] Sathyanarayanan S, Zheng X, Xiao R, Sehgal A. Posttranslational regulation of Drosophila


EP

PERIOD protein by protein phosphatase 2A. Cell 2004;116:603-15.

[132] Curtin KD, Huang ZJ, Rosbash M. Temporally regulated nuclear entry of the Drosophila period
C

protein contributes to the circadian clock. Neuron 1995;14:365-72.


AC

[133] Shafer OT, Rosbash M, Truman JW. Sequential nuclear accumulation of the clock proteins

period and timeless in the pacemaker neurons of Drosophila melanogaster. J Neurosci

2002;22:5946-54.

[134] Lin J-M, Kilman VL, Keegan K, Paddock B, Emery-Le M, Rosbash M, et al. A role for casein

kinase 2α in the Drosophila circadian clock. Nature 2002;420:816-20.

[135] Aschoff J. Circadian control of body temperature. J Therm Biology. 1983;8:143-7.


ACCEPTED MANUSCRIPT
[136] Cameron MA, Barnard AR, Lucas RJ. The electroretinogram as a method for studying circadian

rhythms in the mammalian retina. J Genet 2008;87:459-66.

[137] Eckel-Mahan KL, Storm DR. Circadian rhythms and memory: not so simple as cogs and gears.

EMBO Rep 2009;10:584-91.

[138] Lowrey PL, Takahashi JS. Genetics of circadian rhythms in Mammalian model organisms. Adv

PT
Genet 2011;74:175.

[139] Bollinger T, Schibler U. Circadian rhythms-from genes to physiology and disease. Swiss Med

RI
Wkly 2014;144.

SC
[140] Cuninkova L, Brown SA. Peripheral circadian oscillators. Ann NY Acad Sci 2008;1129:358-70.

[141] Bell D, Go R, Miguel C, Parks W, Bryan J. Unequal treatment access and malaria risk in a

U
community-based intervention program in the Philippines. Southeast Asian J Trop Med Public Health
AN
2005;36:578-86.

[142] Relógio A, Westermark PO, Wallach T, Schellenberg K, Kramer A, Herzel H. Tuning the
M

mammalian circadian clock: robust synergy of two loops. PLoS Comput Biol 2011;7:e1002309.
D

[143] Zhang EE, Kay SA. Clocks not winding down: unravelling circadian networks. Nature reviews
TE

Molec Cell Biol 2010;11:764-76.

[144] Ueda HR, Hayashi S, Chen W, Sano M, Machida M, Shigeyoshi Y, et al. System-level
EP

identification of transcriptional circuits underlying mammalian circadian clocks. Nature Genet

2005;37:187-92.
C

[145] Jones RA, Panda SS, Hall CD. Quinine conjugates and quinine analogues as potential
AC

antimalarial agents. Eur J Med Chem 2015;97:335-55.

[146] O'Neill JS, Reddy AB. Circadian clocks in human red blood cells. Nature 2011;469:498-503.

[147] Herzog ED. Neurons and networks in daily rhythms. Nat Revs Neurosci 2007;8:790-802.

[148] Shigeyoshi Y, Taguchi K, Yamamoto S, Takekida S, Yan L, Tei H, et al. Light-Induced Resetting of

a Mammalian Circadian Clock Is Associated with Rapid Induction of the mPer1Transcript. Cell

1997;91:1043-53.
ACCEPTED MANUSCRIPT
[149] Partch CL, Green CB, Takahashi JS. Molecular architecture of the mammalian circadian clock.

Trends Cell Biol 2014;24:90-9.

[150] Mohawk JA, Takahashi JS. Cell autonomy and synchrony of suprachiasmatic nucleus circadian

oscillators. Trends Neurosci 2011;34:349-58.

[151] Dibner C, Schibler U, Albrecht U. The mammalian circadian timing system: organization and

PT
coordination of central and peripheral clocks. Annu Rev Physiol 2010;72:517-49.

[152] Ralph MR, Foster RG, Davis FC, Menaker M. Transplanted suprachiasmatic nucleus determines

RI
circadian period. Science 1990;247:975.

SC
[153] Stratmann M, Schibler U. Properties, entrainment, and physiological functions of mammalian

peripheral oscillators. J Biol Rhythms 2006;21:494-506.

U
[154] Cho H, Zhao X, Hatori M, Ruth TY, Barish GD, Lam MT, et al. Regulation of circadian behaviour
AN
and metabolism by REV-ERB-[agr] and REV-ERB-[bgr]. Nature 2012;485:123-7.

[155] Schmutz I, Ripperger JA, Baeriswyl-Aebischer S, Albrecht U. The mammalian clock component
M

PERIOD2 coordinates circadian output by interaction with nuclear receptors. Genes Dev
D

2010;24:345-57.
TE

[156] Peirson S, Foster RG. Melanopsin: another way of signaling light. Neuron 2006;49:331-9.

[157] Golombek DA, Rosenstein RE. Physiology of circadian entrainment. Physiol Rev 2010;90:1063-
EP

102.

[158] Travnickova-Bendova Z, Cermakian N, Reppert SM, Sassone-Corsi P. Bimodal regulation of


C

mPeriod promoters by CREB-dependent signaling and CLOCK/BMAL1 activity. Proc Natl Acad Sci U S
AC

A 2002;99:7728-33.

[159] Sonenberg N, Hinnebusch AG. Regulation of translation initiation in eukaryotes: mechanisms

and biological targets. Cell 2009;136:731-45.

[160] Jagannath A, Butler R, Godinho SI, Couch Y, Brown LA, Vasudevan SR, et al. The CRTC1-SIK1

pathway regulates entrainment of the circadian clock. Cell 2013;154:1100-11.


ACCEPTED MANUSCRIPT
[161] Sakamoto K, Norona FE, Alzate-Correa D, Scarberry D, Hoyt KR, Obrietan K. Clock and light

regulation of the CREB coactivator CRTC1 in the suprachiasmatic circadian clock. J Neurosci

2013;33:9021-7.

[162] Cao R, Butcher GQ, Karelina K, Arthur JS, Obrietan K. Mitogen-and stress-activated protein

kinase 1 modulates photic entrainment of the suprachiasmatic circadian clock. Eur J Neurosci

PT
2013;37:130-40.

[163] Gingras A-C, Raught B, Sonenberg N. eIF4 initiation factors: effectors of mRNA recruitment to

RI
ribosomes and regulators of translation. Annu Rev Biochem 1999;68:913-63.

SC
[164] Ueda T, Watanabe-Fukunaga R, Fukuyama H, Nagata S, Fukunaga R. Mnk2 and Mnk1 are

essential for constitutive and inducible phosphorylation of eukaryotic initiation factor 4E but not for

U
cell growth or development. Molecul Cell Biol 2004;24:6539-49.
AN
[165] Buxade M, Parra-Palau JL, Proud CG. The Mnks: MAP kinase-interacting kinases (MAP kinase

signal-integrating kinases). Front Biosci 2008;13:5359-73.


M

[166] McClung CR. Plant circadian rhythms. Plant Cell 2006;18:792-803.


D

[167] Covington MF, Panda S, Liu XL, Strayer CA, Wagner DR, Kay SA. ELF3 modulates resetting of the
TE

circadian clock in Arabidopsis. Plant Cell 2001;13:1305-16.

[168] Daniel X, Sugano S, Tobin EM. CK2 phosphorylation of CCA1 is necessary for its circadian
EP

oscillator function in Arabidopsis. Proc Natl Acad Sci U S A 2004;101:3292-7.

[169] Doyle MR, Davis SJ, Bastow RM, McWatters HG, Kozma-Bognár L, Nagy F, et al. The ELF4 gene
C

controls circadian rhythms and flowering time in Arabidopsis thaliana. Nature 2002;419:74-7.
AC

[170] Abe H, Honma S, Namihira M, Tanahashi Y, Ikeda M, Honma K-i. Circadian rhythm and light

responsiveness of BMAL1 expression, a partner of mammalian clock gene Clock, in the

suprachiasmatic nucleus of rats. Neurosci Lett 1998;258:93-6.

[171] Albrecht U, Sun ZS, Eichele G, Lee CC. A differential response of two putative mammalian

circadian regulators, mper1and mper2, to light. Cell 1997;91:1055-64.


ACCEPTED MANUSCRIPT
[172] Bunney WE, Bunney BG. Molecular clock genes in man and lower animals: possible

implications for circadian abnormalities in depression. Neuropsychopharmacology 2000;22:335-45.

[173] Griffin EA, Staknis D, Weitz CJ. Light-independent role of CRY1 and CRY2 in the mammalian

circadian clock. Science 1999;286:768-71.

[174] Koike N, Hida A, Numano R, Hirose M, Sakaki Y, Tei H. Identification of the mammalian

PT
homologues of the Drosophila timeless gene, Timeless1. FEBS Lett 1998;441:427-31.

[175] Moore RY. A clock for the ages. Science 1999;284:2102-3.

RI
[176] Kitayama Y, Iwasaki H, Nishiwaki T, Kondo T. KaiB functions as an attenuator of KaiC

SC
phosphorylation in the cyanobacterial circadian clock system. EMBO J 2003;22:2127-34.

[177] Iwasaki H, Nishiwaki T, Kitayama Y, Nakajima M, Kondo T. KaiA-stimulated KaiC

U
phosphorylation in circadian timing loops in cyanobacteria. Proc Natl Acad Sci U S A 2002;99:15788-
AN
93.

[178] Horne JA, Ostberg O. A self-assessment questionnaire to determine morningness-eveningness


M

in human circadian rhythms. Int J Chronobiol 1975;4:97-110.


D

[179] Roenneberg T, Chua EJ, Bernardo R, Mendoza E. Modelling biological rhythms. Curr Biol
TE

2008;18:R826-R35.

[180] Forger DB, Peskin CS. A detailed predictive model of the mammalian circadian clock. Proc Natl
EP

Acad Sci U S A 2003;100:14806-11.

[181] Leloup J-C, Goldbeter A. Toward a detailed computational model for the mammalian circadian
C

clock. Proc Natl Acad Sci U S A 2003;100:7051-6.


AC

[182] Nieto PS, Revelli JA, Garbarino-Pico E, Condat CA, Guido ME, Tamarit FA. Effects of different

per translational kinetics on the dynamics of a core circadian clock model. PloS One

2015;10:e0115067.

[183] Smolen P, Baxter DA, Byrne JH. A reduced model clarifies the role of feedback loops and time

delays in the Drosophila circadian oscillator. Biophys J 2002;83:2349-59.


ACCEPTED MANUSCRIPT
[184] Tyson JJ, Hong CI, Thron CD, Novak B. A simple model of circadian rhythms based on

dimerization and proteolysis of PER and TIM. Biophys J 1999;77:2411-7.

[185] Ueda HR, Hagiwara M, Kitano H. Robust oscillations within the interlocked feedback model of

Drosophila circadian rhythm. J Theor Biol 2001;210:401-6.

[186] Pokhilko A, Mas P, Millar AJ. Modelling the widespread effects of TOC1 signalling on the plant

PT
circadian clock and its outputs. BMC Syst Biol 2013;7:23.

[187] Locke JC, Kozma-Bognár L, Gould PD, Fehér B, Kevei E, Nagy F, et al. Experimental validation of

RI
a predicted feedback loop in the multi-oscillator clock of Arabidopsis thaliana. Molec Syst Biol

SC
2006;2:59.

[188] Zeilinger MN, Farré EM, Taylor SR, Kay SA, Doyle FJ. A novel computational model of the

U
circadian clock in Arabidopsis that incorporates PRR7 and PRR9. Molec Syst Biol 2006;2:58.
AN
[189] Thommen Q, Pfeuty B, Morant P-E, Corellou F, Bouget F-Y, Lefranc M. Robustness of circadian

clocks to daylight fluctuations: hints from the picoeucaryote Ostreococcus tauri. PLoS Comput Biol
M

2010;6:e1000990.
D

[190] Troein C, Corellou F, Dixon LE, van Ooijen G, O'Neill JS, Bouget FY, et al. Multiple light inputs to
TE

a simple clock circuit allow complex biological rhythms. Plant J 2011;66:375-85.

[191] Tseng Y-Y, Hunt SM, Heintzen C, Crosthwaite SK, Schwartz J-M. Comprehensive modelling of
EP

the Neurospora circadian clock and its temperature compensation. PLoS Comput Biol

2012;8:e1002437.
C

[192] Leloup J-C, Gonze D, Goldbeter A. Limit cycle models for circadian rhythms based on
AC

transcriptional regulation in Drosophila and Neurospora. J Biol Rhythm 1999;14:433-48.

[193] Ruoff P, Vinsjevik M, Monnerjahn C, Rensing L. The Goodwin oscillator: on the importance of

degradation reactions in the circadian clock. J Biol Rhythm 1999;14:469-79.

[194] Hatakeyama TS, Kaneko K. Generic temperature compensation of biological clocks by

autonomous regulation of catalyst concentration. Proc Natl Acad Sci U S A 2012;109:8109-14.


ACCEPTED MANUSCRIPT
[195] Clodong S, Dühring U, Kronk L, Wilde A, Axmann I, Herzel H, et al. Functioning and robustness

of a bacterial circadian clock. Molec Syst Biol 2007;3:90.

[196] Gallego M, Eide EJ, Woolf MF, Virshup DM, Forger DB. An opposite role for tau in circadian

rhythms revealed by mathematical modeling. Proc Natl Acad Sci U S A 2006;103:10618-23.

[197] Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogenesch JB. A circadian gene expression atlas

PT
in mammals: implications for biology and medicine. Proc Natl Acad Sci U S A 2014;111:16219-24.

[198] Panda S, Singh DV. Whole-Genome Sequences of Staphylococcus haemolyticus Isolated from

RI
Infected Eyes and Healthy Conjunctiva in Bhubaneswar, India. Genome Announc 2016;4.

SC
[199] Storch K-F, Lipan O, Leykin I, Viswanathan N, Davis FC, Wong WH, et al. Extensive and

divergent circadian gene expression in liver and heart. Nature 2002;417:78-83.

U
[200] Wasserman WW, Sandelin A. Applied bioinformatics for the identification of regulatory
AN
elements. Nat Rev Genet 2004;5:276-87.

[201] Goodwin BC. Oscillatory behavior in enzymatic control processes. Adv Enzyme Reg
M

1965;3:425IN1429IN3431-428IN2430IN6437.
D

[202] Achermann P, Kunz H. Modeling circadian rhythm generation in the suprachiasmatic nucleus
TE

with locally coupled self-sustained oscillators: phase shifts and phase response curves. J Biol Rhythm

1999;14:460-8.
EP

[203] Mirsky HP, Liu AC, Welsh DK, Kay SA, Doyle FJ. A model of the cell-autonomous mammalian

circadian clock. Proc Natl Acad Sci U S A 2009;106:11107-12.


C

[204] Korenčič A, Bordyugov G, Rozman D, Goličnik M, Herzel H. The interplay of cis-regulatory


AC

elements rules circadian rhythms in mouse liver. PloS One 2012;7:e46835.

[205] Baker CT, Paul CA, Willé DR. Issues in the numerical solution of evolutionary delay differential

equations. Adv Comput Math 1995;3:171-96.

[206] Akman OE, Watterson S, Parton A, Binns N, Millar AJ, Ghazal P. Digital clocks: simple Boolean

models can quantitatively describe circadian systems. J R Soc Interface 2012:rsif20120080.


ACCEPTED MANUSCRIPT
[207] Minors DS, Waterhouse JM. Mathematical and statistical analysis of circadian rhythms.

Psychoneuroendocrinology 1988;13:443-64.

[208] Meyer P, Saez L, Young MW. PER-TIM interactions in living Drosophila cells: an interval timer

for the circadian clock. Science 2006;311:226-9.

[209] Kondo T, Strayer CA, Kulkarni RD, Taylor W, Ishiura M, Golden SS, et al. Circadian rhythms in

PT
prokaryotes: luciferase as a reporter of circadian gene expression in cyanobacteria. Proc Natl Acad

Sci U S A 1993;90:5672-6.

RI
[210] Halberg F, Tong YL, Johnson E. Circadian system phase – an aspect of temporal morphology;

SC
procedures and illustrative examples. The Cellular Aspects of Biorhythms: Springer; 1967. p. 20-48.

[211] Bäck T, Schwefel H-P. An overview of evolutionary algorithms for parameter optimization. Evol

U
Comput 1993;1:1-23.
AN
[212] Kirkpatrick S, Gelatt C, Vecchi M. Optimization by simulated annealing. Science 1983; 220

(4598): 671–80.
M

[213] Straume M. DNA microarray time series analysis: automated statistical assessment of circadian
D

rhythms in gene expression patterning. Meth Enzymol 2004;383:149-66.


TE

[214] Pizarro A, Hayer K, Lahens NF, Hogenesch JB. CircaDB: a database of mammalian circadian

gene expression profiles. Nucleic Acids Res 2012:gks1161.


EP

[215] Zielinski T, Moore AM, Troup E, Halliday KJ, Millar AJ. Strengths and limitations of period

estimation methods for circadian data. PloS One 2014;9:e96462.


C

[216] Consortium GO. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res
AC

2004;32:D258-D61.

[217] Kanehisa M, Goto S, Kawashima S, Okuno Y, Hattori M. The KEGG resource for deciphering the

genome. Nucleic Acids Res 2004;32:D277-D80.

[218] Lehmann R, Childs L, Thomas P, Abreu M, Fuhr L, Herzel H, et al. Assembly of a comprehensive

regulatory network for the mammalian circadian clock: a bioinformatics approach. PloS One

2015;10:e0126283.
ACCEPTED MANUSCRIPT
[219] Wallach T, Schellenberg K, Maier B, Kalathur RKR, Porras P, Wanker EE, et al. Dynamic circadian

protein–protein interaction networks predict temporal organization of cellular functions. PLoS Genet

2013;9:e1003398.

[220] Bozek K, Relógio A, Kielbasa SM, Heine M, Dame C, Kramer A, et al. Regulation of clock-

controlled genes in mammals. PloS One 2009;4:e4882.

PT
RI
U SC
AN
M
D
TE
C EP
AC

Fig. 1. Phylogenetic tree showing simplest organisms to higher forms exhibiting circadian

regulation.

Fig. 2. Circadian feedback loops in plants.

Fig. 3. Molecular mechanism of Drosophila clock.


ACCEPTED MANUSCRIPT
Fig. 4. Interlocked feedback loops: light–temperature.

Fig. 5. Transcriptional–translational feedback loop in the mammalian clock.

PT
RI
U SC
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
Table 1. Fly Clock genes and proteins.

Gene Protein

period (per) RNA and protein cycle. Binds to TIM. Inhibits dCLK/CYC function.

RNA and protein cycle. Binds to PER and facilitates PER nuclear

timeless (tim) transport. Inhibits dCLK/CYC function. Degrades in response to

PT
light.

RI
Doubletime Constitutively expressed. Ser/Thr kinase (CK1ἐ). Phosphorylates

(dbt) TIM-free PER, promoting its degradation.

SC
RNA and protein cycle. bHLH–PAS protein. Heterodimerizes with
dClock (dClk)
CYC and promotes transcription from E-box.

U
Constitutively expressed. bHLH–PAS protein. Heterodimerizes
AN
cycle (cyc)
with dCLK and promotes transcription from E-box.

cryptochrome RNA cycles. Circadian photoreceptor. Promotes light-dependent


M

(cry) degradation of TIM. May be essential for some peripheral clocks.


D

RNA and protein cycle. bZIP transcription factor. May repress per
vrille
and tim transcription.
TE

Constitutively expressed. Ser/Thr kinase (GSK-3). Promotes TIM


shaggy
phosphorylation and nuclear localization of PER/TIM complex.
C EP
AC
ACCEPTED MANUSCRIPT
Table 2. Mammalian Clock genes and proteins.

Gene Protein
Period 1 (Per1) RNA and proteins cycle. Physically associates with CRY and among
Period 2 (Per2) PER proteins. Activator of BMAL1 function. (Mutation in Per2 is
Period 3 (Per3) associated with FASPS.)
Constitutively expressed. Closest Drosophila relative is timeout.
Timeless (Tim)
Homozygous null mutant is lethal, making it impossible to
conclusively establish clock function.

PT
Casein kinase 1ἐ Constitutively expressed. Protein kinase (CK1 ἐ). Phosphorylates
PER and affects PER stability.
Circadian locomotor

RI
output cycle kaput Constitutively expressed. Heterodimerizes with BMAL1 and binds to
E-box. Promotes transcription of Per and Cry.

SC
(Clock)

Bmal1/MOP3 RNA cycles. Heterodimerizes with CLOCK and binds to E-box.


Promotes transcription of Per and Cry.

U
Cryptochrome 1 AN
(Cry1)
RNA cycles. Mutations alter rhythmicity in mice, implying a central
oscillator function. Physically associates with and stabilizes PER.
Cryptochrome 2
Inhibits transcription of Per and Cry.
M

(Cry2)

Nfil3/E4BP4 In chicken, represses cPer expression78. In mouse, suppresses


D

mPer1 expression in cell-culture assays79


TE
C EP
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT
Highlights

• The genetic circuits regulating the pacemaker are conserved from bacteria to
humans.

• Light is the leading entrainment cue for all the circadian circuits across all taxa.

• Light entrains through peripheral oscillators in lower taxa and via pacemakers in the

PT
higher taxa.

• The per and Cry genes have extensively duplicated to show modulation of function

RI
across the taxa.

SC
The hierarchical mechanisms of an organism’s normal sleep/wake distribution are
intricately organized.

U
AN
M
D
TE
C EP
AC

You might also like