You are on page 1of 7

Review

Development of the Circadian Core


Machinery in Mammals

Yasuhiro Umemura and Kazuhiro Yagita


Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kyoto, 602-
8566, Japan

Correspondence to Kazuhiro Yagita: Fax: þ81 75-241-1499 kyagita@koto.kpu-m.ac.jp


https://doi.org/10.1016/j.jmb.2019.11.026
Edited by Achim Kramer

Abstract
The emergence of circadian molecular oscillation is observed as a gradual process during the development in
mammals. Pluripotent stem cell differentiation cultures recapitulate this process, whereas reprogramming into
an undifferentiated state reverses it. These findings indicate that the circadian clock is tightly coupled to the
state of cellular differentiation. The state of the circadian core machinery in nonrhythmic cells may be different
from that in rhythmic cells. In this review, we describe the circadian rhythm development during ontogeny in
mammals and focus on the molecular mechanisms that suppress circadian molecular oscillations during early
development and in pluripotent stem cells. We also discuss the biological implications of repressing cellular
circadian oscillation in nonrhythmic cells.
© 2020 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://crea
tivecommons.org/licenses/by-nc-nd/4.0/).

Introduction CRY in the nucleus and cytoplasm [5]. Once the


BMAL1eCLOCK heterodimer forms a complex with
The mammalian circadian clock regulates diverse PER and CRY proteins, it loses its transactivation
physiological functions in the whole body, such as activity. PERs and CRYs are reported to inhibit the
the sleepewake cycle, hormone release, body inducible gene expression by promoting the dis-
temperature, and metabolism. The central pace- sociation of the BMAL1eCLOCK heterodimers from
maker of the circadian clock is the suprachiasmatic DNA [6]. In addition, BMAL1eCLOCK heterodimers
nucleus (SCN) in the hypothalamus. The SCN tunes also induce the expression of Rev-erba (Nr1d1) and
the peripheral tissue clocks in anticipation of the Rev-erbb (Nr1d2), whose proteins repress Bmal1
cyclic environmental changes of the rotating earth. expression by binding to the RORE promoter
Each organ and also each cell, in addition to cultured element [7,8]. This process accrues the delay in
cell lines, are endowed with an intrinsic cellular gene expression of the clock components, and this
circadian clock [1e4]. The molecular principle of the delay in the TTFLs leads to circadian rhythmicity.
cellular circadian clock is driven by transcriptional/ Other proteins are involved in the regulation of this
translational feedback loops (TTFLs) composed of a process and have been featured in this special issue
subset of circadian clock genes. At the core of the and other reviews [9,10].
TTFLs are two transcriptional factors, BMAL1 (also
called ARNTL) and CLOCK. BMAL1 and CLOCK
form a heterodimer that actively works in the Circadian Development During
induction of gene transcription by binding to E-box Ontogenesis
enhancer elements. Their induced gene expression
includes their negative regulators Period (Per1, 2, 3) Although almost all somatic cells are equipped
and Cryptochrome (Cry1, 2). The core circadian with an intrinsic circadian clock, it has been reported
clock proteins assemble into a larger than 1-MDa that some cell types, including zygotes, the early
macromolecular complex, which include PER and embryo, and germline cells, do not have a robust

0022-2836/© 2020 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creati
vecommons.org/licenses/by-nc-nd/4.0/). Journal of Molecular Biology (2020) 432, 3611e3617
3612 Development of Circadian Machinery

circadian clock [11e13]. In recent decades, a the core circadian clock. The core circadian clock
gradual circadian development during ontogeny gene network regulates the diverse lineage- and
has been reported [14e17]. In the mouse, embryonic tissue-specific gene expression as a circadian out-
tissues, such as the heart and liver, show circadian put. Consequently, higher-order physiological func-
rhythms at around E13e18 [18e21]. Although the tions are acquired in the whole body. For example,
mouse SCN at E13.5 displays no circadian oscilla- the circadian clock in the mouse embryonic heart
tions, the circadian rhythm of the SCN develops at gradually develops from E10 after organogenesis,
E14.5 [22]. Human circadian rhythms also emerge and a circadian molecular oscillation is observed
gradually. The circadian rhythm of body temperature from E17 [30]. In young adults, clock-controlled
emerges immediately after birth in full-term infants heart-specific genes are expressed [31]. It is also
[23], followed by the hormonal circadian rhythms considered that other tissues acquire a core circa-
between 3 and 6 months of age [23], with the dian molecular oscillation during ontogeny, at which
sleepewake cycle becoming more stable after 6 point clock-controlled tissue-specific output genes
weeks of life [24]. Interestingly, in zebrafish, the are expressed. It is still unknown how tissue-specific
circadian molecular oscillation is developed within gene expression becomes under the control of the
one day of fertilization [25]. The different circadian core circadian clock after cell differentiation. The
clock formation periods may be attributed to species- regulation may be related to the several circadian
specific developmental processes. regulation of mRNA transcripts. Circadian regula-
tions reside not only in the process of transcription
but also mRNA translation and modification such as
Circadian Clock Development Tightly ploy-A modification, RNA methylation, and RNA-
Coupled with Cellular Differentiation editing [32e34]. Recently, it was reported that most
mRNAs of mouse forebrain are oscillating exclu-
State sively in synapse likely by mRNA transport from
Recently, it has been reported that, despite mouse cytosol to synapse, and the mRNA localization and
pluripotent stem cells such as embryonic stem (ES) the local translation are related to sleep/wake cycle
cells having no discernible circadian molecular [35,36].
oscillations of TTFLs, in vitro differentiation of ES
cells presents a circadian oscillation of clock gene
expression cell-autonomously [26,27]. Reprogram- Molecular Mechanisms of Circadian
ming of somatic cells using Yamanaka factors (Oct3/ Clock Development During Ontogenesis
4, Klf4, Sox2, and c-Myc) [28] to produce induced
pluripotent stem (iPS) cells results in the disappear- In vitro differentiated mouse ES and iPS cells
ance of the established circadian rhythm, suggesting require an approximately 14-day culture time for
that the circadian clock development is tightly circadian clock development, and the discernible
coupled with the cellular differentiation state and robust circadian oscillation does not emerge
(Fig. 1) [26]. immediately after the loss of pluripotency
During cell differentiation, genomewide epigenetic [26,30,37,38]. It is suggested that additional
modifications occur, and lineage-specific gene reg- mechanisms are necessary for the development of
ulatory networks are established. Each undifferen- the circadian clock after cell differentiation. Our
tiated cell is considered to follow a lineage-specific previous work [20,26,30,37e39] has proposed a
pathway to differentiate adequately as described in two-step mechanism as a working hypothesis for
Waddington's epigenetic landscape [29]. Almost all circadian clock development during ontogeny [30].
differentiated cells ultimately establish the TTFLs of The first step is the cell-lineage determination

Fig. 1. Circadian clock development is tightly coupled to cellular differentiation. The core TTFLs of the circadian
molecular oscillation in ES cells are not detectable, but the in vitro differentiation of ES cells induces a cell-autonomous
robust circadian oscillation. The circadian oscillation disappears after reprogramming to iPS cells.
Development of Circadian Machinery 3613

process from an undifferentiated to a differentiated This stepwise differentiation process may be


state. The second step is a CLOCK protein rate- required for the development of circadian molecular
limiting TTFL network maturation process. oscillation [30,37]. Perturbations of the differentiation
CLOCK protein expression is suppressed posttran- process by genetic modifications, such as DNA
scriptionally in the first step, and this posttranscrip- methyltransferase (Dnmt1, Dnmt3a, and Dnmt3b)
tional suppression gradually subsides during the knockouts, c-Myc overexpression, or Kpna2 over-
second step [30]. The gradual upregulation in the expression, prevent the development of the circa-
expression of CLOCK proteins is concomitant with dian molecular oscillation. The perturbation of the
the increase in the amplitude of circadian molecular normal differentiation process in the first step may
oscillation. NPAS2, a paralog of CLOCK that can inhibit cells from starting the developmental upregu-
compensate for CLOCK function [40,41], is hardly lation of CLOCK protein expression in the second
expressed in undifferentiated mouse cells [30]; step [30,37].
therefore, posttranscriptional suppression of Interestingly, mouse multipotent germline stem
CLOCK proteins is one of the key mechanisms of (mGS) cells [42], a type of pluripotent stem cells, also
the development of the circadian clock. We have present posttranscriptional suppression of CLOCK
previously reported that Dicer/Dgcr8-dependent protein expression and circadian molecular oscilla-
miRNAs-mediated repression and Clock mRNA tion development similarly to ES cells and iPS cells
export retardation are the main mechanisms of during in vitro differentiation (Fig. 2) [30]. Further-
CLOCK posttranscriptional suppression. These sup- more, we recently suggested that posttranscriptional
pressive mechanisms may be gradually canceled suppression of CLOCK proteins also occurs in
during the second step of the circadian clock human early-stage development by analysis using
development [30]. human iPS cells [39]. Human iPS cells require longer

Fig. 2. CLOCK protein expression is upregulated and PER proteins translocate from the cytoplasm to the nucleus
during in vitro differentiation, correlating with the emergence of the “24-h” oscillation. (A) Morphology of ES cells,
mGS cells, and iPS cells. Scale bars ¼ 100 mm. (B) The circadian oscillations are emerged gradually in the pluripotent stem
cells during in vitro differentiation [30]. Representative bioluminescence traces in undifferentiated pluripotent stem cells
carrying Bmal1-luc reporters (top panels) and averaged bioluminescence traces in vitro differentiated pluripotent stem cells
at day 7, 14, and 28. Data are shown with s.e.m. (n ¼ 4e6). (C) The suppression of CLOCK protein expression and the
cytoplasmic localization of PER are observed in the undifferentiated pluripotent stem cells [30,32]. Immunofluorescence
study of endogenous CLOCK and PER1 proteins in ES cells, mGS cells, iPS cells, and 28-day differentiated ES cells.
Arrowheads indicate feeder cells. Scale bars ¼ 25 mm. Part of data were adapted and modified from Umemura et al. [30].
3614 Development of Circadian Machinery

differentiation times in culture for the development of tion of the complex in the nucleus and stabilizes PER
the circadian molecular oscillations [39]. This may be proteins [54e56]. In future, it will be necessary to
related to the difference in the gestation periods elucidate whether these molecular mechanisms that
between mouse and human. occur in rhythmic cells also function in nonrhythmic
The circadian oscillation suppression in the first cells, such as ES cells.
step cannot be rescued by CLOCK overexpression
[30], suggesting that other mechanisms are
involved. Although further elucidation is needed to CLOCK Suppression in Human
understand the molecular mechanisms, the core Epigenetic Cancers
clock proteins and genes may be in different states
between normal rhythmic cells with circadian clock Interestingly, we found that posttranscriptional
and nonrhythmic cells. The state of most core clock suppression of CLOCK proteins is also observed in
molecules in nonrhythmic cells seems to have some types of human epigenetic cancers, such as
inhibitory effects on the cellular circadian oscillation. Wilms tumor and malignant rhabdoid tumor [57].
Although Per2 gene expression is relatively low in This suggests not only the disruption of the circadian
mouse ES cells, most core clock genes are clock in cancer but also that epigenetic cancers
transcribed at similar expression levels in the might undergo deviation from the normal cellular
rhythmic differentiated cells. As reported previously, differentiation process. Conversely, it could reflect
the knock-down of Clock and Npas2 expression the pathophysiological significance of CLOCK pro-
induced that E-box-driven gene expressions such as tein suppression. Although the relationships
Nr1d1 and Dbp were inhibited in the somatic cells between cancer and circadian clock disruption
[43] , as seen in Bmal1 knockout mice [44]. However, have been reported in several studies [58e67], the
these clock genes are expressed at almost similar viewpoint of cellular differentiation coupled to the
levels between ES cells and the rhythmic differen- circadian clock may be useful to further investigate in
tiated cells. This discrepancy may arise from the these settings.
different epigenetic state of overall genome between
ES cells and the differentiated cells or E-box-binding
transcriptional factors such as MYC, which is highly
expressed in ES cells. On the other hand, it was Carrier Proteins of the Circadian Clock
reported about the effect of dominant-negative Proteins
BMAL1 (BMAL1-DN) overexpression in ES cells
[26]. BMAL1-DN has the lack of the C-terminus and It is important to regulate the subcellular localiza-
strongly suppresses the BMAL1/CLOCK-mediated tion of circadian clock molecules in the 24-h
transcription [45]. Although in NIH3T3 cells with the oscillation period. Many types of carrier proteins
circadian molecular oscillation, the BMAL1-DN over- have been identified as regulators of the subcellular
expression indicated the suppression of the clock localization of clock proteins. These carrier proteins
gene sets including Dbp, the BMAL1-DN suppres- recognize nuclear localization signals in the clock
sive effect in ES cells is not observed in the clock proteins and transport them from the cytoplasm to
gene set. This result supports the CLOCK posttran- the nucleus. KPNB1, also known as importin subunit
scriptional suppression in ES cells. beta-1, is associated with KPNAs (importin subunit
The other components of clock also seem to be alpha) and translocates the carrier substrates from
regulated for the suppression of clock molecular the cytoplasm to the nucleus. KPNB1 interacts
oscillation not at the level of transcription but at the mainly with PERs and mediates PER and CRY
level of translation and/or the protein modification. nuclear transport [68]. Other carrier proteins,
PER1 proteins in mouse pluripotent stem cells such TNPO1, facilitate the nuclear localization of PER1,
as ES cells, iPS cells, and mGS cells, are exclusively but not PER2 [69]. More recently, it was reported that
localized to the cytoplasm (Fig. 2) [37]. In normal noncoding RNAs are also related to the localization
rhythmic differentiated cells, PERs and CRYs can of clock proteins [70].
shuttle dynamically between the nucleus and cyto- One member of the Kpna family, Kpna2, has
plasm independently, but they are mainly localized been identified as an importin alpha subunit, and
to the nucleus. Some modifications, such as its expression is abundant in undifferentiated cells,
phosphorylation and ubiquitination, of these nega- such as ES cells. KPNA2 are reported to regulate
tive regulators modulate the subcellular dynamics the differentiation state of cells by promoting
and protein stability [46e52]. PER2 is phosphory- OCT3/4 nuclear entry and preventing OCT6
lated by CK2 and GSK3beta, and these modifica- nuclear entry, leading to cellular differentiation
tions change its subcellular localization [50,53]. In [71,72]. Recently, we found that Kpna2 over-
addition, PER-CRY binding induces the accumula- expression during in vitro differentiation of ES
Development of Circadian Machinery 3615

cells has inhibitory effects on the development of Keywords:


the circadian clock and the nuclear localization of cellular differentiation;
PERs. However, the binding interaction between posttranscriptional regulation;
KPNA2 and PERs is considered to be very weak. CLOCK;
Therefore, KPNA2 overexpression during in vitro circadian rhythm;
differentiation of ES cells may inhibit the process of cellular circadian clock
cellular differentiation and indirectly affect the
abnormal localization of PER proteins to the
cytoplasm [37]. References

Biological Significance of Arresting the [1] A. Balsalobre, F. Damiola, U. Schibler, A serum shock
TTFLs induces circadian gene expression in mammalian tissue
culture cells, Cell 93 (1998) 929e937.
Clock genes and proteins in nonrhythmic cells [2] K. Yagita, F. Tamanini, G.T. van Der Horst, H. Okamura,
may be regulated for arresting TTFLs, suggesting Molecular mechanisms of the biological clock in cultured
that circadian oscillation is disadvantageous in the fibroblasts, Science 292 (2001) 278e281.
early developmental stages. Although the biologi- [3] S. Yamazaki, R. Numano, M. Abe, A. Hida, R. Takahashi,
cal significance has not yet been elucidated in M. Ueda, et al., Resetting central and peripheral circadian
oscillators in transgenic rats, Science 288 (2000) 682e685.
mammals, in the fruit fly, Drosophila melanogaster,
[4] S.H. Yoo, S. Yamazaki, P.L. Lowrey, K. Shimomura,
Clock (Clk) overexpression induces developmental C.H. Ko, E.D. Buhr, et al., PERIOD2::LUCIFERASE real-
lethality [73], and Clk posttranscriptional regulation time reporting of circadian dynamics reveals persistent
is essential for adequate development [74]. This circadian oscillations in mouse peripheral tissues, Proc.
suggests that regulation of CLOCK expression is Natl. Acad. Sci. U.S.A. 101 (2004) 5339e5346.
essential not only for the circadian clock emer- [5] R.P. Aryal, P.B. Kwak, A.G. Tamayo, M. Gebert, P.L. Chiu,
gence but also for the developmental process as a T. Walz, et al., Macromolecular assemblies of the mamma-
whole. lian circadian clock, Mol. Cell 67 (2017) 770e782 e6.
Interestingly, in mammals, circadian clock arrest [6] Y.Y. Chiou, Y. Yang, N. Rashid, R. Ye, C.P. Selby,
may be not only due to defective TTFLs by A. Sancar, Mammalian Period represses and de-represses
transcription by displacing CLOCK-BMAL1 from promoters in
different states of circadian clock molecules.
a Cryptochrome-dependent manner, Proc. Natl. Acad. Sci.
Although it is believed that the maternal circadian U.S.A. 113 (2016) E6072eE6079.
rhythm entrains the fetus [15,17,18,75], in our [7] H.R. Ueda, W. Chen, A. Adachi, H. Wakamatsu, S. Hayashi,
study, gene expression was not rhythmic in the T. Takasugi, et al., A transcription factor response element
mouse heart at E10e12, suggesting that maternal for gene expression during circadian night, Nature 418
circadian clock entrainment in the embryo is (2002) 534e539.
prevented by unknown mechanisms in early [8] N. Preitner, F. Damiola, L. Lopez-Molina, J. Zakany,
development [30]. This suggests the presence of D. Duboule, U. Albrecht, et al., The orphan nuclear receptor
multilayered mechanisms that suppress both the REV-ERBalpha controls circadian transcription within the
TTFLs of the circadian clock and the maternal positive limb of the mammalian circadian oscillator, Cell 110
(2002) 251e260.
clock-entrained gene cycles during early develop-
[9] J.B. Hogenesch, E.D. Herzog, Intracellular and intercellular
ment and might highlight further the biological processes determine robustness of the circadian clock,
significance of the suppression of the circadian FEBS Lett. 585 (2011) 1427e1434.
clock in early developmental stages. [10] J.S. Takahashi, Transcriptional architecture of the mamma-
lian circadian clock, Nat. Rev. Genet. 18 (2017) 164e179.
[11] J.D. Alvarez, D. Chen, E. Storer, A. Sehgal, Non-cyclic and
developmental stage-specific expression of circadian clock
proteins during murine spermatogenesis, Biol. Reprod. 69
Conflict of interest statement (2003) 81e91.
[12] T. Amano, A. Matsushita, Y. Hatanaka, T. Watanabe,
This work was supported in part by grants-in-aid K. Oishi, N. Ishida, et al., Expression and functional
for scientific research from the Japan Society for the analyses of circadian genes in mouse oocytes and
Promotion of Science to Y.U. (17KT0129) and K.Y. preimplantation embryos: cry1 is involved in the meiotic
process independently of circadian clock regulation, Biol.
(18H02600 and 19K22516).
Reprod. 80 (2009) 473e483.
[13] D. Morse, N. Cermakian, S. Brancorsini, M. Parvinen,
Received 15 October 2019; P. Sassone-Corsi, No circadian rhythms in testis: period1
Received in revised form 26 November 2019; expression is clock independent and developmentally
Accepted 26 November 2019 regulated in the mouse, Mol. Endocrinol. 17 (2003)
Available online 10 January 2020 141e151.
3616 Development of Circadian Machinery

[14] V. Carmona-Alcocer, J.H. Abel, T.C. Sun, L.R. Petzold, [32] S. Kojima, E.L. Sher-Chen, C.B. Green, Circadian control of
F.J. Doyle 3rd, C.L. Simms, et al., Ontogeny of circadian mRNA polyadenylation dynamics regulates rhythmic protein
rhythms and synchrony in the suprachiasmatic nucleus, expression, Genes Dev. 26 (2012) 2724e2736.
J. Neurosci. 38 (2018) 1326e1334. [33] J.M. Fustin, M. Doi, Y. Yamaguchi, H. Hida, S. Nishimura,
[15] F.C. Davis, R.A. Gorski, Development of hamster circadian M. Yoshida, et al., RNA-methylation-dependent RNA pro-
rhythms: role of the maternal suprachiasmatic nucleus, cessing controls the speed of the circadian clock, Cell 155
J. Comp. Physiol. 162 (1988) 601e610. (2013) 793e806.
[16] C. Jud, U. Albrecht, Circadian rhythms in murine pups [34] H. Terajima, H. Yoshitane, H. Ozaki, Y. Suzuki, S. Shimba,
develop in absence of a functional maternal circadian clock, S. Kuroda, et al., ADARB1 catalyzes circadian A-to-I editing
J. Biol. Rhythm. 21 (2006) 149e154. and regulates RNA rhythm, Nat. Genet. 49 (2017) 146e151.
[17] S.M. Reppert, W.J. Schwartz, Maternal suprachiasmatic [35] F. Bruning, S.B. Noya, T. Bange, S. Koutsouli, J.D. Rudolph,
nuclei are necessary for maternal coordination of the S.K. Tyagarajan, et al., Sleep-wake cycles drive daily
developing circadian system, J. Neurosci. 6 (1986) dynamics of synaptic phosphorylation, Science 366 (2019).
2724e2729. [36] S.B. Noya, D. Colameo, F. Bruning, A. Spinnler, D. Mircsof,
[18] A. Sumova, Z. Bendova, M. Sladek, R. El-Hennamy, L. Opitz, et al., The forebrain synaptic transcriptome is
K. Mateju, L. Polidarova, et al., Circadian molecular clocks organized by clocks but its proteome is driven by sleep,
tick along ontogenesis, Physiol. Res. 57 (Suppl 3) (2008) Science 366 (2019).
S139eS148. [37] Y. Umemura, N. Koike, T. Matsumoto, S.H. Yoo, Z. Chen,
[19] D. Landgraf, C. Achten, F. Dallmann, H. Oster, Embryonic N. Yasuhara, et al., Transcriptional program of Kpna2/
development and maternal regulation of murine circadian Importin-alpha2 regulates cellular differentiation-coupled
clock function, Chronobiol. Int. 32 (2015) 416e427. circadian clock development in mammalian cells, Proc. Natl.
[20] Y. Inada, H. Uchida, Y. Umemura, W. Nakamura, T. Sakai, Acad. Sci. U.S.A. 111 (2014) E5039eE5048.
N. Koike, et al., Cell and tissue-autonomous development of [38] Y. Umemura, J. Yoshida, M. Wada, Y. Tsuchiya, Y. Minami,
the circadian clock in mouse embryos, FEBS Lett. 588 H. Watanabe, et al., An in vitro ES cell-based clock
(2014) 459e465. recapitulation assay model identifies CK2alpha as an
[21] H. Dolatshad, A.J. Cary, F.C. Davis, Differential expression endogenous clock regulator, PLoS One 8 (2013), e67241.
of the circadian clock in maternal and embryonic tissues of [39] Y. Umemura, I. Maki, Y. Tsuchiya, N. Koike, K. Yagita,
mice, PLoS One 5 (2010) e9855. Human circadian molecular oscillation development using
[22] V. Carmona-Alcocer, J.H. Abel, T.C. Sun, L.R. Petzold, induced pluripotent stem cells, J. Biol. Rhythm. 34 (2019)
F.J. Doyle 3rd, C.L. Simms, et al., Ontogeny of circadian 525e532.
rhythms and synchrony in the suprachiasmatic nucleus, [40] J.P. DeBruyne, D.R. Weaver, S.M. Reppert, CLOCK and
J. Neurosci. 38 (2018) 1326e1334. NPAS2 have overlapping roles in the suprachiasmatic
[23] S.A. Rivkees, Developing circadian rhythmicity in infants, circadian clock, Nat. Neurosci. 10 (2007) 543e545.
Pediatr. Endocrinol. Rev. 1 (2003) 38e45. [41] J.P. DeBruyne, D.R. Weaver, S.M. Reppert, Peripheral
[24] N. Kleitman, T.G. Engelmann, Sleep characteristics of circadian oscillators require CLOCK, Curr. Biol. 17 (2007)
infants, J. Appl. Physiol. 6 (1953) 269e282. R538eR539.
[25] M.P. Dekens, D. Whitmore, Autonomous onset of the [42] M. Kanatsu-Shinohara, K. Inoue, J. Lee, M. Yoshimoto,
circadian clock in the zebrafish embryo, EMBO J. 27 N. Ogonuki, H. Miki, et al., Generation of pluripotent stem
(2008) 2757e2765. cells from neonatal mouse testis, Cell 119 (2004)
[26] K. Yagita, K. Horie, S. Koinuma, W. Nakamura, 1001e1012.
I. Yamanaka, A. Urasaki, et al., Development of the [43] E.E. Zhang, A.C. Liu, T. Hirota, L.J. Miraglia, G. Welch,
circadian oscillator during differentiation of mouse embryo- P.Y. Pongsawakul, et al., A genome-wide RNAi screen for
nic stem cells in vitro, Proc. Natl. Acad. Sci. U.S.A. 107 modifiers of the circadian clock in human cells, Cell 139
(2010) 3846e3851. (2009) 199e210.
[27] E. Kowalska, E. Moriggi, C. Bauer, C. Dibner, S.A. Brown, [44] H. Yoshitane, H. Ozaki, H. Terajima, N.H. Du, Y. Suzuki,
The circadian clock starts ticking at a developmentally early T. Fujimori, et al., CLOCK-controlled polyphonic regulation
stage, J. Biol. Rhythm. 25 (2010) 442e449. of circadian rhythms through canonical and noncanonical E-
[28] K. Takahashi, S. Yamanaka, Induction of pluripotent stem boxes, Mol. Cell. Biol. 34 (2014) 1776e1787.
cells from mouse embryonic and adult fibroblast cultures by [45] Y.B. Kiyohara, S. Tagao, F. Tamanini, A. Morita,
defined factors, Cell 126 (2006) 663e676. Y. Sugisawa, M. Yasuda, et al., The BMAL1 C terminus
[29] C.H. Waddington, The Strategy of the Genes: A Discussion regulates the circadian transcription feedback loop, Proc.
of Some Aspects of Theoretical Biology, George Allen and Natl. Acad. Sci. U.S.A. 103 (2006) 10074e10079.
Unwin, London, 1957. [46] C. Lee, J.P. Etchegaray, F.R. Cagampang, A.S. Loudon,
[30] Y. Umemura, N. Koike, M. Ohashi, Y. Tsuchiya, Q.J. Meng, S.M. Reppert, Posttranslational mechanisms regulate the
Y. Minami, et al., Involvement of posttranscriptional regula- mammalian circadian clock, Cell 107 (2001) 855e867.
tion of Clock in the emergence of circadian clock oscillation [47] M. Akashi, Y. Tsuchiya, T. Yoshino, E. Nishida, Control of
during mouse development, Proc. Natl. Acad. Sci. U.S.A. intracellular dynamics of mammalian period proteins by
114 (2017) E7479eE7488. casein kinase I epsilon (CKIepsilon) and CKIdelta in cultured
[31] R. Zhang, N.F. Lahens, H.I. Ballance, M.E. Hughes, cells, Mol. Cell. Biol. 22 (2002) 1693e1703.
J.B. Hogenesch, A circadian gene expression atlas in [48] A. Takano, Y. Isojima, K. Nagai, Identification of mPer1
mammals: implications for biology and medicine, Proc. Natl. phosphorylation sites responsible for the nuclear entry,
Acad. Sci. U.S.A. 111 (2014) 16219e16224. J. Biol. Chem. 279 (2004) 32578e32585.
Development of Circadian Machinery 3617

[49] H. Lee, R. Chen, Y. Lee, S. Yoo, C. Lee, Essential roles of [63] P. Janich, G. Pascual, A. Merlos-Sua rez, E. Batlle,
CKIdelta and CKIepsilon in the mammalian circadian clock, J. Ripperger, U. Albrecht, et al., The circadian molecular
Proc. Natl. Acad. Sci. U.S.A. 106 (2009) 21359e21364. clock creates epidermal stem cell heterogeneity, Nature
[50] B. Maier, S. Wendt, J.T. Vanselow, T. Wallach, S. Reischl, (2011) 1e8.
S. Oehmke, et al., A large-scale functional RNAi screen [64] N.M. Kettner, C.A. Katchy, L. Fu, Circadian gene variants in
reveals a role for CK2 in the mammalian circadian clock, cancer, Ann. Med. 46 (2014) 208e220.
Genes Dev. 23 (2009) 708e718. [65] K.C.G. Van Dycke, W. Rodenburg, C.T.M. van Oostrom,
[51] M. Gallego, H. Kang, D.M. Virshup, Protein phosphatase 1 L.W.M. van Kerkhof, J.L.A. Pennings, T. Roenneberg, et al.,
regulates the stability of the circadian protein PER2, Chronically alternating light cycles increase breast cancer
Biochem. J. 399 (2006) 169e175. risk in mice, Curr. Biol. : CB 25 (2015) 1932e1937.
[52] E.J. Eide, E.L. Vielhaber, W.A. Hinz, D.M. Virshup, The [66] T. Papagiannakopoulos, M.R. Bauer, S.M. Davidson,
circadian regulatory proteins BMAL1 and cryptochromes are M. Heimann, L. Subbaraj, A. Bhutkar, et al., Circadian
substrates of casein kinase Iepsilon, J. Biol. Chem. 277 rhythm disruption promotes lung tumorigenesis, Cell Meta-
(2002) 17248e17254. bol. 24 (2016) 324e331.
[53] C. Iitaka, K. Miyazaki, T. Akaike, N. Ishida, A role for [67] R.V. Puram, M.S. Kowalczyk, C.G. de Boer, R.K. Schneider,
glycogen synthase kinase-3beta in the mammalian circadian P.G. Miller, M. McConkey, et al., Core circadian clock genes
clock, J. Biol. Chem. 280 (2005) 29397e29402. regulate leukemia stem cells in AML, Cell 165 (2016)
[54] K. Kume, M.J. Zylka, S. Sriram, L.P. Shearman, 303e316.
D.R. Weaver, X. Jin, et al., mCRY1 and mCRY2 are [68] Y. Lee, A.R. Jang, L.J. Francey, A. Sehgal, J.B. Hogenesch,
essential components of the negative limb of the circadian KPNB1 mediates PER/CRY nuclear translocation and
clock feedback loop, Cell 98 (1999) 193e205. circadian clock function, Elife 4 (2015).
[55] K. Miyazaki, M. Mesaki, N. Ishida, Nuclear entry mechanism [69] S. Korge, B. Maier, F. Bruning, L. Ehrhardt, T. Korte,
of rat PER2 (rPER2): role of rPER2 in nuclear localization of M. Mann, et al., The non-classical nuclear import carrier
CRY protein, Mol. Cell. Biol. 21 (2001) 6651e6659. Transportin 1 modulates circadian rhythms through its effect
[56] K. Yagita, F. Tamanini, M. Yasuda, J.H. Hoeijmakers, on PER1 nuclear localization, PLoS Genet. 14 (2018),
G.T. van der Horst, H. Okamura, Nucleocytoplasmic e1007189.
shuttling and mCRY-dependent inhibition of ubiquitylation [70] Y. Lee, Y. Shen, L.J. Francey, C. Ramanathan, A. Sehgal,
of the mPER2 clock protein, EMBO J. 21 (2002) A.C. Liu, et al., The NRON complex controls circadian clock
1301e1314. function through regulated PER and CRY nuclear transloca-
[57] M. Ohashi, Y. Umemura, N. Koike, Y. Tsuchiya, Y. Inada, tion, Sci. Rep. 9 (2019) 11883.
H. Watanabe, et al., Disruption of circadian clockwork in in [71] N. Yasuhara, N. Shibazaki, S. Tanaka, M. Nagai,
vivo reprogramming-induced mouse kidney tumors, Genes Y. Kamikawa, S. Oe, et al., Triggering neural differentiation
Cells 23 (2018) 60e69. of ES cells by subtype switching of importin-alpha, Nat. Cell
[58] E. Filipski, P.F. Innominato, M. Wu, X.M. Li, S. Iacobelli, Biol. 9 (2007) 72e79.
L.J. Xian, et al., Effects of light and food schedules on liver [72] N. Yasuhara, R. Yamagishi, Y. Arai, R. Mehmood,
and tumor molecular clocks in mice, JNCI J. Natl. Cancer C. Kimoto, T. Fujita, et al., Importin alpha subtypes
Inst. 97 (2005) 507e517. determine differential transcription factor localization in
[59] S. Gery, N. Komatsu, L. Baldjyan, A. Yu, D. Koo, embryonic stem cells maintenance, Dev. Cell 26 (2013)
H.P. Koeffler, The circadian gene Per1 plays an important 123e135.
role in cell growth and DNA damage control in human [73] J. Zhao, V.L. Kilman, K.P. Keegan, Y. Peng, P. Emery,
cancer cells, Mol. Cell 22 (2006) 375e382. M. Rosbash, et al., Drosophila clock can generate ectopic
[60] P.A. Wood, X. Yang, A. Taber, E.-Y. Oh, C. Ansell, circadian clocks, Cell 113 (2003) 755e766.
S.E. Ayers, et al., Period 2 mutation accelerates ApcMin/þ [74] I. Lerner, O. Bartok, V. Wolfson, J.S. Menet, U. Weissbein,
tumorigenesis, Mol. Cancer Res. 6 (2008) 1786e1793. S. Afik, et al., Clk post-transcriptional control denoises
[61] S. Sahar, P. Sassone-Corsi, Metabolism and Cancer: the circadian transcription both temporally and spatially, Nat.
Circadian Clock Connection, 2009, pp. 1e11. Commun. 6 (2015).
[62] S. Lee, L.A. Donehower, A.J. Herron, D.D. Moore, L. Fu, [75] M. Seron-Ferre, N. Mendez, L. Abarzua-Catalan, N. Vilches,
Disrupting circadian homeostasis of sympathetic signaling F.J. Valenzuela, H.E. Reynolds, et al., Circadian rhythms in
promotes tumor development in mice, PLoS One 5 (2010) the fetus, Mol. Cell. Endocrinol. 349 (2012) 68e75.
e10995-13.

You might also like