You are on page 1of 8

Review Article

https://doi.org/10.1038/s41556-019-0384-4

The cell cycle in stem cell proliferation,


pluripotency and differentiation
Lijun Liu 1,2
, Wojciech Michowski 2,3
, Aleksandra Kolodziejczyk 2
and Piotr Sicinski 2
*

Cyclins, cyclin-dependent kinases and other components of the core cell cycle machinery drive cell division. Growing evidence
indicates that this machinery operates in a distinct fashion in some mammalian stem cell types, such as pluripotent embryonic
stem cells. In this Review, we discuss our current knowledge of how cell cycle proteins mechanistically link cell proliferation,
pluripotency and cell fate specification. We focus on embryonic stem cells, induced pluripotent stem cells and embryonic neural
stem/progenitor cells.

T
he core cell cycle machinery operating in the cell nucleus complexes with cyclin–CDK2 and cyclin–CDK1 molecules and
orchestrates cell division. The key components of this machin- inhibit their kinase activities1–4.
ery are proteins called cyclins, which bind, activate and pro-
vide substrate specificity to their associated catalytic partners, the Cell cycle machinery in embryonic stem cells
cyclin-dependent kinases (CDKs)1–4. Cell cycle progression can be The work of several laboratories revealed that the core cell cycle
divided into four phases: gap 1 (G1), DNA synthesis (S), gap 2 (G2) machinery operates differently during early embryonic develop-
and mitosis (M). Depending on the mitogenic environment, cells ment16. In developing flies, fish and frogs, first cell cycles are very
traversing G1 phase either activate a program that will result in cell rapid, lack obvious gap phases and consist of alternating S and M
division or enter a quiescent G0 state1–4 (Fig. 1a). At the molecular phases16. Analyses of peri-implantation mouse embryos demon-
level, stimulation of cells with growth-promoting factors results in strated that murine embryonic cells display remarkably short divi-
upregulation of the D-type cyclins (D1, D2 and D3), which acti- sion times in vivo (~4.4–7.5 h)17–19, with a very small fraction of
vate cyclin-dependent kinase 4 (CDK4) and CDK61–5. In a classi- cells residing in G1 phase20,21. Intriguingly, the first two cell cycles are
cal cell cycle model, cyclin D–CDK4/6 complexes, together with considerably longer, with significant shortening occurring during
E-type cyclins (E1 and E2) and their associated kinases (primarily the third cell division22. Later in development, following gastrula-
CDK2, but also CDK1 and CDK3) phosphorylate and functionally tion and formation of the endodermal, mesodermal and ectodermal
inactivate the retinoblastoma protein RB1, as well as pRB1-related lineages, the cell cycle length substantially increases because of an
RBL1 and RBL2 proteins1–4. This leads to activation or derepres- extension of the gap phases19.
sion of E2F transcription factors, which then transactivate genes Murine embryonic stem ES cells (mESCs), derived from inner-
required for the entry and progression of cells into S phase1–4,6,7. This cell masses of blastocysts and cultured in vitro in conditions that
model has been questioned by the demonstration that throughout favour retention of their pluripotent state, recapitulate this unique
most of G1 phase, RB1 exists in a monophosphosphorylated state, organization of the cell cycle20. They divide very rapidly, although
becoming fully phosphorylated by cyclin E–CDK2 at the end of G1 not as fast as their in vivo counterparts (division time ~12 h), and
phase8. In addition to RB1 phosphorylation, inactivation of Cdh1, a have a short G1 phase that lasts only 3 h16,23–25 (Table 1). These cells
substrate-recognition subunit of the anaphase-promoting complex express high levels of cyclins E, A and B and display elevated levels
(APC/C), contributes to an irreversible commitment to cell divi- of Cdk1 and Cdk2 kinases, greatly exceeding those seen in somatic
sion9. Inhibition of Cdh1 by cyclin E–CDK2 at the end of G1 phase cells20,26,27. Strikingly, high levels of cyclins E and A are observed
inactivates APC/C and allows accumulation of S-phase cyclins that throughout the cell cycle, in contrast to their periodic expression
are normally degraded by Cdh1–APC/C9–11. in somatic cells20,28 (Fig. 1a,b). Consequently, Cdk2, cyclin-E- and
The entry of mammalian cells into S phase is driven by cyclin cyclin-A-associated kinases are constitutively active throughout the
E–CDK2 acting in concert with the DBF4-associated kinase12–14. Later entire cell cycle, in contrast to somatic cells, wherein these kinases
during S phase, cyclin A2 becomes upregulated, pairs with CDK2 and become transiently activated at specific cell cycle phases16,20,28. The
CDK1 and promotes S-phase progression1–4. Following a second gap only cell cycle kinase that retains periodicity in mESCs is cyclin
phase (G2), cyclin B translocates to the nucleus, activates CDK1 and B–Cdk1, although its levels are substantially higher than those
drives the separation of genetic material to daughter cells3,15. in somatic cells20,27. As a consequence of hyperactivated Cdk1
In addition to these positive regulators of the cell cycle, mam- and Cdk2 kinases, RB1 is constitutively phosphorylated and inac-
malian cells also express two classes of cell cycle inhibitors. The tivated throughout the cell cycle, and E2F activity is constitutively
INK proteins (p16INK4a, p15INK4b, p18INK4c and p19INK4d) interact derepressed16,20,23 (Fig. 1; Table 1).
with CDK4 or CDK6 and block their association with D cyclins1–4. Several mechanisms likely contribute to very high levels of Cdk1
The KIP/CIP proteins (p21Cip1, p27Kip1 and p57Kip2) form ternary and Cdk2 kinase activity in ESCs. In contrast to somatic cells, ESCs

1
College of Life and Health Sciences, Key Laboratory of Data Analytics and Optimization for Smart Industry (Ministry of Education), Northeastern
University, Shenyang, Liaoning, China. 2Department of Cancer Biology, Dana–Farber Cancer Institute and Department of Genetics, Blavatnik
Institute, Harvard Medical School, Boston, MA, USA. 3Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.
*e-mail: Peter_Sicinski@dfci.harvard.edu

1060 Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology


NaTuRe Cell BIoloGy Review Article
a b
Cyclin B–Cdk1
INK: p16INK4a
p15 INK4b D-cyclins Cyclin E Cyclin A Cyclin B
G0 D-cyclins–Cdk4/6

Expression level
p18INK4c p19INK4d
M
Mouse RB1
G2 embryonic G1 E2F
fibroblasts
Cyclin A–Cdk1/2 KIP/CIP: p21Cip1
Cyclin E–Cdk2
S P P P p27Kip1 p57Kip2 G0 G1 S G2 M
KIP/CIP: RB1
p21Cip1 p27Kip1 MEFs
E2F
p57Kip2
RB1 hyperphosphorylation
APC/C Cdh1 Cdk2 Cyclin E
D-cyclins Cyclin E Cyclin A Cyclin B

Expression level
Cyclin B–Cdk1

Cyclin E–Cdk2
G1 S G2 M
M RB1 hyperphosphorylation
G1 mESCs
Mouse P P P
embryonic
G2 stem cells RB1
<12 h
E2F
S D-cyclins Cyclin E Cyclin A Cyclin B

Expression level
Cyclin A–Cdk1/2
CDK1/2 p27Kip1 Skp2

G1 S G2 M
S-phase
cyclins APC/C Emi1
hESCs

Fig. 1 | Organization of the cell cycle in somatic cells (MEFs) and in different types of ESCs. a, Differences in activity and expression of cell cycle
components in MEFs and in murine ESCc (mESCs). In contrast to MEFs, mESCs lack expression of D cyclins and continuously express cyclin A and cyclin
E. This allows them to maintain RB1 hyperphosphorylation throughout the cell cycle and results in a very short G1 phase. Cyclin-dependent kinase inhibitors
are absent from mESCs. Upward blue arrows indicate increased expression. Gray cylinders represent histones. b, Oscillations of cyclin levels in MEFs,
mESCs and hESCs. MEFs, mouse embryonic fibroblasts; mESCs, mouse embryonic stem cells; hESCs, human embryonic stem cells.

Table 1 | Comparison of cell cycle features in MEFs, mESCs and hESCs


MEFs mESCs hESCs
Cell cycle length 24 h 4.4–7.5 h in vivo; up to 12 h in vitro 16 h
G1 phase length 11 h 3h 3h
CDK1 and CDK2 activity Periodical Very high and constant Very high and periodical
D-cyclin expression +++ +/− +
RB1 phosphorylation status Hypo- and hyperphosphorylated Hyperphosphorylated Hypo- and hyperphosphorylated
KIP/CIP inhibitor expression Present Absent Present
MEFs, mouse embryonic fibroblasts; mESCs, mouse embryonic stem cells; hESCs, human embryonic stem cells.

do not express KIP/CIP inhibitors, which normally mitigate the of serum and leukaemia inhibitory factor (LIF). Recent studies
activity of Cdk1/216,20,26. ESCs also express high levels of the APC/C indicate that murine cells cultured in a defined medium with inhibi-
inhibitor Emi1, leading to decreased activity of APC/C29,30. This tors of MEK and GSK3 kinases resemble true pluripotent stem cells
mechanism allows cell-cycle-wide accumulation of S-phase cyclins from inner-cell masses and are hence termed to reside in a ‘ground
and another APC/C target, Skp211,29. Skp2, in turn, triggers degra- state’35,36. The cell cycle organization of these ground state cells may
dation of p27Kip1, thereby further contributing to elevated Cdk1/2 be different from that of cells cultured in serum and LIF, including
kinase activity16 (Fig. 1a). a longer G1 phase and the presence of hypophosphorylated RB137.
Initial reports documented that mESCs express low levels of D This surprising finding calls for more side-by-side comparisons of
cyclins and are refractory to cell cycle inhibition by p16INK4a (ref. 31). mESCs cultured in different conditions versus pluripotent cells of
Consistent with these results, another study32 demonstrated that early embryos in vivo.
prior to gastrulation, mouse embryos do not express D cyclins. In The organization of the cell cycle in human ESCs (hESCs) is
contrast to these findings, others reported expression of cyclins D1 slightly different (Fig. 1b; Table 1). This likely reflects the fact that
and D3 in the pluripotent cells of mouse embryonic epiblast33. Cyclin hESCs are more similar to ‘primed’ pluripotent cells derived from
D3 was also shown to be expressed in ESCs, in which it complexes the late epiblast layer of post-implantation embryos (epiblast-
with Cdk6; however, these cyclin D3–Cdk6 complexes are refrac- derived stem cells, EpiSCs)19,38,39. It is now appreciated that various
tory to inhibition by p16INK4a (ref. 33). A similar cell cycle organiza- forms of pluripotent stem cells exist during early development and
tion was reported in ESCs derived from rhesus monkeys34. It should that their cell cycle organization might show some differences19.
be noted that these studies utilized ESCs grown in the presence Like mESCs, hESCs proliferate rapidly, display a short G1 phase and

Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology 1061


Review Article NaTuRe Cell BIoloGy

G1
cyclins

Proteasomal Serial Yamanaka


P
degradation passaging factors
P
Oct4 c-Myc
Oct4 Cyclins
Most reprogramming
P P occurs within a subset of Sox2 Klf4
Sox2 Nanog highly profilerative cells
CDKs

Cyclin B
Self-
renewal
Cyclin E Somatic reprogramming
MEF

P
PI3K–Akt ESC/iPSC Nanog LIN28 Sox2 p21Cip1 p19ARF CDKs
and
P Sox2 Oct4 P p16INK4a p53 cyclins
PP1

CDK1 CDK2
P CDK1
RB1 Cyclin B Cyclin E

Fig. 2 | The cell cycle in somatic reprogramming and pluripotency maintenance. During somatic reprogramming by expression of Oct4, Sox2, Klf4 and
c-Myc, somatic cells rapidly accelerate the cell cycle. Ectopic overexpression of cell cycle proteins or inactivation of cell cycle inhibitors increases the
efficiency of reprogramming. Conversely, serial passaging leads to a decreased reprogramming rate. CDK2-dependent phosphorylation of Sox2 or cyclin
B–CDK1-dependent upregulation of LIN28 was postulated to aid reprogramming. RB1 represses expression of core pluripotency factors. The cell cycle
machinery is also important for the maintenance of ESC pluripotency. G1 cyclins stabilize core pluripotency factors through phosphorylation (P), thereby
preventing their proteasomal degradation. High levels of cyclins and CDK1-dependent phosphorylation of PI3K–Akt pathway components likely contribute
to the maintenance of pluripotency. CDK1 also inhibits Oct4 activity during the M phase, acting through PP1 and Aurkb. Upward blue arrows indicate
increased expression. ESC, embryonic stem cell; iPSC, induced pluripotent stem cell.

express high levels of CDK1 and CDK2 kinases40,41. However, these the critical residues of Oct4, Sox2 and Nanog, thereby triggering
cells express appreciable levels of D cyclins and KIP/CIP inhibitors, their proteasomal degradation and attenuation of the pluripotent
show cell-cycle-dependent fluctuations of CDK2 kinase and contain state25 (Fig. 2).
both hyper- and hypophosphorylated RB119,40–42 (Fig. 1b; Table 1). Although G1 cyclins are dispensable for the proliferation of
mESCs, no ESC colonies were observed upon ablation of cyclin
Molecular links between cell cycle and pluripotency A, revealing an essential role for this cyclin in stem cells51. These
One of the main questions in the field has been whether the unique observations are in stark contrast to the situation seen in fibroblasts,
cell cycle organization of pluripotent stem cells simply reflects the wherein G1 cyclins are essential, but cyclin A is not, as its function
necessity to rapidly expand this cell population or it plays an active can be carried out by cyclin E51. Collectively, these observations
role in enforcing the pluripotent state. Several observations sug- indicate that, in contrast to differentiated cells, ESCs rely on cyclins
gested a role for cell cycle proteins in enforcing pluripotency24,27,40,43–46 A and B for cell cycle progression, whereas G1 cyclins contribute to
(Fig. 2). The knockdown of CDK1, CDK2, cyclin E or B1, and treat- the maintenance of the pluripotent state.
ment with CDK-inhibitors all resulted in the loss of the pluripo- In addition to cell cycle proteins regulating pluripotency factors,
tent state and triggered differentiation24,27,40,43–46, whereas ectopic the reverse was also noted; namely, the ability of Nanog and Oct4
overexpression of cyclins E or B1 promoted ESC self-renewal24,44. proteins to affect proliferation52–54. Specifically in hESCs, Nanog was
However, several of these manipulations resulted in cell cycle arrest, shown to bind the regulatory regions of the CDK6 and CDC25 genes
apoptosis or perturbed cell cycle progression, which confounded and upregulate their expression, thereby promoting cell prolifera-
the interpretation of the results40,44,45,47. At the mechanistic level, tion52. Oct4 was postulated to stimulate cell growth by repressing
Wang et al.27 proposed that CDK1 kinase maintains the pluripotent the expression of p21Cip1, a cell cycle inhibitor gene53. In contrast to
state by regulating the PI3K–Akt pathway. Conversely, Kim et al.48 these findings, another study reported that Oct4 inhibits cell pro-
reported that CDK1 inhibits chromatin binding of Oct4 through an liferation by forming a complex with cyclin–CDK1 and inhibiting
indirect mechanism involving Aurkb and PP1. The notion that the CDK1 activation54. Pluripotency factors can also affect prolifera-
core cell cycle machinery actively regulates pluripotency was fur- tion through indirect mechanisms. For instance, Oct4 and Nanog
ther supported by genetic studies using ESCs derived from cyclin upregulate c-Myc, which in turn regulates expression of several cell
D-null49 or E-null50 mice. Although ablation of all three D-type cycle genes55–57.
(D1−/−D2−/−D3−/−) or both E-type (E1−/−E2−/−) cyclins had no effect
on the pluripotent state, a combined acute shutdown of all five G1 Dissolution of pluripotency and cell fate specification
cyclins resulted in strong attenuation of pluripotency25. Notably, The unusual cell cycle properties of ESCs change upon dissolution
cells lacking all G1 cyclins proliferated in vitro, albeit at a somewhat of pluripotency, when stem cells undergo cell fate specification and
reduced rate, revealing that D and E cyclins are not essential for differentiate into different lineages16. During this process, the dura-
the proliferation of mESCs25. At the molecular level, G1 cyclin–CDK tion of cell division increases, mainly because of an expanded G1
kinases were shown to directly phosphorylate core pluripotency phase16,24,28,34,43,58. D cyclins become upregulated, the activity of CDK1
factors Oct4, Sox2 and Nanog, resulting in their stabilization25. and CDK2 decreases, in part due to expression of KIP/CIP inhibi-
Ablation of all G1 cyclins strongly diminished phosphorylation of tors, and the activation CDK2-, cyclin E- and cyclin A-associated

1062 Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology


NaTuRe Cell BIoloGy Review Article
G1 S G2 M This mechanism prevents endodermal cell fate and renders cells
Cyclin B1
susceptible to neuroectodermal differentiation68. In another study69,
these authors reported that in hESCs D cyclins regulate cell fate
5hmC
H3K4me3 Dissolution of specification via a CDK4/6- and Smad-independent mechanism.
pluripotency According to this model, cyclin D1 recruits transcriptional coactiva-
tors to neuroectoderm genes, thereby promoting neuroectodermal
D-cyclins
differentiation69. D cyclins also bind endodermal gene promoters,
Early G1 Late G1 S G2 M but in this case they recruit transcriptional co-repressors, which
Smad2 silence gene expression69 (Fig. 3). Indeed, cyclin D1 was previously
Cytoplasm
Smad3 shown to interact with gene regulatory regions and regulate gene
expression in vivo in a CDK4/6-independent fashion70,71.
Nucleus P
These two models68,69 are at odds with several observations.
D-cyclin For instance, knockout mice lacking individual D cyclins, all D
Smad2
Cdk4/6 cyclins (D1−/−D2−/−D3−/−), or Cdk4 and Cdk6 (Cdk4−/−Cdk6−/−)
Smad3 D-cyclin can develop at least until midgestation and undergo organogenesis,
Endoderm Endoderm indicating that specification into different lineages is not signifi-
D-cyclin cantly affected49,72–75. In addition, mESCs lacking all three D-type
Neuroectoderm cyclins do not upregulate differentiation markers25. A lack of all five
G1 cyclins in mESCs increases expression of several neuroectoder-
Fig. 3 | The cell cycle during dissolution of pluripotency and cell mal genes and accelerates neural differentiation in vivo in chimae-
differentiation. G1 phase may provide a window of opportunity for the ric embryos and teratoma assays25, which is an opposite outcome
dissolution of pluripotency, as during this phase many developmental compared to the one described for shRNA-mediated silencing of D
genes contain permissive epigenetic marks (H3K4me3 and cyclins in hESCs68. In agreement with the genetic data, several other
5-hydroxymethylcytosine). Cyclin B1 actively prevents pluripotency exit. reports document that cyclin D–CDK4/6 activity inhibits neuro-
Increased expression of D cyclins in late G1 triggers the phosphorylation ectodermal differentiation in vivo, whereas its silencing promotes
and cytoplasmic retention of Smad2 and Smad3, thereby inhibiting neuroectodermal fate76–80. One possible explanation for these dis-
endodermal differentiation. In addition, D cyclins directly repress crepancies is that the molecular mechanism of lineage specification
the expression of endodermal genes and augment the expression of is fundamentally different in mouse and human cells.
neuroectodermal genes. 5hmC, 5-hydroxymethylcytosine. Whereas cells in G1 are prone to differentiation, pathways oper-
ating in S and G2 phases may actively repress the dissolution of the
pluripotent state44. At the molecular level, it was suggested that
cyclin B1 might represent a key component of such pluripotency-
kinases becomes restricted to specific cell cycle phases16,26–29,31,34,45. promoting pathway during G2 phase44 (Fig. 3). Van Oudenhove
Downregulation of Emi1 causes increased APC/C activity, resulting et al.81 indicated that ESCs differentiating into mesodermal or
in enhanced degradation of APC/C targets, such as cyclins29,30. All endodermal lineages pause during G2 phase owing to upregulation
these changes lead to the appearance of hypophosphorylated RB1 of Wee1, a kinase that inhibits CDK182, which may attenuate the
during G1 phase and subject E2F transcriptional activity to a tight pluripotency-promoting mechanism.
cell-cycle-dependent control16,28.
Several studies document that ESCs initiate differentiation in the Cell cycle alterations during somatic reprogramming
G1 phase59–62 (Fig. 3). This observation was best demonstrated using a Expression of a defined set of transcription factors, such as Oct4,
FUCCI system, which allows the sorting of cells in defined cell cycle Sox2, Klf4 and c-Myc, can convert differentiated somatic cells into
phases24,60,61. One possible mechanistic explanation was provided induced pluripotent stem cells (iPSCs) in a process called somatic
by the observation that cells traversing the G1 phase express higher reprogramming83–85 (Fig. 2). One of the early events accompanying
levels of developmentally regulated transcription factors, suggesting this process is a strong acceleration of the cell cycle86–89. Guo et al.88
that G1 phase ESCs may exist in a ‘lineage-primed’ state61,62. Several demonstrated that during reprogramming of mouse fibroblasts, a
developmentally regulated genes in ESCs are marked by overlap- small subpopulation of ‘ultra-fast’ dividing cells (cell division time
ping activatory (H3K4me3) and inhibitory (H3K27me3) histone of ~8 h) accounts for >99% of the reprogramming. Others have
marks63,64. In late G1, the activating H3K4me3 mark increases on shown that by sorting highly proliferative cells, one can substan-
many of these ‘bivalent’ developmental genes, resulting in increased tially increase the efficiency of reprogramming80. These observa-
levels of their transcripts, which might promote differentiation62. tions suggest that an acceleration of the cell cycle, which occurs
In addition, 5-hydroxymethylation of cytosine increases on some only in a fraction of cells, may represent a rate-limiting factor in
developmental genes during G1 phase, and this mechanism may reprogramming88. Indeed, ectopic expression of several cyclins
contribute to their elevated expression61 (Fig. 3). and CDKs increased the reprogramming efficiency, whereas their
Collectively, these observations indicate that G1 phase represents depletion had the opposite effect27,89,90. Moreover, the reprogram-
a permissive phase for the initiation of cell fate decisions19. In light ming potential of primary murine fibroblasts declines after serial
of these findings, it has been argued that a long G1 phase may enable passaging, concomitantly with their reduced proliferation rates91.
accumulation of factors needed for the dissolution of pluripotency Conversely, genetic lesions in somatic cells that increase cell cycling,
and differentiation, and conversely, a short G1 phase minimizes the such as the inactivation of p19ARF/p16INK4a, p53, or p21Cip1, increased
exposure to differentiation-promoting signals and helps to maintain the efficiency of reprogramming91–93.
the pluripotent state65–67. Work by Pauklin et al. has further extended At the molecular level, CDK2-mediated phosphorylation of
this model68. The authors reported that during early G1 phase of Sox2 has shown to promote the ability of Sox2 to establish pluripo-
hESCs, when D cyclins are not yet expressed, Smad2 and Smad3 tency during reprogramming in combination with other factors94,
transcription factors bind and activate endoderm genes, thereby consistent with an observation that cyclin E–CDK2 stabilizes
specifying endodermal differentiation. Upregulation of cyclin the Sox2 protein25. According to another study27, cyclin B1 and
D–CDK4/6 kinases during late G1 phase leads to phosphorylation CDK1 play a rate-limiting role in somatic-cell reprogramming by
of Smad2/3, thereby blocking entry of Smad2/3 into the nucleus. upregulating and maintaining the expression of the reprogramming

Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology 1063


Review Article NaTuRe Cell BIoloGy

p27 Ngn2

p57 Mash1

P
Ngn2 Ngn2
Neurogenic genes
P
Sox2 CDKs Ngn2 CCND1, CCNE2

NSC Differentiation

Sox2 Neurogenic genes

RBL1 HES1 Neuron

Fig. 4 | The cell cycle in neurogenesis. A decrease of CDK activity during neurogenic divisions enables the transactivation of proneural genes by a
truncated form of Sox2 and Ngn2. Ngn2 in turn inhibits the expression of G1 cyclins. A cell cycle inhibitor p27Kip1 stabilizes Ngn2, whereas p57Kip2 interacts
with proneural factor Mash1 and represses its transcriptional activity. RBL1 modulates the Notch pathway and affects the expression of its target genes.
NSC, neural stem cell.

factor LIN2895. Inactivation of RB1 promotes reprogramming via a neural stem/progenitor cells, cyclin A- and B-dependent kinases
cell-cycle-independent mechanism, related to the ability of RB1 to phosphorylate the proneural basic helix-loop-helix transcription
directly bind pluripotency genes to repress their expression96. factor neurogenin 2 (Ngn2), a master regulator of neuronal dif-
The organization of the cell cycle in iPSCs—namely, a rapid divi- ferentiation. This phosphorylation inhibits the ability of Ngn2 to
sion time and a very short G1 phase (~2.5 h)—is similar to that in bind neurogenic genes and promote neurogenesis106. According to
ESCs89,97. Similarly to ESCs, depletion of CDK1 in reprogrammed this model, upon lengthening of G1 phase, decreased CDK activity
iPSCs also attenuates the pluripotent state46. Collectively, these relieves this repression and allows Ngn2 to turn on neurogenesis106.
observations suggest that the achievement of a unique ‘ESC-like’ Interestingly, Ngn2 can repress expression of cyclins D1 and E2107,
cell cycle organization in the process of reprogramming is function- suggesting the presence of a positive-feedback loop that enforces the
ally linked to the acquisition of pluripotency97. However, the exact exit from the cell cycle during neural differentiation.
molecular mechanisms by which cell cycle proteins help to acquire It should be noted that an opposite conclusion has also been
and enforce the pluripotent state in iPSCs remain largely unknown. reported108; namely, in embryonic chick spinal cord, cyclin D1
serves to promote neurogenesis through a cell-cycle-independent
Cell cycle machinery in neural stem cells function mediated by Hes6. Moreover, as mentioned before, Pauklin
Similarly to pluripotent ESCs, embryonic neural stem/progenitor et al. reported that in hESCs, cyclin D1 and CDK4 promote neuro-
cells display rapid cell division times (~8 h)98–100. Interestingly, a sub- genic differentiation68,69.
population of slowly dividing neural progenitors was also identified In addition to cyclins and CDKs, other cell cycle proteins were
that could give rise to adult neural stem cells (NSCs)101. Expression shown to play roles in neurogenesis. For instance, p27Kip1 can pro-
of a cell cycle inhibitor, p57Kip2, is essential to maintain this slow- mote neuronal differentiation by stabilizing Ngn2 protein indepen-
dividing pool and is required for the emergence of adult NSCs101. As dently of its ability to inhibit CDKs109. In contrast, a related inhibitor,
development proceeds, the cell cycle length of the embryonic neu- p57Kip2, interacts with proneural basic helix-loop-helix factor Mash1
ral progenitor cells increases (to up to ~18 h), owing to a four-fold and represses its transcriptional activity, thereby inhibiting neuro-
increase in the length of G180,98,99,102. genesis110. In addition, RBL1 protein regulates neural precursor cells
Manipulations that prevent the physiological lengthening of in the mammalian brain by repressing the Notch1 pathway111. Lastly,
G1 phase, such as ectopic overexpression of cyclin D1, cyclin E1, CDK inhibitors p27Kip1, p57Kip2 and RB1 play cell-cycle-independent
or CDK4, increase self-renewal and inhibit neurogenic differentia- roles in regulating neuronal cell migration109,112–117.
tion76–78. Conversely, depletion of cyclin D1 and CDK4 or treatment In contrast to highly proliferative ESCs, which are the focus
with CDK4- or pan-CDK inhibitors stimulates neurogenesis76,80,103. of this Review, tissue-specific stem cells of adult organisms are
Likewise, NSCs of Cdk2/Cdk4 double-knockout mice display an largely quiescent65,66. It was proposed that their G0 arrest serves the
increased propensity for neuronal differentiation, resulting in same purpose as G1 phase truncation in ESCs, namely to limit the
enhanced neurogenic divisions in the brains of these embryos79. window of opportunity for cell differentiation66. Several cell cycle
Intriguingly, studies of the developing primate cortex revealed that proteins, including CDK inhibitors, can enforce the quiescent state
local variations in levels of cyclin E and p27Kip1 in neuronal precur- in adult stem cells118–124, as detailed in excellent Reviews on this sub-
sors residing in different areas correlate with the lengths of their cell ject published elsewhere125,126.
cycles (and with that of G1), suggesting that area-specific levels of
cyclin-E-associated kinase might locally influence neurogenesis104. Conclusions and outlook
Collectively, these analyses strongly suggest that the activity of One of the major questions in the field is whether the unusual
cyclin–CDK kinases inhibits neuronal differentiation. Lim et al.105 cell cycle organization seen in ESCs and iPSCs plays a direct role
proposed that in self-renewing NSCs, phosphorylation of Sox2 in maintaining pluripotency. In addition, it remains to be resolved
by high levels of cyclin-dependent kinases enhances the ability whether the reorganization of the cell cycle upon pluripotency dis-
of Sox2 to inhibit neuronal differentiation105. When the activity solution represents the cause or consequence of cell differentiation.
of CDKs decreases, the shift towards non-phosphorylated Sox2 The combined evidence to date suggests that cell cycle proteins
allows the proteolytically cleaved, truncated Sox2 species to bind are causally involved in these processes; however, the mechanistic
and transactivate proneural genes, thereby promoting neurogen- underpinnings are only beginning to emerge. The physiological
esis105 (Fig. 4). Ali et al.106 postulated that in rapidly proliferating role of very high CDK1 and CDK2 kinase activities in pluripotent

1064 Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology


NaTuRe Cell BIoloGy Review Article
ESCs remains a mystery. These kinases may likely phosphorylate 14. Yeeles, J. T., Deegan, T. D., Janska, A., Early, A. & Diffley, J. F. Regulated
additional substrates in stem cells that are different from those eukaryotic DNA replication origin firing with purified proteins. Nature 519,
431–435 (2015).
in differentiated cells, and this might contribute to pluripotency 15. Gavet, O. & Pines, J. Progressive activation of CyclinB1-Cdk1 coordinates
through some currently unknown mechanisms. Proteome-wide entry to mitosis. Dev. Cell 18, 533–543 (2010).
identification of cyclin–CDK substrates in pluripotent cells and 16. Boward, B., Wu, T. & Dalton, S. Concise review: control of cell fate through
during pluripotency dissolution will help to decipher the full rep- cell cycle and pluripotency networks. Stem Cells 34, 1427–1436 (2016).
17. Lawson, K. A., Meneses, J. J. & Pedersen, R. A. Clonal analysis of epiblast
ertoire of cyclin–CDK functions in these processes. The role of cell
fate during germ layer formation in the mouse embryo. Development 113,
cycle proteins in cell fate specification and differentiation remains 891–911 (1991).
largely unclear, with different reports proposing different mecha- 18. Snow, M. H. L. Gastrulation in the mouse: growth and regionalization of
nisms and outcomes. Combinations of genome- and proteome- the epiblast. J. Embryol. Exp. Morphol. 42, 293–303 (1977).
wide approaches might help to address this point. The contribution 19. Dalton, S. Linking the cell cycle to cell fate decisions. Trends Cell Biol. 25,
592–600 (2015).
of 3D chromatin conformation, promoter-enhancer looping, insu- 20. Stead, E. et al. Pluripotent cell division cycles are driven by ectopic Cdk2,
lated neighborhoods and topologically associating domains, as well cyclin A/E and E2F activities. Oncogene 21, 8320–8333 (2002).
as enhancer repertoire and decommissioning has recently been 21. Solter, D., Skreb, N. & Damjanov, I. Cell cycle analysis in the mouse
acknowledged in pluripotency and differentiation127–132, however it EGG-cylinder. Exp. Cell Res. 64, 331–334 (1971).
remains unclear whether these processes are linked to cell cycle pro- 22. Smith, R. K. & Johnson, M. H. Analysis of the third and fourth cell cycles
of mouse early development. J. Reprod. Fertil. 76, 393–399 (1986).
teins. Another largely unaddressed issue is the heterogeneity within 23. Savatier, P., Huang, S., Szekely, L., Wiman, K. G. & Samarut, J. Contrasting
the stem cell compartments. Single-cell approaches such as live sin- patterns of retinoblastoma protein expression in mouse embryonic stem
gle-cell microscopy9,133–135 or single-cell RNA sequencing136–139 will cells and embryonic fibroblasts. Oncogene 9, 809–818 (1994).
help to ascribe specific molecular functions to cell cycle proteins at 24. Coronado, D. et al. A short G1 phase is an intrinsic determinant of naïve
discreet stages of fate specification and cell differentiation. embryonic stem cell pluripotency. Stem Cell Res. 10, 118–131 (2013).
25. Liu, L. et al. G1 cyclins link proliferation, pluripotency and differentiation of
An improved understanding of the cell cycle machinery in stem embryonic stem cells. Nat. Cell Biol. 19, 177–188 (2017).
cell compartments will likely have important implications for regen- 26. Fujii-Yamamoto, H., Kim, J. M., Arai, K. & Masai, H. Cell cycle and
erative medicine. For example, a transient expression of cyclin D1 developmental regulations of replication factors in mouse embryonic stem
and CDK4 in NSCs in vivo resulted in increased neurogenesis and cells. J. Biol. Chem. 280, 12976–12987 (2005).
improved neuronal function140. Other work revealed the utility of 27. Wang, X. Q. et al. CDK1-PDK1-PI3K/Akt signaling pathway regulates
embryonic and induced pluripotency. Cell Death Differ. 24, 38–48 (2017).
inhibiting cell cycle proteins to promote hepatic and pancreatic dif- 28. White, J. et al. Developmental activation of the Rb-E2F pathway and
ferentiation68,141. Given the growing armamentarium of specific cell establishment of cell cycle-regulated cyclin-dependent kinase activity during
cycle inhibitors82, these studies offer opportunities to specifically embryonic stem cell differentiation. Mol. Biol. Cell 16, 2018–2027 (2005).
promote cell differentiation into a lineage of interest. Conversely, 29. Bar-On, O., Shapira, M., Skorecki, K., Hershko, A. & Hershko, D. D.
activation of the cell cycle machinery can be used as a mean to aug- Regulation of APC/C (Cdh1) ubiquitin ligase in differentiation of human
embryonic stem cells. Cell Cycle 9, 1986–1989 (2010).
ment somatic reprogramming27,89,90. For these reasons, the elucida- 30. Ballabeni, A. et al. Cell cycle adaptations of embryonic stem cells. Proc. Natl
tion of the full range of molecular functions of cell cycle proteins in Acad. Sci. USA 108, 19252–19257 (2011).
stem cell self-renewal, differentiation and reprogramming should 31. Savatier, P., Lapillonne, H., van Grunsven, L. A., Rudkin, B. B. & Samarut, J.
provide important insights to help increase the efficiency of these Withdrawal of differentiation inhibitory activity/leukemia inhibitory factor
up-regulates D-type cyclins and cyclin-dependent kinase inhibitors in
processes in a therapeutically meaningful way.
mouse embryonic stem cells. Oncogene 12, 309–322 (1996).
32. Wianny, F. et al. G1-phase regulators, cyclin D1, cyclin D2, and cyclin D3:
Received: 12 December 2018; Accepted: 24 July 2019; up-regulation at gastrulation and dynamic expression during neurulation.
Published online: 2 September 2019 Dev. Dyn. 212, 49–62 (1998).
33. Faast, R. et al. Cdk6-cyclin D3 activity in murine ES cells is resistant to
References inhibition by p16(INK4a). Oncogene 23, 491–502 (2004).
1. Malumbres, M. & Barbacid, M. To cycle or not to cycle: a critical decision 34. Fluckiger, A. C. et al. Cell cycle features of primate embryonic stem cells.
in cancer. Nat. Rev. Cancer 1, 222–231 (2001). Stem Cells 24, 547–556 (2006).
2. Sherr, C. J. & Roberts, J. M. Living with or without cyclins and cyclin- 35. Nichols, J. & Smith, A. Naive and primed pluripotent states. Cell Stem Cell
dependent kinases. Genes Dev. 18, 2699–2711 (2004). 4, 487–492 (2009).
3. Morgan, D. O. The Cell Cycle: Principles of Control (OUP/New Science 36. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal.
Press, London, 2007). Nature 453, 519–523 (2008).
4. Satyanarayana, A. & Kaldis, P. Mammalian cell-cycle regulation: several 37. Ter Huurne, M., Chappell, J., Dalton, S. & Stunnenberg, H. G. Distinct
Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene cell-cycle control in two different states of mouse pluripotency. Cell Stem
28, 2925–2939 (2009). Cell 21, 449–455.e4 (2017).
5. Sherr, C. J. Growth factor-regulated G1 cyclins. Stem Cells 12, 38. Brons, I. G. et al. Derivation of pluripotent epiblast stem cells from
47–55 (1994). mammalian embryos. Nature 448, 191–195 (2007).
6. Geng, Y. et al. Rescue of cyclin D1 deficiency by knockin cyclin E. Cell 97, 39. Tesar, P. J. et al. New cell lines from mouse epiblast share defining features
767–777 (1999). with human embryonic stem cells. Nature 448, 196–199 (2007).
7. Hwang, H. C. & Clurman, B. E. Cyclin E in normal and neoplastic cell 40. Neganova, I., Zhang, X., Atkinson, S. & Lako, M. Expression and functional
cycles. Oncogene 24, 2776–2786 (2005). analysis of G1 to S regulatory components reveals an important role for
8. Narasimha, A. M. et al. Cyclin D activates the Rb tumor suppressor by CDK2 in cell cycle regulation in human embryonic stem cells. Oncogene 28,
mono-phosphorylation. eLife 3, e02872 (2014). 20–30 (2009).
9. Cappell, S. D., Chung, M., Jaimovich, A., Spencer, S. L. & Meyer, T. 41. Becker, K. A. et al. Self-renewal of human embryonic stem cells is
Irreversible APC(Cdh1) inactivation underlies the point of no return for supported by a shortened G1 cell cycle phase. J. Cell. Physiol. 209,
cell-cycle entry. Cell 166, 167–180 (2016). 883–893 (2006).
10. Manchado, E., Eguren, M. & Malumbres, M. The anaphase-promoting 42. Conklin, J. F., Baker, J. & Sage, J. The RB family is required for the
complex/cyclosome (APC/C): cell-cycle-dependent and -independent self-renewal and survival of human embryonic stem cells. Nat. Commun. 3,
functions. Biochem. Soc. Trans. 38, 65–71 (2010). 1244 (2012).
11. Pines, J. Cubism and the cell cycle: the many faces of the APC/C. Nat. Rev. 43. Filipczyk, A. A., Laslett, A. L., Mummery, C. & Pera, M. F. Differentiation is
Mol. Cell Biol. 12, 427–438 (2011). coupled to changes in the cell cycle regulatory apparatus of human
12. Heller, R. C. et al. Eukaryotic origin-dependent DNA replication in vitro embryonic stem cells. Stem Cell Res. 1, 45–60 (2007).
reveals sequential action of DDK and S-CDK kinases. Cell 146, 80–91 (2011). 44. Gonzales, K. A. et al. Deterministic restriction on pluripotent state
13. Walter, J. C. Evidence for sequential action of cdc7 and cdk2 protein dissolution by cell-cycle pathways. Cell 162, 564–579 (2015).
kinases during initiation of DNA replication in Xenopus egg extracts. J. Biol. 45. Zhang, W. W. et al. Cdk1 is required for the self-renewal of mouse
Chem. 275, 39773–39778 (2000). embryonic stem cells. J. Cell. Biochem. 112, 942–948 (2011).

Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology 1065


Review Article NaTuRe Cell BIoloGy
46. Neganova, I. et al. CDK1 plays an important role in the maintenance of 78. Pilaz, L. J. et al. Forced G1-phase reduction alters mode of division, neuron
pluripotency and genomic stability in human pluripotent stem cells. number, and laminar phenotype in the cerebral cortex. Proc. Natl Acad. Sci.
Cell Death Dis. 5, e1508 (2014). USA 106, 21924–21929 (2009).
47. Huskey, N. E. et al. CDK1 inhibition targets the p53-NOXA-MCL1 axis, 79. Lim, S. & Kaldis, P. Loss of Cdk2 and Cdk4 induces a switch from
selectively kills embryonic stem cells, and prevents teratoma formation. proliferation to differentiation in neural stem cells. Stem Cells 30,
Stem Cell Reports 4, 374–389 (2015). 1509–1520 (2012).
48. Kim, H. J. et al. Cyclin-dependent kinase 1 activity coordinates the 80. Roccio, M. et al. Predicting stem cell fate changes by differential cell cycle
chromatin associated state of Oct4 during cell cycle in embryonic stem progression patterns. Development 140, 459–470 (2013).
cells. Nucleic Acids Res. 46, 6544–6560 (2018). 81. Van Oudenhove, J. J. et al. Lineage-specific early differentiation of
49. Kozar, K. et al. Mouse development and cell proliferation in the absence of human embryonic stem cells requires a G2 cell cycle pause. Stem Cells 34,
D-cyclins. Cell 118, 477–491 (2004). 1765–1775 (2016).
50. Geng, Y. et al. Cyclin E ablation in the mouse. Cell 114, 431–443 (2003). 82. Otto, T. & Sicinski, P. Cell cycle proteins as promising targets in cancer
51. Kalaszczynska, I. et al. Cyclin A is redundant in fibroblasts but essential in therapy. Nat. Rev. Cancer 17, 93–115 (2017).
hematopoietic and embryonic stem cells. Cell 138, 352–365 (2009). 83. Park, I. H. et al. Reprogramming of human somatic cells to pluripotency
52. Zhang, X. et al. A role for NANOG in G1 to S transition in human with defined factors. Nature 451, 141–146 (2008).
embryonic stem cells through direct binding of CDK6 and CDC25A. 84. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from
J. Cell Biol. 184, 67–82 (2009). mouse embryonic and adult fibroblast cultures by defined factors. Cell 126,
53. Lee, J., Go, Y., Kang, I., Han, Y. M. & Kim, J. Oct-4 controls cell-cycle 663–676 (2006).
progression of embryonic stem cells. Biochem. J. 426, 171–181 (2010). 85. Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent
54. Zhao, R. et al. A nontranscriptional role for Oct4 in the regulation of ES-cell-like state. Nature 448, 318–324 (2007).
mitotic entry. Proc. Natl Acad. Sci. USA 111, 15768–15773 (2014). 86. Cacchiarelli, D. et al. Integrative analyses of human reprogramming reveal
55. Kareta, M. S., Sage, J. & Wernig, M. Crosstalk between stem cell and cell dynamic nature of induced pluripotency. Cell 162, 412–424 (2015).
cycle machineries. Curr. Opin. Cell Biol. 37, 68–74 (2015). 87. Mikkelsen, T. S. et al. Dissecting direct reprogramming through integrative
56. Varlakhanova, N. V. et al. myc maintains embryonic stem cell pluripotency genomic analysis. Nature 454, 49–55 (2008).
and self-renewal. Differentiation 80, 9–19 (2010). 88. Guo, S. et al. Nonstochastic reprogramming from a privileged somatic cell
57. Amati, B., Alevizopoulos, K. & Vlach, J. Myc and the cell cycle. state. Cell 156, 649–662 (2014).
Front. Biosci. 3, d250–d268 (1998). 89. Ruiz, S. et al. A high proliferation rate is required for cell reprogramming
58. Becker, K. A., Stein, J. L., Lian, J. B., van Wijnen, A. J. & Stein, G. S. Human and maintenance of human embryonic stem cell identity. Curr. Biol. 21,
embryonic stem cells are pre-mitotically committed to self-renewal and 45–52 (2011).
acquire a lengthened G1 phase upon lineage programming. J. Cell. Physiol. 90. Edel, M. J. et al. Rem2 GTPase maintains survival of human embryonic
222, 103–110 (2010). stem cells as well as enhancing reprogramming by regulating p53 and cyclin
59. Sela, Y., Molotski, N., Golan, S., Itskovitz-Eldor, J. & Soen, Y. Human D1. Genes Dev. 24, 561–573 (2010).
embryonic stem cells exhibit increased propensity to differentiate during 91. Utikal, J. et al. Immortalization eliminates a roadblock during cellular
the G1 phase prior to phosphorylation of retinoblastoma protein. Stem Cells reprogramming into iPS cells. Nature 460, 1145–1148 (2009).
30, 1097–1108 (2012). 92. Hong, H. et al. Suppression of induced pluripotent stem cell generation by
60. Calder, A. et al. Lengthened G1 phase indicates differentiation status in the p53-p21 pathway. Nature 460, 1132–1135 (2009).
human embryonic stem cells. Stem Cells Dev. 22, 279–295 (2013). 93. Kawamura, T. et al. Linking the p53 tumour suppressor pathway to somatic
61. Singh, A. M. et al. Cell-cycle control of developmentally regulated cell reprogramming. Nature 460, 1140–1144 (2009).
transcription factors accounts for heterogeneity in human pluripotent cells. 94. Ouyang, J. et al. Cyclin-dependent kinase-mediated Sox2 phosphorylation
Stem Cell Reports 1, 532–544 (2013). enhances the ability of Sox2 to establish the pluripotent state. J. Biol. Chem.
62. Singh, A. M. et al. Cell-cycle control of bivalent epigenetic domains 290, 22782–22794 (2015).
regulates the exit from pluripotency. Stem Cell Reports 5, 323–336 (2015). 95. Tanabe, K., Nakamura, M., Narita, M., Takahashi, K. & Yamanaka, S.
63. Bernstein, B. E. et al. A bivalent chromatin structure marks key Maturation, not initiation, is the major roadblock during reprogramming
developmental genes in embryonic stem cells. Cell 125, 315–326 (2006). toward pluripotency from human fibroblasts. Proc. Natl Acad. Sci. USA 110,
64. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent 12172–12179 (2013).
and lineage-committed cells. Nature 448, 553–560 (2007). 96. Kareta, M. S. et al. Inhibition of pluripotency networks by the Rb tumor
65. Lange, C. & Calegari, F. Cdks and cyclins link G1 length and differentiation suppressor restricts reprogramming and tumorigenesis. Cell Stem Cell 16,
of embryonic, neural and hematopoietic stem cells. Cell Cycle 9, 39–50 (2015).
1893–1900 (2010). 97. Ghule, P. N. et al. Reprogramming the pluripotent cell cycle: restoration of
66. Orford, K. W. & Scadden, D. T. Deconstructing stem cell self-renewal: an abbreviated G1 phase in human induced pluripotent stem (iPS) cells.
genetic insights into cell-cycle regulation. Nat. Rev. Genet. 9, 115–128 (2008). J. Cell. Physiol. 226, 1149–1156 (2011).
67. Singh, A. M. & Dalton, S. The cell cycle and Myc intersect with 98. Salomoni, P. & Calegari, F. Cell cycle control of mammalian neural stem
mechanisms that regulate pluripotency and reprogramming. Cell Stem Cell cells: putting a speed limit on G1. Trends Cell Biol. 20, 233–243 (2010).
5, 141–149 (2009). 99. Caviness, V. S. Jr., Takahashi, T. & Nowakowski, R. S. Numbers, time and
68. Pauklin, S. & Vallier, L. The cell-cycle state of stem cells determines cell fate neocortical neuronogenesis: a general developmental and evolutionary
propensity. Cell 155, 135–147 (2013). model. Trends Neurosci. 18, 379–383 (1995).
69. Pauklin, S., Madrigal, P., Bertero, A. & Vallier, L. Initiation of stem cell 100. Takahashi, T., Nowakowski, R. S. & Caviness, V. S. Jr. The cell cycle of the
differentiation involves cell cycle-dependent regulation of developmental pseudostratified ventricular epithelium of the embryonic murine cerebral
genes by Cyclin D. Genes Dev. 30, 421–433 (2016). wall. J. Neurosci. 15, 6046–6057 (1995).
70. Bienvenu, F. et al. Transcriptional role of cyclin D1 in development revealed 101. Furutachi, S. et al. Slowly dividing neural progenitors are an embryonic
by a genetic-proteomic screen. Nature 463, 374–378 (2010). origin of adult neural stem cells. Nat. Neurosci. 18, 657–665 (2015).
71. Casimiro, M. C. et al. ChIP sequencing of cyclin D1 reveals a 102. Arai, Y. et al. Neural stem and progenitor cells shorten S-phase on
transcriptional role in chromosomal instability in mice. J. Clin. Invest. 122, commitment to neuron production. Nat. Commun. 2, 154 (2011).
833–843 (2012). 103. Calegari, F. & Huttner, W. B. An inhibition of cyclin-dependent kinases that
72. Malumbres, M. et al. Mammalian cells cycle without the D-type cyclin- lengthens, but does not arrest, neuroepithelial cell cycle induces premature
dependent kinases Cdk4 and Cdk6. Cell 118, 493–504 (2004). neurogenesis. J. Cell Sci. 116, 4947–4955 (2003).
73. Sicinska, E. et al. Requirement for cyclin D3 in lymphocyte development 104. Lukaszewicz, A. et al. G1 phase regulation, area-specific cell cycle
and T cell leukemias. Cancer Cell 4, 451–461 (2003). control, and cytoarchitectonics in the primate cortex. Neuron 47,
74. Sicinski, P. et al. Cyclin D2 is an FSH-responsive gene involved in gonadal 353–364 (2005).
cell proliferation and oncogenesis. Nature 384, 470–474 (1996). 105. Lim, S. et al. Cyclin-Dependent Kinase-Dependent Phosphorylation
75. Sicinski, P. et al. Cyclin D1 provides a link between development and of Sox2 at Serine 39 Regulates Neurogenesis. Mol. Cell. Biol. 37,
oncogenesis in the retina and breast. Cell 82, 621–630 (1995). e00201-17 (2017).
76. Lange, C., Huttner, W. B. & Calegari, F. Cdk4/cyclinD1 overexpression 106. Ali, F. et al. Cell cycle-regulated multi-site phosphorylation of Neurogenin 2
in neural stem cells shortens G1, delays neurogenesis, and promotes the coordinates cell cycling with differentiation during neurogenesis.
generation and expansion of basal progenitors. Cell Stem Cell 5, Development 138, 4267–4277 (2011).
320–331 (2009). 107. Lacomme, M., Liaubet, L., Pituello, F. & Bel-Vialar, S. NEUROG2 drives
77. Artegiani, B., Lindemann, D. & Calegari, F. Overexpression of cdk4 and cell cycle exit of neuronal precursors by specifically repressing a subset of
cyclinD1 triggers greater expansion of neural stem cells in the adult mouse cyclins acting at the G1 and S phases of the cell cycle. Mol. Cell. Biol. 32,
brain. J. Exp. Med. 208, 937–948 (2011). 2596–2607 (2012).

1066 Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology


NaTuRe Cell BIoloGy Review Article
108. Lukaszewicz, A. I. & Anderson, D. J. Cyclin D1 promotes neurogenesis in 127. Barakat, T. S. et al. Functional dissection of the enhancer repertoire in
the developing spinal cord in a cell cycle-independent manner. Proc. Natl human embryonic stem cells. Cell Stem Cell 23, 276–288.e8 (2018).
Acad. Sci. USA 108, 11632–11637 (2011). 128. Dowen, J. M. et al. Control of cell identity genes occurs in insulated
109. Nguyen, L. et al. p27kip1 independently promotes neuronal differentiation neighborhoods in mammalian chromosomes. Cell 159, 374–387 (2014).
and migration in the cerebral cortex. Genes Dev. 20, 1511–1524 (2006). 129. Hnisz, D., Day, D. S. & Young, R. A. Insulated neighborhoods: structural and
110. Joseph, B. et al. p57Kip2 is a repressor of Mash1 activity and neuronal functional units of mammalian gene control. Cell 167, 1188–1200 (2016).
differentiation in neural stem cells. Cell Death Differ. 16, 1256–1265 (2009). 130. Ji, X. et al. 3D chromosome regulatory landscape of human pluripotent
111. Vanderluit, J. L. et al. p107 regulates neural precursor cells in the cells. Cell Stem Cell 18, 262–275 (2016).
mammalian brain. J. Cell Biol. 166, 853–863 (2004). 131. Kishi, Y. & Gotoh, Y. Regulation of chromatin structure during neural
112. Ferguson, K. L. et al. A cell-autonomous requirement for the cell cycle development. Front. Neurosci. 12, 874 (2018).
regulatory protein, Rb, in neuronal migration. EMBO J. 24, 4381–4391 (2005). 132. Whyte, W. A. et al. Enhancer decommissioning by LSD1 during embryonic
113. Godin, J. D. et al. p27(Kip1) is a microtubule-associated protein that stem cell differentiation. Nature 482, 221–225 (2012).
promotes microtubule polymerization during neuron migration. Dev. Cell 133. Cappell, S. D. et al. EMI1 switches from being a substrate to an inhibitor of
23, 729–744 (2012). APC/CCDH1 to start the cell cycle. Nature 558, 313–317 (2018).
114. Itoh, Y., Masuyama, N., Nakayama, K., Nakayama, K. I. & Gotoh, Y. The 134. Sakaue-Sawano, A. et al. Visualizing spatiotemporal dynamics of
cyclin-dependent kinase inhibitors p57 and p27 regulate neuronal migration multicellular cell-cycle progression. Cell 132, 487–498 (2008).
in the developing mouse neocortex. J. Biol. Chem. 282, 390–396 (2007). 135. Spencer, S. L. et al. The proliferation-quiescence decision is controlled by a
115. Kawauchi, T., Chihama, K., Nabeshima, Y. & Hoshino, M. Cdk5 bifurcation in CDK2 activity at mitotic exit. Cell 155, 369–383 (2013).
phosphorylates and stabilizes p27kip1 contributing to actin organization 136. Cao, J. et al. The single-cell transcriptional landscape of mammalian
and cortical neuronal migration. Nat. Cell Biol. 8, 17–26 (2006). organogenesis. Nature 566, 496–502 (2019).
116. Kawauchi, T., Shikanai, M. & Kosodo, Y. Extra-cell cycle regulatory 137. Guo, H. et al. Single-cell RNA sequencing of human embryonic stem cell
functions of cyclin-dependent kinases (CDK) and CDK inhibitor proteins differentiation delineates adverse effects of nicotine on embryonic
contribute to brain development and neurological disorders. Genes Cells 18, development. Stem Cell Reports 12, 772–786 (2019).
176–194 (2013). 138. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and
117. McClellan, K. A. et al. Unique requirement for Rb/E2F3 in neuronal early organogenesis. Nature 566, 490–495 (2019).
migration: evidence for cell cycle-independent functions. Mol. Cell. Biol. 27, 139. Zhao, T. et al. Single-cell RNA-seq reveals dynamic early embryonic-
4825–4843 (2007). like programs during chemical reprogramming. Cell Stem Cell 23,
118. Cheng, T., Rodrigues, N., Dombkowski, D., Stier, S. & Scadden, D. T. Stem 31–45.e37 (2018).
cell repopulation efficiency but not pool size is governed by p27(kip1). 140. Bragado Alonso, S. et al. An increase in neural stem cells and olfactory bulb
Nat. Med. 6, 1235–1240 (2000). adult neurogenesis improves discrimination of highly similar odorants.
119. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/ EMBO J. 38, e98791 (2019).
waf1. Science 287, 1804–1808 (2000). 141. Azzarelli, R. et al. Multi-site Neurogenin3 Phosphorylation Controls
120. Furutachi, S., Matsumoto, A., Nakayama, K. I. & Gotoh, Y. p57 controls Pancreatic Endocrine Differentiation. Dev. Cell 41, 274–286.e5 (2017).
adult neural stem cell quiescence and modulates the pace of lifelong
neurogenesis. EMBO J. 32, 970–981 (2013).
121. Kippin, T. E., Martens, D. J. & van der Kooy, D. p21 loss compromises the Acknowledgements
This work was supported by grant R01CA202634 (to P.S.).
relative quiescence of forebrain stem cell proliferation leading to exhaustion
of their proliferation capacity. Genes Dev. 19, 756–767 (2005).
122. Matsumoto, A. et al. p57 is required for quiescence and maintenance of Competing interests
adult hematopoietic stem cells. Cell Stem Cell 9, 262–271 (2011). P.S. has been a consultant at Novartis, Genovis, Guidepoint, The Planning Shop, ORIC
123. Viatour, P. et al. Hematopoietic stem cell quiescence is maintained by Pharmaceuticals and Exo Therapeutics; his laboratory receives research funding from
compound contributions of the retinoblastoma gene family. Cell Stem Cell Novartis. W.M. is currently an employee of Cedilla Therapeutics.
3, 416–428 (2008).
124. Yuan, Y., Shen, H., Franklin, D. S., Scadden, D. T. & Cheng, T. In vivo
self-renewing divisions of haematopoietic stem cells are increased in the Additional information
absence of the early G1-phase inhibitor, p18INK4C. Nat. Cell Biol. 6, Reprints and permissions information is available at www.nature.com/reprints.
436–442 (2004). Correspondence should be addressed to P.S.
125. Cheung, T. H. & Rando, T. A. Molecular regulation of stem cell quiescence.
Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013). Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
126. van Velthoven, C. T. J. & Rando, T. A. Stem cell quiescence: dynamism, published maps and institutional affiliations.
restraint, and cellular idling. Cell Stem Cell 24, 213–225 (2019). © Springer Nature Limited 2019

Nature Cell Biology | VOL 21 | SEPTEMBER 2019 | 1060–1067 | www.nature.com/naturecellbiology 1067

You might also like