Sei sulla pagina 1di 8

REVIEW ARTICLE 279

Molecular Mechanisms Involved


in Self-Renewal and Pluripotency
of Embryonic Stem Cells
NA LIU, MIN LU, XUEMEI TIAN, AND ZHONGCHAO HAN*
State Key Laboratory of Experimental Hematology, National Research Center for Stem Cell Engineering and Technology,
Institute of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China

Embryonic stem cells (ES cells) are derived from inner cell mass (ICM). The self-renewal and pluripotency are the main specificities of ES
cells, which are likely to reveal a deeper understanding of human cellular biology and which are considered to be promising sources for cell
therapy to treat patients with degenerative diseases in clinical. Growth of ES cells as a pluripotent population requires a balance between
survival, proliferation, and self-renewal signals. In fact, the precise mechanism that regulates stem cell self-renewal and pluripotency
remains largely unknown. Recently, in vitro and in vivo studies have identified several genetic regulators that may play important roles in
the self-renewal and pluripotency process of human and mouse ES cells, including extracellular signaling factors, transcription factors, cell-
cycle regulators, microRNA, genes implicated in chromosomal stability, and DNA methylation. In this review, we will summarize the
currently known molecular regulators for ES cells self-renewal, and we will propose some possibilities to explain the ways in which these
distinct pathways might interact.
J. Cell. Physiol. 211: 279–286, 2007. ß 2006 Wiley-Liss, Inc.

Embryonic stem cells (ES cells) were first established from the Signaling Transduction Pathways
inner cell mass (ICM) of mouse blastocysts in 1981 (Evans and LIF-STAT3 pathway
Kaufman, 1981). Human ES cells were first derived in 1998
(Thomson et al., 1998), which possess the remarkable property Mouse embryonic stem cells (mES cells) could be expanded
of pluripotency, giving rise to all cells of the organism. For this continuously in culture when co-cultured with mitotic
reason, ES cells are thought to hold great promise for inactivated embryonic fibroblast cells and resulted in stem cells
regenerative medicine (Pera and Trounson, 2004; Liew et al., capable of multi-lineage differentiation (Evans and Kaufman,
2005). Therefore, a detailed understanding of the mechanisms 1981). The factor capable of promoting self-renewal and
that enable propagation of ES cells in a pluripotent state is inhibiting differentiation provided by the feeder cells was later
essential to realize their therapeutic potential. Now, a great identified as leukemia inhibitory factor (LIF), a member of the
deal of effort is focused on experiments aimed at improving IL-6 cytokine family (Smith et al., 1988; Williams et al., 1988).
culture conditions, generating new ES cell lines suitable for LIF stimulates mES cells through the gp130, which works as a
clinical purpose and founding efficient differentiation method to heterodimer together with LIFR. Activation of gp130 leads to
produce human cells suitable for transplantation. All of these the activation of the Janus-associated tyrosine kinase (JAK) and
need to understand the molecular mechanism involved in of signal transducer and activation of transcription (STAT). The
self-renewal and pluripotency in ES cells. Understanding the ability of LIF to maintain the mES cells self-renewal is dependent
molecular mechanism by which pluripotency is maintained in upon activation of STAT3 (Niwa et al., 1998), whose activation
human ES cells is important for the development of improved is sufficient to prevent mES cells differentiation in the presence
methods to derive and culture human ES cells. It is of equal of serum (Matsuda et al., 1999). In addition to the activation of
importance to understand the changes which take place in such STAT3, LIF also stimulates the activation of the
signaling networks as a cause or consequence of ES cells mitogen-activated protein kinase (MAPK), whose activation
differentiation, as this may help us to develop better strategies promotes differentiation. Suppression of ERKs signal can
to direct differentiation into defined populations of cells for promote mES cells self-renewal (Burdon et al., 1999). MAPK
therapeutic use (Armstrong et al., 2006). signal may be a negative regulation mechanism for STAT3. mES
In fact, the precise mechanism that regulates stem cell cells can maintain their property in the presence of LIF due to
self-renewal and pluripotency remains largely unknown. Thus, the balance of the STAT3 activation and MAPK effect.
investigation into the molecular and cellular mechanisms of Although the essential functions of LIF and STAT3 in self-
stem cell self-renewal and pluripotency should help meet these renewal of mES cells have been thoroughly documented, the
challenges and provide the necessary tools to harness the downstream target genes of activated STAT3 in mES cells have
regenerative potential of ES cells for therapeutical purposes. remained elusive. Recently, in order to isolate these genes,
The past few years have seen remarkable progress in our researchers performed a microarray-based kinetic comparison
understanding of ES cells biology. Pluripotency of mouse and of LIF-stimulated (undifferentiated) ES cells versus ES cells
human ES cells is regulated by distinct signaling pathways.
Recently, in vitro and in vivo studies have identified several
genetic regulators that may play important roles in the human
and mouse ES cell self-renewal and pluripotency process. Extra
signaling factors, transcription factors, cell-cycle regulators, *Correspondence to: Zhongchao Han, Institute of Hematology,
microRNA, genes implicated in chromosomal stability, and Chinese Academy of Medical Sciences and Peking Union Medical
College, Tianjin 300020, China. E-mail: tihzchan@public.tpt.tj.cn
DNA methylation. In this review, we will summarize the
currently known molecular regulators for ES cells self-renewal, Received 2 November 2006; Accepted 3 November 2006
and we will propose some possibilities to explain the ways in DOI: 10.1002/jcp.20978
which these distinct pathways might interact.

ß 2 0 0 6 W I L E Y - L I S S , I N C .
280 LIU ET AL.

induced to differentiate by shutting down STAT3 activity maintain the pluripotency of mouse ES cells independent of
through either LIF deprivation or, more specifically, expression Wnt/b-catenin signaling. PI3K/Akt signaling can be activated by
of a STAT3 dominant-negative mutant. In this experiment, the growth factors, which participate in ES cell pluripotency,
some growth factors such as Lefty1 or transcriptional such as LIF and bFGF (Jirmanova et al., 2002; Xu et al., 2005b).
regulators such as Id1 and Id2 and the groucho-like protein Akt controls various cellular processes through a number of
Aes1 were found to be related to STAT3. Aes1 may be the target proteins. The mTOR is a critical downstream effector for
direct target of STAT3 (Sekkai et al., 2005). There may be some PI3K/Akt signaling. But the addition of rapamycin, an inhibitor of
links between LIF-STAT3 and signaling pathways involved in mTOR, to the Akt-expressing mouse ES cells inhibited
the maintenance of stem cell properties such as BMP and Wnt. proliferation, but did not induce differentiation. Additional
Furthermore, Esg-1 (ES-cell-specific protein) may be another activation of mTOR is not sufficient for the LIF-independent
direct or indirect target gene of STAT3, although the actual maintenance of pluripotency, suggesting that mTOR is not a
binding and transcriptional activation of Esg-1 by STAT3 major downstream effector in ES cells pluripotency
remains to be tested (Tanaka et al., 2002). These identified maintenance (Watanabe et al., 2006). Short G1 phase
genes are required to better understand the molecular contributes to the ES cells undifferentiation status, and Akt can
mechanisms involved in ES cells self-renewal. promote a G1 phase to S phase transition by facilitating the
Cartwright indicated a role for the transcription factor c-Myc in formation of cyclin/CDK complexes (Brazil et al., 2004). This
self-renewal by functioning as a key target of LIF-STAT3 mechanism may be the key role of Akt for ES cells self-renewal.
signaling. The expression of c-Myc is rapidly downregulated However, overexpression of cyclin D, regulated by Akt, does
within the first 36 h following LIF withdrawal. Expression of not maintain the undifferentiated status of ES cells. Thus, the
c-Myc renders self-renewal independent of LIF, while critical target molecules remain to be identified (Watanabe
expression of a dominant negative form of c-Myc antagonizes et al., 2006). Beside these, ERK pathway may be another
self-renewal and promotes differentiation (Cartwright et al., mechanism for PI3K effect in maintaining ES cells pluripotency
2005). The induction of c-Myc by STAT3 is in agreement with (Armstrong et al., 2006).
infinite proliferation of ES cells through c-Myc induction of the In ES cells, p53 promotes differentiation by suppression of
regulatory subunit of telomerase (Wang et al., 1998). Nanog expression (Lin et al., 2005). This effect depended on the
It is known that BCR-ABL also can stimulate STAT3 activity. phosphorylation status of Ser315 of p53, which is a substrate of
Further studies reveal that BCR-ABL expressing mESC GSK3b (Watcharasit et al., 2002; Qu et al., 2004). GSK3b is
persistently exhibited a primitive morphology, despite LIF negatively regulated by PI3K and Akt (Hay and Sonenberg,
withdrawal (Coppo et al., 2003). In this progress, MEKK1 2004). Taken together, PI3K may contribute to self-renewal of
plays a key role in self-renewal and STAT3 activation in ES cells through GSK3b, p53, and Nanog (Takahashi et al.,
BCR-ABL-transformed mES cells (Nakamura et al., 2005). 2003).
LIFR and gp130 are also expressed in human embryonic stem Furthermore, PI3K/Akt signaling is required to suppress
cells (hES cells) and functional activation of the STAT3 by human apoptosis in ES cells. The density of ES cells is very important for
LIF (Daheron et al., 2004). However, this is not sufficient to ES cells self-renewal. Thus, this may be another mechanism of
maintain hES cell pluripotency in vitro. Furthermore studies PI3K in ES cell self-renewal (Gross et al., 2005).
found that hES cells can maintain their property in vitro, which is
independent of LIF-STAT3 (Humphrey et al., 2004). The Wnt
different effect of LIF-STAT3 in the hES cells and mES cells may
be due to the different stage in their development. Recently, evidence has been presented that the Wnt pathway
could be involved in the maintenance of pluripotency of both
MAPK-ERK pathway human and mouse ES cells. Activation of Wnt pathway by
6-bromoindirubin-3-oxime (BIO), a specific pharmacological
mES cells show high ERK activity when they are stimulated to inhibitor of glycogen synthase kinase-3 (GSK-3), can maintain
undergo differentiation. Suppression of ERK signaling pathway the undifferentiated phenotype in ES cells and sustain
promotes mES cells self-renewal. And, BMP activation inhibits expression of the ES cells specific markers. Wnt signaling is
mES cells differentiation cooperated with LIF due to Id endogenously activated in ES cells and is downregulated upon
expression and ERK inactivation (Burdon et al., 1999; Qi et al., differentiation (Sato et al., 2004). In contrast to LIF-STAT3 and
2004; Lodge et al., 2005). However, hES cells display high-level BMP signaling, the effect of Wnt/b-catenin signaling in ES
basal activity of ERK in undifferentiation status. Its activation self-renewal has no difference between mouse and human ES
relates to the bFGF signaling which can maintain human ES cells cells. Wnt signaling activation can upregulate STAT3
self-renewal for a long time (Dvorak et al., 2005a; Kang et al., expression, suggesting that it has synergistic effect with
2005; Levenstein et al., 2006). ERK activation may be at LIF-STAT3 (Hao et al., 2006). Furthermore, the Wnt signaling
downstream of PI3K, which is also important in human ES cells pathway has been shown to elevate the level of c-Myc, which is
self-renewal (Armstrong et al., 2006). also the target gene of STAT3 that was described above. Thus,
Wnt and LIF-STAT3 may converge on c-Myc (Cartwright et al.,
PI3K pathway 2005; Kristensen et al., 2005).

Phosphoinositide 3-kinase (PI3K) pathway is important for TGFb pathways


proliferation, survival, and maintenance of pluripotency in ES
cells. Eras is specifically expressed in ES cells, which stimulates TGFb is a prototypic member of a large superfamily of related
PI3K. PI3K activation promotes ES cell proliferation (Takahashi growth and differentiation factors. The family has more than
et al., 2003). In addition to proliferation for ES cells, PI3K activity 40 members, including TGFb, Activin, Nodal, and bone
might be also crucial for self-renewal of ES cells (Takahashi et al., morphogenetic proteins (BMPs). These ligands are all
2005). It has been reported that the inhibition of PI3K and Akt associated with ES cells. The TGFb transduces signal from the
induces differentiation of mouse and human ES cells in the membrane to the nucleus by binding to heteromeric complex of
presence of LIF and feeder cells, respectively (Paling et al., 2004; serine/threonine kinase receptors known as TGFb type I and
Kim et al., 2005), suggesting that P13K/Akt signaling is necessary type II receptors. TGFb and activin have high affinity for the type
for the maintenance of ES cell pluripotency. Watanabe et al. II receptors but do not bind to the type I receptors in the
(2006) also demonstrate that activation of Akt signaling can absence of type II receptor, whereas BMPs have higher affinity

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP


MECHANISMS IN ES CELLS SELF-RENEWAL 281

for their type I receptors than for type II (Valdimarsdottir and maintain human ES cells. Human ES cells have most commonly
Mummery, 2005). been cultured in the presence of basic fibroblast growth factor
BMP4. BMP4 is a member of the TGFb superfamily but it (bFGF) either on fibroblast feeder layers or in
functions via a different signaling pathway with TGFb. fibroblast-conditioned medium. bFGF signaling appears to be of
Activation of STAT3 is sufficient for mES cells self-renewal. But central importance to human ES cells self-renewal (Amit et al.,
4 years later, researchers found that LIF-STAT3 cannot 2000; Xu et al., 2001).
maintain mES cells in serum-free medium. That is to say that Investigations that examined the expression of different
there are many unknown factors present in serum which are elements of signaling pathways in human ES cells have shown the
required to co-affect with LIF-STAT3. This serum factor is likely presence of elements of FGF signaling, including all four FGF
to be BMP4 acting via activation of Smad1/5/8. Addition of BMP4 receptors and certain components of their downstream
to the media enables LIF to maintain mES cells in serum-free cascade, which are enriched in undifferentiated human ES cells
culture (Ying et al., 2003). BMP4 have been shown to in comparison to their differentiated derivatives (Bhattacharya
phosphorylate Smad1/5 in both human and mouse ES cells. et al., 2004; Brandenberger et al., 2004; Dvash et al., 2004; Ginis
Smad1/5 activation results in the expression of inhibitor of et al., 2004). Among the four FGFRs, the expression of FGFR1 is
differentiation (Id) protein, which functionally antagonizes most abundant in undifferentiated human ES cells. SU5402
neurogenic bHLH transcription factors and blocks the neural (inhibitor of FGFR) causes rapid differentiation (Dvorak and
differentiation (Ying et al., 2003; Gerrard et al., 2005). Hampl, 2005).
Exogenous expression of Id mimics the effect of BMP4 in mouse Human ES cells are routinely cultured on feeder layers of
ES cells (Ying et al., 2003). Furthermore, BMP4 can maintain fibroblasts in medium that is supplemented with bFGF at the
mouse ES cells in the presence of LIF, possibly by blocking the concentration of 4 ng/ml (Thomson et al., 1998). Due to
MAPK signaling cascade (Qi et al., 2004). In a word, BMP4 expected clinical requirements, many laboratories have begun
contributes to mESC self-renewal; this effect needs LIF culturing human ES cells in a feeder-free system. In this case,
presence. they increased the concentration of bFGF to 8 ng/ml
In contrast to mES cells, BMP4 does not maintain the hES cells (Carpenter et al., 2004). Recently, researchers have shown that
self-renewal and in fact induces hES cells trophoblast or bFGF (40 ng/ml) combined with noggin (inhibitor of BMP4)
primitive endoderm differentiation (Xu et al., 2002). Noggin, an supports the undifferentiated proliferation of human ES cells in
inhibitor of BMP4 signaling, sustains undifferentiated states of the absence of feeder cells (Dvorak et al., 2005a; Wang et al.,
human ES cells in the presence of basic FGF (Xu et al., 2005a). 2005; Xu et al., 2005a). Furthermore, high concentration bFGF
GDF3 is a secreted factor of the TGFb family. It is expressed at (100 ng/ml) alone is sufficient to maintain human ES cells (Xu
high levels during pluripotency and declines dramatically when et al., 2005b). Although it is likely that exogenous bFGF
ES cells undergo differentiation (Sato et al., 2003). GDF3 may supports growth of undifferentiated human ES cells, the
play a role in the ES cells pluripotency, but recent study found mechanism of bFGF action is not yet clearly established. Some
that GDF3 effect is different in human ES cells and mouse ES researchers found that bFGF can stimulate MAPK signaling
cells. In human ES cells, GDF3 overexpression contributes to cascade and induces the expression of c-fos in human ES cells
the undifferentiation status. By contrast, in mouse ES cells, (Kang et al., 2005; Dvorak et al., 2005a). bFGF also can inhibit
reduced GDF3 levels helped to support pluripotency (Levine BMP activity by preventing the nuclear translocation of
and Brivanlou, 2006). GDF3 may be an inhibitor of BMP4; that is phosphorylated Smad1 (Xu et al., 2005a).
why GDF3 function in ES cells opposes to BMP4. In serum-free media not substituted with bFGF, the cells
TGFb/activin/nodal. Gene expression profile indicates differentiate, while its addition makes the cells morphology
TGFb and its correlate factors, including Nodal Cripto Lefty1 more compact and enables prolonged undifferentiated culture
and Lefty2, all expressed at high levels in undifferentiated human under these conditions. In addition, bFGF enhances the cloning
ES cells (Sato et al., 2003). Activin A, a member of the TGFb efficiency of cells (Amit et al., 2000). But the detailed mechanism
superfamily, is secreted by mouse embryonic feeders, and that of bFGF in ES cells with undifferentiated maintenance requires
cultured medium enriched with activin A maintains human ES to be declared.
cells undifferentiated without condition medium (CM) or
STAT3 activation (Beattie et al., 2005). These data are Transcription Factors
supported by James and colleagues who reported that the
TGFb/Activin/Nodal pathway is activated through the Recent experiments have also elucidated transcriptional
transcription factors Smad2/3 in undifferentiated cells. Smad2/3 regulatory circuitry responsible for ES cells self-renewal and
activation suppresses Smad1/5 activity, thus TGFb/Activin/ differentiation, which involves the transcription factors Oct4,
Noda can negatively regulate BMP4 in human ES cells (Beattie Sox2, Nanog, and et al. Some of these factors are expressed
et al., 2005; James et al., 2005). But in mouse ES cells, upon specifically in pluripotent cells, such as Oct4 and Nanog. These
withdrawal of LIF, levels of Smad2/3 phosphorylation are transcriptional factors may be essential for ES cells self-renewal
reduced, but the SB431542-mediated inhibition of Smad2/3 and differentiation. They are switched ON/OFF by input
signaling in the context of LIF resulted in no significant change in environmental signals, such as the signals we described above.
Oct4 levels, suggesting that the Smad2/3 activity is not And they are also regulated by themselves. When these genes
necessary for maintenance of undifferentiated state in mouse ES are expressed, the self-renewal genes are activated, and the
cells (James et al., 2005). BIO can maintain undifferentiation of differentiated genes are repressed. So the ES cells can maintain
human ES cells by activation of Wnt signaling. In this condition, their pluripotency.
the level of Smad2/3 phosphorylation is also elevated (James
et al., 2005). This signaling may be correlated with Wnt. But the Oct4/Sox2
role of the TGFb superfamily in ES self-renewal is not well
understood. Oct4 (also known as Oct3 and encoded by Pou5f1) is a POU
domain-containing transcription factor that binds to an
bFGF pathway octamer sequence, ATGCAAAT. During mouse
preimplantation development, Oct4 expression is activated at
ESC can maintain their pluripotency with feeder cells. And the four-cell stage and is later restricted to the pluripotent cells,
mouse ES cells can also maintain their undifferentiated status in such as ES cells, ICM, and germ cells. Oct4 is highly expressed in
the presence of LIF without feeder cells. However, LIF cannot human and mouse ES cells, and its expression diminishes when

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP


282 LIU ET AL.

these cells differentiate and lose pluripotency. The precise activation by interfering with the recruitment of coactivators
levels of this gene are required to maintain the ES cells state. (Suzuki et al., 2006), suggesting that Nanog may interact with
Loss of Oct4 causes inappropriate differentiation of ES cells into Wnt and BMP4 signaling pathways. These can explain the
trophectoderm, whereas overexpression of Oct4 results in phenomenon that mouse ES cells can maintain self-renewal
differentiation into primitive endoderm and mesoderm (Yeom when overexpressing Nanog, but the capability is reduced
et al., 1996; Niwa, 2001). Several target genes of Oct4 in ES cells (Chambers et al., 2003; Mitsui et al., 2003). Furthermore,
have been identified, and these include Fgf4, Utf1, Opn, Rex1/ Nanog promoter has been suggested as a direct target of the
Zfp42, Fbx15, and Sox2 (Nishimoto et al., 1999; Tomioka et al., Oct4/Sox2 complex through Chip analysis, in vitro-binding
2002; Zeng et al., 2004). Sox2 is an HMG-family protein that experiments and RNAi-mediated knockdowns (Boyer et al.,
occupies many gene targets with Oct4 and is also required in ES 2005; Kuroda et al., 2005; Rodda et al., 2005).
cells with pluripotent sustenance.
LIF does not appear to regulate Oct4, and Oct4 does not appear Network between these factors
to regulate Jak-STAT signaling, suggesting that the Oct4
pathway is a parallel pathway for maintaining ES self-renewal. Oct4, Sox2, and Nanog may be the main transcriptional factors
Many of Oct4 target genes also contain STAT-binding sites, in regulating ES cells pluripotency. Recent studies have enabled
suggesting that the two transcription factors may cooperate in the construction of transcriptional regulatory networks in ES
ES cells. ESG1 (embryo-specific gene 1, originally called Dppa5) cells that provide a foundation for understanding how these
is highly expressed in human ES cells, and is downregulated as ES factors control pluripotency and influence subsequent
cells differentiate as much as in mouse ES cells. ESG1 appears to differentiation events. These key transcription factors have also
be downstream of both STAT3 and Oct4 pathways. But, how been identified that form an intrinsic core-regulatory circuit
ESG1 integrates Oct4 and STAT3 remains to be determined that maintains ES cells in the pluripotent state in vitro. Using
(Tanaka et al., 2002; Ginis et al., 2004). RNA interference method, microarray analysis, and genome
In contrast with its target genes, little is known about upstream wide chromatin immunoprecipitation experiments, numerous
regulators. Oct4 promoter contains conserved distal and target genes bound by Nanog, Oct4, and Sox2 have been
proximal enhancers that can either repress or activate its identified. These factors appear to form a tight transcriptional
expression depending on the binding factors occupying these regulatory circuit that maintains ES cells in a pluripotent state
sites (Pan et al., 2002). The quantitative level of Oct4 is (Boyer et al., 2005, 2006a; Ivanova et al., 2006; Loh et al., 2006;
important for the fate determination of primitive cells. To Rao and Orkin, 2006).
maintain the pluripotent state, it is required that the cells Oct4 activity is modulated by interactions with other
maintain the Oct4 protein level within the narrow window of transcription factors that include Sox2, FoxD3, and ESG1 which
abundance. Its expression can be regulated by itself (Chew et al., are highly expressed in ES cells. Sox2 (SRY-related HMG box2),
2005; Okumura-Nakanishi et al., 2005). This circuit regulation a member of the HMG-domain DNA-binding protein family,
may contribute to a robust maintenance of the steady levels of forms a ternary complex with Oct4 protein in the enhancer
Oct4 in ES cells. Furthermore, Nanog and FoxD3 also can element of target gene and regulates its expression as a binary
activate Oct4 expression (Pan et al., 2006). complex. Sox2 has an expression pattern similar to Oct4. All of
the Oct4 target genes have octamer and sox heptamer
Nanog elements separated by either 0 or 3 bp. Oct4 interacts with
Sox2 to regulate downstream genes. As we described above,
Nanog is another homeobox-containing transcription factor Oct4/Sox2 complex can regulate the Oct4 and Sox2 expression.
with an essential role in maintaining the pluripotent cells of the Beside these target genes, recently, researchers found that
inner cells mass and ES cells. It is expressed in pluripotent cells, Nanog is another target of Oct4/Sox2 complex (Kuroda et al.,
and is absent from differentiated cells. Nanog disruption in ES 2005; Rodda et al., 2005). There is Oct4/Sox2-binding site in the
cells results in differentiation to endoderm lineages. Nanog promoter. That is to say that Oct4/Sox2 complex plays a
Overexpression of Nanog in mouse ES cells renders ES cells key role in maintaining the expression of essential transcription
self-renewal independent LIF, although the cells self-renewal factors in ES cells through autoregulatory and multicomponent
capability is reduced, suggesting that Nanog is a major regulator loop network motifs. Conserved Oct3/4 and Sox2 co-binding
of the pluripotent state (Chambers et al., 2003; Mitsui et al., domains were identified in most ES expressed genes,
2003). The Nanog overexpression effect is neither dependent highlighting the importance of this transcriptional pathway.
on STAT3 activation nor needs BMP4 presence. But the In addition to Oct-sox cooperative binding, interaction
overexpression of Nanog can maintain Id expression (Ying between FoxD3 (forkhead family member D3) and Oct has also
et al., 2003). been reported. FoxD3 appears to be nonessential, and differing
The mechanism through which Nanog regulates stem cell expression has been reported in various human ES cell lines
pluripotency remains entirely unknown. Based on the (Ginis et al., 2004; Richards et al., 2004). As we described above,
differences in gene expression between wild-type and Nanog Oct4 can regulate Nanog expression. Oct4 maintains Nanog
null cells, it has been proposed that Nanog regulates activity by directly activating its promoter when expressed
pluripotency mainly as a transcription repressor for below steady state, yet represses it at par with or above steady
downstream genes that are important for cell differentiation state concentration in ES cells. This can explain the evidence
such as Gata4 and Gata6 (Chambers et al., 2003; Mitsui et al., that overexpression of Oct4 induces differentiation. FoxD3,
2003). However, Nanog can also activate the genes necessary isolated as a transcription factor with restrictive expression in
for self-renewal such as Rex1. Its C-terminus may contribute to ES cells, is also required for the maintenance of embryonic cells
its transcriptional activity (Pan and Pei, 2005; Shi et al., 2006). As in early mouse embryos, suggesting that it may play a role in
we described above, Rex1 is a target gene of Oct4/Sox2 regulating stem cells self-renewal and pluripotency alongside
complex. Rex1 is also directly regulated by Nanog, suggesting Oct4 and Nanog. FoxD3, a novel activator of Nanog,
that Rex1 may be an intersection of Nanog and Oct4/Sox2 counteracts the repressive activity of Oct4 on Nanog promoter
signaling (Shi et al., 2006). And Nanog can also activate Oct4 at the steady state (Pan et al., 2006). These transcription factors,
promoter (Pan et al., 2006). playing important roles in ES cells, form regulatory circuit. This
In a recent experiment, Nanog may be regulated by STAT3 and regulatory circuit maintains ES cells self-renewal and
Brachyury (Brachyury may be a target of Wnt/b-catenin); then pluripotency by sustaining transcription factors expression at
it directly binds to Smad1 and blocks the transcriptional precise level.

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP


MECHANISMS IN ES CELLS SELF-RENEWAL 283

Factors involved in somatic cells’ reprogramming undergo apoptosis when relatively high levels of DNA damage
have been introduced. p53 plays an important role in ES cells’
Fusion experiments with mouse ES cells have shown the response to low levels of DNA damage. p53 presents in ES cells,
dominance of the ES cell phenotype over that of somatic cells, and its activation can induce ES cells differentiation by
implying that proteins in the nucleus of ES cells are able to repressing Nanog expression (Lin et al., 2005). Furthermore,
reprogram more differentiated cells to an embryo-like state the function of p53 in ES cells may be to trigger apoptosis to
(Cowan et al., 2005). In ES cell–NS cell (neural stem cell) fusion, eliminate the cells with damaged DNA (Fluckiger et al., 2006).
the frequency of reprogramming was markedly enhanced by p53 is mostly cytoplasmic in ES cells but translocated to the
modestly increasing Nanog expression in ES cells. This is nucleus during differentiation. p53 is another mechanism for
consistent with the view that Nanog is a major driver of repressing Nanog expression when ES cells differentiate (Xu,
pluripotency (Rao and Orkin, 2006; Silva et al., 2006). Recently, 2005).
further studies found that forcing the expression of ES
cells-specific genes, particularly transcription factors (Oct4, Telomerase
Sox2, c-Myc, Klf4), in somatic cells might induce them to take on
a more embryonic character (Takahashi and Yamanaka, 2006). Eukaryotic chromosomes are capped by special structures
Unexpectedly, Nanog was dispensable. However, c-Myc and called telomeres, which act to guarantee chromosome
Klf4 is essential in this experiment. The c-Myc may be associated integrity. Synthesis and maintenance of telomeric repeats are
with histone acetyltransferase (HAT) complex, and induces accomplished by telomerase. Analysis of telomerase enzyme
global histone acetylation, thus allowing Oct4 and Sox2 to bind levels or activity has shown that high levels are expressed in ES
to their specific target loci. Klf4 might contribute to activation cells (Thomson et al., 1998). Telomerase pathway is active and
of Nanog and other ES cells-specific genes through p53 indeed is required for prolonged self-renewal in human and
repression (Takahashi and Yamanaka, 2006). These factors mouse ES cells (Kim et al., 1994; Thomson et al., 1998). During
induce somatic cell de-differentiation, always following the ES cells differentiation, telomerase activity is downregulated
chromatin modification. That is to say, chromatin modification, (Miura et al., 2004). In contrast to most somatic cells, stem,
as a consequence of these transcription factors, may play an germ, and tumor cells have high telomerase activity. The
important role in the reprogramming progress. catalytic subunit of telomerase (TERT) promoter contains two
E-boxes surrounding several Sp1-binding sites. E-box sites bind
Cell Cycle the Myc/Mad/Max family of transcription factors. Suggesting
Abbreviated cell cycle that c-Myc may play a role in ES cells self-renewal (Miura et al.,
2004). Overexpression of c-Myc is sufficient to maintain mouse
In contrast to somatic cells cycle, ES cells cell cycle has some ES cells in an undifferentiated state, and blocking c-Myc
specific features, the most striking feature of ES cells cell cycle is expression promotes differentiation (Cartwright et al., 2005).
that ES cells have an abbreviated cell cycle (human ES cells: Telomerase activity can be regulated by two growth factors:
15–16 h; mouse ES cells: 10 h) (Stead et al., 2002; Becker et al., bFGF and TGFb. bFGF can upregulate telomerase activity in
2006). The abbreviated cell cycle of ES cells appears to be due to human endothelial cells (Kurz et al., 2003). Furthermore, bFGF
the short G1 phase as compared to somatic cells. The short G1 plays an important role in human ES cells self-renewal (Xu et al.,
phase in ES cells cell cycle may contribute to the self-renewal. ES 2001). But it remains unclear whether bFGF activates
cells in G1 phase are more vulnerable to differentiation by telomerase in ES cells.
retinoic acid (RA) (Lukaszewicz et al., 2005). ES cells lack the
cyclin-dependent kinase (CDK) 4-associated kinase activity that Chromatin and Epigenetic Modification
characterized all somatic cells. ES cells are known to express
D-type cyclins at very low levels, and these levels increase Differentiation of ES cells from pluripotent to developmentally
dramatically during in vitro and in vivo differentiation. more restricted states is accompanied by global changes in
Expression of D1 type cyclin is regulated by PI3K-dependent genome expression patterns. Genes active in earlier
pathways. And PI3K signaling pathway plays a critical role in the progenitors are gradually silenced at developmentally later
G1/S transition in ES cells. However, PI3K activity is not stages, and subsets of cell type-specific genes are turned on.
dependent on persistent serum stimulation but rather largely This progression is the result of selectivity active expression of
rely both on stimulation of the LIF receptor and expression of transcription factors in concern with chromatin remodeling and
the ES cells-specific Eras factor. ERK activity also regulates modification, which includes covalent histone modification,
cyclin D1 expression in somatic cells, which is dispensable in ES DNA methylation of CpG dinucleotides, and localization of
cells cycle (Schratt et al., 2001; Burdon et al., 2002; Jirmanova chromatin to specialized nuclear domains.
et al., 2002; Takahashi et al., 2003).
Control in ES cells and somatic cells differs in that the ES cells Chromatin and chromatin modification
cell cycle is not dependent on a functional p16ink4a/
cyclinD:Cdk4/pRB:E2F pathway (Savatier et al., 1996; Burdon Gene activity is critically determined by chromatin structure
et al., 2002; White et al., 2005). Rather, the cyclinE:Cdk2 and and interactions of chromatin-binding proteins. To search for
cyclinA:Cdk2 complexes in ES cells are constitutively active the elusive keys for ES cells pluripotency, many researchers
throughout the cell cycle, suggesting that the mitotic cycle is have turned to the study of ES cells chromatin. Some features of
constitutively primed for DNA replication (Stead et al., 2002). chromatin, including nuclear architecture, chromatin structure,
chromatin dynamics, and histone modifications, in ES cells are
p53 different from the somatic cells. For example, ES cells
chromatin displays characteristics of loosely euchromatin, such
After DNA damage, somatic cells undergo cell-cycle arrest in as an abundance of acetylated histone modifications and
the G1 phase to get rid of the damaged DNA, but for ES cells, it increased accessibility to nucleases (Boyer et al., 2006b;
chooses apoptosis and differentiation instead of going through Meshorer and Misteli, 2006). ES cells undergo functionally
the checkpoint between the G1 phase and S phase in response important global and gene-specific remodeling of chromatin
to DNA damage. Mechanisms involved in G1/S checkpoint in structure during in vitro differentiation. Increase in the
somatic cells cycle (chk1/chk2/Cdc25A and p53-p21) are not heterochromatin marker tri-methylated residue K9 of histone
functional in ES cells (Aladjem et al., 1998; Mailand et al., 2000; H3 (H3-triMeK9) and a decrease in the global levels of
Bartek and Lukas, 2001; Hong and Stambrook, 2004). ES cells acetylated histones H3 and H4 (a euchromatin marker) were

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP


284 LIU ET AL.

observed during RA-induced differentiation (Lee et al., 2004; repression of pluripotency genes during ES cells differentiation.
Meshorer et al., 2006). Furthermore, histone deacetylase It can directly repress Oct4 and Nanog expression when ES was
(HDACs) and methyl-CpG-binding protein (MECPs) are induced by RA. The repression mechanism may be that GCNF
expressed in ES and their levels are dynamically regulated as directly binds to the promoter of Oct4 and Nanog (Gu et al.,
cells undergo differentiation (Rao, 2004). Consistent with the 2005). Another mechanism is that GCNF recruits DNA
fact that the chromatins of pluripotent nuclei is in an open methyltransferase to the Oct-3/4 promoter and facilitates its
conformation, recent studies have shown that lineage-specific methylation (Sato et al., 2006).
genes replicated earlier in pluripotent cells than differentiated Nucleosome remodeling and histone deacetylation (NuRD)
cells, and had unexpectedly high levels of acetylated H3K9 and complex also play important role when ES cells differentiate. ES
methylated H3K4. But this modification is also combined with cells lacking Mbd3 (a component of NuRD) display a profound
H3K27 trimethylation. H3K27 methylation is functionally defect in differentiation that results in persistent self-renewal. It
important for preventing these genes expression in ES cells is possible that NuRD is important for silencing Nanog during
(Azuara et al., 2006; Bernstein et al., 2006; Boyer et al., 2006b). ES cells differentiation. Thus, NuRD-mediated gene silencing
has a crucial function in the lineage commitment of ES cells as
Polycomb group proteins this process allows cells to exhibit pluripotency (Kaji et al.,
2006).
Polycomb group proteins (PcG) components are required for
early developmental gene expression (Pasini et al., 2004). In
addition, recent studies found that PcG also play an important microRNA
role in ES cells self-renewal and pluripotency. They are microRNA are a large family of small non-coding RNAs that
chromatin modification factors. They can facilitate consist of more than 200 known members in the mammalian
oligomerization, condensation of chromatin structure, and genomes. They are important in a wide variety of biological
inhibition of chromatin remodeling activity. PcG proteins are processes including cell-cycle regulation, apoptosis, cell
comprised of at least two distinct repressor complexes (PRC1 differentiation, and maintenance of stemness (Ambros, 2004;
and PRC2-PRC3). Studies in ES cells found that PRC1 and PRC2 Griffiths-Jones, 2004). Distinct sets of microRNA are
bind to a large set of genes composed of transcriptional specifically expressed in pluripotent ES cells but not in
regulators and signaling factors, which promote ES cells differentiated embryonic bodies or in adult tissues, suggesting a
differentiation. Many of the target genes were de-repressed in role in ES cells self-renewal. As ES cells differentiate, they
the absence of the PcG components. Thus, PcG proteins make downregulate stem cells maintenance genes and activate
an essential contribution to the ES cell state by repressing the lineage-specific genes (Houbaviy et al., 2003; Suh et al., 2004).
premature expression of differentiation genes. But when ES Loss of mature microRNA in Dicer1 null mouse ES cells results
cells differentiate, these target genes were preferentially in their failure to differentiate into the three germ layers,
activated. That is to say PcG repress differentiated genes suggesting that microRNA are important for ES cells
expression in a flexible manner, which can be reversed later by pluripotency, although the regulatory mechanism is still largely
gene-specific and lineage-specific signals (Bernstein et al., 2006; unknown (Cheng et al., 2005; Kanellopoulou et al., 2005).
Boyer et al., 2006b; Buszczak and Spradling, 2006; Lee et al.,
2006).
Conclusions
DNA methylation This review has made important contributions toward
DNA methylation at CpG islands is another gene silence elucidating the complexity molecular mechanism in ES cells
mechanism. Silencing of certain genes by DNA methylation is self-renewal and pluripotency. Mouse ES cells and human ES
required for induction of differentiation of ES cells. This was cells show similar markers of pluripotent stem cell lines such as
shown in experiments with ES cells deficient either in the DNA Oct4, Nanog, ALP, and high levels of telomerase activity. But,
methyltransferases Dnmt1, both Dnmt3a and Dnmt3b, or the there are notable differences between mouse and human ES
CpG island-binding protein CGBP that binds to non-methylated cells. These differences between mouse and human ES cells
DNA. These cells showed severe DNA hypomethylation and a suggest that although the same pluripotency genes are
complete differentiation block (Jackson et al., 2004; Carlone expressed in both mouse ES and human ES cells, their function
et al., 2005). Hypermethylation of Oct4 promoter/enhancer and downstream signaling pathways may differ between the two
region in differentiated cells correlates with gene silencing, species. An important goal is to understand the mechanism
whereas hypomethylation in ES cells allows cells to maintain involved in pluripotency used by human and mouse ES cells. So,
high level of Oct4 expression, thus keeping them in pluripotent we can reprogram differentiated cells to an embryo-like state,
state. Reprogramming phenomenon can be observed during and then recreate these cells in somatic cells which can be used
somatic cells fusion with ES cells or in the culture containing in clinic. Despite our limited current knowledge, there is little
extracts of ES cells. Oct4 DNA demethylation in thymocyte doubt that we will eventually uncover the molecular
nuclei has been reported after fusion with ES cells (Taranger mechanisms that give rise to the unique properties of ES cells
et al., 2005). and that we will then be able to exploit them as powerful tools in
basic discovery and clinical applications.
From Pluripotency to Differentiation
Literature Cited
ES cells are capable of unlimited symmetrical self-renewal and
Aladjem MI, Spike BT, Rodewald LW, Hope TJ, Klemm M, Jaenisch R, Wahl GM. 1998. ES cells
have the potential to differentiate into any cell type in the body. do not activate p53-dependent stress responses and undergo p53-independent apoptosis
Differentiation of ES cells is accompanied by downregulation of in response to DNA damage. Curr Biol 8:145–155.
the whole series of transcription factors and signaling molecules Ambros V. 2004. The functions of animal microRNAs. Nature 431:350–355.
Amit M, Carpenter MK, Inokuma MS, Chiu CP, Harris CP, Waknitz MA, Itskovitz-Eldor J,
that maintain the pluripotent phenotype, such as Oct4 and Thomson JA. 2000. Clonally derived human embryonic stem cells lines maintain
Nanog, and upregulation of transcription factors involved in pluripotency and proliferative potential for prolonged periods of culture. Dev Biol
227:271–278.
differentiation. Little is known about how determinants of ES Armstrong L, Hughes O, Yung S, Hyslop L, Stewart R, Wappler I, Peters H, Walter T,
cell pluripotency are regulated during differentiation of ES cells. Stojkovic P, Evans J, Stojkovic M, Lako M. 2006. The role of PI3K/AKT, MAPK/ERK and
NFkappabeta signalling in the maintenance of human embryonic stem cell pluripotency and
Recently, studies have shown that orphan nuclear receptor viability highlighted by transcriptional profiling and functional analysis. Hum Mol Genet
Germ Cell Nuclear Receptor (GCNF) is required for the 15:1894–1913.

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP


MECHANISMS IN ES CELLS SELF-RENEWAL 285

Azuara V, Perry P, Sauer S, Spivakov M, Jorgensen HF, John RM, Gouti M, Casanova M, Houbaviy HB, Murray MF, Sharp PA. 2003. Embryonic stem cell-specific MicroRNAs. Dev
Warnes G, Merkenschlager M, Fisher AG. 2006. Chromatin signatures of pluripotent cell Cell 5:351–358.
lines. Nat Cell Biol 8:532–538. Humphrey RK, Beattie GM, Lopez AD, Bucay N, King CC, Firpo MT, Rose-John S, Hayek A.
Bartek J, Lukas J. 2001. Pathways governing G1/S transition and their response to DNA 2004. Maintenance of pluripotency in human embryonic stem cells is STAT3 independent.
damage. FEBS Lett 490:117–122. Stem Cells 22:522–530.
Beattie GM, Lopez AD, Bucay N, Hinton A, Firpo MT, King CC, Hayek A. 2005. Activin A Ivanova N, Dobrin R, Lu R, Kotenko I, Levorse J, DeCoste C, Schafer X, Lun Y, Lemischka IR.
maintains pluripotency of human embryonic stem cells in the absence of feeder layers. Stem 2006. Dissecting self-renewal in stem cells with RNA interference. Nature 442:533–538.
Cells 23:489–495. Jackson M, Krassowska A, Gilbert N, Chevassut T, Forrester L, Ansell J, Ramsahoye B. 2004.
Becker KA, Ghule PN, Therrien JA, Lian JB, Stein JL, van Wijnen AJ, Stein GS. 2006. Severe global DNA hypomethylation blocks differentiation and induces histone
Self-renewal of human embryonic stem cells is supported by a shortened G1 cell cycle hyperacetylation in embryonic stem cells. Mol Cell Biol 24:8862–8871.
phase. J Cell Physiol 209:883–893. James D, Levine AJ, Besser D, Hemmati-Brivan lou A. 2005. TGFbeta/activin/nodal signaling is
Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, necessary for the maintenance of pluripotency in human embryonic stem cells.
Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL, Lander ES. 2006. A bivalent chromatin Development 132:1273–1282.
structure marks key developmental genes in embryonic stem cells. Cell 125:315–326. Jirmanova L, Afanassieff M, Gobert-Gosse S, Markossian S, Savatier P. 2002. Differential
Bhattacharya B, Miura T, Brandenberger R, Mejido J, Luo Y, Yang AX, Joshi BH, Ginis I, contributions of ERK and PI3-kinase to the regulation of cyclin D1 expression and to the
Thies RS, Amit M, Lyons I, Condie BG, Itskovitz-Eldor J, Rao MS, Puri RK. 2004. Gene control of the G1/S transition in mouse embryonic stem cells. Oncogene 21:5515–5528.
expression in human embryonic stem cell lines: Unique molecular signature. Blood Kaji K, Caballero IM, MacLeod R, Nichols J, Wilson VA, Hendrich B. 2006. The NuRD
103:2956–2964. component Mbd3 is required for pluripotency of embryonic stem cells. Nat Cell Biol
Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, 8:285–292.
Murray HL, Jenner RG, Gifford DK, Melton DA, Jaenisch R, Young RA. 2005. Core Kanellopoulou C, Muljo SA, Kung AL, Ganesan S, Drapkin R, Jenuwein T, Livingston DM,
transcriptional regulatory circuitry in human embryonic stem cells. Cell 122:947–956. Rajewsky K. 2005. Dicer-deficient mouse embryonic stem cells are defective in
Boyer LA, Mathur D, Jaenisch R. 2006a. Molecular control of pluripotency. Curr Opin Genet differentiation and centromeric silencing. Genes Dev 19:489–501.
Dev 16:455–462. Kang HB, Kim JS, Kwon HJ, Nam KH, Youn HS, Sok DE, Lee Y. 2005. Basic fibroblast growth
Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, Levine SS, Wernig M, factor activates ERK and induces c-fos in human embryonic stem cell line MizhES1. Stem
Tajonar A, Ray MK, Bell GW, Otte AP, Vidal M, Gifford DK, Young RA, Jaenisch R. 2006b. Cells Dev 14:395–401.
Polycomb complexes repress developmental regulators in murine embryonic stem cells. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE,
Nature 441:349–353. Weinrich SL, Shay JW. 1994. Specific association of human telomerase activity with
Brandenberger R, Wei H, Zhang S, Lei S, Murage J, Fisk GJ, Li Y, Xu C, Fang R, Guegler K, Rao immortal cells and cancer. Science 266:2011–2015.
MS, Mandalam R, Lebkowski J, Stanton LW. 2004. Transcriptome characterization Kim SJ, Cheon SH, Yoo SJ, Kwon J, Park JH, Kim CG, Rhee K, You S, Lee JY, Roh SI, Yoon HS.
elucidates signaling networks that control human ES cell growth and differentiation. Nat 2005. Contribution of the PI3K/Akt/PKB signal pathway to maintenance of self-renewal in
Biotechnol 22:707–716. human embryonic stem cells. FEBS Lett 579:534–540.
Brazil DP, Yang ZZ, Hemmings BA. 2004. Advances in protein kinase B signalling: AKTion on Kristensen DM, Kalisz M, Nielsen JH. 2005. Cytokine signalling in embryonic stem cells.
multiple fronts. Trends Biochem Sci 29:233–242. APMIS 113:756–772.
Burdon T, Stracey C, Chambers I, Nichols J, Smith A. 1999. Suppression of SHP-2 and ERK Kuroda T, Tada M, Kubota H, Kimura H, Hatano SY, Suemori H, Nakatsuji N, Tada T. 2005.
signalling promotes self-renewal of mouse embryonic stem cells. Dev Biol 210:30–43. Octamer and Sox elements are required for transcriptional cis regulation of Nanog gene
Burdon T, Smith A, Savatier P. 2002. Signaling, cell cycle and pluripotency in embryonic stem expression. Mol Cell Biol 25:2475–2485.
cells. Trends Cell Biol 12:432–438. Kurz DJ, Hong Y, Trivier E, Huang HL, Decary S, Zang GH, Luscher TF, Erusalimsky JD. 2003.
Buszczak M, Spradling AC. 2006. Searching chromatin for stem cell identity. Cell 125:233– Fibroblast growth factor-2, but not vascular endothelial growth factor, upregulates
236. telomerase activity in human endothelial cells. Arterioscler Thromb Vasc Biol 23:748–754.
Carlone DL, Lee JH, Young SR, Dobrota E, Butler JS, Ruiz J, Skalnik DG. 2005. Reduced Lee JH, Hart SR, Skalnik DG. 2004. Histone deacetylase activity is required for embryonic
genomic cytosine methylation and defective cellular differentiation in embryonic stem cells stem cell differentiation. Genesis 38:32–38.
lacking CpG binding protein. Mol Cell Biol 25:4881–4891. Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, Chevalier B, Johnstone SE,
Carpenter MK, Rosler ES, Fisk GJ, Brandenberger R, Ares X, Miura T, Lucero M, Rao MS. Cole MF, Isono K, Koseki H, Fuchikami T, Abe K, Murray HL, Zucker JP, Yuan B, Bell GW,
2004. Properties of four human embryonic stem cell lines maintained in a feeder-free Herbolsheimer E, Hannett NM, Sun K, Odom DT, Otte AP, Volkert TL, Bartel DP, Melton
culture system. Dev Dynamics 229:243–258. DA, Gifford DK, Jaenisch R, Young RA. 2006. Control of developmental regulators by
Cartwright P, McLean C, Sheppard A, Rivett D, Jones K, Dalton S. 2005. LIF/STAT3 controls Polycomb in human embryonic stem cells. Cell 125:301–313.
ES cell self-renewal and pluripotency by a Myc-dependent mechanism. Development Levenstein ME, Ludwig TE, Xu RH, Llanas RA, Vandenheuvel-Kramer K, Manning D,
132:885–896. Thomson JA. 2006. Basic fibroblast growth factor support of human embryonic stem cell
Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A. 2003. Functional self-renewal. Stem Cells 24:568–574.
expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell Levine AJ, Brivanlou AH. 2006. GDF3, a BMP inhibitor, regulates cell fate in stem cells and
113:643–655. early embryos. Development 133:209–216.
Cheng LC, Tavazoie M, Doetsch F. 2005. Stem cells: From epigenetics to microRNAs. Liew CG, Moore H, Ruban L, Shah N, Cosgrove K, Dunne M, Andrews P. 2005. Human
Neuron 46:363–367. embryonic stem cells: Possibilities for human cell transplantation. Ann Med 37:521–532.
Chew JL, Loh YH, Zhang W, Chen X, Tam WL, Yeap LS, Li P, Ang YS, Lim B, Robson P, Ng HH. Lin T, Chao C, Saito S, Mazur SJ, Murphy ME, Appella E, Xu Y. 2005. p53 induces
2005. Reciprocal transcriptional regulation of Pou5f1 and Sox2 via the Oct4/Sox2 complex differentiation of mouse embryonic stem cells by suppressing Nanog expression. Nat Cell
in embryonic stem cells. Mol Cell Biol 25:6031–6046. Biol 7:165–171.
Coppo P, Dusanter-Fourt I, Millot G, Nogueira MM, Dugray A, Bonnet ML, Mitjavila-Garcia Lodge P, McWhir J, Gallagher E, Sang H. 2005. Increased gp130 signaling in combination with
MT, Le Pesteur D, Guilhot F, Vainchenker W, Sainteny F, Turhan AG. 2003. Constitutive inhibition of the MEK/ERK pathway facilitates embryonic stem cell isolation from normally
and specific activation of STAT3 by BCR-ABL in embryonic stem cells. Oncogene refractory murine CBA blastocysts. Cloning Stem Cells 7:2–7.
22:4102–4110. Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J,
Cowan CA, Atienza J, Melton DA, Eggan K. 2005. Nuclear reprogramming of somatic cells Wong KY, Sung KW, Lee CW, Zhao XD, Chiu KP, Lipovich L, Kuznetsov VA, Robson P,
after fusion with human embryonic stem cells. Science 309:1369–1373. Stanton LW, Wei CL, Ruan Y, Lim B, Ng HH. 2006. The Oct4 and Nanog transcription
Daheron L, Opitz SL, Zaehres H, Lensch WM, Andrews PW, Itskovitz-Eldor J, Daley GQ. network regulates pluripotency in mouse embryonic stem cells. Nat Genet 38:431–440.
2004. LIF/STAT3 signaling fails to maintain self-renewal of human embryonic stem cells. Lukaszewicz A, Savatier P, Cortay V, Giroud P, Huissoud C, Berland M, Kennedy H, Dehay C.
Stem Cells 22:770–778. 2005. G1 phase regulation, areaspecific cell cycle control, and cytoarchitectonics in the
Dvash T, Mayshar Y, Darr H, McElhaney M, Barker D, Yanuka O, Kotkow KJ, Rubin LL, primate cortex. Neuron 47:353–364.
Benvenisty N, Eiges R. 2004. Temporal gene expression during differentiation of human Mailand N, Falck J, Lukas C, Syljuasen RG, Welcker M, Bartek J, Lukas J. 2000. Rapid
embryonic stem cells and embryoid bodies. Hum Reprod 19:2875–2883. destruction of human Cdc25A in response to DNA damage. Science 288:1425–1429.
Dvorak P, Hampl A. 2005. Basic fibroblast growth factor and its receptors in human Matsuda T, Nakamura T, Nakao K, Arai T, Katsuki M, Heike T, Yokota T. 1999. STAT3
embryonic stem cells. Folia Histochem Cytobiol 43:203–208. activation is sufficient to maintain an undifferentiated state of mouse embryonic stem cells.
Dvorak P, Dvorakova D, Koskova S, Vodinska M, Najvirtova M, Krekac D, Hampl A. 2005a. EMBO J 18:4261–4269.
Expression and potential role of fibroblast growth factor 2 and its receptors in human Meshorer E, Misteli T. 2006. Chromatin in pluripotent embryonic stem cells and
embryonic stem cells. Stem Cells 23:1200–1211. differentiation. Nat Rev Mol Cell Biol 7:540–546.
Evans MJ, Kaufman MH. 1981. Establishment in culture of pluripotential cells from mouse Meshorer E, Yellajoshula D, George E, Scambler PJ, Brown DT, Misteli T. 2006.
embryos. Nature 292:154–156. Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. Dev
Fluckiger AC, Marcy G, Marchand M, Negre D, Cosset FL, Mitalipov S, Wolf D, Savatier P, Cell 10:105–116.
Dehay C. 2006. Cell cycle features of primate embryonic stem cells. Stem Cells 24:547– Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M,
556. Yamanaka S. 2003. The homeoprotein Nanog is required for maintenance of pluripotency
Gerrard L, Rodgers L, Cui W. 2005. Differentiation of human embryonic stem cells to neural in mouse epiblast and ES cells. Cell 113:631–642.
lineages in adherent culture by blocking bone morphogenetic protein signaling. Stem Cells Miura T, Mattson MP, Rao MS. 2004. Cellular lifespan and senescence signaling in embryonic
23:1234–1241. stem cells. Aging Cell 3:333–343.
Ginis I, Luo Y, Miura T, Thies S, Brandenberger R, Gerecht-Nir S, Amit M, Hoke A, Carpenter Nakamura Y, Yujiri T, Nawata R, Tagami K, Tanizawa Y. 2005. MEK kinase 1 is essential for
MK, Itskovitz-Eldor J, Rao MS. 2004. Differences between human and mouse embryonic Bcr-Abl-induced STAT3 and self-renewal activity in embryonic stem cells. Oncogene
stem cells. Dev Biol 269:360–380. 24:7592–7598.
Griffiths-Jones S. 2004. The microRNA Registry. Nucleic Acids Res 32:109–111. Nishimoto M, Fukushima A, Okuda A, Muramatsu M. 1999. The gene for the embryonic stem
Gross VS, Hess M, Cooper GM. 2005. Mouse embryonic stem cells and preimplantation cell coactivator UTF1 carries a regulatory element which selectively interacts with a
embryos require signaling through the phosphatidylinositol 3-kinase pathway to suppress complex composed of Oct-3/4 and Sox-2. Mol Cell Biol 19:5453–5465.
apoptosis. Mol Reprod Dev 70:324–332. Niwa H. 2001. Molecular mechanism to maintain stem cell renewal of ES cells. Cell Struct
Gu P, LeMenuet D, Chung AC, Mancini M, Wheeler DA, Cooney AJ. 2005. Orphan nuclear Funct 26:137–148.
receptor GCNF is required for the repression of pluripotency genes during retinoic Niwa H, Burdon T, Chambers I, Smith A. 1998. Self-renewal of pluripotent embryonic stem
acid-induced embryonic stem cell differentiation. Mol Cell Bio 25:8507–8519. cells is mediated via activation of STAT3. Genes Dev 12:2048–2060.
Hao J, Li TG, Qi X, Zhao DF, Zhao GQ. 2006. WNT/beta-catenin pathway up-regulates Stat3 Okumura-Nakanishi S, Saito M, Niwa H, Ishikawa F. 2005. Oct-3/4 and Sox2 regulate Oct-3/4
and converges on LIF to prevent differentiation of mouse embryonic stem cells. Dev Biol gene in embryonic stem cells. J Biol Chem 280:5307–5317.
290:81–91. Paling NR, Wheadon H, Bone HK, Welham MJ. 2004. Regulation of embryonic stem cell
Hay N, Sonenberg N. 2004. Upstream and downstream of mTOR. Genes Dev 18:1926–1945. self-renewal by phosphoinositide 3-kinase-dependent signaling. J Biol Chem 279:48063–
Hong Y, Stambrook PJ. 2004. Restoration of an absent G1 arrest and protection from 48070.
apoptosis in embryonic stem cells after ionizing radiation. Proc Natl Acad Sci USA Pan G, Pei D. 2005. The stem cell pluripotency factor NANOG activates transcription with
101:14443–14448. two unusually potent subdomains at its C terminus. J Biol Chem 280:1401–1407.

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP


286 LIU ET AL.

Pan GJ, Chang ZY, Scholer HR, Pei D. 2002. Stem cell pluripotency and transcription factor Takahashi K, Mitsui K, Yamanaka S. 2003. Role of ERas in promoting tumour like properties in
Oct4. Cell Res 12:321–329. mouse embryonic stem cells. Nature 423:541–545.
Pan G, Li J, Zhou Y, Zheng H, Pei D. 2006. A negative feedback loop of transcription factors Takahashi K, Murakami M, Yamanaka S. 2005. Role of the phosphoinositide 3-kinase pathway
that controls stem cell pluripotency and self-renewal. FASEB J 20:1730–1732. in mouse embryonic stem (ES) cells. Biochem Soc Trans 33:1522–1525.
Pasini D, Bracken AP, Jensen MR, Lazzerini Denchi E, Helin K. 2004. Suz12 is essential for Tanaka TS, Kunath T, Kimber WL, Jaradat SA, Stagg CA, Usuda M, Yokota T, Niwa H,
mouse development and for EZH2 histone methyltransferase activity. EMBO J 23:4061– Rossant J, Ko MS. 2002. Gene expression profiling of embryo-derived stem cells reveals
4071. candidate genes associated with pluripotency and lineage specificity. Genome Res
Pera MF, Trounson AO. 2004. Human embryonic stem cells: Prospects for development. 12:1921–1928.
Development 131:5515–5525. Taranger CK, Noer A, Sorensen AL, Hakelien AM, Boquest AC, Collas P. 2005. Induction of
Qi X, Li TG, Hao J, Hu J, Wang J, Simmons H, Miura S, Mishina Y, Zhao GQ. 2004. BMP4 dedifferentiation, genomewide transcriptional programming, and epigenetic
supports self-renewal of embryonic stem cells by inhibiting mitogen-activated protein reprogramming by extracts of carcinoma and embryonic stem cells. Mol Biol Cell
kinase pathways. Proc Natl Acad Sci USA 101:6027–6032. 16:5719–5735.
Qu L, Huang S, Baltzis D, Rivas-Estilla AM, Pluquet O, Hatzoglou M, Koumenis C, Taya Y, Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM.
Yoshimura A, Koromilas AE. 2004. Endoplasmic reticulum stress induces p53 cytoplasmic 1998. Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147.
localization and prevents p53-dependent apoptosis by a pathway involving glycogen Tomioka M, Nishimoto M, Miyagi S, Katayanagi T, Fukui N, Niwa H, Muramatsu M, Okuda A.
synthase kinase-3beta. Genes Dev 18:261–277. 2002. Identification of Sox-2 regulatory region which is under the control of Oct-3/4-Sox-2
Rao M. 2004. Conserved and divergent paths that regulate self-renewal in mouse and human complex. Nucleic Acids Res 30:3202–3213.
embryonic stem cells. Dev Biol 275:269–286. Valdimarsdottir G, Mummery C. 2005. Functions of the TGFbeta superfamily in human
Rao S, Orkin SH. 2006. Unraveling the transcriptional network controlling ES cell embryonic stem cells. APMIS 113:773–789.
pluripotency. Genome Biol 7:230. Wang J, Xie LY, Allan S, Beach D, Hannon GJ. 1998. Myc activates telomerase. Genes Dev
Richards M, Tan SP, Tan JH, Chan WK, Bongso A. 2004. The transcriptome profile of human 12:1769–1774.
embryonic stem cells as defined by SAGE. Stem Cells 22:51–64. Wang G, Zhang H, Zhao Y, Li J, Cai J, Wang P, Meng S, Feng J, Miao C, Ding M, Li D, Deng H.
Rodda DJ, Chew JL, Lim LH, Loh YH, Wang B, Ng HH, Robson P. 2005. Transcriptional 2005. Noggin and bFGF cooperate to maintain the pluripotency of human embryonic stem
regulation of nanog by OCT4 and SOX2. J Biol Chem 280:24731–24737. cells in the absence of feeder layers. Biochem Biophys Res Commun 330:934–942.
Sato N, Sanjuan IM, Heke M, Uchida M, Naef F, Brivanlou AH. 2003. Molecular signature of Watanabe S, Umehara H, Murayama K, Okabe M, Kimura T, Nakano T. 2006. Activation of
human embryonic stem cells and its comparison with the mouse. Dev Biol 260:404–413. Akt signaling is sufficient to maintain pluripotency in mouse and primate embryonic stem
Sato N, Meijer L, Skaltsounis L, Greengard P, Brivanlou AH. 2004. Maintenance of cells. Oncogene 25:2697–2707.
pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling Watcharasit P, Bijur GN, Song L, Zhu J, Chen X, Jope RS. 2002. Glycogen synthase kinase-
by a pharmacological GSK-3- specific inhibitor. Nat Med 10:55–63. 3beta (GSK3beta) binds to and promotes the actions of p53. Proc Natl Acad Sci USA
Sato N, Kondo M, Arai K. 2006. The orphan nuclear receptor GCNF recruits DNA 99:7951–7955.
methyltransferase for Oct-3/4 silencing. Biochem Biophys Res Commun 344:845–851. White J, Stead E, Faast R, Conn S, Cartwright P, Dalton S. 2005. Developmental activation of
Savatier P, Lapillonne H, van Grunsven LA, Rudkin BB, Samarut J. 1996. Withdrawal of the Rb-E2F pathway and establishment of cell cycle-regulated cyclin-dependent kinase
differentiation inhibitory activity/leukemia inhibitory factor up-regulates Dtype cyclins and activity during embryonic stem cell differentiation. Mol Biol Cell 16:2018–2027.
cyclin-dependent kinase inhibitors in mouse embryonic stem cells. Oncogene 12:309–322. Williams RL, Hilton DJ, Pease S, Willson TA, Stewart CL, Gearing DP, Wagner EF, Metcalf D,
Schratt G, Weinhold B, Lundberg AS, Schuck S, Berger J, Schwarz H, Weinberg RA, Ruther U, Nicola NA, Gough NM. 1988. Myeloid leukaemia inhibitory factor maintains the
Nordheim A. 2001. Serum response factor is required for immediate-early gene activation developmental potential of embryonic stem cells. Nature 336:684–687.
yet is dispensable for proliferation of embryonic stem cells. Mol Cell Biol 21:2933–2943. Xu Y. 2005. A new role for p53 in maintaining genetic stability in embryonic stem cells. Cell
Sekkai D, Gruel G, Herry M, Moucadel V, Constantinescu SN, Albagli O, Tronik-Le Roux D, Cycle 4:363–364.
Vainchenker W, Bennaceur-Griscelli A. 2005. Microarray analysis of LIF/Stat3 Xu C, Inokuma MS, Denham J, Golds K, Kundu P, Gold JD, Carpenter MK. 2001. Feeder-free
transcriptional targets in embryonic stem cells. Stem Cell 23:1634–1642. growth of undifferentiated human embryonic stem cells. Nat Biotechnol 19:971–974.
Shi W, Wang H, Pan G, Geng Y, Guo Y, Pei D. 2006. Regulation of the pluripotency marker Xu RH, Chen X, Li DS, Li R, Addicks GC, Glennon C, Zwaka TP, Thomson JA. 2002. BMP4
rex-1 by nanog and sox2. J Biol Chem 281:23319–23325. initiates human embryonic stem cell differentiation to trophoblast. Nat Biotechnol
Silva J, Chambers I, Pollard S, Smith A. 2006. Nanog promotes transfer of pluripotency after 20:1261–1264.
cell fusion. Nature 441:997–1001. Xu RH, Peck RM, Li DS, Feng X, Ludwig T, Thomson JA. 2005a. Basic FGF and suppression of
Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, Stahl M, Rogers D. 1988. Inhibition BMP signaling sustain undifferentiated proliferation of human ES cells. Nat Methods 2:185–
of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature 190.
336:688–690. Xu C, Rosler E, Jiang J, Lebkowski JS, Gold JD, O’Sullivan C, Delavan-Boorsma K, Mok M,
Stead E, White J, Faast R, Conn S, Goldstone S, Rathjen J, Dhingra U, Rathjen P, Walker D, Bronstein A, Carpenter MK. 2005b. Basic fibroblast growth factor supports
Dalton S. 2002. Pluripotent cell division cycles are driven by ectopic Cdk2, cyclin A/E and undifferentiated human embryonic stem cell growth without conditioned medium. Stem
E2F activities. Oncogene 21:8320–8333. Cells 23:315–323.
Suh MR, Lee Y, Kim JY, Kim SK, Moon SH, Lee JY, Cha KY, Chung HM, Yoon HS, Moon SY, Yeom YI, Fuhrmann G, Ovitt CE, Brehm A, Ohbo K, Gross M, Hubner K, Scholer HR. 1996.
Kim VN, Kim KS. 2004. Human embryonic stem cells express a unique set of microRNAs. Germline regulatory element of Oct-4 specific for the totipotent cycle of embryonal cells.
Dev Biol 270:488–498. Development 122:881–894.
Suzuki A, Raya A, Kawakami Y, Morita M, Matsui T, Nakashima K, Gage FH, Rodriguez- Ying QL, Nichols J, Chambers I, Smith A. 2003. BMP induction of Id proteins suppresses
Esteban C, Izpisua Belmonte JC. 2006. Nanog binds to Smad1 and blocks bone differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3.
morphogenetic protein-induced differentiation of embryonic stem cells. Proc Natl Acad Cell 115:281–292.
Sci USA 103:10294–10299. Zeng X, Miura T, Luo Y, Bhattacharya B, Condie B, Chen J, Ginis I, Lyons I, Mejido J, Puri RK,
Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from mouse embryonic Rao MS, Freed WJ. 2004. Properties of pluripotent human embryonic stem cells BG01 and
and adult fibroblast cultures by defined factors. Cell 126:663–676. BG02. Stem Cells 22:292–312.

JOURNAL OF CELLULAR PHYSIOLOGY DOI 10.1002/JCP

Potrebbero piacerti anche