Sie sind auf Seite 1von 6

Cell Stem Cell

Perspective

Epigenetic Mechanisms that Regulate Cell Identity


Marı́a J. Barrero,1 Stephanie Boué,1 and Juan Carlos Izpisúa Belmonte1,2,*
1Center of Regenerative Medicine in Barcelona, Dr. Aiguader, 88, 08003 Barcelona, Spain
2Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
*Correspondence: belmonte@salk.edu or izpisua@cmrb.eu (J.C.I.B.)
DOI 10.1016/j.stem.2010.10.009

Individual cell fate decisions can vary according to changes in gene expression in response to environmental,
developmental, or metabolic cues. This plasticity is tightly regulated during embryonic development and
mediated by the exquisitely coordinated activation and repression of groups of genes. Genes that become
repressed are immersed in a condensed chromatin environment that renders them refractory to stimulation.
This mechanism is responsible for both the loss of cell plasticity during differentiation and the preservation of
cell identity. Understanding the molecular events involved in the establishment and maintenance of these
restrictive domains will benefit the design of strategies for cellular reprogramming, differentiation, and cancer
treatment.

Despite sharing the same genome, different cell types from rather than altering the chromatin structure per se. In a similar
a given organism respond differently to environmental, develop- fashion, DNA methylation, which takes place at cytosine resi-
mental, or metabolic cues. This variable property, or plasticity of dues within CpG dinuclotides at gene promoters, is usually
responsiveness, is a defining aspect of a cell’s identity and is correlated with transcriptional repression and mediates its
mainly interpreted at the level of gene expression. Stem and effect by blocking the binding of transcription factors and/or
progenitor cells are a special case, given that their potential for facilitating the assembly of repressor complexes at the methyl-
plasticity is, by definition, more pronounced than for other ated regions. In general, the presence of histone modifications
tissue-specific cells. During differentiation, stem cells lose their involved in gene activation, such as trimethylation of histone
plasticity and narrow down their identity into a particular differen- H3 at lysine 4 (H3K4me3), promotes the recruitment and
tiated cell type. For many years this differentiated state was stabilization of effector complexes with histone acetyltransfer-
thought to be irreversible but recent findings indicate that it is ase (HAT) and ATP-dependent remodeling activities. These
possible to manipulate cells in order to change cell identity or complexes mediate the acetylation of histones and stimulate
to regain plasticity. Understanding how fate decisions are regu- the mobility of the nucleosomes, or even their eviction, to facili-
lated and maintained by different cell types, both during devel- tate the relaxation of chromatin. Ultimately, this change renders
opment and under pathological or experimental conditions, DNA more accessible and facilitates the recruitment of DNA-
represents an important step toward the successful manipula- binding transcription factors to target genes, which in turn
tion of cell plasticity in clinical settings. contribute to the recruitment of RNA polymerase and the stabi-
We have known for a long time that genetic material associ- lization of the preinitiation complex (PIC). Modifications involved
ates with structural proteins to constitute chromatin, which plays in gene repression, such as DNA methylation and trimethylation
important roles structurally and functionally. Hence, genes of histone H3 at lysine 9 (H3K9me3) and at lysine 27 (H3K27me3),
immersed in highly packed areas of chromatin remain largely serve as docking sites for repressor complexes that contain
inaccessible, fail to recruit the RNA polymerase to their pro- histone deacetylase (HDAC) and ATP-dependent remodeling
moters, and thus remain silent and refractory to stimulation. activities. The action of these complexes, together with the
However, genes having a more open and accessible chromatin recruitment of structural nonhistone proteins, leads to chromatin
structure are more likely to recruit and engage the RNA poly- compaction and reduced DNA accessibility.
merase into productive rounds of transcription and their expres-
sion is more prone to be modulated by signaling events. Impor- Molecular Basis of Pluripotency in Embryonic
tantly, the chromatin environment in which a particular gene is Stem Cells
embedded can differ from one cell type to another, contributing The chromatin in pluripotent embryonic stem cells (ESCs) dis-
to cell diversity and identity. plays distinctive features when compared to differentiated cells.
The fundamental unit of chromatin is the nucleosome, which is In ESCs the chromatin structural proteins display hyperdynamic
composed of two copies each of four core histones, H2A, H2B, interactions with chromatin (Meshorer et al., 2006) and the over-
H3, and H4, and wrapped by 146 bp of DNA. The N-terminal tails all transcriptional activity is hyperactive compared to differenti-
of histones are relatively accessible to enzymatic modifications ated cells (Efroni et al., 2008). ESCs also express high levels of
such as acetylation, methylation, phosphorylation, ubiquitina- chromatin-remodeling factors involved in maintaining the open
tion, and sumoylation. Of all the known histone modifications, chromatin status. Moreover, variations in the expression levels
acetylation is the only modification that directly causes a struc- of specific subunits of remodeling complexes, such as the BAF
tural relaxation of chromatin by introducing a negative charge. complex, in ESCs relative to differentiated cells leads to the
Other modifications seem to act as docking sites that promote formation of unique remodeling complexes that differ in subunit
the recruitment and stabilization of effector protein complexes composition and potentially in function (Ho et al., 2009). These

Cell Stem Cell 7, November 5, 2010 ª2010 Elsevier Inc. 565


Cell Stem Cell

Perspective

specialized complexes could contribute to sustain high levels of in the embryo and to appear after fertilization during genomic
transcriptional activity in pluripotent cells. This idea is further activation at the maternal-zygotic transition (Vastenhouw et al.,
supported by the reported loss of self-renewal caused by the 2010). In addition, recent reports describe the presence of
depletion of the remodeling factors Chd1 or Brg1 in ESCs (Efroni microregions of H3K27me3 at the transcriptional start sites of
et al., 2008; Gaspar-Maia et al., 2009). developmental regulators in sperm. The persistence of these
ESCs feature a unique plasticity that allows them to differen- discrete regions after fertilization and the presence of CpG
tiate into virtually any cell type of the adult organism. This prop- islands at the promoters of the marked developmental genes
erty, called pluripotency, relies on the fact that critical genes might serve as primers for the establishment of broad
involved in differentiation, despite remaining silent, have a H3K27me3 domains during genome activation in embryos
permissive chromatin structure that makes them sensitive to (Brykczynska et al., 2010). However, it is likely that specific tran-
differentiation-inducing signals. This permissive chromatin envi- scription factors also contribute to establish and/or maintain
ronment is characterized by the presence of large H3K27me3 these domains. Indeed, genome-wide correlation studies have
domains harboring smaller regions of H3K4me3 around the tran- revealed that the self-renewal transcription factors Oct4, Sox2,
scriptional start site of critical developmental genes. The coexis- and Nanog occupy not only promoters of genes involved in
tence of these two antagonistic marks is the defining feature of self-renewal but also of a high number of genes that contain
so-called bivalent genes and has been suggested to play bivalent domains (Boyer et al., 2005), suggesting that they play
a role in silencing differentiation genes in ESCs while keeping a role in keeping this genes silenced. In agreement with this
them poised for activation upon initiation of specific develop- model, it has been reported that Nanog and Oct4 interact with
mental pathways (Azuara et al., 2006; Bernstein et al., 2006). complexes involved in transcriptional repression, including
Moreover, the poised nature of these bivalent domains is further PcG subunits (Pardo et al., 2010; Wang et al., 2006). Another
reinforced by the absence of DNA methylation, despite the pres- potential transcription factor that could be involved in the molec-
ence of numerous CpG islands (Fouse et al., 2008; Meissner ular arrangement of these domains is JARID2, which has been
et al., 2008) and the presence of poised RNA polymerase II at recently suggested to participate in the recruitment of PcG to
the transcription initiation site of the marked genes (Guenther the regulatory regions of target genes in mouse ESCs (Landeira
et al., 2007). Ultimately, the physiological function of bivalent et al., 2010; Li et al., 2010; Pasini et al., 2010; Peng et al., 2009).
domains might be to maintain important regulatory sequences
accessible and responsive at very early stages of differentiation Chromatin Dynamics during Differentiation
(Gargiulo et al., 2009). Importantly, genes marked with bivalent The permissive chromatin structure present in multipotent cells
domains often encode master transcription factors that are is progressively and selectively closed during differentiation.
induced very early during differentiation, usually in a lineage- Accessible regulatory areas, such as bivalent domains, close
restricted manner, and that are able to orchestrate whole up and are no longer accessible to transcription factors, leading
programs of gene expression in differentiated cells. to a loss of regulatory potential (Gargiulo et al., 2009).
Although the bivalent marks are faithfully transmitted through The analysis of the genome-wide distribution of histone and
cell division in self-renewing cells, they should not be regarded DNA methylation marks and their correlation with gene expres-
as static but as the result of a highly dynamic equilibrium that sion in ESCs, in vitro differentiated ESCs, and adult cells has
is controlled by the balance of histone-modifying enzymes provided a better understanding of gene plasticity in pluripotent
recruited to these areas. The polycomb group of proteins (PcGs) and differentiated tissues (Fouse et al., 2008; Hawkins et al.,
has been described to play an essential role in maintaining the 2010; Meissner et al., 2008; Mikkelsen et al., 2007). These
pluripotent state of ESCs by mediating H3K27 methylation at studies have led researchers to focus on two types of genes:
the bivalent domains (Surface et al., 2010). PcG proteins localize ones with promoters enriched in CpG islands (HCP) and those
at genes encoding developmental regulators and correlate with showing poor representation of CpGs (LCP) (Figure 1). HCP
the presence of H3K27 trimethylation both in mouse and human genes are usually devoid of DNA methylation in ESCs and
ESCs. Moreover, mouse ESCs null for specific PcG proteins include self-renewal and housekeeping genes usually marked
exhibit decreased H3K27 methylation and aberrant expression with H3K4me3 and developmental regulators containing bivalent
of key developmental genes (Boyer et al., 2006; Lee et al., domains. This pattern suggests that the targets of trxG are pro-
2006). On the other hand, the H3K4me3 marks present at the tected against DNA methylation and are transcriptionally active
bivalent domains of silent developmental genes could be cata- unless actively repressed by PcG proteins (Mikkelsen et al.,
lyzed by trithorax (trxG) homologs that belong to the MLL family, 2007). LCP genes are not expressed, show DNA methylation,
although their putative contribution to the establishment of these and are mostly devoid of H3K4 or H3K27 methylation in ESCs
domains remains unknown. As part of a highly dynamic and (Meissner et al., 2008). These genes are likely to be targets of
responsive equilibrium, it is likely that histone lysine demethy- early master transcription factors and thus they become induced
lases also contribute to keep the appropriate balance of marks late during differentiation in a tissue-specific manner to carry out
at these domains. Confirming this idea, is the observation that specialized functions in particular tissues.
the PcGs recruit the H3K4 demethylase RBP2 to its target genes Differentiation to one particular lineage implies the permanent
to maintain the proper balance of H3K4 and H3K27 methylation and irreversible silencing of genes involved in alternative line-
at the bivalent domains in mouse ESCs (Pasini et al., 2008). ages as well as those necessary for pluripotent cells. For exam-
To fully understand the role of bivalent domains in ESCs, it ple, pluripotency genes lose H3K4me3 and gain distinct combi-
is important to know how these domains are first established. nations of repressive marks (H3K27me3, H3K9me3, and DNA
Bivalent marks have been reported to exist in pluripotent cells methylation). Bivalent genes that become irreversibly silenced

566 Cell Stem Cell 7, November 5, 2010 ª2010 Elsevier Inc.


Cell Stem Cell

Perspective

Figure 1. Epigenetic Map of Fate


Genome-wide and correlation studies (Boyer et al., 2005; Doi et al., 2009; Fouse et al., 2008; Hawkins et al., 2010; Meissner et al., 2008; Mikkelsen et al., 2007)
have allowed the classification of genes according to their genetic, epigenetic, and transcriptional status in different cell types and physiological situations. This
classification scheme derives from statistical correlations of broad data sets, and thus not all genes will follow the flowchart precisely. Blue arrows denote tran-
sitions from self-renewal to differentiation, red arrows denote transitions to cancer, and gray arrows indicate critical changes to regain pluripotency during
reprogramming. ON and OFF refer to transcriptional status; DNAme refers to DNA methylation; OSN targets refers to genes occupied by Oct4, Sox2, and Nanog;
and trimethylation at lysine 4, 9, or 27 of histone H3 is referred to as H3K4me3, H3K27me3, and H3K9me3, respectively. Marks separated by a slash indicate
co-occurrence of marks.

lose H3K4me3 and preserve H3K27me3 (Cui et al., 2009; Mikkel- that removal of the H3K27me3 mark and maintenance of the
sen et al., 2007). Usually, repression is further reinforced by the H3K4me3 at bivalent genes that become activated during differ-
expansion of the H3K27 methylated areas or the accumulation entiation are coordinated events. Less is known about the
of a second repressive mark, such as H3K9me3 or DNA methyl- molecular regulation and physiological relevance of a small
ation (Hawkins et al., 2010). This ‘‘double lock’’ mechanism percentage of bivalent domains that remain unresolved upon
ensures permanent repression of developmental and pluripo- differentiation and that are thus bivalent in adult cells (Figure 1).
tency genes and thus preserves cell identity. Perhaps more intriguing is the fact that a few new bivalent
How chromatin modifying and remodeling activities partici- regions are established during differentiation (Mikkelsen et al.,
pate in the process of differentiation is not yet fully understood. 2007). Regarding the activities that participate in repression
The switch of signaling events that characterizes the transition during differentiation, the methyltransferase G9a is needed for
from self-renewal to differentiation induces dramatic changes the proper silencing of genes, among which are self-renewal
in gene expression, including the downregulation of genes factors such as Oct4 (Feldman et al., 2006). G9a catalyzes the
involved in self-renewal and changes in the expression of critical methylation of H3K9 at the regulatory regions of these genes
chromatin regulators (Pardo et al., 2010; Wang et al., 2006). This during differentiation, which in turn facilitates the binding of
last event can lead to variations in the subunit composition of heterochromatin protein 1 (HP1) and de novo DNA methylation.
effector complexes, allowing them to carry out specialized func-
tions during differentiation. Contrary to previous perceptions, in Reprogramming and Cell Plasticity
which chromatin marks could be passively diluted, several For many years the differentiated state was thought to be irre-
reports point out the importance of active and targeted removal versible. However, several experimental strategies have been
of marks. Such is the role of the H3K27 demethylases UTX and developed to induce the reactivation of the embryonic program
Jmjd3 in the activation of Hox genes during development (Agger in differentiated cells, a process known as nuclear reprogram-
et al., 2007) and in neuronal commitment (Burgold et al., 2008). ming. Historically, nuclear transfer experiments showed that
Both demethylases associate with MLL complexes, suggesting the genetic content of adult and embryonic cells is equivalent

Cell Stem Cell 7, November 5, 2010 ª2010 Elsevier Inc. 567


Cell Stem Cell

Perspective
Figure 2. Critical Events during
Reprogramming and Differentiation
Transduction of somatic cells with the four factors
causes stress, apoptosis, and replicative senes-
cence. Cells able to overcome this initial barrier
are able to self-renew but still depend on the
expression of the ectopic factors. A critical second
step consists of overcoming the epigenetic barrier
that prevents the reactivation of the endogenous
pluripotency genes. However, after reactivation
of the pluripotency network, several cell divisions
might be needed to erase the adult cellular
memory, a process that is characterized by the re-
gaining of H3K4 and H3K27 trimethylation and
erasing of DNA methylation at bivalent domains.
Proper reestablishment of these domains ensures
the adequate induction of developmental genes
during differentiation and thus is a critical determi-
nant of the quality of the reprogrammed cells. Blue
and orange backgrounds indicate stages depen-
dent on transgene or endogenous pluripotency
factor expression, respectively.

and that the differences lie in gene expression (Wilmut et al., can be trapped in a partially reprogrammed state in which they
1997). Later, cell fusion experiments demonstrated that the self-renew but are dependent on the expression of the trans-
epigenetic activities present in the nucleus of ESCs are able to genes. A second stage of reprogramming is characterized by
induce the reactivation of the embryonic program in differenti- the activation of the endogenous pluripotency factors Oct4,
ated cells (Do and Scholer, 2004). More recently, it has been Sox2, and Nanog. This event allows the maintenance of pluripo-
described that it is possible to generate induced pluripotent tency in an autonomous way, independent of the expressed
stem cells (iPSCs) from somatic cells by overexpressing specific transgenes. Recent findings suggest that the rare occurrence
transcription factors, most commonly Oct4, Sox2, Klf4, and of late events during reprogramming is due to the inability of
c-Myc (Takahashi and Yamanaka, 2006). This methodology the transfactors to bind and activate the regulatory regions of
enables the generation of patient-specific pluripotent cells that the endogenous pluripotency genes, resulting from the presence
could overcome the risk of rejection in future cell-based thera- of an inaccessible repressive chromatin environment (Sridharan
pies and provide a valuable model for mechanistic studies. In a et al., 2009). Several lines of evidence further support this idea.
similar approach, transdifferentiation of one adult cell type to First, it has been recently demonstrated that the overexpression
another can also be achieved by overexpression of appropriate of members of the chromatin remodeling complex BAF enhance
combinations of transcription factors (Davis et al., 1987). reprogramming by facilitating the binding of Oct4 to target pro-
Both nuclear transfer and transcription factor-induced reprog- moters (Singhal et al., 2010). Second, treatment with DNA
ramming are very inefficient processes, and the few animals that methylation inhibitors, which render chromatin more accessible,
are successfully generated by nuclear transfer often suffer from facilitates the completion of reprogramming in partially reprog-
numerous abnormalities. Importantly, the efficiency of reprog- rammed cells (Mikkelsen et al., 2008). Increased reprogramming
ramming differs depending on the somatic cell type and seems efficiency has also been reported in the presence of HDACs,
to be inversely correlated with its degree of differentiation DNA methyltransferases, and G9a inhibitors (Huangfu et al.,
(Maherali and Hochedlinger, 2008). Overall, these findings sug- 2008; Mikkelsen et al., 2008). Finally, it has been suggested
gest the existence of important epigenetic barriers that are that the increased efficiency of reprogramming observed in
imposed during differentiation, probably to preserve cell identity, cord blood cells compared to adult fibroblasts may be due to
and that need to be overcome to successfully reprogram a cell the presence of a more permissive chromatin organization at
back to the pluripotent state. Certainly, the process of reprog- the promoters of self-renewal genes in this population (Giorgetti
ramming entails dramatic changes in gene expression including et al., 2009). Thus, a more complete understanding of the activ-
the transcriptional activation of endogenous self-renewal factors ities and mechanisms that keep pluripotency genes silenced in
and the silencing of differentiation genes. Both events involve differentiated cells appears critical to develop strategies to
profound changes in the chromatin environment at the regula- improve the efficiency of reprogramming.
tory regions of these genes. Critical differentiation genes must Analysis of genome-wide data shows that important changes
reacquire their poised status, as suggested by the reestablish- at the level of DNA methylation occur during reprogramming in
ment of bivalent domains at their proximal promoters (Hawkins bivalent developmental regulators and more generally in genes
et al., 2010), and self-renewal genes must lose the repressive occupied by Oct4, Sox2, and Nanog in ESCs. These genes
chromatin marks that kept them silent in somatic cells (Takaha- become hypomethylated in iPSCs compared to the fibroblast
shi and Yamanaka, 2006). starting population. Indeed, comparison of the methylomes of
Factor-induced reprogramming of fibroblasts is a gradual ESCs and iPSCs revealed that they are very similar, although
process that includes several critical stages (Figure 2). An early some differentially methylated regions can be found, mainly in
stage is characterized by the appearance of alkaline phospha- genes involved in developmental processes. In these differen-
tase activity and the stem cell marker SSEA-1. At this stage cells tially methylated regions, the methylation of iPSCs was found

568 Cell Stem Cell 7, November 5, 2010 ª2010 Elsevier Inc.


Cell Stem Cell

Perspective

to be intermediate between that of ESCs and fibroblasts, sug- a hallmark of cancer. De novo DNA methylation of CpG islands
gesting incomplete reprogramming, but also different from in tumor cells is associated with the transcriptional silencing of
both ESCs and fibroblasts, which might imply aberrant estab- many cancer-related genes, among them tumor suppressors.
lishment of marks during reprogramming (Doi et al., 2009). It is not clear how this aberrant DNA methylation takes place at
Regarding histone modifications, it appears that H3K9me3 con- the molecular level, but the fact that some cancers present
tributes more than H3K27me3 to the differences between iPSCs abnormal expression of chromatin modifiers and that DNA meth-
and ESCs. Both failure to erase K9me3 methylation marks at yltransferase and histone deacetylase (HDAC) inhibitors have
particular regions and the establishment of aberrant H3K9 been successfully used to treat cancer brings to light the rele-
methylated areas during reprogramming have been reported vance of the epigenetic machinery in this process (Esteller,
(Hawkins et al., 2010). The impact of aberrant epigenetic reprog- 2008).
ramming is clearly illustrated by a recent report describing that Promoters marked with bivalent domains in ESCs frequently
the persistent aberrant silencing of a particular imprinted region undergo DNA methylation in a tissue-specific manner during
after reprogramming renders mouse iPSCs developmentally differentiation (Meissner et al., 2008; Mohn et al., 2008), become
challenged (Stadtfeld et al., 2010). Consistently, aberrant hypomethylated during reprogramming, and have a tendency to
imprinted loci have been also reported in cloned embryos (Liu become hypermethylated in cancer (Doi et al., 2009; Widsch-
et al., 2008). wendter et al., 2007). More striking is the finding that these
In essence, the compact chromatin structure present at self- hypermethylated domains display bivalent marks in certain
renewal genes in differentiated cells seems at least partly cancers (Rodriguez et al., 2008). Cancer-dependent DNA hyper-
responsible for the low efficiency of the reprogramming process. methylation of these regions can be reverted during reprogram-
However, a larger negative impact may stem from the potential ming, highlighting the importance of the epigenetic context in the
establishment of aberrant chromatin marks at developmental control of these domains (Li et al., 2003; Ron-Bigger et al., 2010).
genes during reprogramming, because these mistakes might Despite the heterogeneity that cancer cells display, overall these
affect the quality of the reprogrammed cells. Importantly, a data suggest that bivalent regions are critical epigenetic targets
recent study suggests that the aberrant expression of bivalent for defining cell fate and that deregulation of their epigenetic
genes in iPSCs can be inversely correlated with the degree of state can lead to important disorders such as cancer.
pluripotency that these cells display (Boue et al., 2010). Thus, Although cancer cells might resemble ESCs regarding the
improper establishment of chromatin structure at these develop- silencing of tissue-specific genes and the subsequent adoption
mental genes might lead to defective differentiation, including of a more undifferentiated phenotype, evidence suggests that
the inability to generate a particular differentiated cell type or, these parallel outcomes are mediated by distinct mechanisms.
more importantly, the production of immature cell types in which Although in ESCs developmental genes are repressed through
cell identity is not fully established and perhaps prone to rever- bivalent domains and probably the action of Oct4, Sox2,
sion. In line with this hypothesis, a recent study found that iPSCs and Nanog, in cancer cells these genes become permanently
derived from nonhematopoietic cells display reduced blood- silenced by DNA methylation. However, the bivalent domain
forming potential resulting from residual DNA methylation at and cancer DNA hypermethylation connection makes it tempting
loci required for hematopoietic lineage specification (Kim et al., to speculate about the potential stem cell origin of cancer. Thus,
2010). Interestingly, partial reprogramming of bivalent domains in adult stem cells, bivalent domain-containing genes could be
could bestow iPSCs with an epigenetic memory, which might prone to acquire DNA methylation and as a result, generate
make them more prone to differentiate back into cells of their cancer-like cells able to self-renew, but with impaired differenti-
tissue of origin. Indeed, early passage iPSCs have been proven ation potential (Widschwendter et al., 2007).
to retain degree of expression of cell-of-origin-specific genes, In summary, gene silencing plays critical roles during develop-
which facilitates their differentiation into their original lineage ment, differentiation, reprogramming, and cancer. Thus, a better
(Polo et al., 2010). However, cell-of-origin differences tend to understanding of the activities, mechanisms, and signals that
disappear after successive passages in culture, suggesting control the silencing of critical genes will help to design strate-
that complete epigenetic reprogramming to pluripotency is a gies to improve the efficiency of reprogramming and to treat
slow and gradual process. Thus, after the reactivation of the cancer.
endogenous pluripotency genes, a third critical step, the full
reestablishment of bivalent domains, involving both histone ACKNOWLEDGMENTS
marks and DNA demethylation, is essential to achieve pluripo-
tent status (Figure 2). Although the efficiency of reprogramming We apologize for those authors whose work could not be cited because of
is often reported based on the detection of alkaline phosphatase space limitation. We would like to thank all the members of the J.C.I.B. lab
at The Salk Institute for Biological Studies and the CMRB for their insightful
activity or the reactivation of pluripotency genes, quality will defi- comments and input. This work was partially supported by grants RYC-
nitely need to be judged based on the complete epigenetic 2007-01510 and SAF2009-08588 from the Ministerio de Ciencia e Innovación
reprogramming of critical differentiation genes (Maherali and of Spain to M.J.B. and grants from the G. Harold and Leila Y. Mathers Chari-
table and Cellex Foundations, Sanofi-Aventis, and MICINN to J.C.I.B.
Hochedlinger, 2008).

Differentiation, Dedifferentiation, and Cancer: A Stem REFERENCES


Cell Link to Cancer?
Agger, K., Cloos, P.A., Christensen, J., Pasini, D., Rose, S., Rappsilber, J.,
Although malignant transformation is usually associated with Issaeva, I., Canaani, E., Salcini, A.E., and Helin, K. (2007). Nature 449,
mutations and translocations, aberrant DNA methylation is also 731–734.

Cell Stem Cell 7, November 5, 2010 ª2010 Elsevier Inc. 569


Cell Stem Cell

Perspective
Azuara, V., Perry, P., Sauer, S., Spivakov, M., Jorgensen, H.F., John, R.M., Li, L., Connelly, M.C., Wetmore, C., Curran, T., and Morgan, J.I. (2003). Cancer
Gouti, M., Casanova, M., Warnes, G., Merkenschlager, M., et al. (2006). Nat. Res. 63, 2733–2736.
Cell Biol. 8, 532–538.
Li, G., Margueron, R., Ku, M., Chambon, P., Bernstein, B.E., and Reinberg, D.
Bernstein, B.E., Mikkelsen, T.S., Xie, X., Kamal, M., Huebert, D.J., Cuff, J., Fry, (2010). Genes Dev. 24, 368–380.
B., Meissner, A., Wernig, M., Plath, K., et al. (2006). Cell 125, 315–326.
Liu, J.H., Yin, S., Xiong, B., Hou, Y., Chen, D.Y., and Sun, Q.Y. (2008). Mol.
Boue, S., Paramonov, I., Barrero, M.J., and Izpisua Belmonte, J.C. (2010). Reprod. Dev. 75, 598–607.
PLoS ONE 5, e12664.
Maherali, N., and Hochedlinger, K. (2008). Cell Stem Cell 3, 595–605.
Boyer, L.A., Lee, T.I., Cole, M.F., Johnstone, S.E., Levine, S.S., Zucker, J.P.,
Guenther, M.G., Kumar, R.M., Murray, H.L., Jenner, R.G., et al. (2005). Cell
Meissner, A., Mikkelsen, T.S., Gu, H., Wernig, M., Hanna, J., Sivachenko, A.,
122, 947–956.
Zhang, X., Bernstein, B.E., Nusbaum, C., Jaffe, D.B., et al. (2008). Nature
Boyer, L.A., Plath, K., Zeitlinger, J., Brambrink, T., Medeiros, L.A., Lee, T.I., 454, 766–770.
Levine, S.S., Wernig, M., Tajonar, A., Ray, M.K., et al. (2006). Nature 441,
349–353. Meshorer, E., Yellajoshula, D., George, E., Scambler, P.J., Brown, D.T., and
Misteli, T. (2006). Dev. Cell 10, 105–116.
Brykczynska, U., Hisano, M., Erkek, S., Ramos, L., Oakeley, E.J., Roloff, T.C.,
Beisel, C., Schubeler, D., Stadler, M.B., and Peters, A.H. (2010). Nat. Struct. Mikkelsen, T.S., Ku, M., Jaffe, D.B., Issac, B., Lieberman, E., Giannoukos, G.,
Mol. Biol. 17, 679–687. Alvarez, P., Brockman, W., Kim, T.K., Koche, R.P., et al. (2007). Nature 448,
553–560.
Burgold, T., Spreafico, F., De Santa, F., Totaro, M.G., Prosperini, E., Natoli, G.,
and Testa, G. (2008). PLoS ONE 3, e3034. Mikkelsen, T.S., Hanna, J., Zhang, X., Ku, M., Wernig, M., Schorderet, P.,
Bernstein, B.E., Jaenisch, R., Lander, E.S., and Meissner, A. (2008). Nature
Cui, K., Zang, C., Roh, T.Y., Schones, D.E., Childs, R.W., Peng, W., and Zhao, 454, 49–55.
K. (2009). Cell Stem Cell 4, 80–93.
Mohn, F., Weber, M., Rebhan, M., Roloff, T.C., Richter, J., Stadler, M.B., Bibel,
Davis, R.L., Weintraub, H., and Lassar, A.B. (1987). Cell 51, 987–1000. M., and Schubeler, D. (2008). Mol. Cell 30, 755–766.
Do, J.T., and Scholer, H.R. (2004). Stem Cells 22, 941–949.
Pardo, M., Lang, B., Yu, L., Prosser, H., Bradley, A., Babu, M.M., and Choudh-
Doi, A., Park, I.H., Wen, B., Murakami, P., Aryee, M.J., Irizarry, R., Herb, B., ary, J. (2010). Cell Stem Cell 6, 382–395.
Ladd-Acosta, C., Rho, J., Loewer, S., et al. (2009). Nat. Genet. 41, 1350–1353.
Pasini, D., Hansen, K.H., Christensen, J., Agger, K., Cloos, P.A., and Helin, K.
Efroni, S., Duttagupta, R., Cheng, J., Dehghani, H., Hoeppner, D.J., Dash, C., (2008). Genes Dev. 22, 1345–1355.
Bazett-Jones, D.P., Le Grice, S., McKay, R.D., Buetow, K.H., et al. (2008). Cell
Stem Cell 2, 437–447. Pasini, D., Cloos, P.A., Walfridsson, J., Olsson, L., Bukowski, J.P., Johansen,
J.V., Bak, M., Tommerup, N., Rappsilber, J., and Helin, K. (2010). Nature 464,
Esteller, M. (2008). N. Engl. J. Med. 358, 1148–1159. 306–310.

Feldman, N., Gerson, A., Fang, J., Li, E., Zhang, Y., Shinkai, Y., Cedar, H., and Peng, J.C., Valouev, A., Swigut, T., Zhang, J., Zhao, Y., Sidow, A., and
Bergman, Y. (2006). Nat. Cell Biol. 8, 188–194. Wysocka, J. (2009). Cell 139, 1290–1302.

Fouse, S.D., Shen, Y., Pellegrini, M., Cole, S., Meissner, A., Van Neste, L., Jae- Polo, J.M., Liu, S., Figueroa, M.E., Kulalert, W., Eminli, S., Tan, K.Y., Aposto-
nisch, R., and Fan, G. (2008). Cell Stem Cell 2, 160–169. lou, E., Stadtfeld, M., Li, Y., Shioda, T., et al. (2010). Nat. Biotechnol. 28,
848–855.
Gargiulo, G., Levy, S., Bucci, G., Romanenghi, M., Fornasari, L., Beeson, K.Y.,
Goldberg, S.M., Cesaroni, M., Ballarini, M., Santoro, F., et al. (2009). Dev. Cell
Rodriguez, J., Munoz, M., Vives, L., Frangou, C.G., Groudine, M., and Peinado,
16, 466–481.
M.A. (2008). Proc. Natl. Acad. Sci. USA 105, 19809–19814.
Gaspar-Maia, A., Alajem, A., Polesso, F., Sridharan, R., Mason, M.J., Heiders-
bach, A., Ramalho-Santos, J., McManus, M.T., Plath, K., Meshorer, E., et al. Ron-Bigger, S., Bar-Nur, O., Isaac, S., Bocker, M., Lyko, F., and Eden, A.
(2009). Nature 460, 863–868. (2010). Stem Cells 28, 1349–1354.

Giorgetti, A., Montserrat, N., Aasen, T., Gonzalez, F., Rodriguez-Piza, I., Vas- Singhal, N., Graumann, J., Wu, G., Arauzo-Bravo, M.J., Han, D.W., Greber, B.,
sena, R., Raya, A., Boue, S., Barrero, M.J., Corbella, B.A., et al. (2009). Cell Gentile, L., Mann, M., and Scholer, H.R. (2010). Cell 141, 943–955.
Stem Cell 5, 353–357.
Sridharan, R., Tchieu, J., Mason, M.J., Yachechko, R., Kuoy, E., Horvath, S.,
Guenther, M.G., Levine, S.S., Boyer, L.A., Jaenisch, R., and Young, R.A. Zhou, Q., and Plath, K. (2009). Cell 136, 364–377.
(2007). Cell 130, 77–88.
Stadtfeld, M., Apostolou, E., Akutsu, H., Fukuda, A., Follett, P., Natesan, S.,
Hawkins, R.D., Hon, G.C., Lee, L.K., Ngo, Q., Lister, R., Pelizzola, M., Edsall, Kono, T., Shioda, T., and Hochedlinger, K. (2010). Nature 465, 175–181.
L.E., Kuan, S., Luu, Y., Klugman, S., et al. (2010). Cell Stem Cell 6, 479–491.
Surface, L.E., Thornton, S.R., and Boyer, L.A. (2010). Cell Stem Cell 7,
Ho, L., Ronan, J.L., Wu, J., Staahl, B.T., Chen, L., Kuo, A., Lessard, J., Nesvizh- 288–298.
skii, A.I., Ranish, J., and Crabtree, G.R. (2009). Proc. Natl. Acad. Sci. USA 106,
5181–5186. Takahashi, K., and Yamanaka, S. (2006). Cell 126, 663–676.
Huangfu, D., Maehr, R., Guo, W., Eijkelenboom, A., Snitow, M., Chen, A.E.,
Vastenhouw, N.L., Zhang, Y., Woods, I.G., Imam, F., Regev, A., Liu, X.S., Rinn,
and Melton, D.A. (2008). Nat. Biotechnol. 26, 795–797.
J., and Schier, A.F. (2010). Nature 464, 922–926.
Kim, K., Doi, A., Wen, B., Ng, K., Zhao, R., Cahan, P., Kim, J., Aryee, M.J., Ji,
H., Ehrlich, L.I., et al. (2010). Nature 467, 285–290. Wang, J., Rao, S., Chu, J., Shen, X., Levasseur, D.N., Theunissen, T.W., and
Orkin, S.H. (2006). Nature 444, 364–368.
Landeira, D., Sauer, S., Poot, R., Dvorkina, M., Mazzarella, L., Jorgensen, H.F.,
Pereira, C.F., Leleu, M., Piccolo, F.M., Spivakov, M., et al. (2010). Nat. Cell Biol. Widschwendter, M., Fiegl, H., Egle, D., Mueller-Holzner, E., Spizzo, G., Marth,
12, 618–624. C., Weisenberger, D.J., Campan, M., Young, J., Jacobs, I., et al. (2007). Nat.
Genet. 39, 157–158.
Lee, T.I., Jenner, R.G., Boyer, L.A., Guenther, M.G., Levine, S.S., Kumar, R.M.,
Chevalier, B., Johnstone, S.E., Cole, M.F., Isono, K., et al. (2006). Cell 125, Wilmut, I., Schnieke, A.E., McWhir, J., Kind, A.J., and Campbell, K.H. (1997).
301–313. Nature 385, 810–813.

570 Cell Stem Cell 7, November 5, 2010 ª2010 Elsevier Inc.

Das könnte Ihnen auch gefallen