REVIEW ARTICLE

Epigenetic regulation of adult stem cell function Lorenzo Rinaldi1,2,3 and Salvador Aznar Benitah3,4 1 2 3 4

Centre for Genomic Regulation, Barcelona, Spain Universitat Pompeu Fabra, Barcelona, Spain Institute for Research in Biomedicine, Barcelona, Spain Catalan Institution for Research and Advanced Studies, Barcelona, Spain

Keywords adult stem cells, ageing and cancer, epigenetic regulators, lineage commitment, tissue homeostasis Correspondence S. A. Benitah, Institute for Research in Biomedicine (IRB Barcelona), C/ Baldiri Reixac 10, 08028 Barcelona, Spain Fax: 0034 93 403 71 14 Tel: 0034 93 403 40 21 E-mail: [email protected] (Received 12 June 2014, revised 17 July 2014, accepted 22 July 2014) doi:10.1111/febs.12946

Understanding the cellular and molecular mechanisms that specify cell lineages throughout development, and that maintain tissue homeostasis during adulthood, is paramount towards our understanding of why we age or develop pathologies such as cancer. Epigenetic mechanisms ensure that genetically identical cells acquire different fates during embryonic development and are therefore essential for the proper progression of development. How they do so is still a matter of intense investigation, but there is sufficient evidence indicating that they act in a concerted manner with inductive signals and tissue-specific transcription factors to promote and stabilize fate changes along the three germ layers during development. In consequence, it is generally hypothesized that epigenetic mechanisms are also required for the continuous maintenance of cell fate during adulthood. However, in vivo models in which different epigenetic factors have been depleted in different tissues do not show overt changes in cell lineage, thus not strongly supporting this view. Instead, the function of some of these factors appears to be primarily associated with tissue functionality, and a strong causal relationship has been established between their misregulation and a diseased state. In this review, we summarize our current knowledge of the role of epigenetic factors in adult stem cell function and tissue homeostasis.

Introduction The role of epigenetic mechanisms in establishing different cell fates during embryogenesis is fairly well understood. However, if and how epigenetic mechanisms are necessary to stably maintain the identity of adult stem cells and their progeny during tissue homeostasis and repair is still under intense investigation. In recent years, a considerable number of studies have shown that factors involved in chromatin regulation during embryogenesis have diverse roles in adulthood, which are not always related to epigenetics.

Adult stem cells maintain adult tissue homeostasis by replenishing dying or damaged cells or by regenerating injuries. However, since tissues significantly differ in their turnover rates, each imposes a different demand for cellular input from its resident stem cells. Intriguingly, a variety of mechanisms have evolved in different adult stem cells that involve the interplay between quiescent and actively proliferating stem cell populations, stochastic regulation of symmetric or asymmetric stem cell divisions, neutral competition of the stem cell progeny, or combinations of these, all of which effi-

Abbreviations 5-Mc, 5-methylcytosine; DMR, differentially methylated regions; DNMT, DNA methyltransferase; epSC, epidermal stem cell; ESC, embryonic stem cell; HSC, hematopoietic stem cell; NSC, neural stem cell; PcG, polycomb group; PRC, polycomb repressive complex.

FEBS Journal (2014) ª 2014 FEBS

1

Epigenetic regulation of adult stem cell function

L. Rinaldi and S. A. Benitah

ciently ensure tissue homeostasis [1,2]. In addition, some tissues, such as the keratinocyte compartment of the skin, the mammary gland or the prostate, are highly compartmentalized and rely on distinct populations of stem cells, often located adjacent to each other, that display completely different behaviours [1,3–7]. How different stem cells are stably established during embryogenesis, and how they retain or lose their identity while self-renewing or differentiating respectively during steady state, is still largely unknown. Epigenetic modifications are chemical modifications either of double strand DNA or of the core nucleosome histones, catalysed by different families of enzymes. These modifications alter the structure of chromatin by either facilitating or preventing the binding of transcription factors and other cofactors. Importantly, although the turnover of some of these modifications is far more rapid than previously thought, often they are stably transmitted after a cellular division and can therefore affect the identity of the progeny. In this sense, epigenetic mechanisms allow genetically identical cells to stably adopt different

phenotypes by controlling the transcription availability of different parts of the genome packaging or opening different parts of the chromatin [8,9]. In fact, findings from many laboratories in recent years indicate that the combination of transcription factors and chromatin remodelling factors might be essential for different aspects of the biology of adult stem cells. Importantly, a number of epigenetic factors are mutated in human cancers or deregulated during ageing, suggesting that perturbations in their functions may be causative of disease. This review focuses on recent advances in our understanding of the roles of different epigenetic mechanisms in regulating adult stem cell specification and function during homeostasis (Table 1).

Modification of chromatin and DNA Histone modifications Histones can be post-translationally modified at different amino acid residues through methylation, acetylation, ubiquitination, sumoylation and phosphorylation.

Table 1. Summary of the role of epigenetic factors in different adult stem cells and phenotypes associated with their genetic manipulation in vivo. Main epigenetic complexes PRC1

PRC2

5Mc/DNMTs

5-Hmc/TET G9A/GLP

MLL1/3

2

Function in adult stem cells

Epigenetic mechanism disruption

Reference

Self-renewal of hematopoietic, epidermal and neural stem cells Regulation of cell cycle and inhibition of senescence and differentiation

Defects in blood cell maturation

[50–57,81,82,121,122]

Hematopoietic stem cell self-renewal and differentiation Differentiation of neural and epidermal stem cells Maintenance of hematopoietic stem cell, epidermal stem cell, neural stem cell and muscle stem cell (satellite cell) self-renewal Drives hematopoietic stem cell differentiation upon transplantation (Dnmt3a) Abundant in neural stem cells Drives expression of neuronal genes Hematopoietic stem cell differentiation Specification of neurons during development and under external stress Represses adipogenesis Hematopoietic stem cell self-renewal Promotes adipogenesis

Cell cycle defects and premature differentiation of epidermal stem cells Neurological defects Reduced blood and neural stem cell activity

[58–61,76–79,119,120]

Hair follicle cycling defects, and epidermal differentiation Impaired number of mature blood cells

[62–67,96,97,105–107]

Loss of Dnmt1 results in spontaneous and irreversible differentiation of human epidermal stem cells Impaired neural activity causing cognitive defects Impairment in spatial learning and memory Reduced number of neural stem cells Delayed blood stem cell differentiation Altered neuronal gene expression causing impaired cognitive functions

[111–114]

Increased fat accumulation and tissue weight Loss of hematopoietic stem cell self-renewal potential Reduction of fat accumulation

[71–126]

[69–99,125]

FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

Epigenetic regulation of adult stem cell function

The function and activity of these proteins has been extensively reviewed elsewhere [8–12]. In this section we will briefly introduce just those epigenetic factors whose activity has been shown so far to affect the biology of adult tissues. Amongst the most studied histone modifiers are polycomb group (PcG) proteins. PcG proteins were identified in Drosophila and have since been shown to be essential for early development, X chromosome inactivation, adult homeostasis and reprogramming to pluripotency [10,12–15]. PcG proteins promote chromatin remodelling and compaction that leads to a transcription inhibition mainly through the formation of polycomb repressive complexes (PRCs) [9,11]. The two major PRCs, named PRC1 and PRC2, mediate the ubiquitination of histone H2A and the methylation of histone H3, respectively. The core PRC2 is formed by the subunits EED, Suz12 and the histone methyltransferases Ezh1 and Ezh2. On the other hand, Ring1a/b and members of the Pcgf and PhC family of proteins form the PRC1 complex. The classical model of PRC2–PRC1 action establishes that PRC2 methylates lysine 27 of histone H3 (H3K27me3) and that the PRC1 complex recognizes this modification through the chromodomain of its Cbx subunit. PRC1 in turn ubiquitinates lysine 119 at histone H2A (H2AK119ub) through the ring finger protein RING1A/B (Fig. 1A). However, recent studies indicate that this linear model is not always followed, and that the composition of PRC2 and PRC1 varies according to different possible combinations of their subunits (i.e. whether PRC2 contains Ezh2 or Ezh1, or whether PRC1 contains the proteins Bmi1 or Mel18) and is context dependent (Fig. 1B) [16]. In this sense, PRC1 complexes that do not even contain a Cbx subunit capable of interacting with H3K27me3 have been

identified and shown to play an important role during early embryogenesis [10,17–20]. Furthermore, although PRC2 was originally identified as a repressor associated with the presence of H3K27me3 mark, evidence suggests that EZH1 can locate to actively transcribed genes independently of its histone methyltransferase activity [21]. This interesting complexity in the composition and function of PRC2 and PRC1 might in fact endow them with the ability to contribute to different aspects of embryonic development and adult tissue maintenance in a context-dependent manner. Deletion of essential members of both complexes is incompatible with a proper development, underscoring their importance in early embryogenesis [9]. PcG proteins also interact with other epigenetic factors such as G9a and members of the thritorax family proteins. G9a is responsible for establishing dimethylation of lysine in position 9 on histone 3 (H3K9me2), a mark that is often associated with transcriptional repression. Interestingly, H3K9me2-rich regions are organized in ‘large organized chromatin K9 modification blocks’ (LOCKs), that are highly conserved between mammals and have been proposed to play a crucial role compacting chromatin during the commitment of embryonic cells into diverse somatic lineages such as neurons and hepatocytes [22]. Similar to PRC1 and PRC2, the activity of G9a is indispensable for proper development, and its deletion in embryonic stem cells (ESCs) results in embryonic lethality [23]. Another family of epigenetic factors, the thritorax group proteins, is involved in gene activation and they both interact with PcG proteins and other histone modifications. Thritorax complex allows gene expression by producing the trimethylation of lysine 4 at histone 3 (H3K4me3) [24]. H3K4me3 is well established to be

A

Fig. 1. Schematic representation of the regulation of histone post-translational modifications by PRC1 and PRC2 complexes. (A) PRC2 complex deposits H3K27me3, which is recognized by PRC1 containing a Cbx unit. PRC1 in turn catalyzes the ubiquitination of H2A lys 119, resulting in transcriptional repression. (B) A non-canonical interplay between PRC1 and PRC2 also leads to gene repression, whereby PRC1 can establish H2Aub119, which serves as a docking site for PRC2 and subsequent H3K27 trimethylation.

FEBS Journal (2014) ª 2014 FEBS

B

3

Epigenetic regulation of adult stem cell function

present at the promoter of active genes. The first and most described thritorax member is MLL1, initially noted for its high frequency mutation rate in human cancers. Other MLL proteins are involved in methylating H3K4 at gene regulatory regions such as enhancers, suggesting that their role might also be important for fate decisions during embryogenesis [24–26]. Modifications of double strand DNA DNA methylation occurs on the fifth position of the cytosine (5-Mc) at the dinucleotide cytosine-guanine (CpG), and it is involved in the regulation of several aspects of gene expression such as long-term gene silencing, transcriptional elongation and maintenance of genomic stability. The mechanisms underlying the establishment, maintenance and removal of DNA methylation have been extensively reviewed elsewhere [27,28]. Briefly, during mammalian development, the pattern of DNA methylation is established by the de novo DNA methyltransferases DNMT3A and DNMT3B [29–31]. This pattern of DNA methylation is in turn primarily maintained by DNMT1, although with the help of DNMT3A and DNMT3B. In addition, an accessory inactive DNA methyltransferase (DNMT3L) modulates their activities [32] (Fig. 2). DNA methylation, mainly if clustered, has always been associated with gene repression; however, in the last few years, genome-wide methylation analyses have revealed a large variety of patterns of DNA methylation that do not always correlate with the regulation of gene expression. Generally, however, if the promoter of a gene is methylated, it will not be actively transcribed. Conversely, if the exons of the gene have high levels of 5-Mc, this correlates with an actively transcribed gene. Finally, regions of compacted chromatin such as heterochromatin sites, retrotransposons, microsatellites and telomeres normally have very high levels of methylation [33–35]. However, it is still unclear whether changes in DNA methylation are

L. Rinaldi and S. A. Benitah

causative of regulating gene expression per se or rather important for the long-term stabilization of the threedimensional conformation and/or gene expression activity downstream of other regulatory proteins. Recent studies in adult stem cells support this last hypothesis [36,37]. Interestingly, 5-Mc is a reversible modification, since methylated cytosines can be either actively or passively demethylated [27,28]. Passive demethylation occurs during cell replication when DNA methyltransferases might be inhibited or absent, resulting in a locus-specific or global loss of DNA methylation [28,38,39]. On the other hand, active demethylation relies on the ability of 5-Mc to be oxidized into 5-hydroxymethylcytosine (5Hmc) by TET proteins [40] (Fig. 2). For instance, in the very early stages of the formation of the mammalian embryo, DNA is highly demethylated by TET1/2 with a progressive and transient increase of hydroxymethylation. Subsequently, the de novo DNMTs re-establish the correct pattern of DNA methylation in the second week of the murine embryonic stage. Interestingly, DNA methylation does not appear to occur independently of histone modifications, and their concerted action might be essential for the correct spatial and temporal regulation of cell fates during development [9,10,13,30,31,41,42]. How DNA methylation and hydroxymethylation are dynamically regulated during adulthood and their impact in adult tissue homeostasis will be discussed below. Other epigenetic mechanisms Recently, much attention has been paid to studying the role of other epigenetic mechanisms that might regulate gene expression throughout modifications in the high order three-dimensional conformation of chromatin. Amongst these, distant regulatory regions, also called enhancers, have been defined by a combination of distinct histone marks including acetylation of lysine 27 in histone 3 (H3K27ac) and for having a high

Fig. 2. Schematic representation of the role of de novo DNA methyltransferases, maintenance DNA methyltransferases and TET proteins in regulating the methylation and demethylation of DNA.

4

FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

ratio of monomethylation at lysine 4 in histone H3 (H3K4me1) versus trimethylation of the same residue (H3K4me3) [43–45]. Enhancers are abundant in the mammalian genome and they normally show low levels of DNA methylation [25,46]. The importance of these distant regulatory regions is still under investigation, but it is thought that enhancers are capable of driving and stabilizing gene expression in different types of cells during embryogenesis, although their role in adult stem cell homeostasis is still unknown [45,47].

Epigenetics of adult stem cells and adult progenitors Hematopoietic stem cells The hematopoietic system has one of the highest turnover rates among adult tissues. Hematopoietic stem cells (HSCs) residing at the bone marrow are primarily responsible for the daily maintenance of hematopoietic homeostasis during adulthood [48]. Two types of HSC states coexist, a quiescent population thought to constitute a long-term reservoir for injury repair, and an actively dividing short-term subset mainly responsible for the daily sustenance of haematopoiesis [48]. These two populations also differ in their long-term selfrenewal potential. HSCs can commit to two branches, the common lymphoid progenitor or the common myeloid progenitors. Lymphoid progenitors differentiate into diverse lymphocytes, mostly B- and T-cells, whereas common myeloid progenitors instead give rise to all the other blood cells, including platelets, erythrocytes and other immune cells [49]. PRC1 and PRC2 in HSC self-renewal and differentiation Several studies have shown that chromatin remodelling factors are important regulators of HSC self-renewal and differentiation. For instance, PRC1 is important for self-renewal of HSCs. In this sense, forced expression of Bmi1 in mouse ESCs leads to an upregulation of genes involved in haematopoiesis [50], and its overexpression in adult HSCs enhances their self-renewal potential. Accordingly, deletion of BMI1 impairs HSC function and results in defects in the bone marrow microenvironment that sustains HSC homeostasis [51]. Furthermore, deletion of other PRC1 members such as MEL18, Phc1, Phc2, RING1A/B and Cbx2 results in a phenotype similar to that of the deletion of BMI1 [9,52]. Mechanistically, PRC1 maintains HSC longterm self-renewal through the repression of the cell cycle inhibitors p16INK4a and p19ARF [53–55]. In FEBS Journal (2014) ª 2014 FEBS

Epigenetic regulation of adult stem cell function

accordance with this, the deletion of the p16ink4a or P19Arf genes partially rescues the defects in HSC BMI1-deficient mice [53]. In addition, BMI1 prevents premature lineage specification of both HSCs and multipotent progenitors, repressing directly lymphoid lineage genes such as EBF1 and PAX5 [54]. However, PRC1 is not only involved in HSC selfrenewal but is also required for efficient HSC differentiation. Five different Cbx subunits can interact with the core PRC1 complex and are capable of recognizing the histone mark H3K27me3 deposited by PRC2 [56]. Two independent studies have recently shown that the expression of Cbx proteins, and their incorporation into the PRC1 complex, is dynamically regulated during the process of ESC differentiation [17,19]. Cbx7 is the main Cbx subunit expressed in pluripotent ESCs and is exclusively associated with the PRC1 to repress the expression of lineage commitment genes and the other Cbx subunits, with the exception of Cbx6 [17]. However, when ESCs differentiate, Cbx7 is replaced by Cbx8, Cbx4, Cbx2, resulting in different PRC1 complexes that delineate the commitment of ESCs into the three germ layers, while repressing genes that promote pluripotency, including Cbx7 [17–19]. Thus, an intriguing PRC1 auto-regulatory loop is required to balance ESC pluripotency and differentiation. Interestingly, this dynamic behaviour is conserved also in HSCs. In long-term HSCs, PRC1 contains predominantly Cbx7, whose activity enhances HSC cell proliferation and self-renewal [57]. Conversely, during the commitment of HSCs into the lymphoid or myeloid lineages, expression of Cbx7 decreases and is replaced by Cbx8, Cbx2 or Cbx4 within PRC1, which in turn induce HSC differentiation [57]. Similar to PRC1, PRC2 might act at different levels along the axis of HSC self-renewal and differentiation. For instance, mutations that inactivate PRC2 components including SUZ12, EZH2 or EED enhance HSC activity. In addition, PRC2-deficient HSCs are more capable of repopulating the hematopoietic system than the wild type [52,58]. However, intriguingly, conditional deletion of EED enhances HSC/progenitor activity, as in null mutation conditions, but renders HSCs incapable of properly differentiating, leading to an exhaustion of the long-term HSC pool [59]. The impaired proliferation of EED-deficient HSCs is due to high expression of the cell cycle genes p16ink4a and p19Arf and can only be partially rescued by the deletion of CDKN2A. However, knockout of CDKN2A cannot rescue the inability of EED-null HSCs to differentiate [59], whereas it restores the hematopoietic defects in BMI1 and EZH1 knockout mice [51,60]. In addition, deletion of either EZH1 or EZH2 in HSCs 5

Epigenetic regulation of adult stem cell function

results in different and weaker HSC functional defects than by the deletion of EED. Altogether, this suggests that different PRC2 complexes formed by distinct combinations of its subunits might impinge on HSC function at different levels. It can also be speculated that these different PRC2 subunits might have PRC2independent functions in haematopoiesis [59]. For instance, EED acts as a linker between PRC2 and PRC1 function in ESCs by helping recruit PRC1 to H3K27me3 and enhancing its ubiquitinase activity [61]. Thus, this dual interactive mechanism of EED with PRC2 and PRC1 could explain why its deletion in HSCs results in such a strong phenotypic effect compared with other PRC2 members. DNA methyltransferases in HSCs: self-renewal or differentiation? DNA methylation has been identified as an epigenetic modification important for several aspects of haematopoiesis. For instance, differential regulation of DNA methylation is important for the specification of HSCs into myeloid versus lymphoid progenitors [62]. However, DNA methylation also maintains HSC multipotency, but might also be required for the stable establishment of all blood lineages derived from common lymphoid and myeloid progenitors [62,63]. Accordingly, HSCs deficient in the maintenance DNA methyltransferase Dnmt1 are incapable of differentiating into all the blood lineages, and interestingly also disrupt the bone marrow HSC niche [64]. The role of the de novo DNA methyltransferases Dnmt3a and Dnmt3b in HSC function is less clear. A report by Tadokoro et al. (2007) indicated that both proteins are required for the long-term maintenance of HSCs but not for their differentiation into the myeloid and lymphoid lineages [65]. However, the opposite role has recently been described for DNMT3A in murine HSCs. Deletion of DNMT3A in HSCs resulted in no overt hematopoietic phenotype in unchallenged conditions. On the other hand, upon serial transplantation to reconstitute the hematopoietic system of lethally irradiated mice, DNMT3A-deficient HSCs progressively accumulated in their multipotent state and displayed a significantly reduced potential to differentiate [66]. Interestingly, work from the same group has recently shown that DNMT3A is responsible for maintaining borders between regions with high and low levels of 5-Mc that regulate the expression of genes essential for HSC self-renewal and differentiation [67]. Furthermore, the DNA methylation of these regions is altered in aged HSCs, as discussed below [68]. Despite these facts, the specific role of Dnmt3a and Dnmt3b in 6

L. Rinaldi and S. A. Benitah

regulating DNA methylation and the precise impact over gene expression of the pattern of DNA methylation they establish towards the function of HSCs and all lineages downstream has not been determined. Function of histone methyltransferases in HSCs As mentioned above, the methyltransferase G9a together with its partner GLP is essential to establish and maintain the repressive mark H3K9me2. The expression of G9a/GLP, and the levels of its functional readout H3K9me2, increase during HSC differentiation [69]. Regions containing high levels of H3K9me2 can be organized into large organized chromatin blocks, known as LOCKs [22]. In this sense, it has been hypothesized that the increased enrichment for H3K9me2-containing LOCKs during HSC differentiation might diminish transcriptional noise and increase the robustness of the activity of transcription factors that drive HSC differentiation [69]. In accordance with this, depleting human HSCs of G9a/GLP delays their capacity to differentiate in vitro [69]. On the other hand, deletion of G9a in murine HSCs does not result in any relevant phenotype but is required for the proliferation of leukaemic cells in an H3K9me2-dependent manner, thereby impairing the progression of leukaemia in vivo [70]. Another histone methyltransferase that is involved in haematopoiesis is MLL, a member of the thritorax complex that catalyses the methylation of lysine 4 at histone 3 (H3K4) [71]. Embryonic deletion of MLL results in reduced numbers of foetal HSCs [72]. The long-term quiescent pool of HSCs is reduced, and their ability to restore a functional hematopoietic system when transplanted into lethally irradiated mice is significantly impaired. Interestingly, deletion of MLL in adult hematopoietic cells does not affect HSC numbers, but these MLL-deficient HSCs remain incapable of regenerating blood cells in irradiated recipients [26]. Intriguingly, this phenotype is not dependent on the ability of MLL to methylate histone H3 on MLL target genes but rather on its interaction with the histone H4K16 methyltransferase MOF, suggesting that the cooperation of two chromatin remodelling factors is essential for HSC activity [72]. Epigenetic factors regulating epidermal stem cell function The keratinocyte compartment of the skin is formed by the epidermis and appendages including hair follicles, sebaceous glands and sweat glands [3,73]. The epidermis and the structures physically linking it to the FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

hair follicles are under constant demand of cellular replenishment, imposing a strong pressure over its resident stem cells. Conversely, the hair follicles undergo bouts of hair growth followed by long periods of dormancy. Each compartment is maintained by its own subset of stem cells that accordingly show a very different behaviour [3]. So far several populations of stem cells have been identified in all these compartments, although it is not known how epigenetic mechanisms influence their establishment during development [74] or their maintenance in adulthood [3,73]. Recent reports have nevertheless provided much information on how chromatin remodelling factors regulate the behaviour of some of these stem cell subsets during the homeostasis of their particular compartment in adulthood. These will now be discussed. Histone modifications in stem cells of the epidermis and hair follicles The stem cells of the epidermis must cope with a constant demand of cellular supply due to the loss of cornified cells in the uppermost layer of the epidermis. Interestingly, the choice that actively dividing epidermal stem cells (epSC) make is under a stochastic regulation, whereby it cannot be predicted whether a basal cell in the epidermis will divide symmetrically into two stem cells or two differentiated cells or asymmetrically to produce one daughter stem cell and a differentiated cell [1,75]. Whether these stochastic choices require the influence of chromatin remodelling factors to allow for transcriptional noise and plasticity is not known. Furthermore, similar to HSCs, murine and human epidermis also contains a relatively quiescent population of stem cells thought to contribute primarily to damage repair [1,3]. However, once the stochastic fate of the epidermal stem cell has been determined, its quiescence, proliferation or differentiation does require the concerted activity of different epigenetic complexes [74]. For instance, the activity of the PRC2 complex promotes the proliferation and prevents the differentiation of embryonic and new-born mouse epidermis [76,77]. Accordingly, the expression of Ezh2 and Eed is reduced as epSC differentiate, and the removal of the repressive H3K27me3 mark in differentiation genes by the histone demethylase Jmjd3 is necessary for their efficient differentiation [77,78]. Interestingly, the expression of EZH1 follows the opposite pattern from EZH2 upon differentiation, and deletion of EZH2 results in a reduction of H3K27me3 in basal stem cells but not in suprabasal differentiated cells [78]. This suggests that Ezh1 might be required for the deposition of H3K27me3 in genes that should not be expressed in FEBS Journal (2014) ª 2014 FEBS

Epigenetic regulation of adult stem cell function

suprabasal differentiated epidermal cells [78]. In line with this, deletion of EZH1 and EZH2 results in a hyperproliferative epidermis rather than the hypoproliferation observed in single Ezh2 epidermal knockout mice [79]. Similar to the epidermis, the deletion of EZH2 and EZH1 results in massive defects in the production of mature hair follicle structures, blocking hair follicle stem cells in a non-cycling state due to upregulation of the INK4b (p15) locus [79]. Besides PRC2, PRC1 also affects the function of epSC at different levels. For instance, the PRC1 subunit Cbx4 is highly expressed in basal epSC but, contrary to PRC2, prevents their rapid cycling protecting as well as protecting them from differentiation [80]. Cbx4 also protects basal epSC from entering to a premature senescence through the repression of p16 [80]. Mechanistically, Cbx4 relies on its PRC1 activity and also on its E3-SUMO ligase function, which is PRC1 independent, to regulate epidermal stem cell proliferation and differentiation [80]. On the other hand, BMI1 is expressed both in the basal proliferating and suprabasal epidermal cells and is thought to prevent senescence through repression of the p16ink4A locus [81]. It is not clear, however, whether the interplay between Cbx4, Bmi1 and the PRC2 complex is necessary to balance the quiescence and proliferation of epSC or whether they act on different subsets of stem cells. Future in vivo studies combining loss of function and lineage tracing studies will provide insights into these questions. Epigenetic players interact with several transcription factors to maintain stem cell homeostasis. One example in epSC is the transcription factor p63, a master regulator of epidermal stemness, differentiation and stratification that can act both as an activator and a repressor of gene expression [82]. In doing so, p63 regulates cell cycle proteins and the early and terminal epidermal differentiation markers required for proper epidermal homeostasis. The expression of p63 is regulated by the histone methyltransferase Setd8/PR-SET7/ KMT5a, an enzyme required for the monomethylation of lysine 20 at histone H4 [83]. Accordingly, deletion of Setd8 results in a reduced expression of p63 and in a failure to form stratified epidermis [83]. Another epigenetic mechanism related to p63 involves the histone deacetylases 1/2 (HDAC1/2) [84]. HDAC1/2 remove histone acetylation marks, resulting in chromatin compaction and in repression of transcription. Epidermal deletion of HDAC1/2 impairs the ability of p63 to repress the transcription of its target genes, resulting in a dramatic failure of mature hair follicle structures and in an impairment of epidermal stratification [84]. A third epigenetic mechanism required for regulating 7

Epigenetic regulation of adult stem cell function

p63-dependent transcriptional repression in epSC is lymphocyte-specific helicase (LSH) [85]. LSH is a member of the SNF2 chromatin remodelling factor family whose expression is positively regulated by p63, indicating that both proteins act in a positive feedback manner [85]. p63 also utilizes chromatin remodelling factors to promote epidermal stratification. In this sense, the expression of a cluster of genes necessary for the correct cornification of cells (known as the epidermal differentiation cluster) downstream of p63 requires Satb1 and Brg1, two proteins that open and spatially relocate the chromatin of this region within the nucleus, thereby favouring the transcription of the genes located within [86,87]. Besides p63, the transcription factor Myc is also essential for epidermis homeostasis [88]. When activated, Myc induces proliferation and lineage-specific differentiation of epSC. Myc interacts with several epigenetic factors and its activation is followed by changes in the configuration of euchromatin, specifically depending on the presence of H3K4me3 on its target genes [89]. The overexpression of an activated Myc in the epidermis leads to hyperproliferation followed by strong differentiation of epSC [90,91]. Therefore, activation of Myc must be firmly regulated in order to avoid premature differentiation of epSC [88]. One of the key players of this mechanism is the HDAC Sin3a that not only antagonizes Myc activity by repressing the expression of Myc target genes, but also deacetylates activated Myc to induce its degradation [92]. This explains why deletion of Sin3a in mouse epidermis results in a hyperproliferation and in premature differentiation of epidermal stem cells [92–95]. DNA methylation in epSC The expression of the maintenance DNA methyltransferase DNMT1 is high in the basal layer of the epidermis, and decreases as the epidermis stratifies. DNA methylation follows the same pattern of DNMT1 expression, and analysis of DNA methylation content by MeDIP-ChIP revealed that 5-Mc decreases its amplitude in differentiated keratinocytes compared with epidermal basal cells [96]. Interestingly, DNMT1deficient human epSC cannot fully maintain their pattern of DNA methylation and undergo premature and irreversible differentiation [96], although it is not known to what extent this effect is entirely dependent on the observed changes in DNA methylation. Intriguingly, conditional deletion of DNMT1 in mouse epidermis does not affect the development of the epidermis or its appendages, and only shows signs of uneven epidermal thickness and a reduction of the 8

L. Rinaldi and S. A. Benitah

length of the hair shafts upon ageing [97]. In accordance with this, genome-wide single base pair resolution analysis revealed very similar patterns of DNA methylation between quiescent and active proliferating hair follicle stem cells isolated from young mice [36]. Nevertheless, this study reports more than 2000 differentially methylated regions (DMRs), although these regions were not correlated with changes in gene expression. Considering that deletion of DNMT1 results in hair follicle defects only upon ageing, it will be of interest to study whether changes in DNA methylation become prominent as hair follicle stem cells age, similarly to what has been observed in HSCs. Epigenetics in neural stem cells Neural stem cells (NSCs) constitute a heterogeneous population of adult stem cells that differentiate into mature neurons through a slow maturation process that can last several months until they are all fully connected to local nervous circuits of the central nervous system [7]. As for HSCs, NSCs are nested in specific niches, mainly the subventricular zone of the brain, where they interact with niche components that profoundly affect their behaviour [7]. Several studies have described a fundamental role for epigenetic factors during neurogenesis. We shall now review how epigenetics mechanisms regulate neurogenesis and how they regulate NSC behaviour in the adult brain. Histone modifications in neurons and NSCs Disruption of the histone H3K9 dimethyltransferases G9a and GLP in postnatal neurons alters the transcription of neuronal genes in a way that does not affect neuronal architecture and morphology but results in an impairment of the cognitive functions of the brain and its ability to perceive external stimuli [98]. Interestingly, G9a is also important for the specification of neurons during development and under environmental stress, indicating that it can exert different functions in the brain that are context dependent [99]. Other epigenetic factors play important roles in neural homeostasis. For instance, different members of the PcG proteins are necessary for the in vitro specification of ESCs into neural progenitors [14]. PcG proteins are also responsible for regulating adult NSC self-renewal, or the balance between neurogenesis and gliogenesis. BMI1, as shown in other adult stem cell populations, promotes self-renewal of NSCs by repressing the cell cycle inhibitors p16INK4a and p19ARF [100]. Accordingly, deletion of BMI1 in NSCs reduces their self-renewal potential leading to neurological defects FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

and cognitive impairment [101]. On the other hand, loss of the PRC2 subunit EZH2 in cortical progenitors results in the upregulation of differentiation genes that are normally decorated by H3K27me3 in these cells, leading to a skewing from self-renewal to differentiation in the cerebral cortex [102]. EZH2 also controls the rate of the cell cycle in NSCs by repressing neuronal differentiation genes, and its deletion impairs neurogenesis while favouring neuronal differentiation [103].

Epigenetic regulation of adult stem cell function

pattern of DNA methylation of neurogenesis genes. Functionally, the decrease in the number of NSCs in TET1-deficient mice results in an impairment in spatial learning and memory cognition [115]. These studies therefore reveal that perturbing the machinery that regulates DNA methylation and hydroxymethylation profoundly affects neurogenesis and NSC function. However, future studies will be required to determine whether a direct causal link between DNA methylation and hydroxymethylation with these observed phenotypes exists.

DNA methylation in NSC self-renewal and differentiation

Epigenetic factors in muscle stem cells

The machinery responsible for establishing DNA methylation also plays essential roles during neurogenesis that include the regulation of the cell cycle, cell death and dendritic growth of NSCs. All three DNA methyltransferases DNMT1, DNMT3A and DNMT3B are expressed in neuronal progenitors and play important roles during neurogenesis [104]. Deletion of DNMT1 in NSCs decreases both neural progenitors and mature neurons caused by an aberrant upregulation of astrocyte-specific genes and genes involved in inflammation [105]. DNMT3A exerts a similar role maintaining NSC identity by repressing the expression of glial differentiation genes and enhancing neurogenic factors [106]. Accordingly, its deletion results in a significant reduction in the number of mature neurons. Interestingly, the genes downregulated in DNMT3A-deficient NSCs were enriched for H3K27me3 confirming an antagonism between DNA methylation and polycomb repressive mechanisms in NSCs [106]. Unexpectedly, deletion of DNMT1 and DNMT3A from adult forebrain neurons does not cause any observable effect [107]. However, the deletion of both DNA methylases from adult neurons impairs cognitive functions such as learning and memory caused by an impaired neuronal activity in the hippocampal regions [107]. Although the precise role of DNMT3B in adult neurogenesis is not known, mutation of DNMT3B is linked to the development of immunodeficiency centromere instability syndrome in humans [108–110]. Besides DNA methylation, DNA hydroxymethylation is abundant in the central nervous system compared with all other adult tissues [111–113]. At the genomic level, 5hmC is enriched at gene bodies of neuronal genes and it increases during neuronal differentiation [114]. TET family enzymes catalyse 5-hydroxymethylcytosine (5-Hmc), and deletion of TET1 decreases the number of adult NSCs and their selfrenewal potential, perhaps by producing an abnormal

The satellite cells are quiescent adult stem cells that upon damage become active and generate new muscle fibres while self-renewing [116–118]. Similarly to other tissues, PcG complexes have important roles in regulating myogenesis. EZH2 (PRC2) is expressed exclusively at satellite cells and represses, through its repressive mark H3K27me3, muscle differentiation genes [119]. ChIP sequencing experiments revealed that EZH2 and SUZ12 bind at the promoters of master regulators of myogenesis, such as MYOG, to repress their expression [120]. During maturation of muscle cells, EZH1 replaces EZH2, although SUZ12 remains bound to the MyoG promoter. The EZH1 binding enhances the RNA polymerase II elongation mechanism and promotes myogenesis in satellite cells [21,120]. Therefore two forms of PRC2 containing either EZH2 or EZH1 might play distinct roles in undifferentiated versus differentiated myocytes, similar to what has been observed in the epidermis. In this sense, deletion of EZH2 would promote the recruitment of EZH1 to differentiation genes, explaining why it results in the rapid differentiation of muscle stem cells. Opposite to the effect of deletion of EZH2, loss of BMI1 or RING1B (PRC1) in muscle stem cells results in an impairment of myogenesis [121,122]. Interestingly, a recent report has shown that with the progressive lost of BMI1 the muscle stem cells from geriatric mice enter to a pre-senescence phase that irreversibly prevents their entry into the cell cycle in a p16Ink4a-dependent manner [123]. The machineries involved in DNA methylation and demethylation are also important for myogenesis. The genes encoding the master regulators of myogenesis such as MyoG and MyoD are highly DNA-methylated in satellite cells. Upon myogenesis, the levels of the maintenance DNA methyltransferase DNMT1 is reduced, and DNA methylation levels are decreased in these differentiation genes [124]. Thus loss of DNA methylation during muscle lineage commitment is

FEBS Journal (2014) ª 2014 FEBS

9

Epigenetic regulation of adult stem cell function

caused by passive DNA demethylation, although it can also occur by active DNA demethylation. In this sense, the expression of TET1 and TET2 is upregulated during myogenesis, and the levels of hydroxymethylation levels are globally increased in myoblasts compared with muscle progenitors [124]. Epigenetics mechanisms described in other adult tissues The role of epigenetic factors in the homeostasis of other tissues is starting to be understood. For instance, the histone H3K9 dimethyltransferase G9a is essential for adipose tissue homeostasis. G9a is highly expressed in pre-adipocytes where it represses the expression of adipogenic genes, and its expression is downregulated during adipogenesis. It has been shown that G9a can repress adipogenesis through two independent mechanisms: (a) through the deposition of the H3K9me2 mark on the promoter of the PPAR gene, one of the master regulators of adipogenesis, and (b) by directly promoting the expression of the adipogenesis repressor WNT10a [125]. Accordingly, deletion of G9a in preadipocytes results in an increase of adipogenesis, and deletion in mouse adipose tissue stimulates adipogenic gene expression and increases fat accumulation and tissue weight [125]. Interestingly, the deletion of the histone methyltransferase domain of the thritorax protein MLL3 reduces adipose tissue accumulation in mice [126]. Taken together these two studies suggest that the balance between the activities of MLL3 and G9a regulates adipogenesis and fat accumulation in mammals. Intriguingly, studies aimed at determining changes in DNA methylation at single base resolution have revealed that DNA methylation barely changes as stem cells from the intestinal epithelium differentiate into the different lineages of the villi [37]. As mentioned above, a similar observation was made on comparing quiescent and actively proliferating hair follicle stem cells, suggesting that changes in DNA methylation might not be involved in the function of all adult stem cells [37]. In fact, conditional deletion of DNMT1 results in a modest enlargement of the small intestine crypt zone [127]. However, deletion of DNMT3B in the intestinal epithelium does not result in any phenotype, although DNMT3B expression is required for intestinal tumorigenesis [128,129]. In addition, intestinal stem cells and their differentiated progeny do not show strong differences between histone modifications at enhancers. These sustained active enhancers are thought to allow a continuous cross-talk between adjacent cells promoting differential cell fate [130]. Thus, interestingly these independent studies suggest that 10

L. Rinaldi and S. A. Benitah

DNA methylation and histone modifications are quite static during intestinal stem cell differentiation. That said, there are a few DMRs upon intestinal stem cell differentiation which precisely coincide with enhancers that drive the expression of differentiation genes [37,127]. These cis elements are tissue-specific DMRs, short genome elements that are normally hypomethylated in a tissue-specific manner [46]. Although intestinal deletion of DNA methyltransferases does not result in any overt phenotype, they do profoundly affect intestinal tumorigenesis, suggesting that these DMRs might be important for regulating intestinal stem cell function when tissue homeostasis is perturbed [128,129].

Concluding remarks In the last few years, our understanding of epigenetic function in adult stem cell homeostasis has considerably increased. The advance of next generation sequence technologies is bringing new insights to epigenetic mechanism dynamics. The continuous improvements to next generation sequence techniques are allowing the fields of epigenetics and stem cells to analyse in an unprecedented depth the genetic and epigenetic changes that adult stem cells undergo to sustain homeostasis. Although every tissue-specific stem cell relies on unique epigenetic factors and transcription factors to regulate its function, a common theme is arising from all these studies, one in which the balanced interplay between different epigenetic complexes, or even the same complex formed by combinations of different subunits, is essential for the robustness of the response of adult stem cells to the needs of the tissue at any given time. That said, we are still far from understanding how precisely the marks deposited by these epigenetic factors correlate with the phenotypes observed upon modulation of their expression. Targeted mutations that affect their enzymatic activity in vivo in a tissue-specific manner will be necessary regarding this. In addition, it will be necessary to determine how different chromatin marks established by different complexes are dynamically combined to fine-tune the function of adult stem cells. Notwithstanding this, their importance in tissue homeostasis is not only substantiated by loss and gain of function studies in vivo but is also underscored by the fact that epigenetic regulators are among the most mutated or misregulated genes in human cancers and aged tissues [131]. For instance, changes in the levels of DNA methylation have recently been proposed to be an effective measurement for determining cell ageing [132]. Members of the PRC1 and PRC2 complexes, FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

such as Ezh2 and Bmi1, or other histone methyltransferases including MLL and G9a, show different kinds of mutations in solid tumours and blood malignancies. In fact, some of these proteins such as MLL or Ezh2 are among the most mutated genes in human cancers and strongly act as oncogenes when overexpressed [133–136]. Interestingly, some of these proteins are susceptible to pharmacological modulation, and inhibitors are already available that might provide new promising antitumoral strategies [137,138].

Acknowledgements This work has been funded by an ERC Starting Grant (STEMCLOCK) granted to SAB. LR is supported by the International PhD La Caixa Fellowship.

Author contributions Lorenzo Rinaldi and Salvador Aznar Benitah discussed the topic of the review, wrote and revised the manuscript.

References 1 Blanpain C & Simons BD (2013) Unravelling stem cell dynamics by lineage tracing. Nat Rev Mol Cell Biol 14, 489–502. 2 Li L & Clevers H (2010) Coexistence of quiescent and active adult stem cells in mammals. Science 327, 542–545. 3 Solanas G & Benitah SA (2013) Regenerating the skin: a task for the heterogeneous stem cell pool and surrounding niche. Nat Rev Mol Cell Biol 14, 737– 748. 4 Rios AC, Fu NY, Lindeman GJ & Visvader JE (2015) In situ identification of bipotent stem cells in the mammary gland. Nature 506, 322–327. 5 Doucet YS, Woo S-H, Ruiz ME & Owens DM (2013) The Touch dome defines an epidermal niche specialized for mechanosensory signaling. Cell Rep 3, 1759–1765. 6 Van Keymeulen A, Rocha AS, Ousset M, Beck B, Bouvencourt G, Rock J, Sharma N, Dekoninck S & Blanpain C (2012) Distinct stem cells contribute to mammary gland development and maintenance. Nature 479, 189–193. 7 Gage FH & Temple S (2013) Perspective. Neuron 80, 588–601. 8 Bernstein BE, Meissner A & Lander ES (2007) The mammalian epigenome. Cell 128, 669–681. 9 Sauvageau M & Sauvageau G (2010) Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell 7, 299–313.

FEBS Journal (2014) ª 2014 FEBS

Epigenetic regulation of adult stem cell function

10 Luis NM, Morey L, Di Croce L & Benitah SA (2012) Polycomb in stem cells: PRC1 branches out. Cell Stem Cell 11, 16–21. 11 Richly H, Aloia L & Di Croce L (2011) Roles of the polycomb group proteins in stem cells and cancer. Cell Death Dis 2, e204–e207. 12 Schwartz YB & Pirrotta V (2007) Polycomb silencing mechanisms and the management of genomic programmes. Nat Rev Genet 8, 9–22. 13 Surface LE, Thornton SR & Boyer LA (2010) Polycomb group proteins set the stage for early lineage commitment. Cell Stem Cell 7, 288–298. 14 Aloia L, Di Stefano B, Sessa A, Morey L, Santanach A, Gutierrez A, Cozzuto L, Benitah SA, Graf T, Broccoli V et al. (2014) Zrf1 is required to establish and maintain neural progenitor identity. Genes Dev 28, 182–197. 15 Simon JA & Kingston RE (2013) Occupying chromatin: polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol Cell 49, 808–824. 16 Blackledge NP, Farcas AM, Kondo T, King HW, McGouran JF, Hanssen LLP, Ito S, Cooper S, Kondo K, Koseki Y et al. (2014) Variant PRC1 complexdependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell 157, 1445–1459. 17 Morey L, Pascual G, Cozzuto L, Roma G, Wutz A, Benitah SA & Di Croce L (2012) Nonoverlapping functions of the polycomb group Cbx family of proteins in embryonic stem cells. Cell Stem Cell 10, 47–62. 18 Morey L, Aloia L, Cozzuto L, Benitah SA & Di Croce L (2013) RYBP and Cbx7 define specific biological functions of polycomb complexes in mouse embryonic stem cells. Cell Rep 3, 60–69. 19 O’Loghlen A, Mu~ noz-Cabello AM, Gaspar-Maia A, Wu H-A, Banito A, Kunowska N, Racek T, Pemberton HN, Beolchi P, Lavial F et al. (2012) MicroRNA regulation of Cbx7 mediates a switch of polycomb orthologs during ESC differentiation. Cell Stem Cell 10, 33–46. 20 Cooper S, Dienstbier M, Hassan R, Schermelleh L, Sharif J, Blackledge NP, De Marco V, Elderkin S, Koseki H, Klose R et al. (2014) Targeting polycomb to pericentric heterochromatin in embryonic stem cells reveals a role for H2AK119u1 in PRC2 recruitment. Cell Rep 7, 1456–1470. 21 Mousavi K, Zare H, Wang AH & Sartorelli V (2012) Polycomb protein Ezh1 promotes RNA polymerase II elongation. Mol Cell 45, 255–262. 22 Wen B, Wu H, Shinkai Y, Irizarry RA & Feinberg AP (2009) Large histone H3 lysine 9 dimethylated chromatin blocks distinguish differentiated from embryonic stem cells. Nat Genet 41, 246–250. 23 Tachibana M (2002) G9a histone methyltransferase plays a dominant role in euchromatic histone H3

11

Epigenetic regulation of adult stem cell function

24

25

26

27

28

29

30

31

32

33 34

35 36

37

12

lysine 9 methylation and is essential for early embryogenesis. Genes Dev 16, 1779–1791. Schuettengruber B, Martinez A-M, Iovino N & Cavalli G (2011) Trithorax group proteins: switching genes on and keeping them active. Nat Rev Mol Cell Biol 12, 799–814. Rada-Iglesias A, Bajpai R, Swigut T, Brugmann SA, Flynn RA & Wysocka J (2012) A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283. McMahon KA, Hiew SYL, Hadjur S, VeigaFernandes H, Menzel U, Price AJ, Kioussis D, Williams O & Brady HJM (2007) MLL has a critical role in fetal and adult hematopoietic stem cell selfrenewal. Cell Stem Cell 1, 338–345. Wu SC & Zhang Y (2010) Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 11, 607–620. Wu H & Zhang Y (2014) Reversing DNA methylation: mechanisms, genomics, and biological functions. Cell 156, 45–68. Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos F, Popp C, Thienpont B, Dean W & Reik W (2012) The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol Cell 48, 849–862. Watanabe D, Suetake I, Tada T & Tajima S (2002) Stage- and cell-specific expression of Dnmt3a and Dnmt3b during embryogenesis. Mech Dev 118, 187–190. Okano M, Bell DW, Haber DA & Li E (1999) DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257. Neri F, Krepelova A, Incarnato D, Maldotti M, Parlato C, Galvagni F, Matarese F, Stunnenberg HG & Oliviero S (2013) Dnmt3L antagonizes DNA methylation at bivalent promoters and favors DNA methylation at gene bodies in ESCs. Cell 155, 121–134. Blasco MA (2007) The epigenetic regulation of mammalian telomeres. Nat Rev Genet 8, 299–309. Gonzalo S, Jaco I, Fraga MF, Chen T, Li E, Esteller M & Blasco MA (2006) DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat Cell Biol 8, 416–424. Bird A (2002) DNA methylation patterns and epigenetic memory. Genes Dev 16, 6–21. Bock C, Beerman I, Lien W-H, Smith ZD, Gu H, Boyle P, Gnirke A, Fuchs E, Rossi DJ & Meissner A (2012) DNA methylation dynamics during in vivo differentiation of blood and skin stem cells. Mol Cell 47, 633–647. Kaaij LT, van de Wetering M, Fang F, Decato B, Molaro A, van de Werken HJ, van Es JH, Schuijers J, de Wit E, de Laat W et al. (2013) DNA methylation dynamics during intestinal stem cell differentiation

L. Rinaldi and S. A. Benitah

38

39

40

41

42

43

44

45

46

47

48

49

reveals enhancers driving gene expression in the villus. Genome Biol 14, R50. Sharif J, Muto M, Takebayashi S-I, Suetake I, Iwamatsu A, Endo TA, Shinga J, Mizutani-Koseki Y, Toyoda T, Okamura K et al. (2007) The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA. Nature 450, 908–912. Bostick M, Kim JK, Esteve P-O, Clark A, Pradhan S & Jacobsen SE (2007) UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science 317, 1760–1764. Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, Agarwal S, Iyer LM, Liu DR, Aravind L et al. (2009) Conversion of 5methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935. Vire E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, Morey L, Van Eynde A, Bernard D, Vanderwinden J-M et al. (2005) The polycomb group protein EZH2 directly controls DNA methylation. Nature 439, 871–874. Li E, Bestor TH & Jaenisch R (1992) Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69, 915–926. Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-Andre V, Sigova AA, Hoke HA & Young RA (2013) Superenhancers in the control of cell identity and disease. Cell 155, 934–947. Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH, Rahl PB, Lee TI & Young RA (2013) Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319. Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, Hawkins RD, Barrera LO, Van Calcar S, Qu C, Ching KA et al. (2007) Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet 39, 311–318. Hon GC, Rajagopal N, Shen Y, McCleary DF, Yue F, Dang MD & Ren B (2013) Epigenetic memory at embryonic enhancers identified in DNA methylation maps from adult mouse tissues. Nat Genet 45, 1198– 1206. Plank JL & Dean A (2014) Enhancer function: mechanistic and genome-wide insights come together. Mol Cell 55, 5–14. Wilson A & Trumpp A (2006) Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol 6, 93–106. Yang L (2005) Identification of LinSca1 + kit+CD34 + Flt3 short-term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients. Blood 105, 2717–2723.

FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

50 Ding X, Lin Q, Ensenat-Waser R, Rose-John S & Zenke M (2012) Polycomb group protein Bmi1 promotes hematopoietic cell development from embryonic stem cells. Stem Cells Dev 21, 121–132. 51 Oguro H, Iwama A, Morita Y, Kamijo T, van Lohuizen M & Nakauchi H (2006) Differential impact of Ink4a and Arf on hematopoietic stem cells and their bone marrow microenvironment in Bmi1-deficient mice. J Exp Med 203, 2247–2253. 52 Majewski IJ, Ritchie ME, Phipson B, Corbin J, Pakusch M, Ebert A, Busslinger M, Koseki H, Hu Y, Smyth GK et al. (2010) Opposing roles of polycomb repressive complexes in hematopoietic stem and progenitor cells. Blood 116, 731–739. 53 Lessard J & Sauvageau G (2003) Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 423, 255–260. 54 Oguro H, Yuan J, Ichikawa H, Ikawa T, Yamazaki S, Kawamoto H, Nakauchi H & Iwama A (2010) Poised lineage specification in multipotential hematopoietic stem and progenitor cells by the polycomb protein Bmi1. Cell Stem Cell 6, 279–286. 55 Iwama A, Oguro H, Negishi M, Kato Y, Morita Y, Tsukui H, Ema H, Kamijo T, Katoh-Fukui Y, Koseki H et al. (2004) Enhanced self-renewal of hematopoietic stem cells mediated by the polycomb gene product Bmi-1. Immunity 21, 843–851. 56 Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K et al. (2006) A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326. 57 Klauke K, Radulovic V, Broekhuis M, Weersing E, Zwart E, Olthof S, Ritsema M, Bruggeman S, Wu X, Helin K et al. (2013) Polycomb Cbx family members mediate the balance between haematopoietic stem cell self-renewal and differentiation. Nat Cell Biol 15, 353–362. 58 Majewski IJ, Blewitt ME, de Graaf CA, McManus EJ, Bahlo M, Hilton AA, Hyland CD, Smyth GK, Corbin JE, Metcalf D et al. (2008) Polycomb repressive complex 2 (PRC2) restricts hematopoietic stem cell activity. PLoS Biol 6, e93. 59 Xie H, Xu J, Hsu JH, Nguyen M, Fujiwara Y, Peng C & Orkin SH (2014) Polycomb repressive complex 2 regulates normal hematopoietic stem cell function in a developmental-stage-specific manner. Cell Stem Cell 14, 68–80. 60 Hidalgo I, Herrera-Merchan A, Ligos JM, Carramolino L, Nunez J, Martinez F, Dominguez O, Torres M & Gonzalez S (2012) Ezh1 is required for hematopoietic stem cell maintenance and prevents senescence-like cell cycle arrest. Cell Stem Cell 11, 649–662. 61 Cao Q, Wang X, Zhao M, Yang R, Malik R, Qiao Y, Poliakov A, Yocum AK, Li Y, Chen W et al. (2014)

FEBS Journal (2014) ª 2014 FEBS

Epigenetic regulation of adult stem cell function

62

63

64

65

66

67

68

69

70

71

72

The central role of EED in the orchestration of polycomb group complexes. Nat Commun 5, 1–13. Hodges E, Molaro A, Dos Santos CO, Thekkat P, Song Q, Uren PJ, Park J, Butler J, Rafii S, McCombie WR et al. (2011) Directional DNA methylation changes and complex intermediate states accompany lineage specificity in the adult hematopoietic compartment. Mol Cell 44, 17–28. Br€ oske A-M, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, Kuhl C, Enns A, Prinz M, Jaenisch R et al. (2009) DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet 41, 1207–1215. Trowbridge JJ, Snow JW, Kim J & Orkin SH (2009) DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell 5, 442–449. Tadokoro Y, Ema H, Okano M, Li E & Nakauchi H (2007) De novo DNA methyltransferase is essential for self-renewal, but not for differentiation, in hematopoietic stem cells. J Exp Med 204, 715–722. Challen GA, Sun D, Jeong M, Luo M, Jelinek J, Berg JS, Bock C, Vasanthakumar A, Gu H, Xi Y et al. (2012) Dnmt3a is essential for hematopoietic stem cell differentiation. Nat Genet 44, 23–31. Jeong M, Sun D, Luo M, Huang Y, Challen GA, Rodriguez B, Zhang X, Chavez L, Wang H, Hannah R et al. (2014) Large conserved domains of low DNA methylation maintained by Dnmt3a. Nat Genet 46, 17–23. Sun D, Luo M, Jeong M, Rodriguez B, Xia Z, Hannah R, Wang H, Le T, Faull KF, Chen R et al. (2014) Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688. Chen X, Skutt-Kakaria K, Davison J, Ou YL, Choi E, Malik P, Loeb K, Wood B, Georges G, TorokStorb B et al. (2012) G9a/GLP-dependent histone H3K9me2 patterning during human hematopoietic stem cell lineage commitment. Genes Dev 26, 2499– 2511. Lehnertz B, Pabst C, Su L, Miller M, Liu F, Yi L, Zhang R, Krosl J, Yung E, Kirschner J et al. (2014) The methyltransferase G9a regulates HoxA9dependent transcription in AML. Genes Dev 28, 317–327. Shilatifard A (2012) The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu Rev Biochem 81, 65–95. Mishra BP, Zaffuto KM, Artinger EL, Org T, Mikkola HKA, Cheng C, Djabali M & Ernst P (2014) The histone methyltransferase activityof MLL1 is dispensable for hematopoiesis and leukemogenesis. Cell Rep 7, 1239–1247.

13

Epigenetic regulation of adult stem cell function

73 Blanpain C & Fuchs E (2009) Epidermal homeostasis: a balancing act of stem cells in the skin. Nat Rev Mol Cell Biol 10, 207–217. 74 Frye M & Benitah SA (2012) Chromatin regulators in mammalian epidermis. Semin Cell Dev Biol 23, 897– 905. 75 Jones PH & Watt FM (1993) Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression. Cell 73, 713–724. 76 Mejetta S, Morey L, Pascual G, Kuebler B, Mysliwiec MR, Lee Y, Shiekhattar R, Di Croce L & Benitah SA (2011) Jarid2 regulates mouse epidermal stem cell activation and differentiation. EMBO J 30, 3635–3646. 77 Sen GL, Webster DE, Barragan DI, Chang HY & Khavari PA (2008) Control of differentiation in a selfrenewing mammalian tissue by the histone demethylase JMJD3. Genes Dev 22, 1865–1870. 78 Ezhkova E, Pasolli HA, Parker JS, Stokes N, Su I-H, Hannon G, Tarakhovsky A & Fuchs E (2009) Ezh2 orchestrates gene expression for the stepwise differentiation of tissue-specific stem cells. Cell 136, 1122–1135. 79 Ezhkova E, Lien WH, Stokes N, Pasolli HA, Silva JM & Fuchs E (2011) EZH1 and EZH2 cogovern histone H3K27 trimethylation and are essential for hair follicle homeostasis and wound repair. Genes Dev 25, 485–498. 80 Luis NM, Morey L, Mejetta S, Pascual G, Janich P, Kuebler B, Cozutto L, Roma G, Nascimento E, Frye M et al. (2011) Regulation of human epidermal stem cell proliferation and senescence requires polycombdependent and -independent functions of Cbx4. Cell Stem Cell 9, 233–246. 81 Lee K, Adhikary G, Balasubramanian S, Gopalakrishnan R, McCormick T, Dimri GP, Eckert RL & Rorke EA (2007) Expression of Bmi-1 in epidermis enhances cell survival by altering cell cycle regulatory protein expression and inhibiting apoptosis. J Invest Dermatol 128, 9–17. 82 Vanbokhoven H, Melino G, Candi E & Declercq W (2011) p63, a story of mice and men. J Invest Dermatol 131, 1196–1207. 83 Driskell I, Oda H, Blanco S, Nascimento E, Humphreys P & Frye M (2012) The histone methyltransferase Setd8 acts in concert with c-Myc and is required to maintain skin. EMBO J 31, 616–629. 84 LeBoeuf M, Terrell A, Trivedi S, Sinha S, Epstein JA, Olson EN, Morrisey EE & Millar SE (2010) Hdac1 and Hdac2 act redundantly to control p63 and p53 functions in epidermal progenitor cells. Dev Cell 19, 807–818. 85 Keyes WM, Pecoraro M, Aranda V, VernerssonLindahl E, Li W, Vogel H, Guo X, Garcia EL, Michurina TV, Enikolopov G et al. (2011)

14

L. Rinaldi and S. A. Benitah

86

87

88

89

90

91

92

93

94

95

96

97

DeltaNp63alpha is an oncogene that targets chromatin remodeler Lsh to drive skin stem cell proliferation and tumorigenesis. Cell Stem Cell 8, 164–176. Mardaryev AN, Gdula MR, Yarker JL, Emelianov VN, Poterlowicz K, Sharov AA, Sharova TY, Scarpa JA, Chambon P, Botchkarev VA et al. (2014) p63 and Brg1 control developmentally regulated higher-order chromatin remodelling at the epidermal differentiation complex locus in epidermal progenitor cells. Development 141, 101–111. Fessing MY, Mardaryev AN, Gdula MR, Sharov AA, Sharova TY, Rapisarda V, Gordon KB, Smorodchenko AD, Poterlowicz K, Ferone G et al. (2011) p63 regulates Satb1 to control tissue-specific chromatin remodeling during development of the epidermis. J Cell Biol 194, 825–839. Watt FM, Frye M & Benitah SA (2008) MYC in mammalian epidermis: how can an oncogene stimulate differentiation? Nat Rev Cancer 8, 234–242. Varlakhanova NV & Knoepfler PS (2009) Acting locally and globally: Myc’s ever-expanding roles on chromatin. Cancer Res 69, 7487–7490. Arnold I & Watt FM (2001) c-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny. Curr Biol 11, 558–568. Frye M, Gardner C, Li ER, Arnold I & Watt FM (2003) Evidence that Myc activation depletes the epidermal stem cell compartment by modulating adhesive interactions with the local microenvironment. Development 130, 2793–2808. Nascimento EM, Cox CL, MacArthur S, Hussain S, Trotter M, Blanco S, Suraj M, Nichols J, Kubler B, Benitah SA et al. (2011) The opposing transcriptional functions of Sin3a and c-Myc are required to maintain tissue homeostasis. Nat Cell Biol 13, 1395–1405. Rao G, Alland L, Guida P, Schreiber-Agus N, Chen K, Chin L, Rochelle JM, Seldin MF, Skoultchi AI & DePinho RA (1996) Mouse Sin3A interacts with and can functionally substitute for the amino-terminal repression of the Myc antagonist Mxi1. Oncogene 12, 1165–1172. Laherty CD, Yang WM, Sun JM, Davie JR, Seto E & Eisenman RN (1997) Histone deacetylases associated with the mSin3 corepressor mediate mad transcriptional repression. Cell 89, 349–356. Hassig CA, Fleischer TC, Billin AN, Schreiber SL & Ayer DE (1997) Histone deacetylase activity is required for full transcriptional repression by mSin3A. Cell 89, 341–347. Sen GL, Reuter JA, Webster DE, Zhu L & Khavari PA (2010) DNMT1 maintains progenitor function in self-renewing somatic tissue. Nature 463, 563–567. Li J, Jiang T-X, Hughes MW, Wu P, Widelitz RB, Fan G & Chuong C-M (2012) Progressive alopecia

FEBS Journal (2014) ª 2014 FEBS

L. Rinaldi and S. A. Benitah

98

99

100

101

102

103

104

105

106

107

108

reveals decreasing stem cell activation probability during aging of mice with epidermal deletion of DNA methyltransferase 1. J Invest Dermatol 132, 2681–2690. Schaefer A, Sampath SC, Intrator A, Min A, Gertler TS, Surmeier DJ, Tarakhovsky A & Greengard P (2009) Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex. Neuron 64, 678–691. Maze I, Chaudhury D, Dietz DM, Von Schimmelmann M, Kennedy PJ, Lobo MK, Sillivan SE, Miller ML, Bagot RC, Sun H et al. (2014) G9a influences neuronal subtype specification in striatum. Nat Neurosci 17, 533–539. Molofsky AV, He S, Bydon M, Morrison SJ & Pardal R (2005) Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a and p19Arf senescence pathways. Genes Dev 19, 1432–1437. Molofsky AV, Pardal R, Iwashita T, Park I-K, Clarke MF & Morrison SJ (2003) Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425, 962–967. Pereira JD, Sansom SN, Smith J, Dobenecker M-W, Tarakhovsky A & Livesey FJ (2010) Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex. Proc Natl Acad Sci USA 107, 15957–15962. Yu YL, Chou RH, Chen LT, Shyu WC, Hsieh SC, Wu CS, Zeng HJ, Yeh SP, Yang DM, Hung SC et al. (2011) EZH2 Regulates neuronal differentiation of mesenchymal stem cells through PIP5K1C-dependent calcium signaling. J Biol Chem 286, 9657–9667. Watanabe D, Uchiyama K & Hanaoka K (2006) Transition of mouse de novo methyltransferases expression from Dnmt3b to Dnmt3a during neural progenitor cell development. Neuroscience 142, 727–737. Fan G, Martinowich K, Chin MH, He F, Fouse SD, Hutnick L, Hattori D, Ge W, Shen Y, Wu H et al. (2005) DNA methylation controls the timing of astrogliogenesis through regulation of JAK-STAT signaling. Development 132, 3345–3356. Wu H, Coskun V, Tao J, Xie W, Ge W, Yoshikawa K, Li E, Zhang Y & Sun YE (2010) Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science 329, 444–448. Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, Silva AJ & Fan G (2010) Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat Neurosci 13, 423–430. Moarefi AH & Chedin F (2011) ICF syndrome mutations cause a broad spectrum of biochemical defects in DNMT3B-mediated de novo DNA methylation. J Mol Biol 409, 758–772.

FEBS Journal (2014) ª 2014 FEBS

Epigenetic regulation of adult stem cell function

109 Jin B, Tao Q, Peng J, Soo HM, Wu W, Ying J, Fields CR, Delmas AL, Liu X, Qiu J et al. (2007) DNA methyltransferase 3B (DNMT3B) mutations in ICF syndrome lead to altered epigenetic modifications and aberrant expression of genes regulating development, neurogenesis and immune function. Hum Mol Genet 17, 690–709. 110 Weemaes CM, van Tol MJ, Wang J, van Ostaijen-ten Dam MM, van Eggermond MC, Thijssen PE, Aytekin C, Brunetti-Pierri N, van der Burg M, Davies EG et al. (2013) Heterogeneous clinical presentation in ICF syndrome: correlation with underlying gene defects. Eur J Hum Genet 21, 1219–1225. 111 Kinney SM, Chin HG, Vaisvila R, Bitinaite J, Zheng Y, Esteve PO, Feng S, Stroud H, Jacobsen SE & Pradhan S (2011) Tissue-specific distribution and dynamic changes of 5-hydroxymethylcytosine in mammalian genomes. J Biol Chem 286, 24685–24693. 112 Kim M, Park YK, Kang TW, Lee SH, Rhee YH, Park JL, Kim HJ, Lee D, Lee D, Kim SY et al. (2014) Dynamic changes in DNA methylation and hydroxymethylation when hES cells undergo differentiation toward a neuronal lineage. Hum Mol Genet 23, 657–667. 113 Kriaucionis S & Heintz N (2009) The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324, 929–930. 114 Hahn MA, Qiu R, Wu X, Li AX, Zhang H, Wang J, Jui J, Jin S-G, Jiang Y, Pfeifer GP et al. (2013) Dynamics of 5-hydroxymethylcytosine and chromatin marks in mammalian neurogenesis. Cell Rep 3, 291– 300. 115 Zhang R-R, Cui Q-Y, Murai K, Lim YC, Smith ZD, Jin S, Ye P, Rosa L, Lee YK, Wu H-P et al. (2013) Tet1 regulates adult hippocampal neurogenesis and cognition. Cell Stem Cell 13, 237–245. 116 Mauro A (1961) Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol 9, 493–495. 117 Peault B, Rudnicki M, Torrente Y, Cossu G, Tremblay JP, Partridge T, Gussoni E, Kunkel LM & Huard J (2007) Stem and progenitor cells in skeletal muscle development, maintenance, and therapy. Mol Ther 15, 867–877. 118 Brack AS & Rando TA (2012) Tissue-specific stem cells: lessons from the skeletal muscle satellite cell. Cell Stem Cell 10, 504–514. 119 Caretti G, Di Padova M, Micales B, Lyons GE & Sartorelli V (2004) The polycomb Ezh2 methyltransferase regulates muscle gene expression and skeletal muscle differentiation. Genes Dev 18, 2627–2638. utzer A, 120 Stojic L, Jasencakova Z, Prezioso C, St€ Bodega B, Pasini D, Klingberg R, Mozzetta C, Margueron R, Puri PL et al. (2011) Chromatin regulated interchange between polycomb repressive

15

Epigenetic regulation of adult stem cell function

121

122

123

124

125

126

127

128

129

16

complex 2 (PRC2)-Ezh2 and PRC2-Ezh1 complexes controls myogenin activation in skeletal muscle cells. Epigenetics Chromatin 4, 16. Asp P, Blum R, Vethantham V, Parisi F, Micsinai M, Cheng J, Bowman C, Kluger Y & Dynlacht BD (2011) Genome-wide remodeling of the epigenetic landscape during myogenic differentiation. Proc Natl Acad Sci USA 108, E149–E158. Robson LG, Di Foggia V, Radunovic A, Bird K, Zhang X & Marino S (2011) Bmi1 is expressed in postnatal myogenic satellite cells, controls their maintenance and plays an essential role in repeated muscle regeneration. PLoS One 6, e27116. Sousa-Victor P, Gutarra S, Garcıa-Prat L, RodriguezUbreva J, Ortet L, Ruiz-Bonilla V, Jardı M, Ballestar E, Gonzalez S, Serrano AL et al. (2014) Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 506, 316–321. Tsumagari K, Baribault C, Terragni J, Varley KE, Gertz J, Pradhan S, Badoo M, Crain CM, Song L, Crawford GE et al. (2013) Early de novo DNA methylation and prolonged demethylation in the muscle lineage. Epigenetics 8, 317–332. Wang L, Xu S, Lee J-E, Baldridge A, Grullon S, Peng W & Ge K (2012) Histone H3K9 methyltransferase G9a represses PPARc expression and adipogenesis. EMBO J 32, 45–59. Lee J, Saha PK, Yang QH, Lee S, Park JY, Suh Y, Lee S-K, Lee SK, Chan L, Chan L et al. (2008) Targeted inactivation of MLL3 histone H3–Lys-4 methyltransferase activity in the mouse reveals vital roles for MLL3 in adipogenesis. Proc Natl Acad Sci USA 105, 19229–19234. Sheaffer KL, Kim R, Aoki R, Elliott EN, Schug J, Burger L, Sch€ ubeler D & Kaestner KH (2014) DNA methylation is required for the control of stem cell differentiation in the small intestine. Genes Dev 28, 652–664. Lin H, Yamada Y, Nguyen S, Linhart H, JacksonGrusby L, Meissner A, Meletis K, Lo G & Jaenisch R (2006) Suppression of intestinal neoplasia by deletion of Dnmt3b. Mol Cell Biol 26, 2976–2983. Linhart HG, Lin H, Yamada Y, Moran E, Steine EJ, Gokhale S, Lo G, Cantu E, Ehrich M, He T et al. (2007) Dnmt3b promotes tumorigenesis in vivo by

L. Rinaldi and S. A. Benitah

130

131 132 133

134

135

136

137

138

gene-specific de novo methylation and transcriptional silencing. Genes Dev 21, 3110–3122. Kim T-H, Li F, Ferreiro-Neira I, Ho L-L, Luyten A, Nalapareddy K, Long H, Verzi M & Shivdasani RA (2014) Broadly permissive intestinal chromatin underlies lateral inhibition and cell plasticity. Nature 506, 511–515. Shen H & Laird PW (2013) Interplay between the cancer genome and epigenome. Cell 153, 38–55. Horvath S (2013) DNA methylation age of human tissues and cell types. Genome Biol 14, R115. Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C, Jones AV, Waghorn K, Zoi K, Ross FM, Reiter A et al. (2010) Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 42, 722–726. Lohr JG, Stojanov P, Lawrence MS, Auclair D, Chapuy B, Sougnez C, Cruz-Gordillo P, Knoechel B, Asmann YW, Slager SL et al. (2012) Discovery and prioritization of somatic mutations in diffuse large Bcell lymphoma (DLBCL) by whole-exome sequencing. Proc Natl Acad Sci USA 109, 3879–3884. McCabe MT, Graves AP, Ganji G, Diaz E, Halsey WS, Jiang Y, Smitheman KN, Ott HM, Pappalardi MB, Allen KE et al. (2012) Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27). Proc Natl Acad Sci USA 109, 2989–2994. Nikoloski G, Langemeijer SMC, Kuiper RP, Knops R, Massop M, T€ onnissen ERLTM, van der Heijden A, Scheele TN, Vandenberghe P, de Witte T et al. (2010) Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet 42, 665–667. McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, Liu Y, Graves AP, Pietra Della A III, Diaz E et al. (2013) EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492, 108– 112. Kim W, Bird GH, Neff T, Guo G, Kerenyi MA, Walensky LD & Orkin SH (2013) Targeted disruption of the EZH2–EED complex inhibits EZH2-dependent cancer. Nat Chem Biol 9, 643–650.

FEBS Journal (2014) ª 2014 FEBS

Epigenetic regulation of adult stem cell function.

Understanding the cellular and molecular mechanisms that specify cell lineages throughout development, and that maintain tissue homeostasis during adu...
303KB Sizes 0 Downloads 5 Views