Epigenetic Dynamics of Stem Cells

Embed Size (px)

Citation preview

  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    1/12

    The life cycle of an organism during developmentfrom a zygote to an adult is characterized by phasesof reprogrammingand differentiation. The changes ingene expression that guide these processes are causedby transcription factors and epigenetic modifications(BOX 1), which impose heritable cellular memories thatdefine both specific cell lineages and cell types1. Thus,on the whole, somatic cells progressively acquire anincreasing array of epigenetic marks (by a processknown as epigenetic programming) that are importantfor cell specification because they establish a heritablememory of cellular states and differentiation pathways.By contrast, germ cells and early embryos can reset(or reprogramme) epigenetic marks24. This epigeneticreprogramming of germ cells is an essential characteris-tic of their immortality and occurs in preparation of theeventual acquisition of totipotency: the epigenetic marks

    (termed imprints) that distinguish the parental originof some genes, for example, are erased in primordialgerm cells (PGCs) and then reset in a parent-of-origin-dependent manner, such that they are in place at thetime of fertilization.

    The combination of oocyte and sperm in the zygoterepresents the climax of cellular potency: the zygote isthe only unequivocally totipotent cell in the life cycle.During this temporally unique period immediatelyafter fertilization, the sperm nucleus is extensivelyremodelled, replacing protamineswith histones, andundergoes paternal-specific active demethylation ofDNA57. Subsequent cleavage divisions in the early

    embryo are characterized by a progressive reduc-tion in DNA methylation as a consequence of pas-sive demethylation caused by the exclusion of DNA(cytosine-5)-methyltransferase 1 (DNMT1) from thenucleus8. Genome-wide reprogramming of histonemodifications also occurs during this period. Thisprocess coincides with the gradual commitment ofcells towards the earliest lineages, which is accom-panied by lineage-specific epigenetic programmingevents.

    The steps leading towards the earliest lineagedecisions have been best studied in mice, in whichseparation of the first cell lineages occurs in two suc-cessive, dichotomous differentiation events. First, atthe blastocyststage, the trophectoderm (TE) becomesfixed in its differentiation potential towards theextraembryonic trophoblast lineage and is set apart

    from the inner cell mass (ICM). This is followed bythe separation of the ICM into epiblast (or primitiveectoderm) and primitive endoderm (or hypoblast) inthe late blastocyst (FIG. 1). The epiblast will form theembryo proper, whereas the primitive endoderm givesrise to the parietal and visceral endoderm layers that,together with the trophoblast, contribute to extra-embryonic tissues. It is only in the region overlyingthe epiblast at the egg cylinderstage that some cellsof the visceral endoderm will contribute to the defini-tive embryonic endoderm, such that embryonic endo-dermal structures are composed of cells originating inboth the epiblast and the primitive endoderm9.

    Laboratory of Developmental

    Genetics and Imprinting,

    The Babraham Institute,Babraham Research Campus,

    Cambridge CB22 3AT,

    and Centre for Trophoblast

    Research, University of

    Cambridge, Downing Street,

    Cambridge CB2 3EG, UK.

    Correspondence to

    M.H. and W.R.

    e-mails: myriam.hemberger@

    bbsrc.ac.uk;wolf.reik@bbsrc.

    ac.uk

    *These authors contributed

    equally to this work.

    doi:10.1038/nrm2727

    Published online 15 July 2009

    Reprogramming

    The resetting of epigenetic

    marks, usually to achieve wider

    developmental potency.

    Reprogramming occurs during

    the life cycle in the early

    embryo and in the germ line,

    and also in experimental

    systems in which differentiated

    cells are converted into

    induced pluripotent stem cells.

    Epigenetic dynamics of stem cellsand cell lineage commitment:digging Waddingtons canalMyriam Hemberger*, Wendy Dean* and Wolf Reik

    Abstract | Cells of the early mammalian embryo, including pluripotent embryonic stem (ES)

    cells and primordial germ cells (PGCs), are epigenetically dynamic and heterogeneous.

    During early development, this heterogeneity of epigenetic states is associated withstochastic expression of lineage-determining transcription factors that establish an

    intimate crosstalk with epigenetic modifiers. Lineage-specific epigenetic modification of

    crucial transcription factor loci (for example, methylation of the Elf5promoter) leads to the

    restriction of transcriptional circuits and the fixation of lineage fate. The intersection of

    major epigenetic reprogramming and programming events in the early embryo creates

    plasticity followed by commitment to the principal cell lineages of the early conceptus.

    CHROMATI N DYNAMI CS

    R E V I E W S

    526 |AUGUST 2009 |VOLUME 10 www.nature.com/reviews/molcellbio

    2009 Macmillan Publishers Limited. All rights reserved

    mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]
  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    2/12

    Epiblast

    |

    PGCs

    Developmentalstage

    Stem cells

    Morula

    EG cells

    Midlate blastocyst

    Somatic cell lineagesEndodermMesodermEctoderm

    Dedifferentiationand reprogamming

    ES cellsXEN cells TS cells iPS cellsEpiSCs

    Fertilization

    TEPrimitive

    endoderm

    Totipotency

    The differentiative capacity

    of a cell to form all cell types of

    the conceptus and the adult

    organism, including

    extraembryonic membranes,

    the placenta and germ cells.

    Protamine

    A highly basic non-histone

    protein that replaces histones

    in the sperm to achieve dense

    packaging of DNA.

    Blastocyst

    The embryo before

    implantation that contains at

    least two distinct cell types:

    an outer epithelial cell layer

    (trophectoderm) and an inner

    group of cells (inner cell mass).

    Egg cylinder

    The embryonic stage after

    implantation that comprises a

    double-layered structure of the

    outer endoderm and the inner

    embryonic or extraembryonic

    (trophoblast) ectoderm,

    enclosing a narrow lumen.

    Primitive streak

    A structure that is formed at

    the posterior end of amniote

    embryos at gastrulation stages

    and is the area of mesoderm

    formation.

    After these initial decisions have been made, cells

    of all three lineages (epiblast, primitive endoderm andtrophoblast) undergo successive steps of differentiationto form all cell types of the organism, as well as the pla-centa. A few cells, however, (the germ cells) are set asideearly on in the egg cylinder to close the life cycle. Thegerm cells represent the end point of a differentiationcascade and can be considered highly specialized. Atthe same time, however, they have the capacity to returnto totipotency (as a consequence of germ cell-specificepigenetic reprogramming) when the gametes cometogether in the zygote.

    In this Review, we focus on the epigenetic dynamicsthat underlie differentiation and reprogramming eventsduring two key time points in the life cycle of the mousethat might be particularly important for the regulation oflineage decisions and define the potency of a cell. In thiscontext, we concentrate on recent insights into the rolesof DNA methylation and modifications of histones inthe epigenetic control of developmental pathways.

    Epigenetic dynamics during the life cycle

    The progression of PGC specification, migration anddifferentiation gives rise to mature gametes, spermand oocytes, which come together in the zygote to forma new organism.

    Epigenetic reprogramming in PGCs.PGCs are uniquecells that are designated to regain the capacity to forma new organism. They arise from a few cells expressingB lymphocyte-induced maturation protein 1 (Blimp1; alsoknown as Prdm1) that can first be identified at embryonicday 6.25 (E6.25) of mouse development. PGCs are speci-

    fied at E7.25 as a group of approximately 40 cells in theextraembryonic region posterior to the primitive streak,which are demarcated by expression of Blimp1and Stella(also known as Dppa3)1012. PGCs undergo extensive epi-genetic reprogramming during and after their migrationthrough the hindgut to the developing gonads, culmin-ating in the erasure of parental imprints and many otherepigenetic marks throughout the genome2,13,14.

    Although the mechanisms of erasure and the exacttiming of these reprogramming events are still unclear,recent studies have revealed important details of the

    Box 1 | Epigenetic modifications

    Epigenetic modifications are instructions that are imposed on the DNA sequence

    to provide an additional level of gene regulation and to establish heritable

    transcriptional states. In mammals, DNA methylation and covalent modifications of the

    amino-terminal tails and the core of nucleosomal histones constitute most epigenetic

    modifications. DNA methylation occurs symmetrically on the cytosine base in CpG

    dinucleotides and is generally associated with gene repression and formation of

    heterochromatin. Histone modifications encompass a range of modifications, includingacetylation, methylation, phosphorylation, ADP ribosylation and ubiquitylation, that

    extend the information content of the underlying DNA sequence and confer unique

    transcriptional potential. Histone modifications can have both repressive and

    activating functions. The best-understood modifications are the trimethylation of the

    Lys9 and Lys27 residues of histone H3 (H3K9me3 and H3K27me3), which have

    repressive functions, and H3K4me3 and H3K9 acetylation (H3K9ac), which are

    associated with active genes. Many other modifications on histones have been

    identified that regulate gene activity, and many enzymes are now known that modify

    histones or methylate DNA. More recently, activities have also been discovered that

    can reverse some of these epigenetic marks (see also the epigenetic modifiers in FIG. 4).

    Figure 1 |Key differentiation stages in development and derived stem cell types. Alife cycle diagram of the first

    principal cell lineages of the early embryo and the stem cell populations that can be derived from them. The firstdefinitive differentiative decisions are made at the blastocyst stage, in which the trophectoderm (TE) layer is set

    aside from the inner cell mass (ICM; not shown). The ICM subsequently delaminates a layer of primitive endoderm.

    The remaining cells form the epiblast. All three cell lineages that is, TE, primitive endoderm and epiblast give rise

    to distinct stem cell types: trophoblast stem (TS) cells117, extraembryonic endoderm stem (XEN) cells118and embryonic

    stem (ES) cells, respectively. These three types of stem cell can also be established from earlier morula stage embryos,

    and their lineage allocation programme is presumably recapitulated during the in vitroculturing process47. Immediately

    after implantation, the epiblast can give rise to a different stem cell population, termed epiblast stem cells (epiSCs)48,49.

    The three embryonic germ layers, endoderm, mesoderm and ectoderm, are formed during gastrulation. Primordial germ

    cells (PGCs) originate from a small mesodermal cell population at the posterior tip of the epiblast2,11,12. Embryonic

    germ (EG) cells can be derived from early stage PGCs between embryonic day 8.5 (E8.5) and E11.544,45,50. The three

    embryonic germ layers go on to differentiate into all cell types of the adult organism. Recent breakthroughs in

    dedifferentiation techniques allow the reprogramming of fully differentiated somatic cells into induced pluripotent

    stem (iPS) cells, which closely resemble ES cells in their developmental potency100103.

    R E V I E W S

    NATURE REVIEWS |MOLECULAR CELL BIOLOGY VOLUME 10 |AUGUST 2009 |527

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=12142&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=73708&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=73708&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=12142&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSum
  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    3/12

  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    4/12

    Facultative heterochromatin

    The fraction of chromatin that

    is condensed and inactive in agiven cell lineage, which can be

    decondensed and active in

    another.

    Constitutive

    heterochromatin

    The fraction of

    heterochromatin that stays

    compact throughout the cell

    cycle. It is mainly composed of

    repetitive sequences (satellite

    DNA) and is concentrated in

    characteristic regions, such as

    centromeres.

    Pericentromeric

    heterochromatin

    The heterochromatin structure

    composed mostly of satellite

    repeats that are located

    adjacent to the centric

    satellites. Together these

    comprise the centromeres and

    form an important structural

    element for chromosome

    stability.

    Centromere

    The region of a chromosome

    that is attached to the spindle

    during nuclear division.

    DNA remethylation takes place several days aftermany of the histone marks have been reset and occursfrom E15E16 onwards in the male germ line but onlypostnatally during oocyte growth in the female germline2. This process results in a DNA methylation state thatis globally similar to that of somatic cells but with impor-tant pattern differences, including at differentially meth-ylated regions that serve as imprinting control regions(ICRs). ICRs are marked either by maternal or paternalmethylation in the germ line and regulate parent-of-origin-specific expression of imprinted genes in thenext generation. It is worth noting that several aspects ofreprogramming in PGCs and later stage germ cells (forexample, the erasure and re-establishment of imprints)result in specialized epigenetic programmes in the maturegametes, some of which are carried forward and are likelyto influence epigenetic reorganization in the early embryo,

    potentially with lasting effects on pluripotency andpossibly also lineage commitment events.

    Epigenetic reprogramming in early embryos. The organ-ization of the genome in the two types of gamete differsdramatically. Sperm chromatin is packaged extremelydensely using predominantly protamines in addition toa modest amount of sperm-specific histone variants25.Although the mechanism of protamine removal is stillnot known, it follows that the differences between maleand female chromatin organization should be resolvedshortly after fertilization to ensure correct chromosomesegregation in the first cleavage division of the zygote.

    Remarkably, however, the chromatin organization ofthe two pronuclei remains strikingly different. Thus, thepaternal genome lacks the repressive heterochromaticH3K9me3 and H3K20me3 modifications that normallymark pericentromeric heterochromatinand are essential forcentromeric function during mitosis in somatic cells26,27.The paternal pronucleus retains H3K9me1 in pericen-tromeric regions along with residual DNA methyl-ation. These marks, together with a different repressivemechanism that is mediated by Polycomb group (PcG)repressive complex 1 (PRC1), which is provided by theegg cytoplasm, serve as a specialized repressive mecha-nism in the male pronucleus to mediate pericentromericheterochromatin formation2831.

    This organization ensures that, despite their differentchromatin features, male and female pronuclei display acomparable centromereorganization that allows propermetaphase plate arrangement and chromosome segrega-tion during nuclear division31. Thus, although the genomesare functionally equivalent, the distinction of their paren-tal origin is preserved during the return to totipotency in

    the zygote. It remains to be seen whether preservation ofthese epigenetic parent-of-origin marks (which are dis-tinct from those that mark imprinted genes) is importantfor early development and for the first cell determinationevents, or whether these marks are merely a remnant ofthe paternal or maternal origin of the genomes.

    Fertilization initiates a second wave of epigeneticreprogramming that is characterized by rapid activeDNA demethylation before the onset of DNA replica-tion57and is followed by passive DNA demethylation upto the morulastage3234. In contrast to the reprogrammingof DNA methylation in PGCs, parent-specific methylationat ICRs is retained during this reprogramming phase35,36.

    De novoDNA methylation is initiated before theblastocyst stage and thus coincides with the earliest dif-ferentiation event that separates the embryonic andtrophoblast lineages (represented by the ICM and TE,respectively)7. Intriguingly, global DNA methylation levelsare markedly different between the embryonic and extra-embryonic lineages; the TE is relatively hypomethylatedcompared with the ICM, as revealed by 5-methylcytosinestaining 7 (FIGS 2,3). These global differences persistthroughout development and are retained in the embryoand placenta37,38. However, more recent whole genomeapproaches have revealed that promoters might repre-sent an exception to this pattern, which might account forthe similarity in overall transcriptional activity between the

    lineages39,40. Hence, methylation differences are mostlylocalized to intergenic regions and non-promoter genicsequences. Similar to DNA methylation, several his-tone modifications, including H3K27me1, H3K27me2,H3K27me3 and H3K9ac, also exhibit an asymmetrybetween the ICM and TE and might therefore have arole in lineage allocation and/or lineage commitment, asdiscussed in more detail below4143.

    Epigenetic interface with pluripotency

    Three principal phases in murine development allowthe derivation of pluripotent cell lines in vitro(FIG. 1).These developmental stages include early PGCs between

    Box 2 | A geneticepigenetic network

    A simple binary view of the presence or absence of epigenetic modifiers and

    lineage-specifying transcription factors is insufficient to explain their functions. Their

    activities are additionally modulated on various levels by different mechanisms. DNA

    (cytosine-5)-methyltransferase 1 (DNMT1), for example, can be methylated, and this

    modification reduces its stability. Lys-specific demethylase 1 (LSD1; also known as

    KDM1) is required for demethylation and stabilization of DNMT1 (REF. 112).

    Phosphorylation of heterochromatin protein 1(HP1) by casein kinase II (CK2) causesits release from methylated histone H3 at Lys9 (H3K9) 113. Also, automethylation of the

    euchromatic H3K9 methyltransferase G9A (also known as EHMT2) creates binding sites

    for HP1and HP1, thereby redistributing these chromatin proteins114. Similarly, thedistinction between activating and repressive functions of the pluripotency

    transcription factors might be imposed by post-translational modifications (for

    example, sumoylation of OCT4, which is encoded by Pou5f1) that in turn might alter

    stability, DNA binding, dimerization and protein complex association capacities115.

    Another crucial regulatory mechanism relies on the subcellular shuttling of epigenetic

    modifiers between the nucleus and cytoplasm. This is particularly important in the zygote

    with respect to the activity of DNMT1: nuclear exclusion from the first S phase results in

    the sequential reduction of DNA methylation of much of the genome during the crucial

    period that leads up to lineage determination116. In the germ line, the highly defined

    window of nuclear exclusion of chromatin proteins around embryonic day 11.5 (E11.5), at

    a time when erasure of epigenetic marks takes place, suggests a causal relationship with

    this process. Interestingly, this change of localization might be permanent (for example,histone H2A.Z), temporary (for example, histone macroH2A) or reciprocal (for example,

    the histone chaperone chromatin assembly factor 1 (CAF1), which translocates from the

    nucleus to the cytoplasm, and nucleosome assembly protein 1 (NAP1), which translocates

    from the cytoplasm to the nucleus)15. The exclusion of DNMT3B from the nucleus at E12.5

    might prevent pre-emptive remethylation of the germline genome18.

    These examples establish a range of secondary epigenetic modifiers that are not

    necessarily epigenetic in nature but impose an additional level of regulation and are of

    equal importance to the primary modifiers.

    R E V I E W S

    NATURE REVIEWS |MOLECULAR CELL BIOLOGY VOLUME 10 |AUGUST 2009 |529

    2009 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    5/12

    |

    OCT4

    NANOG

    SOX2

    SALL4

    STELLA

    CDX2

    EOMES

    ELF5

    DNAmethylation

    TS cells

    ES cells

    Lineage fixation

    Trophoblast lineage

    Embryonic lineage

    Absent StrongElf5 Stella

    Egg cylinder

    Zygote 2 cell 4 cell 8 cell 16 cell

    Early blastocyst

    Late blastocystPrimitiveendoderm

    TE

    ICM

    Expression in PGCs

    Levels assumed

    Epiblast

    Epiblast

    Trophoblast

    E8.5 and E12.5 that can give rise to EG cells, morula-to-blastocyst stage embryos from which embryonic stem (ES)cells can be derived, and E5.5E6.5 epiblast tissue that cangive rise to epiblast stem cells (epiSCs) 4449. Interestingly,

    human ES cells might be more similar to mouse epiSCsthan to mouse ES cells, which is an important considera-tion for the interpretation of studies that compare ES celldata from different species48,49.

    Figure 3 |Stochasticity of gene expression, developmental potency and lineage commitment. Thestochasticity

    of gene expression and mutual interactions between lineage-determining transcription factors with antagonizing

    functions gradually lead to the first cell lineage decisions. These transcription factors include OCT4 (which is encoded

    by Pou5f1)74,119, the homeobox protein NANOG74, SRY box-containing factor 2 (SOX2)120, Sal-like protein 4 (SALL4)121,

    STELLA (also known as DPPA3)122, caudal-type homeobox 2 (CDX2)74,123, eomesodermin (EOMES)75,124and ELF5 (REFS 90,91).

    This process is accompanied by epigenetic programming events that impose a permissive (or non-permissive) environment

    for cell fate or might predispose a cell towards a particular lineage. It is feasible that the expression of each transcription

    factor oscillates between a minimum and maximum level, and positive or negative interference of such transcriptional

    noise introduces a developmental bias in individual blastomeres (indicated by the cell shading). Expression of keytranscription factors remains mosaic in individual blastomeres until the blastocyst stage, when the embryonic (the inner

    cell mass (ICM), epiblast and derived embryonic stem (ES) cells) and trophoblast (the trophectoderm (TE) and derived

    trophoblast stem (TS) cells) lineages are established. De novoDNA methylation is initiated between the morula (16-cell

    stage) and blastocyst stage7. Lineage fixation coincides with methylation of Elf5in the ICM, which establishes a tight

    epigenetic boundary between the embryonic and trophoblast lineages91. Methylation of Stellaoccurs in the same overall

    time window (although probably slightly later than Elf5) and might have a similar gatekeeper function to establish the

    post-implantation epiblast77.

    R E V I E W S

    530 |AUGUST 2009 |VOLUME 10 www.nature.com/reviews/molcellbio

    2009 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    6/12

    |

    OCT4

    NANOG

    SOX2

    SALL4

    SUZ12, EED and EZH2

    PHC1 and PHC2

    JMJD1A*

    JMJD2C

    EHMT1 and G9A

    DNMT3A, DNMT3B,DNMT3L and HELLS

    ARID3BARID3AARID5BCDYLCDYL2CHD9JARID1BJARID2JMJD1CJMJD2A

    MYST4PRDM14*RESTSETSETD1BSMARCA2SMARCA3SMARCAD1*SMARCD1

    H3K9me2 andH3K9me3

    H3K27me3

    De novo

    DNA methylation

    Epigenetic modifiers regulatingpluripotency factors

    Pluripotency factors regulatingepigenetic modifiers

    * In human ES cells

    Morula

    A ball of cells that is created

    by the first cleavage divisions,

    before compaction, that

    initiates the formation of the

    blastocyst.

    It is worth pointing out that the stem cell aspect ofthese different types of pluripotent mouse cell lines, inparticular their self-renewing capacity, is a feature thatis only acquired during culture in vitro, presumably bydisrupting the normal context of cellcell interactionsand signalling, and by the preservation or acquisitionof epigenetic states that allow continued proliferation.

    Crucial factors necessary to maintain pluripotency. Akey feature that reflects the developmental capacity ofthese EG, ES and epiSCs cell lines is that, as in the earlyembryo, they express markers of pluripotency suchas OCT4, NANOG and SOX2 (REFS 4751)(FIGS 3,4).

    The genetic basis of pluripotency has been elegantlyelaborated in ES cells, in which a transcriptional net-work driven by the core transcription factors OCT4,NANOG and SOX2, is essential to maintain the undif-ferentiated state. This network activates genes that arerequired for ES cell survival and proliferation whilerepressing target genes that are activated only duringdifferentiation52,53. With growing insights into the fac-tors required for pluripotency, this core triumvirate isexpanding; for example, with the notable addition ofthe zinc finger transcription factor sal-like protein 4(SALL4)5457. It seems that the co-expression and co-regulation of these factors is one of the key featuresof pluripotency. Intriguingly, OCT4, NANOG, SOX2and SALL4 regulate their own expression and controla set of target genes through mutual heterodimeriza-tion and/or shared promoter occupancy, thus forminga self-reinforcing circuit of pluripotency52,5760.

    In addition to these transcription factors, chroma-tin organization and epigenetic modifications are alsokey elements for controlling gene expression during

    ES cell self-renewal and differentiation. Gene repres-sion mediated by PcG proteins and the conferredH3K27me3 mark is required for ES cell pluripotencyand plasticity during embryonic development 61,62.Chromatin immunoprecipitation studies have shownan unexpected but potentially key concept in the biol-ogy of ES cell pluripotency: genes that are repressedin ES cells but are required for later differentiation aremarked by bivalent H3K4me3 and H3K27me3 domainsthat render them poised for activation6365. In fact, theH3K4me3 and H3K27me3 marks can effectively dis-criminate genes that are expressed (H3K4me3), poisedfor expression (H3K4me3 and H3K27me3) or stablyrepressed (H3K27me3), and therefore reflect the cellstate and lineage potential65. Approximately one-third ofgenes, however, are not marked by either H3K4me3 orH3K27me3, yet are mostly repressed in ES cells. Thesegenes tend to be marked by DNA methylation, whichis therefore a complementary mechanism to histonemodifications to ensure appropriate gene expressionand heritable gene repression simultaneously39,66,67.

    Epigenetic regulation of pluripotency networks. Thecombination of global detection methods for transcrip-tion factor target genes and epigenetic modificationshas revealed the existence of an intriguing interplaybetween pluripotency factors and epigenetic modifi-

    ers (FIG. 4). In fact, the dynamic balance between thesetwo regulatory systems probably forms the basis forthe pluripotent state. Thus, several epigenetic modi-fiers that confer gene silencing are bound by OCT4,NANOG, SOX2 and/or SALL4. This includes the genesthat encode the PRC components embryonic ectodermdevelopment (Eed), suppressor of zeste 12 homologue(Suz12) and enhancer of zeste homologue 2 (Ezh2),which confers H3K27me3. Other genes bound byOCT4, NANOG, SOX2 and/or SALL4 include poly-homeotic-like 1 (Phc1), the H3K9 methyltransferaseG9a (also known as euchromatic histone methyl-transferase 2(Ehmt2)) and its cofactor Ehmt1(also

    Figure 4 |Crosstalk between pluripotency factors and epigenetic modifiers.Epigenetic modifiers that are bound and, presumably, transcriptionally regulated by one

    or several of the pluripotency factors OCT4 (encoded by Pou5f1), the homeobox protein

    NANOG, SRY box-containing factor 2 (SOX2) and sal-like protein 4 (SALL4)53,56,69. In turn,

    some of these epigenetic modifiers are known to regulate the activity of pluripotency

    factors. Jumonji domain-containing 1A (JMJD1A; also known as KDM3A) and, in

    particular, JMJD2C (also known as KDM4C) are required to reverse the trimethylation

    at Lys9 of histone H3 (H3K9me3) at the Nanoglocus, which keeps the gene active69.

    On differentiation, Oct4and Nanogare rapidly silenced by H3K9me2 and H3K9me3;

    this is mediated by euchromatic histone methyltransferase 1 (EHMT1; also known as GLP)

    and G9A (also known as EHMT2)7072, leading to de novoDNA methylation by the DNA

    methyltransferases DNMT3A and DNMT3B73, potentially in conjunction with their

    co-regulators DNMT3L and lymphoid-specific helicase (HELLS; also known as LSH).

    Nanogis also silenced by H3K27me3 mediated by the Polycomb repressive complex (PRC)

    component enhancer of zeste homologue 2 (EZH2)61,72. Other components of the PRCs

    are embryonic ectoderm development (EED), suppressor of zeste 12 homologue (SUZ12)and polyhomeotic-like 1 (PHC1) and PHC2. The abundance of other epigenetic modifiers

    as targets of the pluripotency network suggests their importance for maintaining

    pluripotency; these include several AT-rich interactive domain (ARID) and JARID factors,

    the chromodomain CDYL proteins, chromodomain helicase DNA-binding protein 9

    (CHD9), the histone acetyltransferase MYST4, PR domain containing 14 (PRDM14),

    RE1-silencing transcription factor (REST), SET and SET domain-containing 1B (SETD1B)

    and the SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin

    (SMARC) proteins. The co-activation of, and co-regulation by, epigenetic modifiers might

    be a fundamental feature of the metastability of the pluripotent state.

    The genes encoding the proteins in bold are bound by all four pluripotency factors, and

    the asterisk indicates factors identified as targets in human embryonic stem (ES) cells. It is

    likely that similar networks also operate in the trophoblast lineage.

    R E V I E W S

    NATURE REVIEWS |MOLECULAR CELL BIOLOGY VOLUME 10 |AUGUST 2009 |531

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.uniprot.org/uniprot/Q8BX22http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13626&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=52615&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=14056&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13619&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=110147&ordinalpos=5&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=77683&ordinalpos=3&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=77683&ordinalpos=3&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=110147&ordinalpos=5&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13619&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=14056&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=52615&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13626&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.uniprot.org/uniprot/Q8BX22
  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    7/12

    known as Glp), the lymphoid-specific helicase andregulator of DNA methylationHells(also known asLsh) and the de novoDNA methyltransferases Dnmt3aand Dnmt3b52,53,56,57,61,68. At the same time, several epi-genetic modifiers with activating functions, includingthe H3K9 demethylases jumonji domain-containing 1a(Jmjd1a ; also known as Kdm3a)and Jmjd2c (alsoknown as Kdm4c),are also regulated by these pluripo-tency factors and their expression is crucial to preventES cell differentiation69.

    SALL4 seems to have a unique role in this cross-talk between pluripotency factors and epigeneticmodifiers as it binds cooperatively with PRC1 andPRC2 to H3K27me3 at some loci but does not requirethese repressive complexes for binding at other sites57.Although the importance of this observation is notclear, it shows a direct interaction between pluripotencyfactors and epigenetic modifiers in the maintenance ofpluripotency. Notably, a dominant function of the targetgenes of OCT4, NANOG, SOX2 and SALL4 that encodeepigenetic modifiers is to regulate the expression of

    the pluripotency factors themselves, thereby creating aself-reinforcing loop of transcriptional control (FIG. 4).For example, JMJD2C is required to reverse H3K9me3marks at the Nanoglocus and thereby keeps the geneactive. By contrast, on the induction of differentiation,Oct4is rapidly silenced by H3K9me2 and H3K9me3;this is mediated by the G9AEHMT1 complex and sub-sequent DNMT3A- and DNMT3B-dependent de novoDNA methylation. Concomitantly, reduced JMJD2Clevels owing to lower amounts of OCT4 lead to Nanogrepression by H3K9me2, H3K9me3, H3K27me3 andDNA methylation6973. Therefore, it seems that thebalance between epigenetic modifiers with opposingfunctions can maintain pluripotency by reinforcingOct4and Nanogexpression. At the same time, it isobvious that a slight disturbance of this equilibriumcauses repression of these genes and consequentlydifferentiation.

    Such a fine-tuned system provides an explanationfor the lability, or unstable nature, of the pluripotentstate and the tendency of ES cells to differentiatein vitro. The positive regulation of several epigeneticrepressors by OCT4, NANOG, SOX2 and SALL4might also explain why pluripotent cells do not persistin vivo, providing a feed-forward control mechanismto prevent uncontrolled cell proliferation and terato-genesis. This view is supported by recent studies of

    the endogenous protein complexes that OCT4 andNANOG are associated with, which identified pro-teins from the NuRD (nucleosome remodelling andhistone deacetylation), SIN3A (a transcriptional regu-lator) and PML (promyelocytic leukaemia) repressioncomplexes60. These findings highlight the repressiveroles of the key pluripotency factors that might under-lie the normal latency of the pluripotent state duringdevelopment. In addition to the probable integrationbetween genetic and epigenetic elements in the estab-lishment of the pluripotent state, it is also becomingclear that additional regulatory tiers exist that canfurther modify this crosstalk (BOX 2).

    Stochasticity and metastable states of pluripotency.Animplicit characteristic of the crosstalk between transcrip-tion factors and epigenetic modifiers in the establish-ment of pluripotency and the differentiation of early celllineages is that it leaves considerable room for plastic-ity (FIGS 3,4). Such plasticity is greatly enhanced by theepigenetic reprogramming activities that are present inearly embryos. Perhaps as a consequence of this, thereis initially no clearly restricted expression of lineage-determining transcription factors in early development(FIG. 3). OCT4 in particular is observed in all cells until themid-blastocyst stage, gradually disappearing from the TEthereafter. NANOG levels in the morula are independentof cell position, whereas expression of the trophoblast-determining transcription factor caudal-type homeo-box 2 (CDX2) seems somewhat more restricted to outercells, which might correlate with the ability of this factorto contribute to asymmetric cell divisions at the eight-cellstage7476. These findings indicate that the earliest differ-entiation events are not decisions made by single or afew factors that are unique to one cell type, but that they

    are brought about gradually by the relative abundance oftranscription factors with opposing functions.

    Interestingly, this stochasticity of gene expression isnot only true for the early embryo but is also character-istic of ES cells. An intrinsic feature of ES cells is theirfluctuation between different epigenetic states that areassociated with the heterogeneous expression of vari-ous developmentally regulated genes77. Preservation ofthis metastability is presumably the crucial step in thederivation process of ES cells, whereas in the embryothe cells rapidly fall into stable states that might beenergetically favourable, such that they become morerestricted in their potency (FIG. 5).

    The growing list of factors that are heterogeneouslyexpressed in ES cells include NANOG, platelet/endothe-lial cell adhesion molecule 1 (PECAM1), stage-specificembryonic antigen 1 (SSEA1), REX1(also known asZFP42) and STELLA7782. Expression levels of the genesthat encode these proteins in individual ES cells mightcorrelate with their potency, which can be tested experi-mentally by assessing the ability of ES cells to contributeto embryonic tissues (and ultimately the germ line) inchimeras. REX1-negative and OCT4-positive (REX1

    OCT4+) ES cells, for example, exhibit a poor ability tocontribute to chimeras compared with REX1+OCT4+EScells, and their behaviour is in that regard more similar tothat of epiSCs82. This is also in line with the finding that

    the gene expression profile of STELLAOCT4+ES cells ismore similar to that of epiSCs than the STELLA+OCT+population77. The fluctuation between the active andsilent state of Stellais accompanied by epigenetic differ-ences in these ES cell populations, with Stella-expressingcells showing an enrichment of the active chromatinmarks H3K9ac and H3K4me3 at this locus. Crucially,Stella-expressing and non-expressing ES cell subpop-ulations are interconvertible, such that the ES cell popu-lation as a whole seems to adopt similar proportions ofStella-positive and Stella-negative cells. Therefore, anongoing setting and resetting of epigenetic marks mightcontribute to the plasticity of the pluripotent state.

    R E V I E W S

    532 |AUGUST 2009 |VOLUME 10 www.nature.com/reviews/molcellbio

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=15201&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13435&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13436&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=104263&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=76804&ordinalpos=5&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.uniprot.org/uniprot/P43241http://www.uniprot.org/uniprot/Q96MM3http://www.uniprot.org/uniprot/Q96MM3http://www.uniprot.org/uniprot/P43241http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=76804&ordinalpos=5&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=104263&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13436&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13435&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=15201&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSum
  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    8/12

    |

    Potency

    Development pro ression

    TrophoblastlineageEmbryonic

    lineage

    Zygote BlastocystLateblastocyst

    Eggcylinder

    Totipotency

    ICM and ES cells iPS cells

    epiSCs

    TE and TS cells

    Epigeneticreprogramming

    Primitiveendoderm

    Trophoblast

    Epiblast

    Blastomere

    A cell that is generated during

    the first embryonic cleavage

    divisions after fertilization.

    Epigenetics and lineage commitment

    Developmental progression is fundamentally a linearprocess that involves a series of differentiation steps,proceeding from totipotency to pluripotency andmultipotency in committed cell lineages towards ter-minal differentiation (FIGS 3,5). Therefore, the embryoas a whole moves forward on the developmental scale,albeit individual cells exhibit some plasticity (the extentof which depends on the developmental stage). Theprogressive restriction of cellular plasticity is accompa-nied by epigenetic modifications that impose a cellularmemory and thereby stable cell fate.

    The question of precisely when the earliest cell line-age decision event takes place that determines the fateof a blastomeretowards TE or ICM has been studied inconsiderable detail; the most widely accepted consensusis that a first bias might be introduced after the third cellcycle, and possibly even earlier between the four- andeight-cell stages83,84.

    Transcription factors determining cell fate. At thegenetic level, key transcription factors are essentialto establish all three cell lineages of the early embryo.The precise regulation of Oct4seems to be particularlyimportant, and OCT4 levels can effectively distinguish

    between trophoblast, epiblast and primitive endodermfate: Oct4is required for ICM development and embry-onic lineage determination, as in its absence these cellsadopt trophoblast cell fate85. Oct4overexpression, bycontrast, leads to primitive endoderm differentiation.As an Oct4regulating factor, SALL4 is equally impor-tant for epiblast and primitive endoderm developmentand, as with OCT4, its downregulation causes differ-entiation into trophoblast54. Nanog is also essentialfor ICM development, but Nanog deficiency causesdifferentiation towards primitive endoderm86.

    As noted above, a feature of key importance for early

    development and pluripotency is the co-regulation ofthese factors. OCT4 and SOX2 can heterodimerize, andOCT4, NANOG and SALL4 are found in shared proteincomplexes that co-occupy binding sites in promoters,including their own5860. Intriguingly, however, OCT4can also heterodimerize with CDX2, one of the earliestknown trophoblast determinants. The OCT4CDX2complex functions mostly as a transcriptional repressor,and it seems that the reciprocal inhibition of these tran-scription factors with antagonizing lineage-determiningcapacity might be a crucial characteristic of early devel-opment87. This mutual interaction reinforces the factthat it is not the absolute presence or absence, but the

    Figure 5 |Cellular potency in development and reprogramming. The gradual loss of differentiative potency duringdevelopment can be likened to the downstream flow of a river, and epigenetic restrictions can be represented by

    cascades. Distinct states of totipotency, pluripotency and multipotency can be likened to energetic plateaux that are

    located just in front of a cascade (indicated by bars). The first major cascade is encountered at the blastocyst stage.

    Trophectoderm (TE) cells reach this cascade first as they become committed to their lineage slightly earlier than cells of

    the inner cell mass (ICM). Multipotent trophoblast stem (TS) cells are present in the conceptus until embryonic day 8.5

    (E8.5), retaining their full differentiation potential for longer than cells of the embryonic lineage125. The epiblast of egg

    cylinder-stage embryos gives rise to epiblast stem cells (epiSCs). EpiSCs are on a lower energetic plateau than embryonic

    stem (ES) cells owing to their decreased potency to produce chimeras. Once such a plateau is reached, it can tolerate

    complete (for example, NANOG in ES cells78) or temporary (for example, stochastically expressed genes) absence of some

    factors that are essential to establish this state in the first instance. These findings indicate that a particular activation

    energy is required to surmount the cascades in an upstream, counter-current direction to create states of increased

    potency. This activation energy probably reflects the necessity to initiate epigenetic reprogramming events. Such an

    interpretation has important implications for reprogramming strategies, indicating that short-term overexpression or

    activation of additional factors is needed to reach an ES cell-like state in induced-pluripotent stem (iPS) cells.

    R E V I E W S

    NATURE REVIEWS |MOLECULAR CELL BIOLOGY VOLUME 10 |AUGUST 2009 |533

    2009 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    9/12

    relative abundance of these transcription factors in pro-portion to each other that is likely the main determinantof cell fate.

    In conjunction with CDX2, the T box factor eomeso-dermin (EOMES) is required for establishment of afunctional TE and for normal trophoblast prolifera-tion and differentiation in vivo, and has trophoblast-determining functions when overexpressed in ES cells87.Notably, despite their importance for the trophoblastlineage, genetic ablation of either of these genes does notprevent initial TE formation. Therefore, there must befactors upstream of Cdx2(and Eomes) that can functionas initial determinants of trophoblast fate. One such fac-tor, TEA domain family member 4 (TEAD4), has beenrecently identified88,89. After this initial determination,reinforcement and maintenance of the trophoblast lin-eage depends on the ETS-related family transcriptionfactor ELF5. Elf5ablation causes an absence of extra-embryonic ectoderm (normally made up from diploid,proliferating trophoblast cells) and an inability to deriveself-renewing trophoblast stem (TS) cell lines in vitro90.

    It was recently found that ELF5 forms the crucial link ina self-reinforcing circuitry with CDX2 and EOMES thatis intriguingly similar to the SALL4OCT4NANOGSOX2 transcriptional network in ES cells91. Thus, CDX2,EOMES and ELF5 activate each other to establish theTS cell compartment.

    Epigenetic environment for cell fate decisions. Inaddition to these crucial transcription factors, theepigenetic environment is similarly important todetermine the potential of a cell towards a particularlineage. The euchromatic H3K9 methyltransferase SETdomain-containing B1 (SETDB1) and the enzymati-cally active component of the PRC2 complex, EZH2,which confers H3K27me3, are important for the estab-lishment of the embryonic lineage92,93. The earliestrole to date of an epigenetic modifier in cell lineagedetermination has been described for co-activator-associated arginine methyltransferase 1 (CARM1),which confers H3R2, H3R17 and H3R26 methyl-ation. These marks are generally associated with geneactivity. Blastomeres with higher endogenous CARM1levels or, in particular, those that have been injectedwith Carm1mRNA to mediate overexpression, showa bias for preferentially localizing to the ICM94. Thus,not only can the epigenetic landscape impose a per-missive (or non-permissive) environment for cell fate,

    but it might be able to predispose a cell towards aparticular lineage.

    As is evident from the preceding examples and fromthe intricate interrelationship between pluripotencygenes and epigenetic modifiers, it is difficult to deter-mine whether key transcription factors with lineage-determining capacity set up a lineage-restrictingepigenetic environment, or whether pre-imposed epi-genetic restrictions determine the activity state of thesekey transcription factors and therefore determine line-age potential. It is probably the interplay between bothmechanisms combined with an element of stochastic-ity brought about by epigenetic reprogramming that,

    in conjunction with cellcell interactions and cell-ular polarization processes95, eventually leads to cellcommitment. This model is further supported by therecent finding that cellcell contact and specific sig-nalling pathways regulate the nuclear accumulationof Yes-associated protein 1 (YAP1), a transcriptionalco-activator of TEAD4, thereby leading to the cellposition-dependent activation of lineage-specifyingtranscriptional networks in the early embryo96.

    Fixation of cell lineage fate.The presence of a lineagepreference is followed by a loss of the ability to switchlineages, resulting in fixation of cell fate at the blasto-cyst stage. A crucial epigenetic lineage barrier betweenthe embryonic and trophoblast lineage compartmentsis established by DNA methylation91. HypomethylatedES cells owing to a deficiency of, for example, Dnmt1,Dnmt3a andDnmt3b, or ubiquitin-like PHD and RINGfinger domain-containing protein 1 (Uhrf1; also knownas Np95) lose their stable lineage restriction, can adopttrophoblast cell fate in vitroand contribute to the TE

    in chimeras.The restriction of lineage potency is mediated

    through epigenetic regulation of Elf5, which is meth-ylated and silenced in the embryonic lineage buthypomethylated and expressed in the trophoblastlineage. By forming a positive-feedback loop with theTS cell genes Cdx2 and Eomes, ELF5 reinforces com-mitment to the trophoblast lineage. By contrast, thisdifferentiation pathway is aborted in the embryoniclineage owing to epigenetic silencing of Elf5. Thus, Elf5functions as a gatekeeper between these earliest twodiverging cell lineages in development91.

    An important feature of this circuitry is the positionof Elf5downstream of initial lineage determination thatcorrelates perfectly with the developmental fixation oflineage fate. It specifically allows the plasticity and regu-lation that is characteristic of early mammalian develop-ment in which expression of lineage markers remainsmosaic and individual blastomeres can still cross line-ages until the mid-blastocyst stage74. Only thereafter,activation of Elf5 in the TE of the late blastocyst and itshigh expression in the extraembryonic ectoderm rein-forces trophoblast cell fate, whereas the inability to acti-

    vate Elf5 in the epiblast owing to promoter methylationrestricts its descendants to the embryonic cell lineage.Thus, a major role of DNA methylation is to impose aspecific cellular memory through the regulation of Elf5.

    Methylation of Elf5coincides with increased Dnmt3bexpression in a time window when de novomethylationin the embryonic lineage becomes crucial for furtherdevelopment97,98. Recent findings also indicate that Stellais marked by DNA methylation in this developmentalperiod. Thus, whereas Stellais expressed in the ICM ofblastocysts, it becomes methylated and stably repressedin the epiblast of post-implantation embryos77. Thiscoincides with a clear restriction of potency in epiblastcells: ICM-derived ES cells readily populate embryonictissues when introduced into blastocysts, but epiblast-derived epiSCs cannot (or can only extremely rarely)contribute to chimeras48,49.

    R E V I E W S

    534 |AUGUST 2009 |VOLUME 10 www.nature.com/reviews/molcellbio

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.uniprot.org/uniprot/O95936http://www.uniprot.org/uniprot/Q15561http://www.uniprot.org/uniprot/Q9UKW6http://www.uniprot.org/uniprot/Q9UKW6http://www.uniprot.org/uniprot/Q15561http://www.uniprot.org/uniprot/O95936
  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    10/12

    The emerging picture from these observations is thatthe progressive reduction of developmental potency pro-ceeds in a stepwise manner in which, first, permissivechromatin states for lineage determination are establishedby histone modifications in a tight interplay with trans-criptional regulators, including OCT4, NANOG, SOX2,SALL4 as well as TEAD4, CDX2 and EOMES. Once thelineages have been specified, DNA methylation of a fewcrucial loci, notably Elf5and Stella,is then required torestrict their differentiation potential and to establishlineage-committed cell populations, the fate allocationof which is stably inherited by all descendants (FIGS 3,5).

    Reversion of cellular restrictions

    The stepwise acquisition of epigenetic marks culminatingin the fixation of lineage fate by differential DNA methyl-ation of gatekeeper genes, such as Elf5, forms the molec-ular basis for Waddingtons concept of the canalization ofdevelopmental pathways. Waddington compared the pathof a cell (lineage) towards terminal differentiation with aball travelling downwards along branching valleys; once

    in its final valley, the ball cannot easily cross the mountaininto neighbouring valleys or return to the beginning99.This canalization explains how cellular differentiationpathways become stable and potentially irreversible.It also depicts the unidirectionality of differentiationthat dictates developmental progression and preventsteratogenesis during a time of exponential growth.

    The ability to derive induced pluripotent stem (iPS)cells from fully differentiated cell types (FIG. 1)is thegroundbreaking reversion of this strict developmentalprogression, creating bidirectional developmental path-ways by experimental manipulation100103. Thus, tem-porally limited overexpression of the four transcriptionfactors OCT4, kruppel-like factor 4 (KLF4), SOX2 andMYCis sufficient to reactivate endogenous pluripotencygenes and to regain a developmental potency akin to thatof ES cells. Other combinations of factors that have beensuccessfully used for the derivation of iPS cells includethe oestrogen-related receptor- (ESRRB) in conjunctionwith OCT4 and NANOG in mouse fibroblasts104, andOCT4, SOX2, NANOG and the Caenorhabditis elegansLIN-28homologue in human fibroblasts105.

    Reflecting the necessity for extensive epigeneticreprogramming in the iPS cell generation process, theefficiency of iPS cell derivation can be enhanced inthe presence of inhibitors of histone deacetylases, the his-tone methyltransferase G9A and DNA methylation106108,

    and these components can substitute for some repro-gramming factors. In efforts to understand how suchdevelopmental reversion can be achieved in principle,recent computational modelling has produced importantinsights109,110. In these models, transcriptional feedbackloops create molecular switches that result in successivegene restrictions that correspond to a controlled differ-entiation cascade. Although cells are robustly resistantto reprogramming, the model also predicts that amplifi-cation of low-level fluctuations in expression (so-calledtranscriptional noise) might be sufficient to trigger thereactivation of the core pluripotency switch. The impor-tant insights from such studies are that reprogramming

    towards pluripotency depends on the positive inter-ference of several factors, each with a certain noise ampli-tude of expression, thus explaining the stochasticity ofexperimental reprogramming as well as the certain extentto which reprogramming factors can be exchanged.

    It is also becoming clear that the reversion of a cell intoa state of increased potency requires temporally higherlevels of gene expression, equivalent to an activationenergy, than those required once that state is reached. Itis even the case that cells on these plateaux can toleratecomplete (such as NANOG in ES cells78) or temporary(as in stochastically expressed genes) absence of somefactors that are essential to establish this state in the firstinstance without loss of developmental potency. We com-bine these data in a model in which development is like-ned to a stream and epigenetic restrictions to cascades;reprogramming is only possible by lifting a cell back upto the original level (FIG. 5). With better knowledge of theprecise molecular processes during reprogramming, itwill be possible to define the exact composition of thefactors that are required for each step.

    Conclusions and future perspectives

    Recent results highlight the characteristics and impor-tance of genome-wide epigenetic reprogramming thatoccurs both in the germ line and in the early embryo.The mechanistic aspects of reprogramming, whichinvolve histone marks, histone exchange and the use ofhistone variants along with demethylation of DNA, arecurrently under intense investigation. Reprogramming inPGCs is likely to be important for the erasure of genomicimprints and of epimutations, and could also be inte-gral for the control of transposon silencing or mobil-ity111. Epigenetic reprogramming in the early embryois crucial to erase further gametic gene expressionprogrammes and thus might be partly responsible forstochastic expression of lineage-determining transcrip-tion factors. The combinatorial stochastic expression oftranscription factors becomes subsequently restrictedto establish lineage-specific transcriptional circuits.This transcriptional focusing involves epigenetic mark-ing of gatekeeper genes, such as Elf5, that function toreinforce the trophoblast-specific transcriptional circuit.Perhaps there are similar gatekeepers that are involved inother lineage decisions, for example in maintaining theepiblast lineage. Thus, it is possible that methylation ofStella, which occurs during the transition from ES cells(and ICM) to epiSCs (and epiblast tissue), fulfils a similar

    gatekeeper function.Future work will focus on finding out when and how

    the first lineage-specific epigenetic marks arise. This willbe aided by the identification of major epigenetic modifi-ers and how they act in both a genome-wide and targetedmanner. The development of epigenomics technologiesthat can be applied to small numbers of cells will be ofcrucial importance to unravel key features of epi geneticplasticity and lineage commitment in mammaliandevelopment. Establishing the basic principles on whichcell lineages are defined and maintained will be crucialfor developing safe and succesful cell therapies to realizethe full potential of regenerative medicine.

    R E V I E W S

    NATURE REVIEWS |MOLECULAR CELL BIOLOGY VOLUME 10 |AUGUST 2009 |535

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.uniprot.org/uniprot/O43474http://www.uniprot.org/uniprot/P01106http://www.uniprot.org/uniprot/O95718http://www.uniprot.org/uniprot/P92186http://www.uniprot.org/uniprot/P92186http://www.uniprot.org/uniprot/O95718http://www.uniprot.org/uniprot/P01106http://www.uniprot.org/uniprot/O43474
  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    11/12

    1. Reik, W. Stability and flexibility of epigenetic gene

    regulation in mammalian development. Nature447,

    425432 (2007).

    2. Sasaki, H. & Matsui, Y. Epigenetic events in

    mammalian germ-cell development: reprogramming

    and beyond. Nature Rev. Genet.9, 129140 (2008).

    3. Dean, W., Santos, F. & Reik, W. Epigenetic

    reprogramming in early mammalian development and

    following somatic nuclear transfer. Semin. Cell Dev.

    Biol.14, 93100 (2003).

    4. Surani, M. A., Hayashi, K. & Hajkova, P. Genetic and

    epigenetic regulators of pluripotency. Cell128,747762 (2007).

    5. Oswald, J. et al.Active demethylation of the paternal

    genome in the mouse zygote. Curr. Biol.10, 475478

    (2000).6. Mayer, W., Niveleau, A., Walter, J., Fundele, R. &

    Haaf, T. Demethylation of the zygotic paternal

    genome. Nature403, 501502 (2000).

    7. Santos, F., Hendrich, B., Reik, W. & Dean, W.

    Dynamic reprogramming of DNA methylation in the

    early mouse embryo. Dev. Biol.241, 172182

    (2002).

    A detailed analysis of the DNA methylation

    dynamics in pre-implantation mouse development.

    8. Howell, C. Y. et al.Genomic imprinting disrupted by a

    maternal effect mutation in the Dnmt1gene. Cell104,

    829838 (2001).

    9. Kwon, G. S., Viotti, M. & Hadjantonakis, A. K. The

    endoderm of the mouse embryo arises by dynamic

    widespread intercalation of embryonic and

    extraembryonic lineages. Dev. Cell15, 509520

    (2008).

    10. Vincent, S. D. et al.The zinc finger transcriptional

    repressor Blimp1/Prdm1 is dispensable for early axis

    formation but is required for specification of

    primordial germ cells in the mouse. Development132,

    13151325 (2005).

    11. Hayashi, K., de Sousa Lopes, S. M. & Surani, M. A.

    Germ cell specification in mice. Science316,

    394396 (2007).

    12. Saitou, M., Barton, S. C. & Surani, M. A. A molecular

    programme for the specification of germ cell fate in

    mice. Nature418, 293300 (2002).

    13. Yamazaki, Y. et al.Reprogramming of primordial

    germ cells begins before migration into the genital

    ridge, making these cells inadequate donors for

    reproductive cloning. Proc. Natl Acad. Sci. USA100,

    1220712212 (2003).

    14. Lee, J. et al.Erasing genomic imprinting memory in

    mouse clone embryos produced from day 11.5

    primordial germ cells. Development129, 18071817

    (2002).15. Hajkova, P. et al.Chromatin dynamics during

    epigenetic reprogramming in the mouse germ line.

    Nature452, 877881 (2008).

    A comprehensive study of the dynamic changes of

    a range of epigenetic modifications during germ

    cell development.

    16. Seki, Y. et al.Cellular dynamics associated with the

    genome-wide epigenetic reprogramming in migrating

    primordial germ cells in mice. Development134,

    26272638 (2007).

    17. Seki, Y. et al.Extensive and orderly reprogramming of

    genome-wide chromatin modifications associated with

    specification and early development of germ cells in

    mice. Dev. Biol.278, 440458 (2005).

    18. Hajkova, P. et al.Epigenetic reprogramming in mouse

    primordial germ cells. Mech. Dev.117, 1523

    (2002).

    19. Ancelin, K. et al.Blimp1 associates with Prmt5 and

    directs histone arginine methylation in mouse germ

    cells. Nature Cell Biol.8, 623630 (2006).20. Morgan, H. D., Dean, W., Coker, H. A., Reik, W. &

    Petersen-Mahrt, S. K. Activation-induced cytidine

    deaminase deaminates 5-methylcytosine in DNA and

    is expressed in pluripotent tissues: implications for

    epigenetic reprogramming.J.Biol. Chem.279,

    5235352360 (2004).

    21. Barreto, G. et al.Gadd45a promotes epigenetic gene

    activation by repair-mediated DNA demethylation.

    Nature445, 671675 (2007).

    22. Gehring, M., Reik, W. & Henikoff, S. DNA

    demethylation by DNA repair. Trends Genet.25,

    8290 (2009).23. Huh, J. H., Bauer, M. J., Hsieh, T. F. & Fischer, R. L.

    Cellular programming of plant gene imprinting.

    Cell132, 735744 (2008).

    24. Rai, K. et al.DNA demethylation in zebrafish involves

    the coupling of a deaminase, a glycosylase, and

    gadd45. Cell135, 12011212 (2008).

    25. Govin, J. et al.Pericentric heterochromatin

    reprogramming by new histone variants during mouse

    spermiogenesis.J. Cell Biol.176, 283294 (2007).

    26. Martens, J. H. et al.The profile of repeat-associated

    histone lysine methylation states in the mouse

    epigenome. EMBO J.24, 800812 (2005).

    27. Peters, A. H. et al.Loss of the Suv39h histone

    methyltransferases impairs mammalian

    heterochromatin and genome stability. Cell107,

    323337 (2001).

    28. Santos, F., Peters, A. H., Otte, A. P., Reik, W. &

    Dean, W. Dynamic chromatin modificationscharacterise the first cell cycle in mouse embryos.

    Dev. Biol.280, 225236 (2005).

    29. van der Heijden, G. W. et al.Asymmetry in histone H3

    variants and lysine methylation between paternal

    and maternal chromatin of the early mouse zygote.

    Mech. Dev.122, 10081022 (2005).

    30. Puschendorf, M. et al.PRC1 and Suv39h specify

    parental asymmetry at constitutive heterochromatin in

    early mouse embryos. Nature Genet.40, 411420

    (2008).

    31. Probst, A. V., Santos, F., Reik, W., Almouzni, G. &

    Dean, W. Structural differences in centromeric

    heterochromatin are spatially reconciled on

    fertilisation in the mouse zygote. Chromosoma116,

    403415 (2007).

    32. Monk, M., Boubelik, M. & Lehnert, S. Temporal and

    regional changes in DNA methylation in the

    embryonic, extraembryonic and germ cell lineages

    during mouse embryo development. Development99,

    371382 (1987).

    33. Rougier, N. et al.Chromosome methylation patterns

    during mammalian preimplantation development.

    Genes Dev.12, 21082113 (1998).

    34. Howlett, S. K. & Reik, W. Methylation levels of

    maternal and paternal genomes during

    preimplantation development. Development113,

    119127 (1991).

    35. Edwards, C. A. & Ferguson-Smith, A. C. Mechanisms

    regulating imprinted genes in clusters. Curr. Opin. Cell

    Biol.19, 281289 (2007).

    36. Lane, N. et al.Resistance of IAPs to methylation

    reprogramming may provide a mechanism for

    epigenetic inheritance in the mouse. Genesis35,

    8893 (2003).

    37. Chapman, V., Forrester, L., Sanford, J., Hastie, N. &

    Rossant, J. Cell lineage-specific undermethylation of

    mouse repetitive DNA. Nature307, 284286 (1984).

    38. Rossant, J., Sanford, J. P., Chapman, V. M. &

    Andrews, G. K. Undermethylation of structural gene

    sequences in extraembryonic lineages of the mouse.

    Dev. Biol.117, 567573 (1986).39. Farthing, C. R. et al.Global mapping of DNA

    methylation in mouse promoters reveals epigenetic

    reprogramming of pluripotency genes. PLoS Genet.4,

    e1000116 (2008).40. Tanaka, T. S. et al.Gene expression profiling of

    embryo-derived stem cells reveals candidate genes

    associated with pluripotency and lineage specificity.

    Genome Res.12, 19211928 (2002).

    41. Morgan, H. D., Santos, F., Green, K., Dean, W. &

    Reik, W. Epigenetic reprogramming in mammals.

    Hum. Mol. Genet.14, R47R58 (2005).42. Erhardt, S. et al.Consequences of the depletion of

    zygotic and embryonic enhancer of zeste 2 during

    preimplantation mouse development. Development

    130, 42354248 (2003).

    43. Sarmento, O. F. et al.Dynamic alterations of specific

    histone modifications during early murine

    development.J. Cell Sci.117, 44494459 (2004).44. Matsui, Y., Zsebo, K. & Hogan, B. L. Derivation of

    pluripotential embryonic stem cells from murineprimordial germ cells in culture. Cell70, 841847

    (1992).

    45. Resnick, J. L., Bixler, L. S., Cheng, L. & Donovan, P. J.

    Long-term proliferation of mouse primordial germ

    cells in culture. Nature359, 550551 (1992).

    46. Evans, M. J. & Kaufman, M. H. Establishment in

    culture of pluripotential cells from mouse embryos.

    Nature292, 154156 (1981).

    Pioneering work that describes the first derivation

    of pluripotent, self-renewing embryonic stem cells

    in mice.

    47. Tesar, P. J. Derivation of germ-line-competent

    embryonic stem cell lines from preblastocyst mouse

    embryos. Proc. Natl Acad. Sci. USA102, 82398244

    (2005).48. Brons, I. G. et al.Derivation of pluripotent epiblast

    stem cells from mammalian embryos. Nature448,

    191195 (2007).

    49. Tesar, P. J. et al.New cell lines from mouse epiblast

    share defining features with human embryonic stem

    cells. Nature448, 196199 (2007).

    References 48 and 49 identify a pluripotent cell

    population in the post-implantation epiblast that

    can give rise to self-renewing epiblast stem cells

    with unique characteristics compared with ES cells.

    50. Durcova-Hills, G., Tang, F., Doody, G., Tooze, R. &

    Surani, M. A. Reprogramming primordial germ cells

    into pluripotent stem cells. PLoS ONE3, e3531

    (2008).

    51. Niwa, H. How is pluripotency determined andmaintained? Development134, 635646 (2007).

    52. Loh, Y. H. et al.The Oct4 and Nanog transcription

    network regulates pluripotency in mouse embryonic

    stem cells. Nature Genet.38, 431440 (2006).

    One of the first large-scale chromatin immuno-

    precipitation screens identifying the transcriptional

    network established by the pluripotency factors

    OCT4 and NANOG in mouse ES cells.53. Sharov, A. A. et al.Identification of Pou5f1, Sox2, and

    Nanogdownstream target genes with statistical

    confidence by applying a novel algorithm to time

    course microarray and genome-wide chromatin

    immunoprecipitation data. BMC Genomics9, 269

    (2008).

    54. Zhang, J. et al.Sall4 modulates embryonic stem cell

    pluripotency and early embryonic development by the

    transcriptional regulation of Pou5f1. Nature Cell Biol.

    8, 11141123 (2006).55. Wu, Q. et al.Sall4 interacts with Nanog and

    co-occupies Nanog genomic sites in embryonic stem

    cells.J. Biol. Chem.281, 2409024094 (2006).

    56. Lim, C. Y. et al.Sall4 regulates distinct transcription

    circuitries in different blastocyst-derived stem cell

    lineages. Cell Stem Cell3, 543554 (2008).

    57. Yang, J. et al.Genome-wide analysis reveals Sall4 to

    be a major regulator of pluripotency in murine-

    embryonic stem cells. Proc. Natl Acad. Sci. USA105,

    1975619761 (2008).

    58. Chew, J. L. et al.Reciprocal transcriptional regulation

    of Pou5f1 and Sox2 via the Oct4/Sox2 complex in

    embryonic stem cells. Mol. Cell. Biol.25, 60316046

    (2005).

    59. Rodda, D. J. et al.Transcriptional regulation of

    Nanogby OCT4 and SOX2.J. Biol. Chem.280,

    2473124737 (2005).

    60. Liang, J. et al.Nanog and Oct4 associate with unique

    transcriptional repression complexes in embryonic

    stem cells. Nature Cell Biol.10, 731739 (2008).

    61. Boyer, L. A. et al.Polycomb complexes repress

    developmental regulators in murine embryonic stem

    cells. Nature441, 349353 (2006).Genome-wide profile of the target genes of PcG

    proteins that identifies that PcG proteins repress a

    large cohort of developmental regulators in ES

    cells, the expression of which would otherwise

    promote differentiation.62. Lee, T. I. et al.Control of developmental regulators by

    polycomb in human embryonic stem cells. Cell125,

    301313 (2006).

    63. Azuara, V. et al.Chromatin signatures of pluripotent

    cell lines. Nature Cell Biol.8, 532538 (2006).

    64. Bernstein, B. E. et al.A bivalent chromatin structure

    marks key developmental genes in embryonic stem

    cells. Cell125, 315326 (2006).

    References 63 and 64 found independently that

    genes required for later development are in a

    poised state for activation, whichis characterized

    by bivalent chromatin domains that consist of active

    (H3K4me3) and repressive (H3K27me3) marks.

    65. Mikkelsen, T. S. et al.Genome-wide maps of chromatin

    state in pluripotent and lineage-committed cells.Nature448, 553560 (2007).

    66. Fouse, S. D. et al.Promoter CpG methylation

    contributes to ES cell gene regulation in parallel with

    Oct4/Nanog, PcG complex, and histone H3 K4/K27

    trimethylation. Cell Stem Cell2, 160169 (2008).

    67. Meissner, A. et al.Genome-scale DNA methylation

    maps of pluripotent and differentiated cells. Nature

    454, 766770 (2008).

    68. Ura, H. et al.STAT3 and Oct-3/4 control histone

    modification through induction of Eed in embryonic

    stem cells.J. Biol. Chem.283, 97139723 (2008).

    69. Loh, Y. H., Zhang, W., Chen, X., George, J. & Ng, H. H.

    Jmjd1a and Jmjd2c histone H3 Lys 9 demethylases

    regulate self-renewal in embryonic stem cells. Genes

    Dev.21, 25452557 (2007).

    70. Feldman, N. et al.G9a-mediated irreversible

    epigenetic inactivation of Oct-3/4during early

    embryogenesis. Nature Cell Biol.8, 188194 (2006).

    R E V I E W S

    536 |AUGUST 2009 |VOLUME 10 www.nature.com/reviews/molcellbio

    2009 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Epigenetic Dynamics of Stem Cells

    12/12

    71. Epsztejn-Litman, S. et al.De novoDNA methylation

    promoted by G9a prevents reprogramming of

    embryonically silenced genes. Nature Struct. Mol.

    Biol.15, 11761183 (2008).

    72. Hattori, N. et al.Epigenetic regulation of Nanoggene

    in embryonic stem and trophoblast stem cells. Genes

    Cells12, 387396 (2007).

    73. Li, J. Y.et al.Synergistic function of DNA

    methyltransferases Dnmt3a and Dnmt3b in the

    methylation of Oct4and Nanog. Mol. Cell. Biol.27,

    87488759 (2007).

    74. Dietrich, J. E. & Hiiragi, T. Stochastic patterning in themouse pre-implantation embryo. Development134,

    42194231 (2007).

    A detailed investigation of the expression of line-

    age-determining transcription factors in the early

    mouse embryo that shows the stochasticity of their

    expression until the mid-to-late blastocyst stage.

    75. Ralston, A. & Rossant, J. Cdx2 acts downstream of

    cell polarization to cell-autonomously promote

    trophectoderm fate in the early mouse embryo. Dev.

    Biol.313, 614629 (2008).

    76. Jedrusik, A. et al.Role of Cdx2 and cell polarity in cell

    allocation and specification of trophectoderm and

    inner cell mass in the mouse embryo. Genes Dev.22,

    26922706 (2008).

    77. Hayashi, K., Lopes, S. M., Tang, F. & Surani, M. A.

    Dynamic equilibrium and heterogeneity of mouse

    pluripotent stem cells with distinct functional and

    epigenetic states. Cell Stem Cell3, 391401 (2008).

    ES cells fluctuate in their expression of

    developmental key genes, such as Stella, and

    these STELLA-positive and STELLA-negative

    subpopulations, although epigenetically and

    functionally distinct, are interconvertible.78. Chambers, I. et al.Nanog safeguards pluripotency

    and mediates germline development. Nature450,

    12301234 (2007).

    79. Cui, L. et al.Spatial distribution and initial changes

    of SSEA-1 and other cell adhesion-related molecules

    on mouse embryonic stem cells before and during

    differentiation.J. Histochem. Cytochem.52,

    14471457 (2004).

    80. Furusawa, T., Ohkoshi, K., Honda, C., Takahashi, S. &

    Tokunaga, T. Embryonic stem cells expressing both

    platelet endothelial cell adhesion molecule-1 and

    stage-specific embryonic antigen-1 differentiate

    predominantly into epiblast cells in a chimeric embryo.

    Biol. Reprod.70, 14521457 (2004).

    81. Payer, B. et al.Generation of stellaGFPtransgenic

    mice: a novel tool to study germ cell development.

    Genesis44, 7583 (2006).

    82. Toyooka, Y., Shimosato, D., Murakami, K.,Takahashi, K. & Niwa, H. Identification and

    characterization of subpopulations in

    undifferentiated ES cell culture. Development135,

    909918 (2008).

    83. Zernicka-Goetz, M. The first cell-fate decisions in the

    mouse embryo: destiny is a matter of both chance

    and choice. Curr. Opin. Genet. Dev.16, 406412

    (2006).

    84. Rossant, J. & Tam, P. P. Blastocyst lineage formation,

    early embryonic asymmetries and axis patterning in

    the mouse. Development136, 701713 (2009).

    85. Niwa, H., Miyazaki, J. & Smith, A. G. Quantitative

    expression of Oct-3/4 defines differentiation,

    dedifferentiation or self-renewal of ES cells. Nature

    Genet.24, 372376 (2000).

    86. Mitsui, K. et al.The homeoprotein Nanog is required

    for maintenance of pluripotency in mouse epiblast and

    ES cells. Cell113, 631642 (2003).

    87. Niwa, H. et al.Interaction between Oct3/4 and Cdx2

    determines trophectoderm differentiation. Cell123,917929 (2005).

    An important concept in the cell lineage

    specification process is the mutual inhibitory

    interaction of transcription factors with opposing

    functions.

    88. Nishioka, N. et al.Tead4 is required for specification of

    trophectoderm in pre-implantation mouse embryos.

    Mech. Dev.125, 270283 (2008).

    89. Yagi, R. et al.Transcription factor TEAD4 specifies

    the trophectoderm lineage at the beginning of

    mammalian development. Development134,

    38273836 (2007).

    90. Donnison, M. et al.Loss of the extraembryonic

    ectoderm in Elf5mutants leads to defects in

    embryonic patterning. Development132,

    22992308 (2005).

    91. Ng, R. K. et al.Epigenetic restriction of embryonic cell

    lineage fate by methylation of Elf5. Nature Cell Biol.

    10, 12801290 (2008).

    DNA methylation establishes a major epigenetic

    restriction of cell lineage fate by regulating the

    trophoblast-reinforcing gatekeeper gene Elf5.92. Dodge, J. E., Kang, Y. K., Beppu, H., Lei, H. & Li, E.

    Histone H3-K9 methyltransferase ESET is essential forearly development. Mol. Cell. Biol.24, 24782486

    (2004).

    93. OCarroll, D. et al.The polycomb-group gene Ezh2is

    required for early mouse development. Mol. Cell. Biol.

    21, 43304336 (2001).94. Torres-Padilla, M. E., Parfitt, D. E., Kouzarides, T. &

    Zernicka-Goetz, M. Histone arginine methylation

    regulates pluripotency in the early mouse embryo.

    Nature445, 214218 (2007).

    95. Yamanaka, Y., Ralston, A., Stephenson, R. O. &

    Rossant, J. Cell and molecular regulation of the mouse

    blastocyst. Dev. Dyn.235, 23012314 (2006).

    96. Nishioka, N. et al.The Hippo signaling pathway

    components Lats and Yap pattern Tead4 activity to

    distinguish mouse trophectoderm from inner cell

    mass. Dev. Cell16, 398410 (2009).

    97. Hirasawa, R. & Sasaki, H. Dynamic transition of

    Dnmt3bexpression in mouse pre- and early post-

    implantation embryos. Gene Expr. Patterns9, 2730

    (2009).

    98. Watanabe, D., Suetake, I., Tada, T. & Tajima, S.

    Stage- and cell-specific expression of Dnmt3aand

    Dnmt3bduring embryogenesis. Mech. Dev.118,

    187190 (2002).

    99. Waddington, C. H. Organisers and Genes(Cambridge

    Univ. Press, Cambridge, UK, 1940).

    100. Takahashi, K. & Yamanaka, S. Induction of pluripotent

    stem cells from mouse embryonic and adult fibroblast

    cultures by defined factors. Cell126, 663676

    (2006).

    The first groundbreaking report of the derivation

    of ES cell-like iPS cells from terminally

    differentiated adult fibroblasts.101. Takahashi, K. et al.Induction of pluripotent stem cells

    from adult human fibroblasts by defined factors.

    Cell131, 861872 (2007).102. Meissner, A., Wernig, M. & Jaenisch, R. Direct

    reprogramming of genetically unmodified fibroblasts

    into pluripotent stem cells. Nature Biotechnol.25,

    11771181 (2007).

    103. Okita, K., Nakagawa, M., Hyenjong, H., Ichisaka, T. &Yamanaka, S. Generation of mouse induced

    pluripotent stem cells without viral vectors. Science

    322, 949953 (2008).

    104. Feng, B. et al.Reprogramming of fibroblasts into

    induced pluripotent stem cells with orphan nuclear

    receptor Esrrb. Nature Cell Biol.11, 197203 (2009).

    105.Yu, J. et al.Induced pluripotent stem cell lines derived

    from human somatic cells. Science318, 19171920

    (2007).

    106. Mikkelsen, T. S. et al.Dissecting direct reprogramming

    through integrative genomic analysis. Nature454,

    4955 (2008).107. Shi, Y. et al.A combined chemical and genetic

    approach for the generation of induced pluripotent

    stem cells. Cell Stem Cell2, 525528 (2008).

    108. Huangfu, D. et al.Induction of pluripotent stem cells

    by defined factors is greatly improved by small-

    molecule compounds. Nature Biotechnol.26,

    795797 (2008).

    109. Chang, H. H., Hemberg, M., Barahona, M., Ingber,D. E. & Huang, S. Transcriptome-wide noise controls

    lineage choice in mammalian progenitor cells.

    Nature453, 544547 (2008).

    Gene expression noise causes fluctuations in

    protein levels that produce persistent cell

    individuality in clonal populations. This

    stochasticity can drive lineage specification.

    110. MacArthur, B. D., Please, C. P. & Oreffo, R. O.

    Stochasticity and the molecular mechanisms of

    induced pluripotency. PLoS ONE3, e3086 (2008).

    Computational modelling of how the positive

    interference of stochastic gene expression levels

    can lead to lineage choice and enables

    reprogramming to iPS cells.

    111. Slotkin, R. K. et al.Epigenetic reprogramming and

    small RNA silencing of transposable elements in

    pollen. Cell136, 461472 (2009).

    112. Wang, J. et al.The lysine demethylase LSD1 (KDM1)

    is required for maintenance of global DNA

    methylation. Nature Genet.41, 125129 (2009).

    113.Ayoub, N., Jeyasekharan, A. D., Bernal, J. A. &

    Venkitaraman, A. R. HP1-mobilization promotes

    chromatin changes that initiate the DNA damage

    response. Nature453, 682686 (2008).114. Chin, H. G. et al.Automethylation of G9a and its

    implication in wider substrate specificity and HP1

    binding. Nucleic Acids Res.35, 73137323

    (2007).

    115. Wei, F., Scholer, H. R. & Atchison, M. L. Sumoylation

    of Oct4 enhances its stability, DNA binding, and

    transactivation. J. Biol. Chem.282, 2155121560

    (2007).116. Cardoso, M. C. & Leonhardt, H. DNA methyltransferase

    is actively retained in the cytoplasm during early

    development.J. Cell Biol.147, 2532 (1999).

    117. Tanaka, S., Kunath, T., Hadjantonakis, A. K., Nagy, A.

    & Rossant, J. Promotion of trophoblast stem cell

    proliferation by FGF4. Science282, 20722075

    (1998).

    The successful derivation and maintenance of

    stem cells of the trophoblast lineage that retain

    the established lineage restriction and contribute

    exclusively to placental structures in chimeras.118. Kunath, T. et al.Imprinted X-inactivation in extra-

    embryonic endoderm cell lines from mouse

    blastocysts. Development132, 16491661 (2005).119.Yeom, Y. I. et al.Germline regulatory element of Oct-4

    specific for the totipotent cycle of embryonal cells.

    Development122, 881894 (1996).

    120.Avilion, A. A. et al.Multipotent cell lineages in early

    mouse development depend on SOX2 function. Genes

    Dev.17, 126140 (2003).

    121. Elling, U., Klasen, C., Eisenberger, T., Anlag, K. &

    Treier, M. Murine inner cell mass-derived lineages

    depend on Sall4 function. Proc. Natl Acad. Sci. USA

    103, 1631916324 (2006).

    122. Payer, B. et al.Stellais a maternal effect gene required

    for normal early development in mice. Curr. Biol.13,

    21102117 (2003).

    123. Strumpf, D. et al.Cdx2 is required for correct cell fate

    specification and differentiation of trophectoderm in

    the mouse blastocyst. Development132, 20932102

    (2005).

    124. McConnell, J., Petrie, L., Stennard, F., Ryan, K. &

    Nichols, J. Eomesodermin is expressed in mouseoocytes and pre-implantation embryos. Mol. Reprod.

    Dev.71, 399404 (2005).

    125. Uy, G. D., Downs, K. M. & Gardner, R. L. Inhibition of

    trophoblast stem cell potential in chorionic ectoderm

    coincides with occlusion of the ectoplacental cavity in

    the mouse. Development129, 39133924 (2002).

    AcknowledgementsThe authors thank all their colleagues for advice and discus-

    sion. Work in the authors laboratories is supported by the

    Biotechnology and Biological Sciences Research Council,

    the Medical Research Council, the European Union Epigenome

    Network of Excellence, the Technology Strategy Board and

    CellCentric.

    DATABASESEntrez Gene:http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene

    Blimp1| Dnmt3a|Dnmt3b| Eed| Ehmt1| Ezh2| G9a| Hells|Jmjd1a|Jmjd2c|Phc1| Stella| Suz12

    UniProtKB:http://www.uniprot.org

    CDX2| ELF5|EOMES|ESRRB| KLF4|LIN-28|MYC|NANOG

    | OCT4| REX1| SALL4| SOX2| TEAD4

    FURTHER INFORMATIONWolf Reiks homepage:

    http://www.babraham.ac.uk/devgen/reik.html

    Myriam Hembergers homepage: http://www.babraham.

    ac.uk/devgen/hemberger.html

    ALL LINKS ARE ACTIVE IN THE ONLINE PDF

    R E V I E W S

    NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 10 | AUGUST 2009 | 537

    http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=genehttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=genehttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=12142&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13435&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13436&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13436&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=13626&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=77683&ordinalpos=3&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=14056&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=14056&ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=110147&ord