Download Control of plant cell differentiation by histone modification

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cytokinesis wikipedia , lookup

Extracellular matrix wikipedia , lookup

Cell growth wikipedia , lookup

Mitosis wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Cell culture wikipedia , lookup

Cell cycle wikipedia , lookup

SULF1 wikipedia , lookup

Histone acetylation and deacetylation wikipedia , lookup

Amitosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Cellular differentiation wikipedia , lookup

Epigenetics in stem-cell differentiation wikipedia , lookup

Transcript
Available online at www.sciencedirect.com
ScienceDirect
Control of plant cell differentiation by histone
modification and DNA methylation
Momoko Ikeuchi, Akira Iwase and Keiko Sugimoto
How cells differentiate and acquire diverse arrays of
determined states in multicellular organisms is a fundamental
and yet unanswered question in biology. Molecular genetic
studies over the last few decades have identified many
transcriptional regulators that activate or repress gene
expression to promote cell differentiation in plant development.
What has recently emerged as an additional important
regulatory layer is the control at the epigenetic level by which
locus-specific DNA methylation and histone modification alter
the chromatin state and limit the expression of key
developmental regulators to specific windows of time and
space. Accumulating evidence suggests that histone
acetylation is commonly linked with active transcription and
this mechanism is adopted to control sequential progression of
cell differentiation. Histone H3 trimethylation at lysine 27 and
DNA methylation are both associated with gene repression,
and these mechanisms are often utilised to promote and/or
maintain the differentiated status of plant cells.
Address
RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho,
Tsurumi, Yokohama, Kanagawa 230-0045, Japan
Corresponding author: Sugimoto, Keiko ([email protected])
Current Opinion in Plant Biology 2015, 28:60–67
This review comes from a themed issue on Cell biology
Edited by Hiroo Fukuda and Zhenbiao Yang
mechanical injuries or hormonal stimuli, undergo cellular
reprogramming to regenerate new tissues or organs [3].
Given that each differentiation step entails global
changes in gene expression, transcription factors have
instructive roles in cellular differentiation and reprogramming. Indeed, recent studies have greatly advanced our
understanding of how key developmental regulators govern a cascade of transcriptional networks to control cell
differentiation and reprogramming [3–6].
Epigenetic modifications refer to mitotically heritable
changes in chromatin without making changes in DNA
sequences and well-characterised examples include DNA
methylation and histone modification. These epigenetic
modifications alter the chromatin environment where
transcription factors and basic transcription machinery
are in play, and influence target gene expression positively or negatively. Earlier studies uncovered vital roles of
epigenetic regulation in controlling key developmental
transitions, that is, embryonic-to-vegetative and vegetative-to-reproductive growth phases [7–9]. In addition,
accumulating evidence implicates epigenetic modifications in the control of cellular differentiation at broader
stages of plant development. In this review we discuss
emerging concepts on the epigenetic regulation of cellular differentiation and reprogramming. Epigenetic regulations governed by other mechanisms such as non-coding
RNA have been recently reviewed in other articles
[10,11], hence we focus our attention on the role of
histone modification and DNA methylation in this article.
http://dx.doi.org/10.1016/j.pbi.2015.09.004
1369-5266/# 2015 Elsevier Ltd. All rights reserved.
Introduction
Cellular differentiation is a sequential process by which
cells that derive from pluripotent stem cells gradually lose
their fate options and acquire a certain determined state.
In plant shoots, for example, stem cells in the shoot apical
meristem (SAM) first give rise to organ founder cells
which subsequently differentiate into leaves or stems
[1]. Cells that have acquired the leaf identity differentiate
into epidermis, ground tissue or vascular tissues, and
many of these cells undergo further specification to gain
terminally differentiated state, for instance, pavement
cells, trichomes and guard cells in the leaf epidermis
[2]. Mature plant organs are also known to maintain a
group of relatively undifferentiated cells, which upon
Current Opinion in Plant Biology 2015, 28:60–67
Histone acetylation in cellular differentiation
Acetylation of the lysine residues within the N-terminal
tail of histones brings the chromatin into a more relaxed
state and often promotes gene expression [12]. The level
of histone acetylation is modulated by counteracting
enzymes called histone acetyltransferases (HATs) and
histone deacetylases (HDACs) that incorporate and remove acetyl groups, respectively. In Arabidopsis 13 HAT
genes and 18 HDACs have been described [13], and
recent studies have shown their key roles in cellular
differentiation. In the Arabidopsis root apical meristem
(RAM), histone acetylation level is highest in mitotically
dividing cells and subsequently drops as cells transit to
differentiate [14]. High histone acetylation is important
to maintain the meristematic activity as mutants in the
HAT complex subunits GENERAL CONTROL NONDEREPRESSIBLE 5 (GCN5) and TRANSCRIPTIONAL ADAPTOR 2B (ADA2b) have shorter roots with
smaller RAM [15]. Conversely, pharmacological inhibition of HDAC activity by Trichostatin A delays cell
www.sciencedirect.com
Epigenetic control of cell differentiation in plants Ikeuchi, Iwase and Sugimoto 61
Figure 1
(a)
promotion of
cell differentiation
HDAC
(b)
maintenance of
cell differentiation
PRC2
HAT
WIND3
LEC2
HDA19
TPL
WOX5
CDF4
leaf
guard cell
BP, KNAT2
PRC2
maintenance of
stem cell fate
PRC2
PRC2
?
PRC2
AS1 AS2
promotion of
cell proliferation
maintenance of
cell differentiation
repression of
meristem cell fate
FAMA RBR1
SPCH, MUTE
shoot meristem
(c)
suppression of
cell proliferation
histone acetylation
histone deacetylation
H3K27me3
CYCD4, APC10
DNA methylation
Current Opinion in Plant Biology
Epigenetic regulation of cellular differentiation. (a) At the Arabidopsis root meristem, histone acetylation levels are higher in mitotically dividing cells
compared to post-mitotic differentiating cells. In the columella stem cells, WOX5 recruits HDA19 via its interaction with TPL and represses a
differentiation-inducing factor CDF4 to maintain the stem cell fate. (b) In the Arabidopsis leaves the AS1-AS2 complex maintains the leaf fate by
repressing meristem regulators, BP and KNAT2, through the recruitment of PRC2. In mature guard cells PRC2 represses key determinants of the
precursor fate, SPCH and MUTE, to prevent reinitiation of stomata differentiation. FAMA-RBR may recruit PRC2 to these target loci. (c) In
developing maize leaves, DNA methylation levels increase at the loci of several cell cycle regulators, CYCD4 and APC10, as cells switch from
proliferation to differentiation. Correspondingly, the level of these transcripts decreases in differentiating cells.
differentiation [14], suggesting that controlling the level
of histone acetylation is fundamental for the timely
transition into differentiation (Figure 1a).
The WUSCHEL (WUS) and its close homologs
WUSCHEL-RELATED HOMEOBOX (WOX) transcription factors are the key determinants of stem cell
niche in SAM and RAM, respectively [16,17]. Pi et al.
(2015) have recently reported that WOX5 is transcribed in
the quiescent center cells and moves to adjacent columella stem cells where it represses a differentiationinducing factor CYCLING DOF FACTOR 4 (CDF4)
to keep stem cells undifferentiated [18] (Figure 1a).
Interestingly, WOX5 recruits HDA19 to the CDF4 locus
through its interaction with a co-repressor TOPLESS
(TPL) and suppresses CDF4 expression by deacetylating
histones. The WOX5-mediated deacetylation and CDF4
repression appear to be highly dynamic as this repression
is already abolished after a single round of stem cell
division to allow CDF4 expression in columella cells
[18]. Given that WUS also interacts with TPL [19],
the WUS/WOX-TPL-dependent recruitment of HDACs
might be a general mechanism to maintain stem cell fate
in both shoots and roots.
recent studies have shown that some of auxin-mediated
transcriptional outputs are mediated by histone acetylation and deacetylation. Auxin-responsive genes contain
the AuxRE motif in their promoter sequences that is
bound by auxin response factors (ARFs) and their interacting partners Aux/IAA [21]. In the absence of auxin,
Aux/IAA forms a protein complex with TPL which then
recruits HDA19 to repress ARF-driven gene expression
[22]. As cellular auxin levels increase, Aux/IAA is degraded by the 26S proteosome via the SCFTIR1 pathway, thus
freeing ARF transcription factors to activate target gene
expression [23]. A new study by Weiste and Droge-Laser
(2014) has shown that for a subset of auxin-response genes,
the ARF-mediated transcription is further enhanced
through the GCN5-mediated histone acetylation [24].
The GCN5 complex is recruited to these ARF target
genes by group S1 bZIP transcription factors that recognise the G-box related cis-elements (GRE motif) in their
promoter sequences. Developmental roles of this elegant
epigenetic switch have yet to be elucidated but it would be
interesting to see how this mechanism controls auxindependent cell differentiation and other auxin-mediated
developmental transitions.
Histone methylation in cellular differentiation
The plant hormone auxin forms concentration gradients
along plant bodies, thereby providing positional cues to
promote cell proliferation and differentiation [20]. Several
www.sciencedirect.com
Several lysine residues in the N-terminal tails of Histone
H3 can be methylated and affect gene expression either
positively or negatively, depending on the position and
Current Opinion in Plant Biology 2015, 28:60–67
62 Cell biology
number of methyl groups added to each lysine residue.
Trimethylation of Histone H3 at lysine 4 (H3K4me3) is
typically associated with active gene expression while
trimethylation at lysine 27 (H3K27me3) is linked with
gene repression [25,26]. Several methyltransferases that
undertake H3K4me3 have been identified and SET DOMAIN GROUP 2 (SDG2) is responsible for H3K4me3 in
vast majority of genes [27]. Loss-of-function mutants in
SDG2 display pleiotropic developmental phenotypes and
some of these defects, such as reduced leaf and root
growth, are linked with impaired cell differentiation
[27–29]. ARABIDOPSIS TRITHORAX 1 (ATX1)-mediated H3K4me3 has been implicated in the control of
flower development [30,31] and recent studies have uncovered its additional function in the establishment of
organ polarity and root development [32,33]. These observations thus suggest that both SDG2 and ATX1 might
play broad regulatory roles in cellular differentiation.
The H3K27me3 mark is incorporated by a Polycomb
group (PcG) protein complex called POLYCOMB REPRESSIVE COMPLEX2 (PRC2), which together with
PRC1 establishes and maintains the repressive chromatin
state [9,34–37]. As in animals, plant PRC2 is composed of
four subunits and three different forms of PRC2 function
at different developmental stages [38]. The H3K27me3
mark is enriched in genes with tissue-specific expression
profiles [39], and differential methylation patterns between shoot apices and leaves are associated with tissuespecific gene expression [34], suggesting the functional
importance of PcG-mediated gene repression in cellular
differentiation. Several lines of evidence indeed demonstrate that PcG-dependent gene repression is pivotal in
preventing the re-acquisition of stem cell fate in determinate lateral organs in shoots. The KNOTTED-1 like
homeobox class I (KNOXI) genes such as SHOOT MERISTEMLESS (STM), BREVIPEDICELLUS (BP) and
KNOTTED-LIKE FROM ARABIDOPSIS THALIANA 2
(KNAT2) establish and maintain SAM in both monocot
and dicot plants [40]. A study by Lodha et al. (2013)
demonstrated that ASYMMETRIC LEAVES 1 (AS1)
and AS2 recruit PRC2 to the KNOXI genes to repress
their expression outside the meristem [41] (Figure 1b).
When this recruitment is abolished, as found in the maize
mutant rough sheath 2 (rs2) defective in the maize AS1
homolog, KNOX genes are ectopically expressed in developing leaves [42]. Similarly, Arabidopsis PRC1
mutants atring1a atring1b misexpress KNOX1 genes and
develop ectopic SAM in leaves [43]. These results therefore highlight how PcG-mediated histone methylation
prevents attainment of stem cell fate in lateral organs.
PcG-dependent suppression of WUS, which also acts as a
stem cell regulator in flowers, is pivotal for the timely
termination of stem cell activity in floral meristems that
exhibit determinate growth [44]. A floral homeotic regulator AGAMOUS (AG) plays dual roles in this repression
Current Opinion in Plant Biology 2015, 28:60–67
and at an early stage AG first recruits PcG to the WUS
locus to repress its expression [45]. At later stages AG in
turn targets the KNUCKLE (KNU) locus and activates its
expression by competing off PcG that shares the same cisregulatory elements in the KNU promoter [46,47]. Interestingly, H3K27me3 marks at KNU are diluted after
two rounds of cell division to allow the KNU expression
and consequently to shut down the WUS expression
completely [47].
PcG-mediated repression is also required for the maintenance of cellular differentiation in some lineage-committed cells. During stomata development precursor cells
undergo scheduled asymmetric division, fate change and
symmetric division to generate a pair of guard cells [48].
Each step of these sequential differentiation processes is
governed by the bHLH transcription factors SPEECHLESS (SPCH), MUTE and FAMA [49–51]. Two recent
studies have reported that the final differentiated status of
stomata requires active maintenance and failure in this
leads to the reiteration of stomata differentiation within
already existing guard cells. Lee et al. (2014) discovered
that the expression of FAMA-GREEN FLUORESCENT PROTEIN (GFP) fusion protein from its own
promoter causes the ‘stomata-in-stomata’ phenotype
[52]. This phenotype is associated with the heterochronic expression of SPCH and MUTE, which are normally
suppressed by PRC2 at late stages of wild-type stomata
development. The authors further showed that the stomata-in-stomata phenotype can be rescued by overexpressing a subunit of PRC2, CURLY LEAF (CLF),
suggesting that PRC2-dependent suppression of SPCH
and MUTE prevents the reinitiation of stomata differentiation. Matos et al. (2014) found that FAMA interacts
with RETINOBLASTOMA-RELATED (RBR) via a
canonical RBR binding motif (LxCxE) and amino acid
substitutions in this motif cause strong stomata-in-stomata phenotypes [53]. These phenotypes are also accompanied by the ectopic expression of several early stomata
regulators including SPCH and MUTE. Interestingly, ectopic expression of SPCH alone does not mimic the
stomata-in-stomata phenotypes, suggesting that FAMARBR controls gene expression more broadly. Combined
with previous results suggesting tight links between RBR
and PRC2 [54], one plausible hypothesis would be that
FAMA-RBR recruits PRC2 to target loci to block the
reversion of terminal differentiated state in guard cells
(Figure 1b).
An important question is whether other cell lineages also
require similar mechanisms to maintain the differentiated
state, and our recent study indeed demonstrates the
requirement of PcG-mediated gene repression to prevent
reprogramming of fully differentiated root cells [55].
The null PRC2 mutants, such as fie, clf-28 swn-7 and emf23 vrn2-1 [8,9], initially develop relatively normal roots
and, for instance, form root hairs indistinguishable from
www.sciencedirect.com
Epigenetic control of cell differentiation in plants Ikeuchi, Iwase and Sugimoto 63
Histone methylation in cellular
reprogramming
Figure 2
PcG-mediated gene repression plays important roles during cellular reprogramming in in vitro tissue culture.
Arabidopsis shoot or root explants form callus when
incubated on the auxin-rich callus inducing media
(CIM) [59], and this process is governed by a genetic
pathway underlying lateral root development [60,61].
Interestingly, PRC2 is required for callus formation from
leaf explants but not from roots [62], suggesting that
repressing leaf traits is the prerequisite for callus formation from leaves. The leaf to callus transition is accompanied by the acquisition of H3K27me3 at 186 genes, some
of which encode known regulators of leaf development
such as SAWTOOTH 1 (SAW1). This transition is also
characterised by the loss of H3K27me3 at 434 genes
including those that encode auxin regulators such as
GH3.2. These observations support the idea that the leaf
to callus transition requires both the suppression of leaf
identity and the activation of auxin pathway (Figure 3a)
although exact targets responsible for this reprogramming
need to be identified in future studies.
(a)
(b)
DNA methylation in cellular differentiation
Current Opinion in Plant Biology
2 PRC2-mediated gene repression is required for the maintenance of
cellular differentiation.
Arabidopsis PRC2 mutants first develop normal root hairs
indistinguishable from wild-type but fail to maintain their differentiated
status, resulting in the callus formation from single root hairs. (a) Wildtype roots (left) and emf2-3 vrn2-1 roots with callus forming hairs
(right). (b) Fluorescence microscopy of DAPI-stained nuclei shows
multicellular root hairs in fie. Bars = 0.5 mm (a), 0.1 mm (b).
wild-type. Later in development these root hairs, which
undergo several rounds of endocycle and increase ploidy
up to 16C [56], revert back to the mitotic cell cycle and
turn into multicellular callus, some of which subsequently
generate somatic embryos (Figure 2). In PRC2 mutants,
several reprogramming regulators, such as WOUND-INDUCED DEDIFFERENTIATION 3 (WIND3) [57], and
embryonic regulators, such as LEAFY COTYLEDON 2
(LEC2) [58], are ectopically expressed, and their overexpression phenocopies multicellular root hairs in wildtype plants, indicating that PcG-mediated suppression of
these regulators is essential to maintain the differentiated
state (Figure 1a). This study also reports dedifferentiation
of other root cells, thus demonstrating the general requirement of this epigenetic regulation in roots.
www.sciencedirect.com
Methylation of cytosine in DNA maintains genome integrity and regulates gene expression. In plant genomes,
methylated cytosine is found in any sequence context,
that is, CG, CHG and CHH (H = A, C, or T). DNA
methylation in promoter regions generally has repressive
effects on gene expression but how gene body methylation impacts gene expression appears more complex. For
instance, a tissue-specific methylome analysis using Arabidopsis flowers has associated DNA methylation in the
first exon with gene repression but CG methylation in the
following exon with gene activation [63]. The role of
DNA methylation during plant embryogenesis and floral
development is well documented [64,65] but whether it
plays instructive roles in cell proliferation or differentiation remains elusive. A genome-wide study that investigated the developmentally regulated DNA methylation
in maize leaves uncovered remarkable alterations of DNA
methylation patterns as cells transitioned from the proliferative to differentiation stage [66]. This study identified
a subset of cell cycle genes, such as CYCLIN D4 (CYCD4)
and ANAPHASE-PROMOTING COMPLEX 10 (APC10),
which become both de novo DNA methylated and downregulated in differentiating cells (Figure 1c). Whether
these DNA modifications are causal for cell differentiation needs to be further explored, but these observations
may add another layer of epigenetic regulation in the
control of cell differentiation.
A recent study by Yamamuro et al. (2014) provides evidence that the expression of key regulators in stomata
differentiation is controlled by DNA methylation [67].
Genes targeted by DNA methylation include EPIDERMAL PATTERNING FACTOR 2 (EPF2) which encodes a
Current Opinion in Plant Biology 2015, 28:60–67
64 Cell biology
Figure 3
(a)
callus formation
CIM
repression of leaf cell fate
PRC2
SAW1
SAW1
promotion of auxin signalling
PRC2
GH3.2
GH3.2
shoot
regeneration
(b)
CIM
SIM
MET1
WUS
promotion of auxin signalling
MET1
ARF3
DNA methylation in cellular reprogramming
Stabilised gene repression mediated by DNA methylation contributes to the maintenance of cellular identity in
normal plant development but it interferes with shoot
regeneration from Arabidopsis root explants. Mutants
defective in DNA METHYLTRANSFERASE1
(MET1) display enhanced efficiency of shoot regeneration on shoot inducing media (SIM), and this phenotype
is associated with accelerated WUS induction [69]. The
WUS locus is marked with DNA methylation in wild-type
root explants but its methylation levels decrease after
transfer to SIM to allow its expression. In addition, many
auxin signaling regulators such as AUXIN RESPONSE
FACTOR3 (ARF3) are repressed by MET1-dependent
DNA methylation and released from this repression on
SIM to permit their expression and thus shoot regeneration (Figure 3b).
Conclusions and future perspectives
promotion of shoot meristem formation
WUS
peptide hormone responsible for stomata patterning.
DNA demethylation mediated by REPRESSOR OF
SILENCING 1 (ROS1), DEMETER-LIKE 2 (DML2)
and DML39 is required for the correct EPF2 expression
and thus patterning of stomata cells in Arabidopsis leaves.
DNA methylation also regulates the expression of SPCH
and FAMA, both of which are methylated de novo under
low humidity conditions where it is beneficial for plants to
make less stomata [68].
ARF3
H3K27me3
DNA methylation
Current Opinion in Plant Biology
Epigenetic regulation of cellular reprogramming. (a) Incubation of
Arabidopsis leaf explants on auxin-rich callus inducing media (CIM)
promotes callus formation. PRC2 is required for this process to
incorporate H3K27me3 marks to leaf development genes such as
SAW1 and thereby repress leaf cell fate. The leaf to callus transition is
also associated with the loss of H3K27me3 at several auxin signaling
genes such as GH3.2. (b) Successive incubation of Arabidopsis root
explants on CIM and shoot inducing media (SIM) induces DNA
demethylation of WUS, a key stem cell regulator of SAM, as well as
auxin signal components such as ARF3.
Current Opinion in Plant Biology 2015, 28:60–67
Recent research has made substantial progress in unveiling how epigenetic regulation is intertwined with genetic
transcriptional regulation to assist faithful progression of
plant development. Cellular differentiation is instructed
by the action of key transcriptional regulators, many of
which are under epigenetic control to spatially and temporally regulate their expression during development. It
is also now evident that some of these transcription factors
recruit or evict epigenetic regulators to activate or repress
target gene expression through locus-specific chromatin
modifications. These multi-layered mechanisms that link
epigenetic and genetic regulations likely serve as a safeguard to control developmental stage-specific gene expression. The discoveries that these mechanisms are still
needed even after cells fully differentiate highlight the
striking plasticity of plant cells.
Epigenomic studies in Arabidopsis and maize show that
both histone modification and DNA methylation profiles
undergo dynamic genome-wide changes as cells differentiate. These studies also highlight the complexity of
these modifications, and one of the major challenges in
future studies will be to understand how plant cells
decode various types of epigenetic modifications and
translate these signals into single transcriptional outputs.
The answer to these questions may come from more
advanced methodology for studying chromatin structures,
for example using and Hi-C seq [70] and ATAC-seq [71],
www.sciencedirect.com
Epigenetic control of cell differentiation in plants Ikeuchi, Iwase and Sugimoto 65
by which various combinations of epigenetic modifications can be linked to the three-dimensional structure and
overall accessibility of chromatin. Following epigenetic
changes at higher cellular resolutions, for example, using
single cells or single cell types in plant tissues, will also be
a key to untangle the causal relationships between chromatin modification and cell differentiation.
Acknowledgements
We thank Bart Rymen, Anna Franciosini, Natalie Clark and Lewis Watt for
providing comments on the manuscript. Research in the authors’ laboratory
on the topic of this review is supported by a grant from Scientific
Technique Research Promotion Program for Agriculture, Forestry,
Fisheries and Food Industry and grants from the Ministry of Education,
Culture, Sports and Technology of Japan to MI (15K18564), AI (15K18565)
and to KS (26291064, 15H05961). MI is a recipient of the RIKEN Special
Postdoctoral Researcher Program.
References and recommended reading
Papers of particular interest, published within the period of review,
have been highlighted as:
of special interest
of outstanding interest
1.
Laux T: The stem cell concept in plants: a matter of debate. Cell
2003, 113:281-283.
2.
Sinha N: Leaf development in angiosperms. Ann Rev Plant
Physiol Plant Mol Biol 1999, 50:419-446.
3.
Ikeuchi M, Sugimoto K, Iwase A: Plant callus: mechanisms of
induction and repression. Plant Cell 2013, 25:3159-3173.
4.
La Rota C, Chopard J, Das P, Paindavoine S, Rozier F, Farcot E,
Godin C, Traas J, Monéger F: A data-driven integrative model of
sepal primordium polarity in Arabidopsis. Plant Cell 2011,
23:4318-4333.
14. Rosa S, Ntoukakis V, Ohmido N, Pendle A, Abranches R, Shaw P:
Cell differentiation and development in Arabidopsis are
associated with changes in histone dynamics at the single-cell
level. Plant Cell 2014, 26:4821-4833.
The authors examined the nucleosome dynamics by FRAP (Fluorescent
Recovery After Photobleaching) analysis in roots. They found that nucleosomes in mitotic cells are more dynamic than those in post-mitotic cells.
Mitotic cells have higher global histone acetylation, which was indeed shown
by pharmacological analysis to be instructive for nucleosome dynamism.
15. Kornet N, Scheres B: Members of the GCN5 histone
acetyltransferase complex regulate PLETHORA-mediated
root stem cell niche maintenance and transit amplifying cell
proliferation in Arabidopsis. Plant Cell 2009, 21:1070-1079.
16. Mayer KF, Schoof H, Haecker A, Lenhard M, Jürgens G, Laux T:
Role of WUSCHEL in regulating stem cell fate in the
Arabidopsis shoot meristem. Cell 1998, 95:805-815.
17. Sarkar AK, Luijten M, Miyashima S, Lenhard M, Hashimoto T,
Nakajima K, Scheres B, Heidstra R, Laux T: Conserved factors
regulate signalling in Arabidopsis thaliana shoot and root
stem cell organizers. Nature 2007, 446:811-814.
18. Pi L, Aichinger E, van der Graaff E, Llavata-Peris CI, Weijers D,
Hennig L, Groot E, Laux T: Organizer-derived WOX5 signal
maintains root columella stem cells through chromatinmediated repression of CDF4 expression. Dev Cell 2015, 8:576588.
In this paper the authors nicely demonstrate how stem cell niche maintains adjacent stem cells. They identified a novel WOX5 target, CDF4,
which promotes columella cell differentiation. Similar to wox5 mutants,
ectopically activated CDF4 abolishes columella stem cell fate in tpl or
hda19 mutants. Biochemical evidence shows that TPR1 and HDA19 are
associated with the CDF4 promoter in the WOX5-dependent manner,
suggesting that niche cell-derived WOX5 represses a differentiationinducing factor CDF4 via HDAC recruitment.
19. Kieffer M, Stern Y, Cook H, Clerici E, Maulbetsch C, Laux T,
Davies B: Analysis of the transcription factor WUSCHEL and its
functional homologue in Antirrhinum reveals a potential
mechanism for their roles in meristem maintenance. Plant Cell
2006, 18:560-573.
20. Sabatini S, Beis D, Wolkenfelt H, Murfett J, Guilfoyle T, Malamy J,
Benfey P, Leyser O, Bechtold N, Weisbeek P et al.: An auxindependent distal organizer of pattern and polarity in the
Arabidopsis root. Cell 1999, 99:463-472.
5.
Bruex A, Kainkaryam RM, Wieckowski Y, Kang YH: A gene
regulatory network for root epidermis cell differentiation in
Arabidopsis. PLoS Genet 2012 http://dx.doi.org/10.1371/
journal.pgen.1002446.s018.
6.
Taylor-Teeples M, Lin L, de Lucas M, Turco G, Toal TW,
Gaudinier A, Young NF, Trabucco GM, Veling MT, Lamothe R et al.:
An Arabidopsis gene regulatory network for secondary cell
wall synthesis. Nature 2014, 517:571-575.
22. Long JA, Ohno C, Smith ZR, Meyerowitz EM: TOPLESS regulates
apical embryonic fate in Arabidopsis. Science 2006, 312:15201523.
7.
Goodrich J, Puangsomlee P, Martin M, Long D, Meyerowitz EM,
Coupland G: A Polycomb-group gene regulates homeotic gene
expression in Arabidopsis. Nature 1997, 386:44-51.
23. Gray WM, Kepinski S, Rouse D, Leyser O, Estelle M: Auxin
regulates SCF(TIR1)-dependent degradation of AUX/IAA
proteins. Nature 2001, 414:271-276.
8.
Chanvivattana Y, Bishopp A, Schubert D, Stock C, Moon Y-H,
Sung ZR, Goodrich J: Interaction of Polycomb-group proteins
controlling flowering in Arabidopsis. Development 2004,
131:5263-5276.
9.
Bouyer D, Roudier F, Heese M, Andersen ED, Gey D, Nowack MK,
Goodrich J, Renou J-P, Grini PE, Colot V: Polycomb repressive
complex 2 controls the embryo-to-seedling phase transition.
PLoS Genet 2011, 7:e1002014.
24. Weiste C, Dröge-Laser W: The Arabidopsis transcription factor
bZIP11 activates auxin-mediated transcription by recruiting
the histone acetylation machinery. Nat Commun 2014, 5:3883.
In this paper the authors reveal histone acetylation regulates expression
of auxin-responsive genes. They show that bZIP11 and other related
bZIPs recruit a SAGA-like complex via interaction with ADA2b. Furthermore, bZIP11 activates the GH3.3 expression in the GRE elementdependent manner. The proposed model suggests that AuxRE is a
qualitative cis-element where HDAC is recruited to block gene expression
under low auxin conditions, and GRE is a quantitative cis-element to
boost gene expression under high auxin conditions.
10. Heo JB, Lee Y-S, Sung S: Epigenetic regulation by long
noncoding RNAs in plants. Chromosome Res 2013, 21:685-693.
11. Baulcombe DC, Dean C: Epigenetic regulation in plant
responses to the environment. Cold Spring Harb Perspect Biol
2014, 6:a019471.
12. Struhl K: Histone acetylation and transcriptional regulatory
mechanisms. Genes Dev 1998, 12:599-606.
13. Pandey R, Müller A, Napoli CA, Selinger DA, Pikaard CS,
Richards EJ, Bender J, Mount DW, Jorgensen RA: Analysis of
histone acetyltransferase and histone deacetylase families of
Arabidopsis thaliana suggests functional diversification of
chromatin modification among multicellular eukaryotes.
Nucleic Acids Res 2002, 30:5036-5055.
www.sciencedirect.com
21. Guilfoyle TJ, Hagen G: Auxin response factors. Curr Opin Plant
Biol 2007, 10:453-460.
25. Zhang X, Bernatavichute YV, Cokus S, Pellegrini M, Jacobsen SE:
Genome-wide analysis of mono-, di- and trimethylation of
histone H3 lysine 4 in Arabidopsis thaliana. Genome Biol 2009,
10:R62.
26. Roudier FCO, Ahmed I, rard CBE, Sarazin A, Mary-Huard T,
Cortijo S, Bouyer D, Caillieux E, Duvernois-Berthet E, Al-Shikhley L
et al.: Integrative epigenomic mapping defines four main
chromatin states in Arabidopsis. EMBO J 2011, 30:1928-1938.
27. Guo L, Yu Y, Law JA, Zhang X: SET DOMAIN GROUP2 is the
major histone H3 lysine 4 trimethyltransferase in Arabidopsis.
Proc Natl Acad Sci U S A 2010, 107:18557-18562.
Current Opinion in Plant Biology 2015, 28:60–67
66 Cell biology
28. Berr A, McCallum EJ, Ménard R, Meyer D, Fuchs J, Dong A,
Shen W-H: Arabidopsis SET DOMAIN GROUP2 is required for
H3K4 trimethylation and is crucial for both sporophyte and
gametophyte development. Plant Cell 2010, 22:3232-3248.
29. Yao X, Feng H, Yu Y, Dong A, Shen W-H: SDG2-mediated H3K4
methylation is required for proper Arabidopsis root growth
and development. PLOS ONE 2013, 8:e56537.
30. Alvarez-Venegas R, Pien S, Sadder M, Witmer X, Grossniklaus U,
Avramova Z: ATX-1 an Arabidopsis homolog of trithorax.
Activates flower homeotic genes. Curr Biol 2003, 13:627-637.
31. Carles CC, Fletcher JC: The SAND domain protein
ULTRAPETALA1 acts as a trithorax group factor to regulate
cell fate in plants. Genes Dev 2009, 23:2723-2728.
32. Pires HR, Monfared MM, Shemyakina EA, Fletcher JC:
ULTRAPETALA trxG genes interact with KANADI transcription
factor genes to regulate Arabidopsis gynoecium patterning.
Plant Cell 2014, 26:4345-4361.
33. Napsucialy-Mendivil S, Alvarez-Venegas R, Shishkova S,
Dubrovsky JG: Arabidopsis homolog of trithorax1 (ATX1) is
required for cell production, patterning, and morphogenesis in
root development. J Exp Bot 2014, 65:6373-6384.
34. Lafos M, Kroll P, Hohenstatt ML, Thorpe FL, Clarenz O,
Schubert D: Dynamic regulation of H3K27 trimethylation during
Arabidopsis differentiation. PLoS Genet 2011, 7:e1002040.
35. Beh LY, Colwell LJ, Francis NJ: A core subunit of Polycomb
repressive complex 1 is broadly conserved in function but not
primary sequence. Proc Natl Acad Sci U S A. 2012, 109:E1063E1071.
36. Kim S-Y, Lee J, Eshed-Williams L, Zilberman D: EMF1 and PRC2
cooperate to repress key regulators of Arabidopsis
development. PLoS Genet 2012 http://dx.doi.org/10.1371/
journal.pgen.1002512.t002.
37. Yang C, Bratzel F, Hohmann N, Koch M, Turck F, Calonje M: VAL-:
AtBMI1-mediated H2Aub initiate the switch from embryonic to
postgerminative growth in Arabidopsis. Curr Biol 2013,
23:1324-1329.
38. Holec S, Berger F: Polycomb group complexes mediate
developmental transitions in plants. Plant Physiol 2012, 158:3543.
39. Zhang X, Clarenz O, Cokus S, Bernatavichute YV, Pellegrini M,
Goodrich J, Jacobsen SE: Whole-genome analysis of histone
H3 lysine 27 trimethylation in Arabidopsis. PLoS Biol 2007,
5:e129.
40. Hay A, Tsiantis M: KNOX genes: versatile regulators of plant
development and diversity. Development 2010, 137:3153-3165.
41. Lodha M, Marco CF, Timmermans MCP: The ASYMMETRIC
LEAVES complex maintains repression of KNOX homeobox
genes via direct recruitment of Polycomb-repressive
complex2. Genes Dev 2013, 27:596-601.
The AS1-AS2 complex has long been suggested to be a epigenetic
repressor of KNOX1 genes. The authors show that the AS complex
directly recruits PRC2 to the KNOXI loci to repress their expression.
42. Timmermans MC, Hudson A, Becraft PW, Nelson T: ROUGH.
SHEATH2: a Myb protein that represses knox homeobox
genes in maize lateral organ primordia. Science 1999, 284:151153.
43. Xu L, Shen W-H: Polycomb silencing of KNOX genes confines
shoot stem cell niches in Arabidopsis. Curr Biol 2008, 18:19661971.
44. Sun B, Ito T: Regulation of floral stem cell termination in
Arabidopsis. Front Plant Sci 2015, 6:17.
45. Liu X, Kim YJ, Muller R, Yumul RE, Liu C, Pan Y, Cao X, Goodrich J,
Chen X: AGAMOUS terminates floral stem cell maintenance in
Arabidopsis by directly repressing WUSCHEL through
recruitment of Polycomb group proteins. Plant Cell 2011,
23:3654-3670.
46. Sun B, Xu Y, Ng K-H, Ito T: A timing mechanism for stem cell
maintenance and differentiation in the Arabidopsis floral
meristem. Genes Dev 2009, 23:1791-1804.
Current Opinion in Plant Biology 2015, 28:60–67
47. Sun B, Looi LS, Guo S, He Z, Gan ES, Huang J, Xu Y, Wee WY,
Ito T: Timing mechanism dependent on cell division is invoked
by Polycomb eviction in plant stem cells. Science 2014,
343:1248559.
The authors previously reported the timelag between the onset of AG
expression and KNU expression. In this work they demonstrate that the
observed delay is dependent on cycles of cell division. In addition, they
found that FIE binding to the KNU promoter is reduced by AG which
shares the binding sites with FIE. Based on these results, they hypothesized the epigenetic timer model and further confirmed this model by
synthetic approaches.
48. Pillitteri LJ, Torii KU: Mechanisms of stomatal development.
Annu Rev Plant Biol 2012, 63:591-614.
49. MacAlister CA, Ohashi-Ito K, Bergmann DC: Transcription factor
control of asymmetric cell divisions that establish the
stomatal lineage. Nature 2007, 445:537-540.
50. Pillitteri LJ, Sloan DB, Bogenschutz NL, Torii KU: Termination of
asymmetric cell division and differentiation of stomata. Nature
2007, 445:501-505.
51. Ohashi-ito K, Bergmann DC: Arabidopsis FAMA controls the
final proliferation/differentiation switch during stomatal
development. Plant Cell 2006, 18:2493-2505.
52. Lee E, Lucas JR, Goodrich J, Sack FD: Arabidopsis guard cell
integrity involves the epigenetic stabilization of the FLP and
FAMA transcription factor genes. Plant J 2014, 78:566-577.
The authors find the stomata-in-stomata phenotype in transgenic plants
expressing FAMA cDNA or FAMA-GFP under the control of the FAMA
promoter (proFAMA-cFAMA or proFAMA-cFAMA-GFP). The phenotype
is not recapitulated by expressing the FAMA genomic fragment and the
exact reason why these transgenes induce the stomata-in-stomata
phenotype remains unsolved. They found that misexpression of SPCH
and MUTE in proFAMA-cFAMA plants is associated with the loss of
H3K27me3, revealing that in wild-type plants these genes are repressed
by a PcG-mediated mechanism.
53. Matos JL, Lau OS, Hachez C, Cruz-Ramirez A, Scheres B,
Bergmann DC: Irreversible fate commitment in the Arabidopsis
stomatal lineage requires a FAMA and retinoblastoma-related
module. eLife 2014 http://dx.doi.org/10.7554/eLife.03271.019.
In this paper the authors demonstrate that the FAMA-RBR interaction in
young guard cells is required to prevent the fate reversal back to
meristemoid mother cells.
54. Kuwabara A, Gruissem W: Arabidopsis Retinoblastoma-related
and Polycomb group proteins: cooperation during plant cell
differentiation and development. J Exp Bot 2014, 65:2667-2676.
55. Ikeuchi M, Iwase A, Rymen B, Harashima H, Shibata M,
Ohnuma M, Breuer C, Morao AK, de Lucas M, De Veylder L et al.:
PRC2 represses dedifferentiation of mature somatic cells in
Arabidopsis. Nat Plants 2015 http://dx.doi.org/10.1038/
NPLANTS.2015.89.
The authors find striking phenotypes, such as multicellularisation, callus
formation and embryogenesis, in root hairs of PRC2 mutants. The mutant
root hairs initially complete normal differentiation program including
endocycles and cell expansion and subsequently turn multicellular,
indicating that PRC2 maintains the differentiated status of mature root
cells. Furthermore, they show that reprogramming factors such as WIND3
and LEC2 are among the PRC2 targets relevant in the observed cellular
reprogramming.
56. Sugimoto-Shirasu K, Roberts GR, Stacey NJ, Mccann MC,
Maxwell A, Roberts K: RHL1 is an essential component of the
plant DNA topoisomerase VI complex and is required for
ploidy-dependent cell growth. Proc Natl Acad Sci U S A 2005,
102:18736-18741.
57. Iwase A, Mitsuda N, Koyama T, Hiratsu K, Kojima M, Arai T,
Inoue Y, Seki M, Sakakibara H, Sugimoto K et al.: The AP2/ERF
transcription factor WIND1 controls cell dedifferentiation in
Arabidopsis. Curr Biol 2011, 21:508-514.
58. Stone SL, Kwong LW, Yee KM, Pelletier J, Lepiniec L, Fischer RL,
Goldberg RB, Harada JJ: LEAFY COTYLEDON2 encodes a B3
domain transcription factor that induces embryo
development. Proc Natl Acad Sci U S A 2001, 98:11806-11811.
59. Valvekens D, Montagu MV, Van Lijsebettens M: Agrobacterium
tumefaciens-mediated transformation of Arabidopsis thaliana
www.sciencedirect.com
Epigenetic control of cell differentiation in plants Ikeuchi, Iwase and Sugimoto 67
root explants by using kanamycin selection. Proc Natl Acad Sci
U S A 1988, 85 5536–5540.46.
60. Atta R, Laurens L, Boucheron-Dubuisson E, Guivarc’h A,
Carnero E, Giraudat-Pautot V, Rech P, Chriqui D: Pluripotency of
Arabidopsis xylem pericycle underlies shoot regeneration
from root and hypocotyl explants grown in vitro. Plant J 2009,
57:626-644.
61. Sugimoto K, Jiao Y, Meyerowitz EM: Arabidopsis regeneration
from multiple tissues occurs via a root development pathway.
Dev Cell 2010, 18:463-471.
62. He C, Chen X, Huang H, Xu L: Reprogramming of H3K27me3 is
critical for acquisition of pluripotency from cultured
Arabidopsis tissues. PLoS Genet 2012, 8:e1002911.
63. Yang H, Chang F, You C, Cui J, Zhu G, Wang L, Zheng Y, Qi J,
Ma H: Whole-genome DNA methylation patterns and complex
associations with gene structure and expression during
flower development in Arabidopsis. Plant J 2015, 81:268-281.
The authors present base-resolution, tissue-specific DNA methylation
profiles during flower formation. They found that overall methylation level
is increased during flower formation from meristems.
64. Choi Y, Gehring M, Johnson L, Hannon M, Harada JJ,
Goldberg RB, Jacobsen SE, Fischer RL: DEMETER, a DNA
glycosylase domain protein, is required for endosperm gene
imprinting and seed viability in Arabidopsis. Cell 2002, 110:3342.
65. Hsieh T-F, Ibarra CA, Silva P, Zemach A, Eshed-Williams L,
Fischer RL, Zilberman D: Genome-wide demethylation of
Arabidopsis endosperm. Science 2009, 324:1451-1454.
www.sciencedirect.com
66. Candaele J, Demuynck K, Mosoti D, Beemster GTS, Inzé D,
Nelissen H: Differential methylation during maize leaf growth
targets developmentally regulated genes. Plant Physiol 2014,
164:1350-1364.
The authors examine dynamic alterations of DNA methylation in developing maize leaves. In dividing cells DNA methylation is maintained by
DNA methytransferases but this pattern is dynamically modified as cells
transit to differentiate.
67. Yamamuro C, Miki D, Zheng Z, Ma J, Wang J, Yang Z, Dong J,
Zhu J-K: Overproduction of stomatal lineage cells in
Arabidopsis mutants defective in active DNA demethylation.
Nat Commun 2014, 5:4062.
68. Tricker PJ, Gibbings JG, Rodrı́guez López CM, Hadley P,
Wilkinson MJ: Low relative humidity triggers RNA-directed de
novo DNA methylation and suppression of genes controlling
stomatal development. J Exp Bot 2012, 63:3799-3813.
69. Li W, Liu H, Cheng ZJ, Su YH, Han HN, Zhang Y, Zhang XS: DNA
methylation and histone modifications regulate de novo shoot
regeneration in Arabidopsis by modulating WUSCHEL
expression and auxin signaling. PLoS Genet 2011, 7:e1002243.
70. Dixon JR, Jung I, Selvaraj S, Shen Y, Antosiewicz-Bourget JE,
Lee AY, Ye Z, Kim A, Rajagopal N, Xie W et al.: Chromatin
architecture reorganization during stem cell differentiation.
Nature 2015, 518:331-336.
71. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ:
Transposition of native chromatin for fast and sensitive
epigenomic profiling of open chromatin, DNA-binding
proteins and nucleosome position. Nat Methods 2013, 10:12131218.
Current Opinion in Plant Biology 2015, 28:60–67