DNA methylation at the fifth position of cytosine is a major epigenetic mark conserved in plants and mammals. Genome-wide DNA methylation patterns are dynamically controlled by integrated activities of establishment, maintenance, and removal. In both plants and mammals, a pattern of global DNA hypomethylation coupled with increased methylation levels at some specific genomic regions arises at specific developmental stages and in certain abnormal cells, such as mammalian aging cells and cancer cells as well as some plant epigenetic mutants. Here we provide an overview of this distinct DNA methylation pattern in mammals and plants, and propose that a methylstat, which is a cis-element responsive to both DNA methylation and active demethylation activities and controlling the transcriptional activity of a key DNA methylation regulator, can help to explain the enigmatic DNA methylation patterns in aging cells and cancer cells.

DNA methylation at the fifth position of cytosine is a major epigenetic mark present in plants and mammals. Through regulation of transcription activities and genome stability, 5-mC DNA methylation is involved in many important biological processes, such as fruit ripening in plants and aging in humans [1,2]. Both plants and mammals use S-adenosyl-methionine as the methyl donor for DNA methylation that is catalyzed by DNA methyltransferases with conserved catalytic domains. The landscape of DNA methylome is dynamically shaped by integrated activities of establishment, maintenance, and removal [1,2]. In both plants and mammals, alterations in genomic DNA methylation patterns can be triggered by intrinsic developmental programs and by certain environmental factors. For instance, aging in mammals is accompanied by a general loss of DNA methylation particularly at repeat sequences and transposable elements on the autosomal chromosomes [3], while a recent study reported a trend of acceleration in Y-chromosomal DNA methylation with increasing age [4]. Besides, locus-specific DNA hypermethylation and the concomitant gene silencing are being increasingly recognized as potential biomarkers for diseases such as cancer [5,6].

In mammals, DNMT3 (DNA methyltransferase 3) family members catalyze de novo DNA methylation [7]. Mammalian piRNAs, which are small RNAs of 25–30 nucleotides, are thought to pair with nascent transcripts and recruit de novo DNA methyltransferases [8]. In another proposed model, DNMT3A is activated or recruited to its target loci via protein interaction with DNMT3L, a noncatalytic DNMT3 paralog that binds unmethylated histone 3 lysine 4 (H3K4) [9,10]. Once established, mammalian DNA methylation is maintained by DNMT1, which restores hemimethylated DNA to a fully methylated status during DNA replication [7].

Plant DNA methylation occurs in all cytosine contexts and can be established through the RNA-directed DNA methylation (RdDM) pathway, in which complementary pairing between 24-nt siRNAs and nascent scaffold RNAs, together with protein–protein interactions, recruits the protein machinery for DNA methylation [1,11]. Once established, DNA methylation in the CG context is maintained by the DNMT1 ortholog MET1 (Methyltransferase 1), while CHG (H represents A, T, or C) methylation is maintained by CMT2 (Chromomethylase 2) and CMT3 through a reinforcing loop that involves KYP4-mediated histone H3K9 methylation [12]. The asymmetric CHH methylation is maintained through persistent de novo methylation catalyzed either by DRM2 (Domains Rearranged Methylase 2) through the RdDM pathway or by CMT2 that requires the chromatin remodeling protein DDM1 (Decreased DNA Methylation 1) [13].

Active DNA demethylation is accomplished through a base excision repair pathway. In contrast with mammals, which initiate active DNA demethylation with oxidation and/or deamination of 5-mC, plants directly excise the 5-mC base utilizing 5-mC DNA glycosylases [1416].

In some mammals including humans, the DNA methylome undergoes global alterations at certain developmental stages. After fertilization, the zygote experiences genome-wide demethylation in the paternal pronuclei, followed by a replication-dependent passive DNA demethylation in the maternal genome [1618]. At the stage of blastocyst formation, de novo DNA methyltransferases reestablish genomic DNA methylation in both parental origins [16,19]. Global demethylation also occurs in primordial germ cells and is followed by extensive chromatin rearrangement, presumably allowing for epigenetic reprogramming toward totipotency [20]. Incomplete and aberrant DNA methylation reprogramming has been postulated as a cause of the high frequency of cloning failure and of developmental abnormalities in cloned animals [14,21,22].

In mammalian cells, aging is characterized by a general loss of DNA methylation and repressive histone marks in heterochromatic regions [23]. In addition to the global hypomethylation, locus-specific DNA hypermethylation also occurs during the process of aging. The age-dependent hypermethylation often exists in actively transcribed genomic regions, including regulatory genes for differentiation, apoptosis, and transcription [23,24]. Aging-associated genome hypomethylation may result from the gradual reduction in the levels of the DNA methyltransferase DNMT1 [25]. Aging-associated DNA methylome is also subject to the impacts of certain environmental factors such as dietary folate and oxidative stress. Aging with dietary deficiency of folate, which is a key source of the methyl group for DNA methylation, causes not only promoter hypomethylation of a couple of proto-oncogenes but also promoter hypermethylation of some tumor suppressor genes including p53 [26]. Oxidative stress due to mitochondria dysfunction occurs during aging and the stress can be enhanced by environmental toxins such as heavy metals and ozone [27,28]. During oxidative stress, accumulation of reactive oxygen species (ROS) can either directly cause DNA lesion or disrupt cellular metabolism and thereby affect TET-mediated DNA demethylation, leading to alterations in epigenomic landscapes [27,29,30]. The locus-specificity and the reproducibility of aging-associated DNA methylation changes indicate that DNA methylation may play a regulatory role in certain biological processes during aging, instead of simply being a stochastic consequence of cellular deterioration. The gradual changes in DNA methylation during aging have been proposed to be an ‘epigenetic clock' [31,32]. Indeed, in certain cases such as human blood DNA, changes in DNA methylation levels are clearly associated with the aging process, and are so highly reproducible that they can be used for age prediction [33].

Besides aging, cancer cells also generally display not only global hypomethylation at heterochromatic regions, but also loci-specific DNA hypermethylation [34]. Increased methylation of tumor suppressor genes is an early event in many tumors, suggesting that altered DNA methylation patterns could be one of the first detectable neoplastic changes associated with tumorigenesis [35,36]. ctDNA bearing cancer-specific methylation patterns have been investigated as feasible biomarkers in cancers [37]. In both aging cells and cancer cells, DNA hypermethylation commonly occurs at loci that are targeted by Polycomb-group (PcG) proteins [3840]. Because PcG-mediated transcriptional silencing can be antagonized by active histone modifications such as H3K4me3, additional DNA methylation at PcG-targeted loci seems to serve a double-locking role to secure transcriptional silencing. In the min mouse model of cancer that displays tumor at ages of 3–5 months, it was shown that the appearance of cancer can be prevented by consecutive chemical inhibition of DNA methylation starting from birth, but not by treatments that started from 3 months of age [41]. These observations suggest that DNA methylation, possibly through developmental accumulation, contributes to tumorigenesis. In addition, DNA methylation abnormalities at gene promoters in elderly cells phenocopy cancer cells; particularly, aging-associated DNA hypermethylation can occur at the promoter regions of some tumor suppressor genes [24,4244]. Thus cancer formation appears to be tightly correlated with the changes in DNA methylation during the aging process.

Similar to mammals, plants also go through massive alterations in their DNA methylome at specific developmental stages. In Arabidopsis thaliana, both the male and female gamete companion cells undergo DME (DEMETER)-mediated global demethylation, resulting in genome-wide DNA hypomethylation with reinforced CHH methylation at transposons in the endosperm [1]. During ripening, tomato fruits display DML2 (DEMETER Like 2)-mediated progressive DNA demethylation at hundreds of gene loci, many of which are known to regulate fruit ripening [45,46]. Chemical inhibition of DNA methylation causes premature ripening of tomato fruits, whereas a loss of DML2 function results in non-ripening fruits [4547]. Thus the changes in DNA methylome during fruit ripening serve a crucial function.

Genetic disruption of key methylation regulators leads to abnormal DNA methylome in plants. Notably, dysfunction of Arabidopsis MET1 or one of the RdDM core factors results in not only global hypomethylation but also hypermethylation at specific loci across the genome [48,49]. The locus-specific increase in DNA methylation is attributed to the activity of the remaining DNA methyltransferases combined with the reduced gene expression of the DNA demethylase ROS1 (Repressor of Silencing 1), whose promoter region harbors a methylation monitoring sequence (MEMS) [50]. DNA methylation at MEMS positively regulates ROS1 gene transcription through a transcription activation complex that contains the SUVH (Suppressor of Variegation 3–9 Homolog) DNA methylation readers [51,52]. Similar to the observations that changes in DNA methylation levels are highly correlated with age changes [33], DNA methylation level of the MEMS is quantitatively associated with the extent of binding of the SUVHs [51]. ROS1 antagonizes MET1 and RdDM activities at several thousands of genomic regions [49], thereby providing a coordination between DNA methylation and demethylation. Similar to the observations that DNA hypermethylation in aging cells and cancer cells preferentially occurs at regions with H3K27me3, ROS1-targeted regions are enriched with this repressive histone modification [49]. Because the methylation level of MEMS is co-regulated by MET1, the RdDM pathway, and ROS1, this unique element is considered as a methylstat that maintains homeostasis of ROS1-dependent DNA methylation across the genome of Arabidopsis thaliana [1,50]. Methylation-sensitive regulation of demethylase gene expression has also been observed in rice, maize, and A. lyrata [5355]. Thus, the methylstat may be a conserved mechanism for regulating DNA methylation dynamics in plants.

Methylstats may also exist in mammals and contribute to shaping the DNA methylome. In such a scenario, the increased DNA methylation at specific loci in aging cells and cancer cells could result from DNA hypomethylation of a methylstat, which controls the expression of a key regulator of DNA methylation or demethylation reactions. In the methylstat model (Figure 1), DNA hypomethylation in aging cells and cancer cells leads to transcriptional activation of the methylstat-controlled DNA establishment or maintenance enzyme or its regulator; alternatively, DNA hypomethylation in aging cells and cancer cells may lead to transcriptional repression of the methylstat-controlled DNA demethylation enzyme or its regulator.

A model for the altered DNA methylome in aging cells and cancer cells.

Figure 1.
A model for the altered DNA methylome in aging cells and cancer cells.

Regulation of the DNA methylome in mammalian cells may involve a methylstat, which responds to DNA methylation levels and regulates gene expression in cis. The methylstat may contribute to the formation of DNA methylation patterns in aging cells and cancer cells in two possible ways. Left upper panel, aging or tumorigenesis causes DNA hypomethylation and consequently transcription activation of the methylstat. The methylstat controls a positive regulator of DNA methylation, the expression of which results in DNA hypermethylation. Right upper panel, DNA methylation in cis is required for transcriptional activation of the methylstat. The methylstat controls a positive regulator of DNA demethylation, whose expression results in the active removal of DNA methylation. Aging or tumorigenesis causes DNA hypomethylation and consequently transcriptional repression of the methylstat-controlled demethylation regulator. As a result, DNA methylation accumulates in regions that are normally pruned by the demethylation machinery. Methylation at the fifth position of cytosine is denoted by a red letter m.

Figure 1.
A model for the altered DNA methylome in aging cells and cancer cells.

Regulation of the DNA methylome in mammalian cells may involve a methylstat, which responds to DNA methylation levels and regulates gene expression in cis. The methylstat may contribute to the formation of DNA methylation patterns in aging cells and cancer cells in two possible ways. Left upper panel, aging or tumorigenesis causes DNA hypomethylation and consequently transcription activation of the methylstat. The methylstat controls a positive regulator of DNA methylation, the expression of which results in DNA hypermethylation. Right upper panel, DNA methylation in cis is required for transcriptional activation of the methylstat. The methylstat controls a positive regulator of DNA demethylation, whose expression results in the active removal of DNA methylation. Aging or tumorigenesis causes DNA hypomethylation and consequently transcriptional repression of the methylstat-controlled demethylation regulator. As a result, DNA methylation accumulates in regions that are normally pruned by the demethylation machinery. Methylation at the fifth position of cytosine is denoted by a red letter m.

Close modal

Unlike the autosomal chromosomes that show a general hypomethylation pattern during aging, the Y chromosome shows age-dependent increases in CG methylation [4]. It is possible that the Y chromosome is intensively targeted either by methylstat-mediated demethylation that is anti-correlated with aging, or by methylstat-mediated methylation that is positively correlated with aging. While the methylstat does not necessarily locate in the promoter region of the methylation/demethylation factor that it controls, its methylation levels should be quantitatively correlated with the transcription levels of the methylation/demethyaltion factor. This expectation may help with the searching for the methylstat, whose methylation levels decline in an age-dependent manner, as well as for the methylation/demethylation factor that it controls. In addition, abnormal gene expression levels of the methylation/demethylation factor would be common in the aging-related diseases that are associated with aberrant DNA methylation patterns, as well as in the individuals that show abnormal rates of aging. DNA hypermethylation in both aging cells and cancer cells preferentially occurs at PcG-targeted loci [3840]. It would be interesting to determine whether DNA methylation at these loci is pruned by active demethylation, and if so, whether the corresponding demethylation factors themselves are subject to methylation-dependent transcription regulation. An EZH2 (Enhancer of Zeste Homolog 2)-containing PcG complex was reported to recruit the methyltransferases for DNA methylation in cancer cells [56]. Thus it would be also interesting to examine whether DNA methylation negatively regulates the PcG-recruited DNA methylation machinery including the methyltransferases.

Summary
  • Importance of the field: DNA methylation confers epigenetic regulation on many important biological processes. The genome-wide hypomethylation coupled with locus-specific hypermethylation is a hallmark of aging and tumorigenesis. Understanding the dynamic regulation of DNA methylome may help us understand and eventually manage aging and tumorigenesis.

  • Summary of current thinking: Because the regulation of DNA methylation shares conserved features in plants and mammals, we propose that, similar to plants, mammalian cells also dynamically control genome-wide DNA methylation through a methylstat-dependent mechanism, and that this dynamic regulation contributes to the seemingly contradictory changes in DNA methylome in aging cells and cancer cells. This methylstat-based model implies that the locus-specific methylation increases observed in aging cells are not due to random epigenetic drift. Importantly, targeted intervention of the DNA methylation of the methylstat may slow down or prevent the DNA methylation increases in genes important for senescence and other old-age disorders and thus avert aging.

  • Comments on future directions: While the methylstat model is enticing, the putative methylstats in mammalian cells have yet to be identified. Future investigations on this methylstat model may be aided by clues from the specific features of euchromatic regions where abnormal DNA methylation occurs.

MEMS

methylation monitoring sequence

ROS

reactive oxygen species

ROS1

Repressor of Silencing 1

RdDM

RNA-directed DNA methylation

SUVH

Suppressor of Variegation 3–9 Homologue

The authors apologize to those colleagues whose work is not cited owing to space constraints. H.Z. and J.K.Z. are supported by the Chinese Academy of Sciences.

The Authors declare that there are no competing interests associated with the manuscript.

1
Zhang
,
H.
,
Lang
,
Z.
and
Zhu
,
J.K.
(
2018
)
Dynamics and function of DNA methylation in plants
.
Nat. Rev. Mol. Cell Biol.
19
,
489
506
2
Luo
,
C.
,
Hajkova
,
P.
and
Ecker
,
J.R.
(
2018
)
Dynamic DNA methylation: in the right place at the right time
.
Science
361
,
1336
1340
3
Wilson
,
A.S.
,
Power
,
B.E.
and
Molloy
,
P.L.
(
2007
)
DNA hypomethylation and human diseases
.
Biochim. Biophys. Acta, Rev Cancer
1775
,
138
162
4
Lund
,
J.B.
,
Li
,
S.
,
Christensen
,
K.
,
Mengel-From
,
J.
,
Soerensen
,
M.
,
Marioni
,
R.E.
et al (
2019
)
Age-dependent DNA methylation patterns on the Y chromosome in elderly males
.
Aging Cell
21
,
e12907
5
Morgan
,
A.E.
,
Davies
,
T.J.
and
Mc Auley
,
M.T.
(
2018
)
The role of DNA methylation in ageing and cancer
.
Proc. Nutr. Soc.
77
,
412
422
6
Xie
,
W.
,
Baylin
,
S.B.
and
Easwaran
,
H.
(
2019
)
DNA methylation in senescence, aging and cancer
.
Oncoscience
6
,
291
293
7
Goll
,
M.G.
and
Bestor
,
T.H.
(
2005
)
Eukaryotic cytosine methyltransferases
.
Annu. Rev. Biochem.
74
,
481
514
8
Watanabe
,
T.
,
Tomizawa
,
S.
,
Mitsuya
,
K.
,
Totoki
,
Y.
,
Yamamoto
,
Y.
,
Kuramochi-Miyagawa
,
S.
et al (
2011
)
Role for piRNAs and noncoding RNA in de novo DNA methylation of the imprinted mouse Rasgrf1 locus
.
Science
332
,
848
852
9
Ooi
,
S.K.
,
Qiu
,
C.
,
Bernstein
,
E.
,
Li
,
K.
,
Jia
,
D.
,
Yang
,
Z.
et al (
2007
)
DNMT3 l connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA
.
Nature
448
,
714
717
10
Cheng
,
X.
and
Blumenthal
,
R.M.
(
2008
)
Mammalian DNA methyltransferases: a structural perspective
.
Structure
16
,
341
350
11
Matzke
,
M.A.
and
Mosher
,
R.A.
(
2014
)
RNA- directed DNA methylation: an epigenetic pathway of increasing complexity
.
Nat. Rev. Genet.
15
,
394
408
12
Du
,
J.M.
,
Johnson
,
L.M.
,
Jacobsen
,
S.E.
and
Patel
,
D.J.
(
2015
)
DNA methylation pathways and their crosstalk with histone methylation
.
Nat. Rev. Mol. Cell Biol.
16
,
519
532
13
Zemach
,
A.
,
Kim
,
M.Y.
,
Hsieh
,
P.H.
,
Coleman-Derr
,
D.
,
Eshed-Williams
,
L.
,
Thao
,
K.
et al (
2013
)
The Arabidopsis nucleosome remodeler DDM1 allows DNA methyltransferases to access H1-containing heterochromatin
.
Cell
153
,
193
205
14
Zhu
,
J.K.
(
2009
)
Active DNA demethylation mediated by DNA glycosylases
.
Annu. Rev. Genet.
43
,
143
166
15
Wu
,
X.
and
Zhang
,
Y.
(
2017
)
TET-mediated active DNA demethylation: mechanism, function and beyond
.
Nat. Rev. Genet.
18
,
517
534
16
Zhang
,
H.
and
Zhu
,
J.K.
(
2012
)
Active DNA demethylation in plants and animals
.
Cold Spring Harb. Symp. Quant. Biol.
77
,
161
173
17
Oswald
,
J.
,
Engemann
,
S.
,
Lane
,
N.
,
Mayer
,
W.
,
Olek
,
A.
,
Fundele
,
R.
et al (
2000
)
Active demethylation of the paternal genome in the mouse zygote
.
Curr. Biol.
10
,
475
478
18
Mayer
,
W.
,
Niveleau
,
A.
,
Walter
,
J.
,
Fundele
,
R.
and
Haaf
,
T.
(
2000
)
Demethylation of the zygotic paternal genome
.
Nature
403
,
501
502
19
Wu
,
S.C.
and
Zhang
,
Y.
(
2010
)
Active DNA demethylation: many roads lead to Rome
.
Nat. Rev. Mol. Cell Biol.
11
,
607
620
20
Hajkova
,
P.
,
Ancelin
,
K.
,
Waldmann
,
T.
,
Lacoste
,
N.
,
Lange
,
U.C.
,
Cesari
,
F.
et al (
2008
)
Chromatin dynamics during epigenetic reprogramming in the mouse germ line
.
Nature
452
,
877
881
21
Morgan
,
H.D.
,
Santos
,
F.
,
Green
,
K.
,
Dean
,
W.
and
Reik
,
W.
(
2005
)
Epigenetic reprogramming in mammals
.
Hum. Mol. Genet.
14
,
R47
R58
22
Yang
,
X.
,
Smith
,
S.L.
,
Tian
,
X.C.
,
Lewin
,
H.A.
,
Renard
,
J.P.
and
Wakayama
,
T.
(
2007
)
Nuclear reprogramming of cloned embryos and its implications for therapeutic cloning
.
Nat. Genet.
39
,
295
302
23
Booth
,
L.N.
and
Brunet
,
A.
(
2016
)
The aging epigenome
.
Mol. Cell
62
,
728
744
24
Maegawa
,
S.
,
Hinkal
,
G.
,
Kim
,
H.S.
,
Shen
,
L.
,
Zhang
,
L.
,
Zhang
,
J.
et al (
2010
)
Widespread and tissue specific age-related DNA methylation changes in mice
.
Genome Res.
20
,
332
340
25
Jung
,
M.
and
Pfeifer
,
G.P.
(
2015
)
Aging and DNA methylation
.
BMC Biol.
13
,
7
26
Najar
,
R.A.
,
Wani
,
N.A.
,
Bhat
,
J.A.
,
Dar
,
N.J.
,
Rahat
,
B.
,
Gupta
,
A.P.
et al (
2018
)
Modulation of dietary folate with age confers selective hepatocellular epigenetic imprints through DNA methylation
.
J. Nutr. Biochem.
53
,
121
132
27
Chia
,
N.
,
Wang
,
L.
,
Lu
,
X.
,
Senut
,
M.C.
,
Brenner
,
C.
and
Ruden
,
D.M.
(
2011
)
Hypothesis: environmental regulation of 5-hydroxymethylcytosine by oxidative stress
.
Epigenetics
6
,
853
856
28
Kudryavtseva
,
A.V.
,
Krasnov
,
G.S.
,
Dmitriev
,
A.A.
,
Alekseev
,
B.Y.
,
Kardymon
,
O.L.
,
Sadritdinova
,
A.F.
et al (
2016
)
Mitochondrial dysfunction and oxidative stress in aging and cancer
.
Oncotarget
7
,
44879
44905
29
Lewandowska
,
J.
and
Bartoszek
,
A.
(
2011
)
DNA methylation in cancer development, diagnosis and therapy–multiple opportunities for genotoxic agents to act as methylome disruptors or remediators
.
Mutagenesis
26
,
475
487
30
Guillaumet-Adkins
,
A.
,
Yañez
,
Y.
,
Peris-Diaz
,
M.D.
,
Calabria
,
I.
,
Palanca-Ballester
,
C.
and
Sandoval
,
J.
(
2017
)
Epigenetics and oxidative stress in aging
.
Oxid. Med. Cell Longev.
2017
,
9175806
31
Hannum
,
G.
,
Guinney
,
J.
,
Zhao
,
L.
,
Zhang
,
L.
,
Hughes
,
G.
,
Sadda
,
S.
et al (
2013
)
Genome-wide methylation profiles reveal quantitative views of human aging rates
.
Mol. Cell
49
,
359
367
32
Field
,
A.E.
,
Robertson
,
N.A.
,
Wang
,
T.
,
Havas
,
A.
,
Ideker
,
T.
and
Adams
,
P.D.
(
2018
)
DNA methylation clocks in aging: categories, causes, and consequences
.
Mol. Cell
71
,
882
895
33
Weidner
,
C.I.
,
Lin
,
Q.
,
Koch
,
C.M.
,
Eisele
,
L.
,
Beier
,
F.
,
Ziegler
,
P.
et al (
2014
)
Aging of blood can be tracked by DNA methylation changes at just three CpG sites
.
Genome Biol.
15
,
R24
34
Kulis
,
M.
,
Merkel
,
A.
,
Heath
,
S.
,
Queirós
,
A.C.
,
Schuyler
,
R.P.
,
Castellano
,
G.
et al (
2015
)
Whole-genome fingerprint of the DNA methylome during human B cell differentiation
.
Nat. Genet.
47
,
746
756
35
Baylin
,
S.B.
and
Jones
,
P.A.
(
2016
)
Epigenetic determinants of cancer
.
Cold Spring Harb. Perspect. Biol.
8
,
a019505
36
Hao
,
X.
,
Luo
,
H.
,
Krawczyk
,
M.
,
Wei
,
W.
,
Wang
,
W.
,
Wang
,
J.
et al (
2017
)
DNA methylation markers for diagnosis and prognosis of common cancers
.
Proc. Natl Acad. Sci. U.S.A.
114
,
7414
7419
37
Xu
,
R.H.
,
Wei
,
W.
,
Krawczyk
,
M.
,
Wang
,
W.
,
Luo
,
H.
,
Flagg
,
K.
et al (
2017
)
Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma
.
Nat. Mater.
11
,
1155
1161
38
Schlesinger
,
Y.
,
Straussman
,
R.
,
Keshet
,
I.
,
Farkash
,
S.
,
Hecht
,
M.
,
Zimmerman
,
J.
et al (
2007
)
Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer
.
Nat. Genet.
39
,
232
236
39
Teschendorff
,
A.E.
,
Menon
,
U.
,
Gentry-Maharaj
,
A.
,
Ramus
,
S.J.
,
Weisenberger
,
D.J.
,
Shen
,
H.
et al (
2010
)
Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer
.
Genome Res.
20
,
440
446
40
Beerman
,
I.
,
Bock
,
C.
,
Garrison
,
B.S.
,
Smith
,
Z.D.
,
Gu
,
H.
,
Meissner
,
A.
et al (
2013
)
Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging
.
Cell Stem Cell
12
,
413
425
41
Laird
,
P.W.
,
Jackson-Grusby
,
L.
,
Fazeli
,
A.
,
Dickinson
,
S.L.
,
Jung
,
W.E.
,
Li
,
E.
et al (
1995
)
Suppression of intestinal neoplasia by DNA hypomethylation
.
Cell
81
,
197
205
42
Gautrey
,
H.E.
,
van Otterdijk
,
S.D.
,
Cordell
,
H.J.
;
Newcastle 85+ Study Core Team
,
Mathers
,
J.C.
and
Strathdee
,
G.
(
2014
)
DNA methylation abnormalities at gene promoters are extensive and variable in the elderly and phenocopy cancer cells
.
FASEB J.
28
,
3261
3272
43
Belshaw
,
N.J.
,
Pal
,
N.
,
Tapp
,
H.S.
,
Dainty
,
J.R.
,
Lewis
,
M.P.
,
Williams
,
M.R.
et al (
2010
)
Patterns of DNA methylation in individual colonic crypts reveal aging and cancer-related field defects in the morphologically normal mucosa
.
Carcinogenesis
31
,
1158
1163
44
Tamura
,
G.
(
2004
)
Promoter methylation status of tumor suppressor and tumor-related genes in neoplastic and non-neoplastic gastric epithelia
.
Histol. Histopathol.
19
,
221
228
45
Liu
,
R.
,
How-Kit
,
A.
,
Stammitti
,
L.
,
Teyssier
,
E.
,
Rolin
,
D.
,
Mortain-Bertrand
,
A.
et al (
2015
)
A DEMETER- like DNA demethylase governs tomato fruit ripening
.
Proc. Natl Acad. Sci. U.S.A.
112
,
10804
10809
46
Lang
,
Z.
,
Wang
,
Y.
,
Tang
,
K.
,
Tang
,
D.
,
Datsenka
,
T.
,
Cheng
,
J.
et al (
2017
)
Critical roles of DNA demethylation in the activation of ripening- induced genes and inhibition of ripening- repressed genes in tomato fruit
.
Proc. Natl Acad. Sci. U.S.A.
114
,
E4511
E4519
47
Zhong
,
S.
,
Fei
,
Z.
,
Chen
,
Y.R.
,
Zheng
,
Y.
,
Huang
,
M.
,
Vrebalov
,
J.
et al (
2013
)
Single- base resolution methylomes of tomato fruit development reveal epigenome modifications associated with ripening
.
Nat. Biotechnol.
31
,
154
159
48
Mathieu
,
O.
,
Reinders
,
J.
,
Čaikovski
,
M.
,
Smathajitt
,
C.
and
Paszkowski
,
J.
(
2007
)
Transgenerational stability of the Arabidopsis epigenome is coordinated by CG methylation
.
Cell
130
,
851
862
49
Tang
,
K.
,
Lang
,
Z.
,
Zhang
,
H.
and
Zhu
,
J.K.
(
2016
)
The DNA demethylase ROS1 targets genomic regions with distinct chromatin modifications
.
Nat. Plants
2
,
16169
50
Lei
,
M.G.
,
Zhang
,
H.
,
Julian
,
R.
,
Tang
,
K.
,
Xie
,
S.
and
Zhu
,
J.K.
(
2015
)
Regulatory link between DNA methylation and active demethylation in Arabidopsis
.
Proc. Natl Acad. Sci. U.S.A.
112
,
3553
3557
51
Xiao
,
X.
,
Zhang
,
J.
,
Li
,
T.
,
Fu
,
X.
,
Satheesh
,
V.
,
Niu
,
Q.
, et al (
2019
)
A group of SUVH methyl-DNA binding proteins regulate expression of the DNA demethylase ROS1 in Arabidopsis
.
J. Integr. Plant Biol.
61
,
110
119
52
Zhao
,
Q.Q.
,
Lin
,
R.N.
,
Li
,
L.
,
Chen
,
S.
and
He
,
X.J.
(
2019
)
A methylated-DNA-binding complex required for plant development mediates transcriptional activation of promoter methylated genes
.
J. Integr. Plant Biol.
61
,
120
139
53
Erhard
,
K.F.
,
Talbot
,
J.E.
,
Deans
,
N.C.
,
McClish
,
A.E.
and
Hollick
,
J.B.
(
2015
)
Nascent transcription affected by RNA polymerase IV in Zea mays
.
Genetics
199
,
1107
1125
54
Hu
,
L.
,
Li
,
N.
,
Xu
,
C.
,
Zhong
,
S.
,
Lin
,
X.
,
Yang
,
J.
et al (
2014
)
Mutation of a major CG methylase in rice causes genome- wide hypomethylation, dysregulated genome expression, and seedling lethality
.
Proc. Natl Acad. Sci. U.S.A.
111
,
10642
10647
55
Williams
,
B.P.
,
Pignatta
,
D.
,
Henikoff
,
S.
and
Gehring
,
M.
(
2015
)
Methylation- sensitive expression of a DNA demethylase gene serves as an epigenetic rheostat
.
PLoS Genet.
11
,
e1005142
56
Viré
,
E.
,
Brenner
,
C.
,
Deplus
,
R.
,
Blanchon
,
L.
,
Fraga
,
M.
,
Didelot
,
C.
et al (
2006
)
The Polycomb group protein EZH2 directly controls DNA methylation
.
Nature
439
,
871
874
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND).