ArticlePDF AvailableLiterature Review

Do age-related changes in DNA methylation play a role in the development of age-related diseases?

Authors:

Abstract

DNA methylation is an important epigenetic mechanism in mammalian cells. It occurs almost exclusively at CpG sites and has a key role in a number of biological processes. It plays an important part in regulating chromatin structure and has been best studied for its role in controlling gene expression. In particular, hypermethylation of gene promoters which have high levels of CpG sites, known as CpG islands, leads to gene inactivation. In healthy cells, however, it appears that only a small number of genes are controlled through promoter hypermethylation, such as genes on the inactivated X-chromosome or at imprinted loci, and most promoter-associated CpG islands remain methylation-free regardless of gene expression status. However, a large body of evidence has now shown that this protection from methylation not only breaks down in a number of pathological conditions (e.g. cancer), but also already occurs during the normal process of aging. The present review focuses on the methylation changes that occur during healthy aging and during disease development, and the potential links between them. We focus especially on the extent to which the acquisition of aberrant methylation changes during aging could underlie the development of a number of important age-related pathological conditions.
Biochemical Society Annual Symposium No. 80
Biochemical Society Annual
Symposium No. 80
Do age-related changes in DNA methylation play a
role in the development of age-related diseases?
Sanne D. van Otterdijk*1, John C. Mathers† and Gordon Strathdee*
*Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE1 4LP, U.K., and Human Nutrition Research Centre, Centre for Brain
Ageing and Vitality, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, U.K.
Abstract
DNA methylation is an important epigenetic mechanism in mammalian cells. It occurs almost exclusively at
CpG sites and has a key role in a number of biological processes. It plays an important part in regulating
chromatin structure and has been best studied for its role in controlling gene expression. In particular,
hypermethylation of gene promoters which have high levels of CpG sites, known as CpG islands, leads to
gene inactivation. In healthy cells, however, it appears that only a small number of genes are controlled
through promoter hypermethylation, such as genes on the inactivated X-chromosome or at imprinted loci,
and most promoter-associated CpG islands remain methylation-free regardless of gene expression status.
However, a large body of evidence has now shown that this protection from methylation not only breaks
down in a number of pathological conditions (e.g. cancer), but also already occurs during the normal process
of aging. The present review focuses on the methylation changes that occur during healthy aging and during
disease development, and the potential links between them. We focus especially on the extent to which
the acquisition of aberrant methylation changes during aging could underlie the development of a number
of important age-related pathological conditions.
Introduction
Epigenetic mechanisms play an important role in numerous
cellular processes, such as genomic imprinting [1], X-
chromosome inactivation [2] and cell differentiation [3]
(Figure 1). One of the primary epigenetic mechanisms
is DNA methylation, which involves the addition of a
methyl group to the fifth position of a cytosine base by
enzymes called DNMTs (DNA methyltransferases). This
occurs almost exclusively in cytosines that are immediately
followed by a guanine, forming so-called CpG dinucleotides
[4]. CpG dinucleotides are underrepresented throughout the
genome, except for short stretches of DNA known as CpG
islands [5], which are frequently associated with human genes.
The state of methylation of CpG islands near gene promoters
has been associated with the transcriptional activity of a gene
[6], although it has been shown that even methylation levels
in CpG islands further away from the gene promoter, so-
called CpG island shores, are related to gene expression
[7]. However, it remains uncertain whether methylation is
Key words: age-related disease, aging, cancer, DNA methylation, epigenetics.
Abbreviations used: CLL, chronic lymphoblastic leukaemia; MMR, mismatch repair.
1To whom correspondence should be addressed (email s.d.van-otterdijk@ncl.ac.uk).
involved directly in silencing gene expression or whether it
plays a role in maintaining the silenced state which has been
induced by the associated epigenetic marks on histones and
other epigenetic molecules [8].
In healthy individuals, CpG islands remain mostly
methylation-free, whereas most of the non-island-associated
CpG sites in the bulk of the genome are methylated [5].
Even though patterns of methylation are inherited through
cell divisions, the copying of methylation patterns from
parental to daughter strand is not 100% efficient and
methylation errors accumulate [9]. The rate at which these
errors are accumulated increases with age and during disease
development. In the present review, we focus on the changes
in methylation levels that occur both during healthy aging and
during the development of age-related diseases. In particular,
to what extent do methylation changes during aging influence
the susceptibility to develop age-related diseases?
Methylation levels change during healthy
aging
Because of the important demographic changes occurring
across most of the world which are leading to an increasingly
Biochem. Soc. Trans. (2013) 41, 803–807; doi:10.1042/BST20120358 C
The Authors Journal compilation C
2013 Biochemical Society 803
804 Biochemical Society Transactions (2013) Volume 41, part 3
Figure 1 Functional roles of DNA methylation
DNA methylation plays important roles in key biological processes, such
as differentiation, which allows stem cells and multipotent progenitor
cells to differentiate into multiple different cell lineages, imprinting,
which allows certain genes to be expressed in a parent-of-origin specific
matter, and silencing of large chromosomal domains, such as inactivation
of the X-chromosome in females.
elderly population [10], aging and age-related diseases are
becoming a major health priority. Life expectancy has
increased steadily over the last two centuries [11], but a
significant proportion of the extra life years is associated
with morbidity. Age-related diseases, including cancers,
cardiovascular disease and dementia, are now the dominant
health problems in most countries. Identifying the underlying
molecular changes that occur as part of the aging process
and how this contributes to the development of age-related
diseases will be critical for improving the health outcome
for elderly patients and also in underpinning potential
preventative strategies.
Alterations in the patterns of DNA methylation are one of
the hallmarks of aging [12]. These changes include reduced
levels of global DNA methylation across the genome, in
conjunction with local areas of hypermethylation, often
centring on promoter-associated CpG islands. Furthermore,
DNA methylation levels have been observed to change
with age in multiple tissue types in both mice and human
studies [13–17], implying that altered patterns of methylation
are an inevitable consequence of aging in mammalian cells.
The exact timing of these changes in methylation, and the
biological driving forces behind them, remains unclear, but
several human and animal studies have suggested that it is
not a process solely associated with old age, but that it
is an ongoing process over the course of life [16,18]. A
previous study by Teschendorff et al. [15] indicated that
increased levels of DNA methylation in promoter regions
are steadily acquired during the course of life and it has
been observed by Talens et al. [19] that epigenetic variation
in the population also increases gradually with age. The
rate at which these changes occurred differed between
different loci and these changes can be substantial at loci
regulating transcription of nearby genes [19]. Whereas several
studies reported that regions near gene promoters become
hypermethylated with age, several repetitive sequences lose
methylation with increasing age, such as the Alu elements
[20], Line-1 [21] and HERV-K [22]. Even though age-related
methylation changes are observed in many tissues, patterns of
DNA methylation are tissue-specific [23], and, during aging,
individual genes acquire differential methylation patterns
in different tissue types [14,24]. However, although it is
clear that altered DNA methylation is linked strongly
with aging, the biological consequences of the observed
DNA methylation changes are far less clear. The advent of
methods for more precise delineation of DNA methylation
levels, such as the use of pyrosequencing, have made age-
related methylation changes at many loci readily detectable.
However, the extent to which this actually alters expression
levels of genes, and, as a consequence, cellular function,
remains largely unknown. Tissue- and cell-type-specific
differences in DNA methylation patterns cause a number
of difficulties in clearly understanding age-related alterations
in DNA methylation. Almost all studies are carried out using
tissue samples, which are a mixture of different cell types. This
means interpretation of the results obtained for methylation
patterns in the tissues may be confounded by age-related
changes in the cellular make-up of tissues. In addition, the
extent of age-related changes may be underestimated if
the changes occur only in one specific cell type in the tissue
being assessed.
Aberrant patterns of methylation are
observed in diseases
An important role for altered DNA methylation in the
development of disease is strongly suggested by the growing
number of human diseases that are known to occur when
the epigenetic information is not properly established during
embryonic and fetal development or maintained later in life.
The importance of DNA methylation was first discovered
in genetic disorders, such as Prader–Willi syndrome and
Angelman syndrome, that result from loss of expression of a
cluster of imprinted genes [25]. Recently, DNA methylation
is most extensively studied in cancers, although changes in
DNA methylation have been described in multiple other
diseases, such as cardiovascular, neurological and metabolic
disorders, and autoimmune diseases.
During cancer development, reduced levels of global DNA
methylation is observed, together with hypermethylation of
some CpG islands in gene promoters [26]. Hypermethylation
is frequently observed in the same genes that are often
mutated in familial cancers, emphasizing their causal
importance in tumorigenesis. For example, loss of function
of DNA MMR (mismatch repair) genes, including MLH1,
are causal for HNPCC (hereditary non-polyposis colorectal
cancer) [27], whereas, in sporadic colorectal cancer, loss
of microsatellite instability (an effect of MMR) is due to
hypermethylation of MLH1 [28]. Another example is the
congenital BRCA1 mutation, which is a cause of breast and
ovarian cancer. It is the same gene, however, that becomes
frequently hypermethylated in sporadic breast and ovarian
cancers [29,30]. In some cases, frequent hypermethylation
of genes can be used to identify novel tumour-suppressor
genes, as was the case for RASSF1 [31], HACE1 [32]
and TWIST2 [33]. Hypermethylation of tumour-suppressor
genes is a feature of several cancers, including the p15 gene
in leukaemia [34], the Rb gene in retinoblastoma [35] and
C
The Authors Journal compilation C
2013 Biochemical Society
Biochemical Society Annual Symposium No. 80: Epigenetic Mechanisms in Development and Disease 805
the VHL gene in renal tumours [36]. On the other hand,
hypomethylation was observed in several cancers in tumour-
promoting or metastasis-promoting genes, such as the uPA
gene in breast cancer [37] and the MAGEB2 gene in head and
neck squamous cell carcinomas [38].
Whereas many genes that have similar functional roles
are methylated during cancer development, some of the
specific genes which are methylated are tumour-type-specific,
whereas others are shared by multiple tumour types [39], such
as the p16/CDKN2 gene [40], the RASSF1 gene and HIC1
gene [41].
Aberrant methylation patterns occur in other pathologies
in addition to cancers. And, whereas in tumours, the causal
methylation changes are in tumour-suppressor genes and
in oncogenes, in cardiovascular diseases, the genes that
change methylation patterns are thought to be involved
in lipid oxidation, inhibition of endothelial cell migration
and formation, the control of cell proliferation and
angiogenesis [42–45]. However, whereas for cancers, the
causal role of DNA methylation is well established, for
cardiovascular diseases, the direct functional link of these
altered methylation patterns is not as clear. The role of DNA
methylation in cardiovascular diseases is discussed in a recent
review [46], and is not reviewed further in the present article.
Neither do we expand on the role of DNA methylation in
other diseases, such as neurological disorders [47], metabolic
disorders [48] and autoimmune disorders [49].
Cross-talk between methylation changes
during aging and disease susceptibility
The changes in DNA methylation patterns observed during
aging are reminiscent of the methylation alterations seen
during cancer development, i.e. loss of DNA methylation
at the genome-wide level in combination with gains in
methylation levels in CpG islands in or near gene promoters.
This raises the possibility that the acquisition of methylation
changes during healthy aging and during the development of
a disease might be linked to each other. The methylation
patterns of functionally important promoter regions are
reported to be more stable as opposed to non-promoter
regions and the corresponding genes might be involved
in longevity [16,50]. Global DNA methylation levels were
reported to correlate negatively with frailty measurements in
individuals over 65 years of age [51]. Indeed, there is some
evidence that genes that are differentially methylated during
healthy aging and during the development of diseases show
a degree of overlap. Teschendorff et al. [15] showed that
there was an overlap between the genes that showed age-
related methylation changes and genes that were reported
previously to be methylated in cancers, such as the genes
TP73 and SFRP1, two genes that were shown to become
methylated in different types of cancers. Rakyan et al.
[52] showed a significant correlation specifically between
bivalent chromatin domain DMRs that showed age-related
hypermethylation and aberrantly methylated promoters in
primary AML (acute myeloid leukaemia).
Before being able to understand the link between aging
and disease development, it is first necessary to understand
the precise role that DNA methylation plays during disease
development. Currently, this remains uncertain, although
several plausible hypotheses can be suggested. The first
hypothesis describes the changes in methylation levels as
events that occur during the process of clonal expansion. This
suggests that methylation changes are not the driving force
behind the development of the disease, but rather a side effect
of disease development. In the second hypothesis, methyla-
tion changes occur during the process of clonal expansion.
However, the methylation event could be aetiologically
important, e.g. in producing further growth and proliferative
advantages of the targeted cells over the non-targeted cells
and so promote disease development. A third hypothesis
states that diseases develop in cells that are pre-primed by
aberrant patterns of DNA methylation. In this hypothesis,
altered patterns of methylation pre-exist in a subset of appar-
ently normal cells. If cancer-driving mutations occur in a cell
with a pre-existing methylation pattern, this cell can rapidly
proliferate and lead to disease (Figure 2). This hypothesis
is supported by the observation that methylation patterns
are already present in apparently normal tissues from those
at higher cancer risk [53] and in pre-cancerous tissues [54].
However, these three hypotheses are not mutually exclusive
and a combination of these hypotheses might be possible.
Two possible hypotheses can explain the link between
the methylation changes that occur during aging and during
disease development. First, both age-related and cancer-
related DNA methylation changes might be driven by similar
mechanisms, which is causing the similar patterns of altered
methylation during cancer and aging. This hypothesis is
supported by the evidence that the same lifestyle factors
which influence the aging process influence the risk of cancer
and of other age-related diseases and modulate patterns of
DNA methylation [55,56]. The second hypothesis states that
age-related DNA methylation may underlie the development
of cancer, and possibly other age-related diseases. This
suggests that methylation is already present in apparently
healthy individuals before the development of the disease and
the disease then develops from cells with altered methylation
patterns (Figure 2). This hypothesis is supported by the
observation that in CLL (chronic lymphoblastic leukaemia),
global DNA methylation was shown to be relatively stable
over time and similar within different CLL compartments
[57], suggesting that cancer cells are not necessarily highly
methylation unstable. However, more research is necessary to
determine what the precise link is between DNA methylation
changes seen in aging and disease development.
Conclusion
DNA methylation levels change during healthy aging.
These changes in methylation levels during aging are
reminiscent of methylation alterations observed during
disease development. The precise mechanisms behind this
link remain unclear. In the present review, we have suggested
C
The Authors Journal compilation C
2013 Biochemical Society
806 Biochemical Society Transactions (2013) Volume 41, part 3
Figure 2 Possible role of DNA methylation during disease development and the link with aging
Three possible hypotheses can explain the role of DNA methylation in disease development. (A) Methylation changes are
an effect of the clonal expansion of cells before disease development, but it does not play a role in the development of
the disease. (B) Methylation changes occur during the clonal expansion of cells that occur before disease development. The
methylation events will then produce further growth advantages, promoting development of disease. (C) Methylation levels
are already present in a subset of normal cells. Disease develops when these ‘methylation-primed’ cells clonally expand.
These three hypotheses lead to new hypotheses explaining the link between DNA methylation changes during healthy
aging and disease: (i) in the case of (A)or(B), both aging and disease-related DNA methylation changes are driven by
similar mechanisms; (ii) in the case of hypothesis (C), age-related DNA methylation underlies the development of cancer,
and possibly other diseases.
two possible hypotheses in understanding the link that exists
between healthy aging and disease development. First, both
age-related and cancer-related DNA methylation changes
are driven by similar mechanisms. The second possible
hypothesis is that age-related DNA methylation underlies
the development of cancer, and possibly other diseases.
Funding
The work of G.S. and S.D.v.O. is supported by the Newcastle National
Institute for Health Research (NIHR) Biomedical Research Centre,
Tyneside Leukaemia Research Association, Dunhill Medical Trust,
Biotechnology and Biological Sciences Research Council and Children
with Cancer. The work of J.C.M. is supported by the Biotechnology
and Biological Sciences Research Council [grant number BH090948]
and through the Centre for Brain Ageing and Vitality which is funded
through the Lifelong Health and Wellbeing cross-council initiative by
the Medical Research Council, Biotechnology and Biological Sciences
Research Council, Engineering and Physical Sciences Research
Council and Economic and Social Research Council and by the U.K.
Department of Health.
References
1 Fedoriw, A., Mugford, J. and Magnuson, T. (2012) Genomic imprinting
and epigenetic control of development. Cold Spring Harbor Perspect.
Biol. 4, a008136
2 Li, Y., Tan, T., Zong, L., He, D., Tao, W. and Liang, Q. (2012) Study of
methylation of histone H3 lysine 9 and H3 lysine 27 during X
chromosome inactivation in three types of cells. Chromosome Res. 20,
769–778
3 Bock, C., Beerman, I., Lien, W.H., Smith, Z.D., Gu, H., Boyle, P., Gnirke, A.,
Fuchs, E., Rossi, D.J. and Meissner, A. (2012) DNA methylation dynamics
during in vivo differentiation of blood and skin stem cells. Mol. Cell 47,
633–647
4 Lister, R., Pelizzola, M., Dowen, R.H., Hawkins, R.D., Hon, G.,
Tonti-Filippini, J., Nery, J.R., Lee, L., Ye, Z., Ngo, Q.M. et al. (2009) Human
DNA methylomes at base resolution show widespread epigenomic
differences. Nature 462, 315–322
5 Cooper, D.N. and Krawczak, M. (1989) Cytosine methylation and the fate
of CpG dinucleotides in vertebrate genomes. Hum. Genet. 83, 181–188
6 Bird, A.P. (1986) CpG-rich islands and the function of DNA methylation.
Nature 321, 209–213
7 Irizarry, R.A., Ladd-Acosta, C., Wen, B., Wu, Z., Montano, C., Onyango, P.,
Cui, H., Gabo, K., Rongione, M., Webster, M. et al. (2009) The human
colon cancer methylome shows similar hypo- and hypermethylation at
conserved tissue-specific CpG island shores. Nat. Genet. 41, 178–186
8 Feng, Y.Q., Desprat, R., Fu, H., Olivier, E., Lin, C.M., Lobell, A., Gowda,
S.N., Aladjem, M.I. and Bouhassira, E.E. (2006) DNA methylation supports
intrinsic epigenetic memory in mammalian cells. PLoS Genet. 2,e65
9 Ushijima, T., Watanabe, N., Okochi, E., Kaneda, A., Sugimura, T. and
Miyamoto, K. (2003) Fidelity of the methylation pattern and its variation
in the genome. Genome Res. 13, 868–874
10 Christensen, K., Doblhammer, G., Rau, R. and Vaupel, J.W. (2009) Ageing
populations: the challenges ahead. Lancet 374, 1196–1208
11 Vaupel, J.W. (2010) Biodemography of human ageing. Nature 464,
536–542
12 Mathers, J.C. (2006) Nutritional modulation of ageing: genomic and
epigenetic approaches. Mech. Ageing Dev. 127, 584–589
13 Wilson, V.L. and Jones, P.A. (1983) DNA methylation decreases in aging
but not in immortal cells. Science 220, 1055–1057
14 Maegawa, S., Hinkal, G., Kim, H.S., Shen, L., Zhang, L., Zhang, J., Zhang,
N., Liang, S., Donehower, L.A. and Issa, J.P. (2010) Widespread and
tissue specific age-related DNA methylation changes in mice. Genome
Res. 20, 332–340
15 Teschendorff, A.E., Menon, U., Gentry-Maharaj, A., Ramus, S.J.,
Weisenberger, D.J., Shen, H., Campan, M., Noushmehr, H., Bell, C.G.,
Maxwell, A.P. 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
16 Bell, J.T., Tsai, P.C., Yang, T.P., Pidsley, R., Nisbet, J., Glass, D., Mangino,
M., Zhai, G., Zhang, F., Valdes, A. et al. (2012) Epigenome-wide scans
identify differentially methylated regions for age and age-related
phenotypes in a healthy ageing population. PLoS Genet. 8, e1002629
17 Issa, J.P., Ottaviano, Y.L., Celano, P., Hamilton, S.R., Davidson, N.E. and
Baylin, S.B. (1994) Methylation of the oestrogen receptor CpG island
links ageing and neoplasia in human colon. Nat. Genet. 7, 536–540
18 Takasugi, M. (2011) Progressive age-dependent DNA methylation
changes start before adulthood in mouse tissues. Mech. Ageing Dev.
132, 65–71
19 Talens, R.P., Christensen, K., Putter, H., Willemsen, G., Christiansen, L.,
Kremer, D., Suchiman, H.E., Slagboom, P.E., Boomsma, D.I. and Heijmans,
B.T. (2012) Epigenetic variation during the adult lifespan: cross-sectional
and longitudinal data on monozygotic twin pairs. Aging Cell 11, 694–703
20 Bollati, V., Schwartz, J., Wright, R., Litonjua, A., Tarantini, L., Suh, H.,
Sparrow, D., Vokonas, P. and Baccarelli, A. (2009) Decline in genomic
DNA methylation through aging in a cohort of elderly subjects. Mech.
Ageing Dev. 130, 234–239
21 Zhu, Z.Z., Hou, L., Bollati, V., Tarantini, L., Marinelli, B., Cantone, L., Yang,
A.S., Vokonas, P., Lissowska, J., Fustinoni, S. et al. (2012) Predictors of
global methylation levels in blood DNA of healthy subjects: a combined
analysis. Int. J. Epidemiol. 41, 126–139
C
The Authors Journal compilation C
2013 Biochemical Society
Biochemical Society Annual Symposium No. 80: Epigenetic Mechanisms in Development and Disease 807
22 Jintaridth, P. and Mutirangura, A. (2010) Distinctive patterns of
age-dependent hypomethylation in interspersed repetitive sequences.
Physiol. Genomics 41, 194–200
23 Davies, M.N., Volta, M., Pidsley, R., Lunnon, K., Dixit, A., Lovestone, S.,
Coarfa, C., Harris, R.A., Milosavljevic, A., Troakes, C. et al. (2012)
Functional annotation of the human brain methylome identifies
tissue-specific epigenetic variation across brain and blood. Genome Biol.
13,R43
24 Thompson, R.F., Atzmon, G., Gheorghe, C., Liang, H.Q., Lowes, C., Greally,
J.M. and Barzilai, N. (2010) Tissue-specific dysregulation of DNA
methylation in aging. Aging Cell 9, 506–518
25 Nicholls, R.D. and Knepper, J.L. (2001) Genome organization, function,
and imprinting in Prader–Willi and Angelman syndromes. Annu. Rev.
Genomics Hum. Genet. 2, 153–175
26 Gama-Sosa, M.A., Slagel, V.A., Trewyn, R.W., Oxenhandler, R., Kuo, K.C.,
Gehrke, C.W. and Ehrlich, M. (1983) The 5-methylcytosine content of
DNA from human tumors. Nucleic Acids Res. 11, 6883–6894
27 Crepin, M., Dieu, M.C., Lejeune, S., Escande, F., Boidin, D., Porchet, N.,
Morin, G., Manouvrier, S., Mathieu, M. and Buisine, M.P. (2012) Evidence
of constitutional MLH1 epimutation associated to transgenerational
inheritance of cancer susceptibility. Hum. Mutat. 33, 180–188
28 Wheeler, J.M., Loukola, A., Aaltonen, L.A., Mortensen, N.J. and Bodmer,
W.F. (2000) The role of hypermethylation of the hMLH1 promoter region
in HNPCC versus MSI+sporadic colorectal cancers. J. Med. Genet. 37,
588–592
29 Bal, A., Verma, S., Joshi, K., Singla, A., Thakur, R., Arora, S. and Singh, G.
(2012) BRCA1-methylated sporadic breast cancers are BRCA-like in
showing a basal phenotype and absence of ER expression. Virchows
Arch. 461, 305–312
30 Press, J.Z., De Luca, A., Boyd, N., Young, S., Troussard, A., Ridge, Y.,
Kaurah, P., Kalloger, S.E., Blood, K.A., Smith, M. et al. (2008) Ovarian
carcinomas with genetic and epigenetic BRCA1 loss have distinct
molecular abnormalities. BMC Cancer 8,17
31 Dammann, R., Yang, G. and Pfeifer, G.P. (2001) Hypermethylation of the
cpG island of Ras association domain family 1A (RASSF1A), a putative
tumor suppressor gene from the 3p21.3 locus, occurs in a large
percentage of human breast cancers. Cancer Res. 61, 3105–3109
32 K ¨uk, C., Hu, X., Iqbal, J., Gaulard, P., Klinkebiel, D., Cornish, A., Dave, B.J.
and Chan, W.C. (2013) HACE1 is a tumor suppressor gene candidate in
natural killer cell neoplasms. Am. J. Pathol. 182, 49–55
33 Thathia, S.H., Ferguson, S., Gautrey, H.E., van Otterdijk, S.D., Hili, M.,
Rand, V., Moorman, A.V., Meyer, S., Brown, R. and Strathdee, G. (2012)
Epigenetic inactivation of TWIST2 in acute lymphoblastic leukemia
modulates proliferation, cell survival and chemosensitivity.
Haematologica 97, 371–378
34 Wong, I.H., Ng, M.H., Huang, D.P. and Lee, J.C. (2000) Aberrant p15
promoter methylation in adult and childhood acute leukemias of nearly
all morphologic subtypes: potential prognostic implications. Blood 95,
1942–1949
35 Sakai, T., Toguchida, J., Ohtani, N., Yandell, D.W., Rapaport, J.M. and
Dryja, T.P. (1991) Allele-specific hypermethylation of the retinoblastoma
tumor-suppressor gene. Am. J. Hum. Genet. 48, 880–888
36 Prowse, A.H., Webster, A.R., Richards, F.M., Richard, S., Olschwang, S.,
Resche, F., Affara, N.A. and Maher, E.R. (1997) Somatic inactivation of
the VHL gene in von Hippel–Lindau disease tumors. Am. J. Hum. Genet.
60, 765–771
37 Pakneshan, P., Szyf, M., Farias-Eisner, R. and Rabbani, S.A. (2004)
Reversal of the hypomethylation status of urokinase (uPA) promoter
blocks breast cancer growth and metastasis. J. Biol. Chem. 279,
31735–31744
38 Pattani, K.M., Soudry, E., Glazer, C.A., Ochs, M.F., Wang, H., Schussel, J.,
Sun, W., Hennessey, P., Mydlarz, W., Loyo, M. et al. (2012) MAGEB2 is
activated by promoter demethylation in head and neck squamous cell
carcinoma. PLoS ONE 7, e45534
39 Hansen, K.D., Timp, W., Bravo, H.C., Sabunciyan, S., Langmead, B.,
McDonald, O.G., Wen, B., Wu, H., Liu, Y., Diep, D. et al. (2011) Increased
methylation variation in epigenetic domains across cancer types. Nat.
Genet. 43, 768–775
40 Esteller, M. (2005) Aberrant DNA methylation as a cancer-inducing
mechanism. Annu. Rev. Pharmacol. Toxicol. 45, 629–656
41 Teng, I.W., Hou, P.C., Lee, K.D., Chu, P.Y., Yeh, K.T., Jin, V.X., Tseng, M.J.,
Tsai, S.J., Chang, Y.S., Wu, C.S. et al. (2011) Targeted methylation of two
tumor suppressor genes is sufficient to transform mesenchymal stem
cells into cancer stem/initiating cells. Cancer Res. 71, 4653–4663
42 Friso, S., Lotto, V., Choi, S.W., Girelli, D., Pinotti, M., Guarini, P., Udali, S.,
Pattini, P., Pizzolo, F., Martinelli, N. et al. (2012) Promoter methylation in
coagulation F7 gene influences plasma FVII concentrations and relates to
coronary artery disease. J. Med. Genet. 49, 192–199
43 Hiltunen, M.O., Turunen, M.P., H ¨akkinen, T.P., Rutanen, J., Hedman, M.,
akinen, K., Turunen, A.M., Aalto-Set¨al ¨a, K. and Yl ¨a-Herttuala, S. (2002)
DNA hypomethylation and methyltransferase expression in
atherosclerotic lesions. Vasc. Med. 7,511
44 Movassagh, M., Choy, M.K., Goddard, M., Bennett, M.R., Down, T.A. and
Foo, R.S. (2010) Differential DNA methylation correlates with differential
expression of angiogenic factors in human heart failure. PLoS ONE 5,
e8564
45 Post, W.S., Goldschmidt-Clermont, P.J., Wilhide, C.C., Heldman, A.W.,
Sussman, M.S., Ouyang, P., Milliken, E.E. and Issa, J.P. (1999) Methylation
of the estrogen receptor gene is associated with aging and
atherosclerosis in the cardiovascular system. Cardiovasc. Res. 43,
985–991
46 Udali, S., Guarini, P., Moruzzi, S., Choi, S.W. and Friso, S. (2012)
Cardiovascular epigenetics: from DNA methylation to microRNAs. Mol.
Aspects Med., doi:10.1016/j.mam.2012.08.001
47 Jakovcevski, M. and Akbarian, S. (2012) Epigenetic mechanisms in
neurological disease. Nat. Med. 18, 1194–1204
48 Bruce, K.D. and Cagampang, F.R. (2011) Epigenetic priming of the
metabolic syndrome. Toxicol. Mech. Methods 21, 353–361
49 Grolleau-Julius, A., Ray, D. and Yung, R.L. (2010) The role of epigenetics
in aging and autoimmunity. Clin. Rev. Allergy Immunol. 39, 42–50
50 Kaminsky, Z.A., Tang, T., Wang, S.C., Ptak, C., Oh, G.H., Wong, A.H.,
Feldcamp, L.A., Virtanen, C., Halfvarson, J., Tysk, C. et al. (2009) DNA
methylation profiles in monozygotic and dizygotic twins. Nat. Genet. 41,
240–245
51 Bellizzi, D., P. D’Aquila, Montesanto, A., Corsonello, A., Mari, V., Mazzei,
B., Lattanzio, F. and Passarino, G. (2012) Global DNA methylation in old
subjects is correlated with frailty. Age 34, 169–179
52 Rakyan, V.K., Down, T.A., Maslau, S., Andrew, T., Yang, T.P., Beyan, H.,
Whittaker, P., McCann, O.T., Finer, S., Valdes, A.M. et al. (2010) Human
aging-associated DNA hypermethylation occurs preferentially at bivalent
chromatin domains. Genome Res. 20, 434–439
53 Belshaw, N.J., Elliott, G.O., Foxall, R.J., Dainty, J.R., Pal, N., Coupe, A.,
Garg, D., Bradburn, D.M., Mathers, J.C. and Johnson, I.T. (2008) Profiling
CpG island field methylation in both morphologically normal and
neoplastic human colonic mucosa. Br. J. Cancer 99, 136–142
54 Sproul, D., Kitchen, R.R., Nestor, C.E., Dixon, J.M., Sims, A.H., Harrison,
D.J., Ramsahoye, B.H. and Meehan, R.R. (2012) Tissue of origin
determines cancer-associated CpG island promoter hypermethylation
patterns. Genome Biol. 13,R84
55 Tapp, H.S., Commane, D.M., Bradburn, D.M., Arasaradnam, R., Mathers,
J.C., Johnson, I.T. and Belshaw, N.J. (2013) Nutritional factors and gender
influence age-related DNA methylation in the human rectal mucosa.
Aging Cell 12, 148–155
56 Mathers, J.C., Strathdee, G. and Relton, C.L. (2010) Induction of
epigenetic alterations by dietary and other environmental factors. Adv.
Genet. 71,339
57 Cahill, N., Bergh, A.C., Kanduri, M., Goransson-Kultima, H., Mansouri, L.,
Isaksson, A., Ryan, F., Smedby, K.E., Juliusson, G., Sundstrom, C. et al.
(2013) 450K-array analysis of chronic lymphocytic leukemia cells reveals
global DNA methylation to be relatively stable over time and similar in
resting and proliferative compartments. Leukemia 27, 150–158
Received 21 December 2012
doi:10.1042/BST20120358
C
The Authors Journal compilation C
2013 Biochemical Society
... In this study, we made several meaningful findings toward a bet- Past studies have established that aging is accompanied by a global decrease in DNA methylation in aging humans (Bjornsson, 2008) and mice (Singhal et al., 1987). However, DNA methylation modifications occur in a tissue and/or gene-specific manner and loss of DNA methylation of genome-wide is accompanied by a gain of methylation in CpG islands in or near gene promoters (van Otterdijk et al., 2013). DNMT inhibitor 5-Azacytidine reverses the aging phenotypes of mesenchymal stem cells (Kornicka et al., 2016), suggesting that gain of DNMT function promotes aging. ...
Article
Full-text available
DNA methylation alterations play mechanistic roles in aging; however, the epigenetic regulators/mediators causally involved in renal aging remain elusive. Here, we report that natural and D‐galactose (D‐gal)‐induced aging kidneys display marked suppression of antiaging factor NRF2 (nuclear factor erythroid‐derived 2‐like 2) and KLOTHO, accompanied by upregulations of DNA methyltransferase (DNMT) 1/3a/3b and NRF2/KLOTHO gene promoter hypermethylations. Administration of a DNMT inhibitor SGI‐1072 effectively hypomethylated the promoters, derepressed NRF2/KLOTHO, and mitigated the structural and functional alterations of renal aging in D‐gal mice. Moreover, oleuropein (OLP), an olive‐derived polyphenol, also displayed similar epigenetic modulation and antiaging effects. OLP inhibited the epigenetic NRF2/KLOTHO suppressions in a gain of DNMT‐sensitive manner in cultured renal cells, demonstrating a strong DNA‐demethylating capacity. In NRF2 knockout and KLOTHO knockdown D‐gal mice, OLP exhibited reduced antiaging effects with KLOTHO displaying a prominent gene effect and effect size; consistently in KLOTHO knockdown mice, the antiaging effects of SGI‐1027 were largely abrogated. Therefore, the KLOTHO recovery is critical for the antiaging effects of DNA demethylation. Collectively, our data indicate that aberrant DNMT1/3a/3b elevations and the resultant suppression of antiaging factors contribute significantly to epigenetic renal aging, which might be targeted for epigenetic intervention by synthetic or natural DNA‐demethylating agents. Epigenetic cues upregulate DNMT1, DNMT3a, and DNMT3b, resulting in the promoter hypermethylation and expression suppressions of KLOTHO and NRF2, which accelerate D‐gal‐induced renal aging. DNA hypomethylations by DNA‐demethylating agents correct the epigenetic alterations, recover the of KLOTHO and NRF2 losses, and reduce the renal aging alterations.
... Consistent with these data, oocytes lack DGCR8 (Pasha), which is necessary for microRNA pathways [239]. It was also demonstrated that many environmental factors contribute to the variations in the epigenome, but diet and early life experiences are key modulators of epigenome, which may initiate the development of the disease [234,[249][250][251][252][253][254][255][256][257]. ...
Article
Full-text available
Gestational diabetes mellitus (GDM), one of the major pregnancy-related complications, characterized as a transitory form of diabetes induced by insulin resistance accompanied by a low/absent pancreatic beta-cell compensatory adaptation to the increased insulin demand, causes the acute, long-term, and transgenerational health complications. The aim of the study was to assess if alterations in gene expression of microRNAs associated with diabetes/cardiovascular/cerebrovascular diseases are present in whole peripheral blood of children aged 3–11 years descending from GDM complicated pregnancies. A substantially altered microRNA expression profile was found in children descending from GDM complicated pregnancies. Almost all microRNAs with the exception of miR-92a-3p, miR-155-5p, and miR-210-3p were upregulated. The microRNA expression profile also differed between children after normal and GDM complicated pregnancies in relation to the presence of overweight/obesity, prehypertension/hypertension, and/or valve problems and heart defects. Always, screening based on the combination of microRNAs was superior over using individual microRNAs, since at 10.0% false positive rate it was able to identify a large proportion of children with an aberrant microRNA expression profile (88.14% regardless of clinical findings, 75.41% with normal clinical findings, and 96.49% with abnormal clinical findings). In addition, the higher incidence of valve problems and heart defects was found in children with a prior exposure to GDM. The extensive file of predicted targets of all microRNAs aberrantly expressed in children descending from GDM complicated pregnancies indicates that a large group of these genes is involved in ontologies of diabetes/cardiovascular/cerebrovascular diseases. In general, children with a prior exposure to GDM are at higher risk of later development of diabetes mellitus and cardiovascular/cerebrovascular diseases, and would benefit from dispensarisation as well as implementation of primary prevention strategies.
... Epigenetic changes can be triggered by several environmental factors, such as diet (Mathers et al., 2010), pollution (Christensen and Marsit, 2011), smoking (Talikka et al., 2012), that can be labeled generically as ''stressors,'' referring to the neutral, adaptive meaning of the term (Cabib and Puglisi-Allegra, 2012). Epigenetic aberrations have been implicated in many diseases, primarily cancer but also cardiovascular, autoimmune, metabolic and neurodegenerative diseases, often with particular regard to aging (van Otterdijk et al., 2013;Jung and Pfeifer, 2015). ...
Article
Full-text available
Parents’ stressful experiences can influence an offspring’s vulnerability to many pathological conditions, including psychopathologies, and their effects may even endure for several generations. Nevertheless, the cause of this phenomenon has not been determined, and only recently have scientists turned to epigenetics to answer this question. There is extensive literature on epigenetics, but no consensus exists with regard to how and what can (and must) be considered to study and define epigenetics processes and their inheritance. In this work, we aimed to clarify and systematize these concepts. To this end, we analyzed the dynamics of epigenetic changes over time in detail and defined three types of epigenetics: a direct form of epigenetics (DE) and two indirect epigenetic processes—within (WIE) and across (AIE). DE refers to changes that occur in the lifespan of an individual, due to direct experiences with his environment. WIE concerns changes that occur inside of the womb, due to events during gestation. Finally, AIE defines changes that affect the individual’s predecessors (parents, grandparents, etc.), due to events that occur even long before conception and that are somehow (e.g., through gametes, the intrauterine environment setting) transmitted across generations. This distinction allows us to organize the main body of epigenetic evidence according to these categories and then focus on the latter (AIE), referring to it as a faster route of informational transmission across generations—compared with genetic inheritance—that guides human evolution in a Lamarckian (i.e., experience-dependent) manner. Of the molecular processes that are implicated in this phenomenon, well-known (methylation) and novel (non-coding RNA, ncRNA) regulatory mechanisms are converging. Our discussion of the chief methods that are used to study epigenetic inheritance highlights the most compelling technical and theoretical problems of this discipline. Experimental suggestions to expand this field are provided, and their practical and ethical implications are discussed extensively.
... This epigenetic phenomenon, in which the addition of a methyl group to cytosine forms 5-methylcytosine, occurs mainly at CpG dinucleotides [1]. Biological ageing is reflected in the epigenome and is characterised by a global hypomethylation [2], but certain regions in CpG islands undergo hypermethylation [3]. Throughout the genome, many CpG sites have been uncovered of which the methylation status is highly correlated with chronological age. ...
Article
DNA methylation is a promising biomarker for forensic age prediction. A challenge that has emerged in recent studies is the fact that prediction errors become larger with increasing age due to interindividual differences in epigenetic ageing rates. This phenomenon of non-constant variance or heteroscedasticity violates an assumption of the often used method of ordinary least squares (OLS) regression. The aim of this study was to evaluate alternative statistical methods that do take heteroscedasticity into account in order to provide more accurate, age-dependent prediction intervals. A weighted least squares (WLS) regression is proposed as well as a quantile regression model. Their performances were compared against an OLS regression model based on the same dataset. Both models provided age-dependent prediction intervals which account for the increasing variance with age, but WLS regression performed better in terms of success rate in the current dataset. However, quantile regression might be a preferred method when dealing with a variance that is not only non-constant, but also not normally distributed. Ultimately the choice of which model to use should depend on the observed characteristics of the data.
... Various disturbances of the epigenetic program may have a negative effect, increasing the risk of developing various chronic diseases and developmental disorders. Thus, it has been shown that DNA methylation changes are associated with many common chronic and age-related diseases (Calvanese et al., 2009;Schneider et al., 2015;van Otterdijk et al., 2013). ...
Article
Full-text available
Epigenetic regulation plays an important role in development, at the embryonic stages and later during the lifespan. Some epigenetic marks are highly conserved throughout the lifespan whereas others are closely associated with specific age periods and/or particular environmental factors. Little is known about the dynamics of epigenetic regulation during childhood, especially during the period of rapid early development. Our study was aimed to determine whether the developmental program at the early stages of human development is accompanied by significant changes in the systems of genome regulation, specifically, by genome-wide changes in DNA methylation. Using a sequencing approach (MBD-seq) we investigated genome-wide DNA methylation patterns in the T-lymphocytes of three healthy toddlers at two timepoints within the second year of life. Pairwise comparisons of the methylation patterns across the individuals and time points was conducted to determine common longitudinal changes in the DNA methylation patterns. Despite relatively high interindividual variability in their epigenetic profiles and the dynamics of these profiles during the second year of life, all children showed consistent changes in the DNA methylation patterns of genes involved in the control of the immune system and genes related to the development of the CNS. Thereby, we provide evidence that early development might be accompanied by epigenetic changes in specific functional groups of genes; many such epigenetic changes appear to be related to the rapid development of the CNS.
Article
DNA methylation clocks estimate biological age based on DNA methylation profiles. This study developed a mathematical model to describe DNA methylation aging and the establishment of a pan-tissue DNA methylation clock. The model simulates the aging dynamics of DNA methylation profiles based on passive demethylation as well as the process of cross-sectional bulk data acquisition. As a result, this study identified two conditions under which the pan-tissue DNA methylation clock can successfully predict biological age: one condition is that the target tissues are sufficiently well represented in the training dataset, and the other condition is that the target sample contains cell subsets that are common among different tissues. When either of these conditions is met, the clock performs well. It is also suggested that the epigenetic age of all samples in the target tissue tends to be either over or underestimated when biological age prediction fails. The model can reveal the statistical characteristics of DNA methylation clocks.
Article
Dioxins and dioxin-like compounds are persistent organic pollutants (POPs) and technogenic ecotoxicants, the most dangerous of which is 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). A peculiar feature of the considered genotoxicants that distinguishes them from other POPs is hormone-like activity, which is realized through binding to a special cellular protein, the Aryl hydrocarbon Receptor (AhR). In the present study, the phenomenological aspects of DNA methylation changes induced by dioxins and dioxin-like compounds and revealed in the studies in vitro and in vivo are considered. In animal models, multigenerational and transgenerational effects of dioxin-induced locus-specific DNA methylation changes and their association with reproductive dysfunctions and congenital malformations were firmly established. The importance of investigations of the long-term epigenetic consequences of human exposure to dioxins and the potential value of such studies for preventive diagnostics of somatic and reproductive pathologies are discussed.
Article
BACKGROUND: Melatonin (MT) is an endogenous indoleamine that regulates senescence progression and stress response in plants. OBJECTIVE: Here, we investigated the effect of MT on the shelf-life and postharvest quality of table grapes (Vitis labrusca L. cv. ‘Fengzao’). METHODS: After harvesting, ‘Fengzao’ grapes were immersed in MT solution at various concentrations (0 [as control], 0.05, 0.1, 0.5 and 1.0 mM for 2 h and stored at 24±1 for 15 days. Physiological indicators including weight loss rate, firmness, contents of total soluble solids (TSSs), ascorbic acid (AsA), malondialdehyde (MDA), hydrogen peroxide (H2O2), and activities of catalase (CAT), ascorbate peroxidase (APX), and superoxide dismutase (SOD) were investigated. Additionally, the DNA methylation rate of ‘Fengzao’ grapes were measured using the methylation-sensitive amplification polymorphism (MSAP) technique. RESULTS: Application of MT effectively delayed grape senescence in all treatment groups compared with the control, with the longest delay observed in the 0.5 mM treatment. Additionally, the rate of DNA methylation decreased in all the 0.5 mM MT treatment groups, indicating a potential role of MT in demethylation. CONCLUSIONS: Our results suggest that the exogenous application of MT can delay the senescence of grapes during postharvest.
Article
Full-text available
Aberrant methylation of CpG islands (CGI) occurs in many genes expressed in colonic epithelial cells, and may contribute to the dysregulation of signalling pathways associated with carcinogenesis. This cross-sectional study assessed the relative importance of age, nutritional exposures and other environmental factors in the development of CGI methylation. Rectal biopsies were obtained from 185 individuals (84 male, 101 female) shown to be free of colorectal disease, and for whom measurements of age, body size, nutritional status and blood cell counts were available. We used quantitative DNA methylation analysis combined with multivariate modelling to investigate the relationships between nutritional, anthropometric and metabolic factors and the CGI methylation of 11 genes, together with LINE-1 as an index of global DNA methylation. Age was a consistent predictor of CGI methylation for 9/11 genes but significant positive associations with folate status and negative associations with vitamin D and selenium status were also identified for several genes. There was evidence for positive associations with blood monocyte levels and anthropometric factors for some genes. In general, CGI methylation was higher in males than in females and differential effects of age and other factors on methylation in males and females were identified. In conclusion, levels of age-related CGI methylation in the healthy human rectal mucosa are influenced by gender, the availability of folate, vitamin D and selenium, and perhaps by factors related to systemic inflammation. © 2012 The Authors Aging Cell © 2012 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland.
Article
Full-text available
HACE1 is an E3 ubiquitin ligase located in 6q21, the genomic region frequently deleted in natural killer (NK) cell malignancies. Here, we report HACE1 as a candidate tumor suppressor gene silenced through a combination of deletion and cytosine phosphate guanine island hypermethylation. We detected deletion of HACE1 in malignant NK cell lines (6 of 9, 67%) and primary biopsies (5 of 15, 33%) by quantitative PCR, with most of the specimen showing cytosine phosphate guanine island hypermethylation in the remaining allele, leading to low mRNA transcription. The ectopic expression of HACE1 in an HACE1-null NK cell line led to apoptosis and G(2)/M cell cycle arrest. Moreover, HACE1 expression was up-regulated in IL-2-activated normal NK cells and NK cells cocultured with an engineered NK cell target, K562 Clone 9.mbIL21, suggesting its role in the regulation of NK cell homeostasis. In conclusion, HACE1 is another potent tumor suppressor gene located within the 6q21 region, and loss of function of multiple tumor suppressor genes within 6q21 may be a critical determinant of NK cell lymphomagenesis.
Article
Full-text available
Background Aberrant CpG island promoter DNA hypermethylation is frequently observed in cancer and is believed to contribute to tumor progression by silencing the expression of tumor suppressor genes. Previously, we observed that promoter hypermethylation in breast cancer reflects cell lineage rather than tumor progression and occurs at genes that are already repressed in a lineage-specific manner. To investigate the generality of our observation we analyzed the methylation profiles of 1,154 cancers from 7 different tissue types. Results We find that 1,009 genes are prone to hypermethylation in these 7 types of cancer. Nearly half of these genes varied in their susceptibility to hypermethylation between different cancer types. We show that the expression status of hypermethylation prone genes in the originator tissue determines their propensity to become hypermethylated in cancer; specifically, genes that are normally repressed in a tissue are prone to hypermethylation in cancers derived from that tissue. We also show that the promoter regions of hypermethylation-prone genes are depleted of repetitive elements and that DNA sequence around the same promoters is evolutionarily conserved. We propose that these two characteristics reflect tissue-specific gene promoter architecture regulating the expression of these hypermethylation prone genes in normal tissues. Conclusions As aberrantly hypermethylated genes are already repressed in pre-cancerous tissue, we suggest that their hypermethylation does not directly contribute to cancer development via silencing. Instead aberrant hypermethylation reflects developmental history and the perturbation of epigenetic mechanisms maintaining these repressed promoters in a hypomethylated state in normal cells.
Article
Full-text available
Although promoter hypermethylation has been an accepted means of tumor suppressor gene inactivation, activation of otherwise normally repressed proto-oncogenes by promoter demethylation has been infrequently documented. In this study we performed an integrative, whole-genome analysis for discovery of epigenetically activated proto-oncogenes in head and neck cancer tumors. We used the 47K GeneChip U133 Plus 2.0 Affymetrix expression microarray platform to obtain re-expression data from 5-aza treated normal cell line and expression data from primary head and neck squamous cell carcinoma (HNSCC) tumor tissues and normal mucosa tissues. We then investigated candidate genes by screening promoter regions for CpG islands and bisulfite sequencing followed by QUMSP and RT PCR for the best candidate genes. Finally, functional studies were performed on the top candidate gene. From the top 178 screened candidates 96 had CpG islands in their promoter region. Seven candidate genes showed promoter region methylation in normal mucosa samples and promoter demethylation in a small cohort of primary HNSCC tissues. We then studied the demethylation of the top 3 candidate genes in an expanded cohort of 76 HNSCC tissue samples and 17 normal mucosa samples. We identified MAGEB2 as having significant promoter demethylation in primary head and neck squamous cell carcinoma tissues. We then found significantly higher expression of MAGEB2 in tumors in a separate cohort of 73 primary HNSCC tissues and 31 normal tissues. Finally, we found that MAGEB2 has growth promoting effects on minimally transformed oral keratinocyte cell lines but not a definite effect on HNSCC cell lines. In conclusion, we identified MAGEB2 as activated by promoter demethylation in HNSCCand demonstrates growth promoting effects in a minimally transformed oral keratinocyte cell line. More studies are needed to evaluate MAGBE2's exact role in HNSCC.
Article
Full-text available
In chronic lymphocytic leukemia (CLL), the microenvironment influences gene expression patterns; however, knowledge is limited regarding the extent to which methylation changes with time and exposure to specific microenvironments. Using high-resolution 450K arrays, we provide the most comprehensive DNA methylation study of CLL to date, analyzing paired diagnostic/follow-up samples from IGHV-mutated/untreated and IGHV-unmutated/treated patients (n=36) and patient-matched peripheral blood and lymph node samples (n=20). On an unprecedented scale, we revealed 2239 differentially methylated CpG sites between IGHV-mutated and unmutated patients, with the majority of sites positioned outside annotated CpG islands. Intriguingly, CLL prognostic genes (for example, CLLU1, LPL, ZAP70 and NOTCH1), epigenetic regulator (for example, HDAC9, HDAC4 and DNMT3B), B-cell signaling (for example, IBTK) and numerous TGF-β and NF-κB/TNF pathway genes were alternatively methylated between subgroups. Contrary, DNA methylation over time was deemed rather stable with few recurrent changes noted within subgroups. Although a larger number of non-recurrent changes were identified among IGHV-unmutated relative to mutated cases over time, these equated to a low global change. Similarly, few changes were identified between compartment cases. Altogether, we reveal CLL subgroups to display unique methylation profiles and unveil methylation as relatively stable over time and similar within different CLL compartments, implying aberrant methylation as an early leukemogenic event.Leukemia advance online publication, 18 September 2012; doi:10.1038/leu.2012.245.
Article
Epigenetic phenomena are defined as heritable mechanisms that establish and maintain mitotically stable patterns of gene expression without modifying the base sequence of DNA. The major epigenetic features of mammalian cells include DNA methylation, post-translational histone modifications and RNA-based mechanisms including those controlled by small non-coding RNAs (miRNAs). The impact of epigenetic mechanisms in cardiovascular pathophysiology is now emerging as a major player in the interface between genotype to phenotype variability. This topic of research has strict implications on disease development and progression, and opens up possible novel preventive strategies in cardiovascular disease. An important aspect of epigenetic mechanisms is that they are potentially reversible and may be influenced by nutritional-environmental factors and through gene-environment interactions, all of which have an important role in complex, multifactorial diseases such as those affecting the cardiovascular system. Gene expression regulation through the interplay of DNA methylation and histone modifications is well-established, although the knowledge about the function of epigenetic signatures in cardiovascular disease is still largely unexplored. The study of epigenetic markers is, therefore, a very promising frontier of science which may aid in a deeper understanding of molecular mechanisms underlying the modulation of gene expression in the biomolecule pathways linked to cardiovascular diseases. This review focuses on up-to-date knowledge pertaining to the role of epigenetics, from DNA methylation to miRNAs, in major cardiovascular diseases such as ischemic heart disease, hypertension, heart failure and stroke.
Article
Histone methylation is one epigenetic modification of an inactive X chromosome (Xi). Histone H3 lysine 9 dimethylation (H3K9me) and histone H3 lysine 27 trimethylation (H3K27me) are both associated with the chromatin of gene-silenced regions in the X chromosome and with X inactivation. Studies have shown that H3K9me is supposedly an early mark on the X chromosome during inactivation. Here, we examined the distribution and enrichment profiles of H3K9me and H3K27me by indirect immunofluorescence. We found that H3K9me appears to have a broad distribution throughout the whole genome, but is specific, to a certain extent, to the Xi in WI38 cells. In contrast, H3K27me is highly specific to the entire Xi, which differs significantly from other areas of the nucleus. Thus, H3K27me is more suitable as an epigenetic mark than H3K9me. The chromatin immunoprecipitation analyses also showed that H3K27me predominates on the inactive genes of the X chromosome. Additionally, we compared the levels of H3K9me and H3K27me in four X-linked genes and two autosomal genes between the normal cells (WI38) and the tumor cells (HeLa). The results revealed that the methylation levels of the inactive genes (POLA and OCRL) in tumor cells (HeLa) were lower than those in normal cells (WI38) and that the methylation levels of the Xi inactivation-avoidance genes (SMCX and ZFX) and autosomal genes (Myc and β-actin) varied widely in tumor cells (HeLa). These events may be significant for cancer cell development and contribute to the characteristics of tumor cells.
Article
The exploration of brain epigenomes, which consist of various types of DNA methylation and covalent histone modifications, is providing new and unprecedented insights into the mechanisms of neural development, neurological disease and aging. Traditionally, chromatin defects in the brain were considered static lesions of early development that occurred in the context of rare genetic syndromes, but it is now clear that mutations and maladaptations of the epigenetic machinery cover a much wider continuum that includes adult-onset neurodegenerative disease. Here, we describe how recent advances in neuroepigenetics have contributed to an improved mechanistic understanding of developmental and degenerative brain disorders, and we discuss how they could influence the development of future therapies for these conditions.
Article
DNA methylation is a mechanism of epigenetic regulation that is common to all vertebrates. Functional studies underscore its relevance for tissue homeostasis, but the global dynamics of DNA methylation during in vivo differentiation remain underexplored. Here we report high-resolution DNA methylation maps of adult stem cell differentiation in mouse, focusing on 19 purified cell populations of the blood and skin lineages. DNA methylation changes were locus specific and relatively modest in magnitude. They frequently overlapped with lineage-associated transcription factors and their binding sites, suggesting that DNA methylation may protect cells from aberrant transcription factor activation. DNA methylation and gene expression provided complementary information, and combining the two enabled us to infer the cellular differentiation hierarchy of the blood lineage directly from genome-scale data. In summary, these results demonstrate that in vivo differentiation of adult stem cells is associated with small but informative changes in the genomic distribution of DNA methylation.