Anda di halaman 1dari 12

Pflugers Arch - Eur J Physiol (2010) 459:247258

DOI 10.1007/s00424-009-0730-7

MOLECULAR AND GENOMIC PHYSIOLOGY

Epigenetic factors in aging and longevity


Silvia Gravina & Jan Vijg

Received: 4 August 2009 / Accepted: 26 August 2009 / Published online: 19 September 2009
# Springer-Verlag 2009

Abstract Epigenetics refers to phenotypic changes caused disease and aging. During various stages of mammalian
by mechanisms that are unrelated to changes in the development, epigenetic patterns, most notably DNA
underlying DNA sequence, most notably chromatin remod- methylation, are reprogrammed genome-wide, generating
eling driven by histone modifications, and DNA methyla- cells with a broad potential of differentiating into various
tion. Such variation is transmitted by cell division, but lineages [82, 88]. While epigenetic changes are essential for
generally not passed on through the germ line. An development and differentiation, they can also arise later in
increasing body of evidence supports a role for epigenetic life either by non-random mechanisms, such as responses to
changes in the etiology of aging and its associated disease environmental change, or through stochastic errors in main-
sequelae. Here, we review the role of epigenetics in aging taining fixed patterns of DNA or histone modification.
and longevity with a focus on DNA methylation. Increased Epigenetic modifications, such as changes in DNA methyla-
understanding of those aging-related processes that are tion or histone modification, can impact on chromatin folding
driven by epigenetic mechanisms will allow for the or non-coding RNAs. Probably the best-documented example
development of novel epigenetic-based diagnostic, preven- of epigenetic modification and the main topic of this review is
tive, and therapeutic strategies for age-related diseases. DNA methylation, which consists of a covalent addition of a
methyl group to the cytosine.
Keywords Aging . Epigenetics . DNA methylation . DNA methylation is carried out by DNA methyltrans-
Lifespan ferases (DNMTs); at least three functional DNMTs have
been identified in eukaryotic systems. DNMT1 is the
enzyme primarily involved in the maintenance of DNA
Introduction methylation patterns during replication, and it preferentially
methylates hemimethylated DNA [83]. Other known
Epigenetic changes involve transmissible alterations in functional methyltransferases are DNMT3a and DNMT3b,
gene expression caused by mechanisms other than changes with higher de novo methylation activity than DNMT1.
in DNA sequence. While, in the past, human biology was DNMT3a and DNMT3b contribute to de novo methylation
considered the interplay between the DNA sequence of during embryogenesis [77].
one's genome and environmental variation, evidence has Clusters of unmethylated CpG pairs, termed CpG
been accumulating that such epigenetic modifications as islands, are located in approximately 40% of promoters of
DNA methylation and histone modification can have a mammalian genes, most notably housekeeping genes, and
primary role in phenotypic outcomes, including human are targets of transcription factors binding the unmethylated
DNA and initiating transcription. By contrast, methylated
S. Gravina (*) : J. Vijg (*) CpGs are usually linked to silent DNA. DNA methylation
Department of Genetics, Albert Einstein College of Medicine, represses transcription directly by preventing the binding of
1301 Morris Park Ave, Bronx,
New York, NY 10461, USA
transcription factors to promoters [108] or indirectly by
e-mail: silvia.gravina@einstein.yu.edu recruiting methyl-CpG binding proteins [73]. While both
e-mail: jan.vijg@einstein.yu.edu mechanisms may contribute to methylation-mediated gene
248 Pflugers Arch - Eur J Physiol (2010) 459:247258

silencing, it has been shown that many fully methylated Indeed, epigenetic errors can cause disease by chromatin
genes can be transcribed at nearly normal rates under remodelling, leading to aberrant gene expression. This is
conditions where methyl-CpG binding proteins are effec- especially important during development, where mistakes
tively absent [9]. in the series of carefully orchestrated changes in epigenetic
DNA methylation patterns are intricately related to patterns can lead to congenital disorders and multisystem
changes in histone modification. Indeed, these two epige- pediatric syndromes [101]. However, epigenetic abnormal-
netic control systems are two sides of the same coin: ities have also been found to be causative factors in adult-
chromatin remodeling. The amino-terminal region of onset diseases, including neurologic disorders, cancer, and
histones is a hot spot for posttranslational modifications autoimmune disease [84, 92]. Interestingly, there is evidence
that affect their interactions with DNA, other histones, or that epigenetic defects can also contribute to segmental
chromatin remodeling proteins. The most studied of the progeroid syndromes and, possibly, to aging itself [49]. Here,
posttranslational modifications of histones are methylation we review the epigenetics of aging and lifespan, with a
and acetylation of lysine residues in the amino-terminal particular focus on DNA methylation, the best understood
tails of histones H3 and H4 [31]. Histone methylation is example of epigenetics. We discuss the emerging role of
catalyzed by histone lysine methyltransferase (HLMTase) epigenetics in susceptibility to age-related disease and
[96], while histone acetyltransferase (HAT) and histone consider the possibility that aging in part is driven by an
deacetylases (HDACs) regulate, respectively, the acetyla- epigenetic-mediated loss of phenotypic plasticity (Fig. 1).
tion and deacetylation of lysine residues [19]. In general,
increased acetylation of the histones in gene control regions
correlates with transcriptional activity, while methylation of Chromatin properties and systematic vs stochastic
histones is primarily found in transcriptionally silenced change
heterochromatin regions. Methylation of histone H3 at
lysine 9 (H3K9) usually leads to transcriptional repression It is increasingly being appreciated that chromatin is a
and can inhibit acetylation of the H3 tail at several lysines highly dynamic entity. Chromatin remodeling is most
[104]. The processes of histone modification and DNA dramatic during development, when genome-wide changes
methylation are interdependent and both contribute to the in epigenetic marks orchestrate chromatin in a way that
overall state of chromatin and its epigenetic control of gene allows it to generate different organs and tissues providing
expression. For example, DNA methylation acts synergis- the broad array of functions needed by the organism to
tically with histone deacetylation to repress transcription. maintain itself and reproduce. Such changes are systematic
The N-terminal domain of DNMT1 can bind to the and are clearly purposeful. As will be discussed below,
HDACs, thereby promoting histone deacetylating and similar changes may occur during normal aging for reasons
suppressing gene transcription [87]. Fuks et al. have shown that may include signals from the environment, resulting in
that DNMT3a can also recruit HDACs, thus leading to a stress response. However, like the genome, the epigenome
transcriptional silencing [35]. is also subject to stochastic changes, called epimutations by
One example of the power of epigenetics in regulating Holliday [48]. As we will see later, such epimutations have
gene expression is the phenomenon of position effect been demonstrated to be critically important as causal
variegation (PEV) [40]. PEV refers to the mosaic expres- factors in the age-related increase in cancer, but could also
sion of a gene in otherwise homogeneous cell populations. play a key role in driving other aging processes.
PEV was initially described in X-ray-irradiated fruit flies Stochastic changes in the epigenome could be due to
undergoing a chromosomal inversion, which places the errors in maintaining established patterns of DNA methyl-
white gene, normally located on the distal tip of the X ation or histone modification. How frequent are such
chromosome, close to the pericentric heterochromatin. mistakes? For DNA methylation, direct analysis is difficult
Normally, white is expressed in the adult eye resulting in since methylation patterns need to be derived from the two
a red eye phenotype, but due to the inversion, the eye complementary strands of an individual DNA molecule.
contains patches of red and white tissue because the Using hairpin-bisulfite PCR, specifically developed for this
expression of white is variegating [72]. PEV has also been purpose, Laird et al. analyzed the fidelity of methylation
documented in other model organisms [23]. transmission in the CpG island of the X-linked FMR1 gene
As it now appears, the epigenome is much more in human lymphocytes [61]. Their results indicate that the
dynamic than its sequence-based counterpart, undergoing fidelity of inheritance of the methylated state of each
extensive alterations during development, differentiation, cytosine in the hypermethylated allele is 0.96 per cell
and a variety of cellular responses that are essential for life. division. When looking at the active, unmethylated X
The large volume of epigenomic transactions, including chromosomal locus, they found that the fidelity of
maintenance, suggests a fairly high chance of errors. inheritance of the unmethylated state was as high as 0.99.
Pflugers Arch - Eur J Physiol (2010) 459:247258 249

Fig. 1 Epigenetics of aging.


DNA hypomethylation can
initiate chromosome instability
while DNA hypermethylation in
promoter regions suppresses
expression of normal genes
(e.g., tumor suppressor genes).
Age-related hypermethylation or
hypomethylation could
theoretically impair or enhance
normal gene responsiveness to
environmental signals, thus in
turn contributing to a
generalized functional decline

However, for the hypermethylated locus on the inactive X somatic functions. This makes it a more likely candidate for
chromosome, the fidelity of inheritance of the unmethylated errors than its more stable DNA sequence counterpart,
state was only 0.83 (a de novo methylation efficiency of changes in which have been well-documented and increase
0.17). These results clearly show that errors in maintaining during aging [17, 25, 37, 67, 102]. Indeed, epigenomic
DNA methylation occur. Thus far, similar information for alterations are now increasingly recognized as part of aging
histone modification patterns is lacking. and its associated pathologic phenotypes. Most of the
The stochastic nature of epigenetic changes is consistent available data on epigenetic changes in human or animal
with new findings demonstrating a stochastic component of cells and tissues involve systematic changes, i.e., affecting
gene expression regulation. In the past few years, through the exact same site in a large enough fraction of the cell
work carried out in our laboratory [2, 12], it was shown that population to be detectable through current assays. Atten-
aging is associated with increased stochastic deregulation of tion has thus far been focused on changes in DNA
cellular gene expression, giving rise to cellular mosaics of methylation at cytosines, primarily in the aforementioned
diverging transcriptomes. This could reflect epigenetic drift, CpG islands, which are usually unmethylated in normal
affecting function and cellular responses to environmental cells. Early studies indicated a general demethylation in
challenges (Fig. 2). most vertebrate tissues with aging [86, 111]. In mitotically
Hence, epigenetic alterations during aging could be active tissue, it is possible that this reflects some deficiency
systematic, as well as stochastic. The former can be in maintenance remethylation, as this normally occurs after
addressed using advanced platforms based on genome- DNA replication. Such a general loss of 5-methylcytosine
wide analysis. However, stochastic changes need to be during aging may de-suppress silenced retrotransposons,
analyzed at the single-cell or single-molecule level, and such as L1, to increase genome instability [3, 68].
suitable, genome-wide technology for that purpose is still There are numerous reports in the literature on changes
lacking. Indeed, cell-to-cell heterogeneity is considered in methylation status of individual genes during aging. Tra
one of the major stumbling blocks of biological inves- et al. [100] compared more than 2,000 loci in T
tigations of aging [66]. Below, we will also discuss the lymphocytes isolated from newborn, middle-aged, and elderly
need for single-cell epigenomics and the various people, and found that 29 loci (approximately 1%) changed
approaches being taken to overcome current limitations. DNA methylation status with age: 23 cases of hypermethyla-
tion and six cases of hypomethylation. Most of these
methylation changes happened before middle age. Interestingly,
The emerging role of epigenetics in aging and longevity the same CpG islands affected in human T cells also changed
with age in other human tissues (esophagus, lung, and
Epigenetic information is only partially stable and destined pancreas), suggesting that methylation instability is locus-
to change during development and in driving essential specific. The study was carried out using restriction landmark
250 Pflugers Arch - Eur J Physiol (2010) 459:247258

Errors in DNA methylation levels across the lifespan for eight out of 50 loci,
Diet
methylation with another 18 loci defined by a sharp rise within the first
Environment maintenance
months or years after birth. The authors also observed that
ALU and other repetitive elements either showed a
significant decrease in DNA methylation during the first
decade of life, followed by relatively little change during
subsequent maturation and aging, or showed relatively little
change across the lifespan [95].
Genes that might be more vulnerable to epigenetic changes
could be those of which one allele is normally suppressed
while the other one is active. For example, imprinted genes, a
SYSTEMATIC STOCHASTIC subset of mammalian genes that is monoallelically expressed
in a parent-of-origin manner due to epigenetic mechanisms
that confer a parent-specific memory to individual cells, could
suffer from derepression of the inactive allele [30]. A list of
all human imprinted genes can be found in the database
http://igc.otago.ac.nz/Search.html. Another special case of
allele-specific epigenetic suppression is X-chromosome
inactivation, a mechanism by which one of the two copies
of the X chromosome present in female mammals is silenced
by packaging into transcriptionally inactive heterochromatin
[76]. Hence, it is conceivable that, during aging, genes on
the silent chromosome are derepressed by demethylation or
incomplete maintenance of methylation patterns. Indeed,
Homogeneous gene Transcriptional multiple authors have reported evidence for a relaxation of
expression profile noise epigenetic control of X-linked and imprinted gene expres-
Fig. 2 Epigenetic changes and effects of stochasticity. Recent findings sion during aging [7, 106].
support the idea that aging is characterized by transcriptional instability, For studying the reactivation of inactive X-linked genes,
which may be due in part to the loss of normal epigenetic patterns. In the
mice are often used harboring an X-autosomal translocation.
past, we have shown that aging is associated with increased molecular
randomization of cellular genomes and transcriptomes, which is In such mice, it is always the normal, non-translocated X
consistent with one of the most attractive concepts in the field: the idea chromosome that is inactivated. Using various approaches for
that aging is a time-dependent process of dysdifferentiation detecting allele-specific transcripts, this makes it possible to
verify whether, during aging, a suppressed gene is re-
genome scanning (RLGS), a genome-wide method based on activated. For example, in the mouse, an approximately 50-
two-dimensional separation of DNA fragments obtained by fold age-related increase in liver cells that had re-activated the
NotI digestion, which is methylation-sensitive. In the same gene ornithine transcarbamoylase on the inactive X chromo-
study, the authors also evaluated methylation patterns in T cells some has been observed. This was ascribed to an age-related
and various tissues from aging mice, but could not detect sig- reduction in DNA methylation [106]. Age-associated loss of
nificant changes in the murine tissues, thus revealing a possible epigenetic repression of X-linked genes has been analyzed
difference in stability between mice and humans. However, quantitatively for the gene Atp7a in various tissues of the
other studies, as we will discuss later, in mice and rats, did find same X-autosomal translocation mouse model [7]. The
evidence for epigenetic changes during aging [7, 106]. results indicated a mean organ-specific relaxation of gene
In a recent study, Siegmund et al. examined DNA repression in the oldest group of mice of up to 2.2% (this is
methylation changes during the course of development, the fraction of transcripts from the inactive X relative to its
maturation, and aging of the human cerebral cortex. The active sister chromosome). Using the same quantitative
authors looked at DNA methylation status at 50 loci in the methodology, Fu et al. observed a similar relaxation of gene
temporal neocortex of 125 subjects ranging in age from repression for the imprinted Igf2 gene, the expression of
17 weeks of gestation to 104 years old, including two which increases in aging mouse liver up to 6.7% [34].
psychiatric disease cohorts, i.e., Alzheimer's disease and Another model ideally suited for studying age-related
schizophrenia. Most of the loci analyzed represent promoter variation in the epigenome is the monozygotic twin. In a
CpG islands of genes expressed in the cerebral cortex; a recent study, global and locus-specific epigenetic differ-
portion of these genes is also implicated in cancer biology. ences between identical twins were reported to arise with
The authors observed a robust and progressive rise in DNA age [32]. In this study, involving 80 twins varying in age
Pflugers Arch - Eur J Physiol (2010) 459:247258 251

from 3 to 74 years, a variety of epigenetic markers, This epigenetic deregulation could provide the tumor cell with
including histone modification, X-chromosome inactiva- a growth advantage, allowing it to metastasize and further
tion, total genomic 5-methylcytosine content, sequence- increase its genetic instability, which in turn will lead to
specific DNA methylation and global DNA methylation further changes advantageous to the tumor.
status (by RLGS), were assessed in blood in comparison Interestingly, Wang et al. [105] provided evidence for a
with measures of gene expression (using microarrays and link between epigenomic relaxation and Alzheimer's disease.
quantitative real-time PCR). The results indicate that, while Using MALDI-TOF mass spectrometry (see further below for
these monozygotic twins are epigenetically indistinguish- a description of this method), these investigators analyzed the
able early in life, at older age, they start to exhibit promoter methylation of 12 potential susceptibility genes for
remarkable differences in overall content and genomic Alzheimer's disease in post-mortem late-onset Alzheimer's
distribution of 5-methylcytosine DNA and histone acetyla- disease (LOAD) brain samples and lymphocytes, as com-
tion, affecting their gene-expression profiles. This diver- pared to normal, healthy controls, of an age range of between
gence of genetically identical individuals over time could 65 and 85 years. The results indicated only minor epigenetic
be stochastic (due, for example, to early, random epimuta- changes in a subset of genes. However, deviations for
tions) or systematic when driven by different cellular individuals from the norm (obtained from the average in the
responses to environmental changes. However, the study control samples) were significantly higher in the LOAD
did not examine the same individuals serially over time, so patients, suggesting an age-specific epigenetic drift that may
we cannot know from these data whether DNA methylation influence age-of-onset and disease progression.
changed or was different historically. Hence, it may be difficult to find epigenetic changes that
In a recent longitudinal study, Bjornsson et al. [8] looked can be associated with defined phenotypic consequences.
at epigenomic changes in blood collected from the same Instead, aging could be associated not with all or nothing
individuals in two separate populations (one from Iceland DNA methylation changes, but with a generalized loss of
and one from Utah, USA) over time. The Iceland cohort was normal epigenetic patterns. This deregulation may contribute
composed of unrelated samples while the Utah samples were to the aging phenotype and perhaps once a critical threshold of
family-based. From measuring global DNA methylation by a epimutations is reached, the tissue or organ, may start to
luminometric methylation assay, a quantitative measurement malfunction. Such decreased epigenetic control during aging
of genome-wide methylation, they observed time-dependent would be in keeping with the so-called dysdifferentiation
changes in global methylation within the same individuals in theory of aging proposed by Richard Cutler [57]. According
the two separate populations, showing decreases, as well as to this hypothesis, the primary process of aging is the time-
increases, in methylation greater than 20% over an 11- to 16- dependent drifting away of cells from their proper state of
year span. Interestingly, in the Utah population, the observed differentiation [78]. In order to test this hypothesis, Cutler
changes in DNA methylation showed a familial clustering. and co-workers assessed whether, during aging, genes that
This clustering occurred for both decreased and increased are normally not expressed in a tissue are gradually
methylation, although it was most striking for decreased derepressed. Indeed, they found an age-dependent increase
methylation. While shared family environment could explain in the expression of the globin gene in non-erythroid tissues
this clustering, most family members did not share house- such as liver and brain, thus suggesting a relaxation of
holds during the majority of the time between samplings. normal gene repression [78].
Therefore, this familiality suggests that methylation stability One of the best examples of the possible clinical
is genetically determined. DNA sequence variation at loci consequences of an accumulation of epigenetic errors during
involved in DNA methylation maintenance may contribute aging is cancer. Aging is one of the highest risk factors for
to this familial clustering. cancer: a hallmark of cancer is that the accumulation of
In conclusion, most or all studies on DNA methylation mutations and epimutations provides the tumor cells with the
during aging support the hypothesis that aging is associated attributes to escape growth control, evade the immune system,
with a relaxation of epigenetic control. However, while this and invade tissues to finally lead to metastatic disease [103].
may contribute to some of the well-documented age-related In his epigenetic progenitor model, Feinberg et al. [29]
phenotypes, such as functional decline and the development proposed that cancer originates from stem cells that have
of age-related diseases, the functional relevance of age-related been primed to become neoplastic through a series of global
epigenetic changes remains largely unknown, possibly with epigenetic disruptions. This is then followed by stochastic,
the exception of cancer. As we will discuss later in the review, genetic, and epigenetic changes that drive tumor progression
epigenetic deregulation has been clearly linked to the etiology [29].
of cancer; for example, hypermethylation has been associated From an epigenetic perspective, cancer is commonly
with the silencing of genes involved in cell cycle regulation, characterized as showing global DNA hypomethylation and
tumor-cell invasion, DNA repair, apoptosis, and cell signaling. site-specific promoter hypermethylation. DNA hypomethy-
252 Pflugers Arch - Eur J Physiol (2010) 459:247258

lation initiates chromosome instability and increased tumor young ones. These results with cells from HGPS patients,
frequency, as has been shown in mouse models [26, 36, as well as normal aged individuals, suggest a causal
91]. It is possible that one of the early epigenetic events, as connection between disrupted nuclear architecture, reduced
proposed by Feinberg et al. [29], consists of DNA hyper- heterochromatin, and human aging.
methylation of tumor suppressor genes, which has been Another interesting example of a possible relationship
described for RB, p16, APC, and MLH1 [5, 6, 54]. For between aging and epigenetic changes comes from studies
example, promoter hypermethylation of p16 has been in the yeast Saccharomyces cerevisiae (Brewer's yeast). In
documented in 15 cancer types, ranging from 9% to 49% this species, a progressive accumulation of extrachromo-
hypermethylation, while the BRCA1 promoter was found somal rDNA circles (ERCs) has been demonstrated to be
hypermethylated in 10%15% of women with no familial the cause of replicative aging [97]. Sir2, a NAD-dependent
breast cancer [53]. Other gene targets are some of the HDAC necessary for transcriptional silencing near telo-
aforementioned imprinted genes, which are expressed only meres and mating type loci [42], has emerged as a key
from one parental allele, representing 1% of autosomal regulator of health and lifespan [55]. It has been shown
genes. Deregulation of imprinted genes has been linked to that, in yeast, deletion of Sir2 shortens while overexpres-
cancer risk, which is due to the haploid state of the sion increases replicative lifespan [55]. The anti-aging
imprinted genes that eliminates any protection against effects of Sir2 in yeast are, at least in part, due to
deleterious effects of recessive mutations [29]. translocation of the Sir2 from telomeres and mating type
Another interesting example of how increased genomic loci to rDNA. These repeats are prone to recombination to
instability and epigenetic defects may contribute to the generate ERCs; the resulting ERCs are segregated to the
aging phenotype is represented by segmental progeroid mother cell, accumulate, and ultimately cause senescence,
syndromes, a group of diseases in which patients develop thus shortening the lifespan. Sir2 prevents recombination
features of aging early in life. One of the best-known and the formation of ERCs, thereby extending lifespan.
progeroid syndromes is Hutchinson-Gilford Progeroid An important function of heterochromatin, therefore,
Syndrome (HGPS, so called childhood progeria) [46]. seems to be the suppression of recombination. This would
HGPS is a rare syndrome characterized by some traits suggest an age-related loss of heterochromatin as a causal
typical of the aging phenotype, such as alopecia, reduced factor in aging, which is in keeping with reported increases
bone density, poor muscle development, and premature in nucleosome spacing with age (in rats) and a lower
death (at age 1215), usually by myocardial infarction or sensitivity to nucleases [56]. Orthologs of Sir2 have anti-
stroke [21]. HGPS is most often caused by a splicing defect aging functions in other species, such as nematodes and
in exon 11 of the LMNA gene, which encodes lamins A flies [85]. The role of the human ortholog SIRT1, in
and C, major structural elements of the nucleus [90]. Cells longevity is not yet known, but there is some evidence
from HGPS patients show dramatic defects in nuclear suggesting that, in mammals, the sirtuins (as the Sir2
envelope structure [60], increased genomic instability, proteins are called) play key roles in many important
DNA damage, and defects in the DNA repair machinery physiological functions, as well as in age-related diseases
[63, 94]. In particular, it appears that, in the cells of [43]. Interestingly, Oberdoerffer et al. [75] have recently
individuals with HGPS, the repair machinery is not as shown that, in mouse embryonic stem (ES) cells treated
efficiently recruited to sites of damage and, consequently, with hydrogen peroxide, the Sir2 ortholog SIRT1 relocates
damage such as DNA breaks is not as efficiently repaired from repeat sequences and gene promoters to sites of DNA
[63]. HGPS cells exhibit an increased amount of nuclei double-strand breaks (DSBs) promoting DNA repair and,
with foci of phorphorylated histone H2AX (-H2AX), hence, genomic stability. Transgenic mice overexpressing a
which is usually considered a marker for DNA double- truncated form of p63 (a homolog of p53) show signs of
strand breaks and also for senescence [15, 63]. HGPS cells premature aging and decreased levels of SIRT1 [98]. This is
also show epigenetics defects, including a decrease in the in keeping with a study by Baur et al., which showed that
heterochromatic markers histone H3 lysine methylation resveratrol, a Sirt1 activator, extends the healthy lifespan of
(H3K9Me) and HP1 protein [89, 90, 94]. Interestingly, mice on a high-fat diet [4].
Scaffidi et al., through a quantitative single-cell analysis,
showed that the percentage of cells with reduced H3K9Me
in cells from old donors (>80 years) was between 40% and New methods for measuring epigenetic changes
90%, comparable to that found in HGPS patient cells and
approximately more than two times higher than the Most of the work reviewed above is based on classical,
percentage found in young donors. In the same study, they single-locus assays for analyzing epigenetic marks. More
showed that the percentage of -H2AX foci is about 3.5 recently, studies on epigenetics have been greatly facilitated
times higher in cells from old individuals compared with by the development of improved methods for the analysis
Pflugers Arch - Eur J Physiol (2010) 459:247258 253

of epigenetic modifications on a global basis. For instance, bases (U or C). Cleavage products are spotted on a chip
it is now possible to profile the sites of DNA methylation or containing a matrix compound to assist in laser deioniza-
various covalent modifications to the histone tails through tion. Within the cleavage products, methylation-dependent
genome-wide analysis either by hybridization to tiling C to T variation appears as G/A generated from the reverse
microarrays (ChIP-chip) [10] or using next-generation strand by base-specific cleavage. These G/A variations
sequencing (ChIP-Seq) [79]. result in a mass difference of 16 Da per CpG site, which is
A substantial effort is underway within the epigenomics easily detected by the MassArray analyzer, resulting in a
community to identify DNA methylation patterns and signal pair pattern from the methylated and non-methylated
histone marks on a genome-wide scale using those methods template DNA (http://www.sequenom.com/). Each assay
in order to provide new insights into the regulation and can read several CpG dinucleotides so that, for example,
dynamics of DNA methylation in genomes. Here, we will a whole promoter can be analyzed in one experiment
focus on DNA methylation assays we have now begun to (Fig. 3).
use routinely in our studies of aging and age-related It has to be noted that, while the methods of epigenetic
diseases. analysis have been greatly improved in the past decade,
The assay we use for the analysis of global DNA allowing the generation of single-base resolution maps of
methylation is termed the Hpa II-tiny fragment Enrichment the entire epigenome, thereby greatly facilitating the
by Ligation-mediated PCR (HELP) assay [58]. The detection of systematic changes, stochastic variation needs
technique compares genome representations generated by to be analyzed at the single cell level. Suitable assays for
the restriction enzyme Hpa II, which will not cut methyl- this purpose are still lacking.
ated CCGG sites, with those generated by its methylation
insensitive isoschizomer MspI, followed by ligation-
mediated PCR. These fragment mixtures are used as probes Epigenetic changes during cell senescence
to hybridize tiling microarrays for assessing the presence or
absence of Hpa II fragments at specific loci, a measure of Cellular senescence, the irreversible cessation of mitotic
hypo- or hypermethylation, respectively. Its built-in control activity, was originally considered as an in vitro model
(Msp I fragments) and the possibility of using customized system for in vivo aging [45]. Currently, cellular senes-
chips make this technique reliable and versatile, i.e., it can cence is known as a stress response and a major tumor
be adapted to examine DNA methylation in certain regions suppressor mechanism [14, 15]. The senescence response is
or in entire genomes. An analytical pipeline for HELP data essentially a DNA damage response and can be induced by
processing written in the R programming language was clastogenic agents, such as ionizing radiation, or activated
recently published [99]. John Greally, the inventor of the oncogenes, i.e., through replicative stress [13, 15, 16].
assay, is presently developing next-generation sequencing Importantly, cell senescence can be triggered by an
approaches to substitute for current tiling array read-outs excessive number of cell divisions, thereby shortening
(personal communication). telomeres and generating double-strand breaks [44, 112].
Many techniques for DNA methylation analysis are This is called replicative senescence and explains why
based on bisulfite treatment of DNA to allow PCR-based human cells in culture have a finite lifespan.
detection of methylated or unmethylated cytosines either Senescence has been linked to a variety of morpholog-
globally or in specific sequences. Sodium bisulfite is used ical changes, such as an enlarged cell shape, decreased
to convert cytosine residues to uracil in single-stranded protein synthesis and degradation, resistance to apoptosis,
DNA, under conditions whereby 5-methylcytosine remains and increased activity of some lysosomal enzymes includ-
non-reactive. The converted DNA can be amplified with ing senescence-associated -galactosidase (SA-gal) [44,
specific primers targeting specific regions of interest (such 50]. Senescent cells can be identified by cytological
as promoter regions), followed by downstream detection markers of senescence-associated DNA damage foci
techniques (Sanger sequencing, MassArray, microarrays, (SDFs); SDFs contain proteins that are associated with
next-generation sequencing). For example, the MassArray DNA damage (such as -H2AX and p53-binding protein-1
technology allows precise and quantitative DNA methyla- (53BP1)) [22, 47].
tion detection in selected target regions (300500 bp) based Cell senescence entails widespread changes in gene
on base-specific cleavage of single-stranded nucleic acids expression [20, 74]. Narita et al. [74] have shown that the
coupled with MALDI-TOF MS detection [27]. After the senescence phenotype is also associated with changes in
bisulfite treatment of the DNA, a PCR amplification step is chromatin structure. Senescent cells tend to form special-
carried out to yield an amplicon with a T7 promoter tag. ized domains of heterochromatin, called senescence-
Next, in vitro RNA transcription is performed on the associated heterochromatin foci (SAHF). SAHF have been
reverse strand followed by RNase A cleavage at specific proposed to play a role in the silencing of proliferation-
254 Pflugers Arch - Eur J Physiol (2010) 459:247258

Fig. 3 EpiGram tab for the gene MKRN3. The EpiGram is a graphical units. The example above is an EpiGram obtained for the studied
representation of methylation ratios found in each sample for the imprinted gene MKRN3 in liver of wild-type mice and mice harboring a
amplicon studied. Each sample's nucleotide sequence is displayed as a defect in non-homologous DNA end joining, a DNA double-strand break
series of individual CpGs, which are color-coded circles on the same line. repair process (Ku80/). The first ten lines represent five wild-type
The color within the circle denotes the level of methylation found at this samples (in duplicates), while lines 11 to 20 represent the Ku80/
particular site in the selected sample. The color spectrum ranges from samples. The last two lines correspond to a universal unmethylated
yellow (or 0% methylated) CpG units to blue (100% methylated) CpG DNA sample, used as a conversion control

promoting genes, thus contributing to the senescence- Environmental effects on the epigenome:
associated proliferation arrest. impact on aging
Cellular senescence is an important barrier to the
development of cancer in mammals due to its ability to Recent findings support the idea that the epigenome is a
arrest proliferation and prevent neoplastic progression of key target of environmental modifications with long-term
cells harboring oncogenic lesions [14, 93]. At the same effects on disease susceptibility and aging. The aforemen-
time, there is increasing evidence that an accumulation of tioned study on monozygotic twins [32] suggests that the
dysfunctional senescent cells contributes to mammalian emerging differences in the epigenome as twins get older
aging [15]. Indeed, senescent stromal cells may actually may reflect environmental factors and lifestyle. One aspect
promote the proliferation and tumorigenesis of epithelial of the lifestyle is the diet; for example, a diet poor in folate
cells [13, 16, 59]. This is, in part, due to the capability of has been linked to genomic instability [52] and hypome-
senescent cells to secrete growth factors, as well as extracel- thylation [33]. Interestingly, a deficiency in folate and
lular matrix components, matrix-degrading enzymes, and methionine has been shown as the cause of aberrant IGF2
inflammatory cytokines that can disrupt tissue integrity and/ imprinting [107].
or stimulate nearby cells to proliferate, thus creating a pro- Several studies suggest the mother's behavior or diet can
oncogenic tissue environment [13, 16]. Furthermore, the affect epigenetic patterns in her offspring. For example, a
senescent phenotype has been linked to some age-related study carried out on Agouti pregnant mice showed that
diseases such as osteoarthritis, atherosclerosis, and liver feeding the mice with a diet rich in methyl donors
cirrhosis [69, 110]. influences coat color, body weight, and health of their
At the moment, very little is known about the progeny [24]. Another study conducted in rats showed that,
relationship between DNA methylation and cell senes- in the absence of appropriate nurturing, there is a
cence. Overall, 5-methylcytosine content and DNMT1 demethylation of the glucocorticoid receptor (GR) in the
activity diminish when cells senesce [39]. Young and hippocampus of the offspring and, as a consequence,
Smith have shown that, in response to the demethylating increased expression of the receptor gene later in life,
agent 5-azacytidine (5AZA), human fibroblasts undergo thereby leading to an increased stress sensitivity of the
senescence. Interestingly, human fibroblasts lacking p21 adolescent rats [109].
fail to undergo senescence after 5AZA treatment, Those findings are in keeping with a new theory called
indicating that p21 is an essential component of this the fetal basis of origins of adult-onset disease [71]. This
5AZA-induced arrest [113, 114]. Nothing is so far known theory posits that the effects of in utero exposures can cause
about the methylation status of genes involved in permanent functional changes that result in increased
processes found associated with senescence. susceptibility to disease/dysfunction later in the life span.
Pflugers Arch - Eur J Physiol (2010) 459:247258 255

An example of this is the so-called Second World War control would be detrimental and its relaxation can be the
Dutch Hunger Winter from 19441945; children of cause of diseases. We have provided an overview of the
pregnant women exposed to famine were more susceptible role of the relaxation of this control in the development of
to diabetes, obesity, and cardiovascular disease later in life age-related diseases, giving emphasis to the concept that
[64]. This finding suggests that transient environmental aging, at least in part, is a time-dependent, epigenetically
conditions early in human gestation can be recorded as mediated loss of phenotypic plasticity.
persistent changes in epigenetic information and have life- One of the most fascinating aspects of possible therapeutic
long phenotypic consequences. Indeed, as discussed previ- intervention is that epimutations can be corrected because
ously, early embryonic development is of special interest in epigenetic marks are, by definition, reversible and dynamic.
this respect because this is a crucial period for establishing To date, DNMT inhibitors (such as decitabine) and histone
and maintaining epigenetic marks [82]. deacetylase inhibitors (SAHA) are in clinical trials in cancer.
Several studies in mammals [11, 62, 70, 80, 81] are now Many companies are currently focusing on HDAC inhibitors.
supporting the hypothesis that epigenetic alterations might be The first one, Vorinostat, has been approved by the FDA in
inherited transgenerationally and affect the health of future October 2006 for second-line therapy of cutaneous T-cell
generations. For example, embryonic rats exposed to the lymphoma [65], while the combination of classic anti-cancer
antiandrogenic compound vinclozolin show decreased sper- treatment has been promising in reversing acquired chemo-
matogenesis, and this trait is passed to several subsequent therapy resistance [38].
generations [1]. Transgenerational epigenetic inheritance has Many questions regarding epigenetics and its role in age-
now also been demonstrated in other eukaryotic organisms, related diseases still remain open. Do epimutations accu-
for example, plants and yeast [18, 41]. The fact that an mulate stochastically with aging in different tissue types
epigenetically induced adaptation to the environment can be and what are, eventually, the consequences of this accumu-
passed through generations has a distinctly Lamarckian lation? Which epigenetic marks confer identity to the
flavor, which is a topic of discussion [51]. cell, which genes are responsible for enhanced disease
susceptibility when epigenetically deregulated, which envi-
ronmental factors are deleterious for the epigenome and,
Conclusions and future prospects finally, which epigenetic biomarkers could be used for
detection of early-stage diseases?
It is becoming evident that epigenetics plays a key role in The elucidation of the interaction of the environment
the development of diseases associated with aging. Epi- with the epigenome and their role in aging will allow for
genetics has been considered to be at the epicenter of the development of novel epigenetic-based diagnostic,
modern medicine [28] because it explains the relationship preventive, and therapeutic strategies. Manipulating and
between an individual's genetic background, the environ- understanding the epigenome holds promises for preventing
ment, aging, and disease. The concept of DNA as a static, and treating age-related diseases.
fixed entity has been replaced by a new vision where DNA
is highly dynamic, capable of large fluctuations, and able to Acknowledgements We thank the SENS Foundation, the National
interact actively with the environment. According to the Institute on Aging, and the Ellison Medical Foundation for supporting
new findings, DNA responds to the environmental stimuli the research of the authors.
by modifying its properties in adapting to changes, in order
to maintain an internal balance and proper functionality.
We have reviewed the pivotal role of epigenetics in References
conferring to DNA this plasticity; thanks to the epigenetic
marks, cells know their role in life. Without its epigenetic 1. Anway MD, Cupp AS, Uzumcu M, Skinner MK (2005)
Epigenetic transgenerational actions of endocrine disruptors
memory, cells would lack identity and function. In addition and male fertility. Science 308:14661469
to that, genes can have a memory and can learn by 2. Bahar R, Hartmann CH, Rodriguez KA, Denny AD, Busuttil
experience, and the (epigenetic) marks left by such RA, Doll ME, Calder RB, Chisholm GB, Pollock BH, Klein
experiences can be passed to daughter cells and to future CA, Vijg J (2006) Increased cell-to-cell variation in gene
expression in ageing mouse heart. Nature 441:10111014
generations. This gives rise to a fascinating concept: health 3. Barbot W, Dupressoir A, Lazar V, Heidmann T (2002) Epigenetic
and general physiology can be affected not only by the regulation of an IAP retrotransposon in the aging mouse:
interplay of our own genes and conditions of life but also progressive demethylation and de-silencing of the element by its
by the inherited effects of the interplay of our own genes repetitive induction. Nucleic Acids Res 30:23652373
4. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A,
and the environment of our ancestors. Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala
Despite the fact that the epigenome is highly dynamic, it S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S,
is also strictly controlled. As discussed, the loss of this Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ,
256 Pflugers Arch - Eur J Physiol (2010) 459:247258

Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA (2006) 25. Doll ME, Giese H, van Steeg H, Vijg J (2000) Mutation
Resveratrol improves health and survival of mice on a high-calorie accumulation in vivo and the importance of genome stability in
diet. Nature 444:337342 aging and cancer. Results Probl Cell Differ 29:165180
5. Baylin SB, Herman JG (2001) Promoter hypermethylationcan 26. Eden A, Gaudet F, Waghmare A, Jaenisch R (2003) Chromo-
this change alone ever designate true tumor suppressor gene somal instability and tumors promoted by DNA hypomethyla-
function? J Natl Cancer Inst 93:664665 tion. Science 300:455
6. Baylin SB, Herman JG (2000) DNA hypermethylation in 27. Ehrich M, Nelson MR, Stanssens P, Zabeau M, Liloglou T,
tumorigenesis: epigenetics joins genetics. Trends Genet Xinarianos G, Cantor CR, Field JK, van den Boom D (2005)
16:168174 Quantitative high-throughput analysis of DNA methylation
7. Bennett-Baker PE, Wilkowski J, Burke DT (2003) Age- patterns by base-specific cleavage and mass spectrometry. Proc
associated activation of epigenetically repressed genes in the Natl Acad Sci USA 102:1578515790
mouse. Genetics 165:20552062 28. Feinberg AP (2008) Epigenetics at the epicenter of modern
8. Bjornsson HT, Sigurdsson MI, Fallin MD, Irizarry RA, Aspelund medicine. JAMA 299:13451350
T, Cui H, Yu W, Rongione MA, Ekstrom TJ, Harris TB, Launer 29. Feinberg AP, Ohlsson R, Henikoff S (2006) The epigenetic
LJ, Eiriksdottir G, Leppert MF, Sapienza C, Gudnason V, progenitor origin of human cancer. Nat Rev Genet 7:2133
Feinberg AP (2008) Intra-individual change over time in DNA 30. Ferguson-Smith AC, Surani MA (2001) Imprinting and the
methylation with familial clustering. JAMA 299:28772883 epigenetic asymmetry between parental genomes. Science
9. Boyes J, Bird A (1991) DNA methylation inhibits transcription 293:10861089
indirectly via a methyl-CpG binding protein. Cell 64:11231134 31. Fischle W, Wang Y, Jacobs SA, Kim Y, Allis CD, Khorasanizadeh S
10. Buck MJ, Lieb JD (2004) ChIP-chip: considerations for the (2003) Molecular basis for the discrimination of repressive methyl-
design, analysis, and application of genome-wide chromatin lysine marks in histone H3 by Polycomb and HP1 chromodomains.
immunoprecipitation experiments. Genomics 83:349360 Genes Dev 17:18701881
11. Buiting K, Gross S, Lich C, Gillessen-Kaesbach G, el-Maarri O, 32. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML,
Horsthemke B (2003) Epimutations in Prader-Willi and Angel- Heine-Suner D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet
man syndromes: a molecular study of 136 patients with an M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A,
imprinting defect. Am J Hum Genet 72:571577 Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M (2005)
12. Busuttil R, Bahar R, Vijg J (2007) Genome dynamics and Epigenetic differences arise during the lifetime of monozygotic
transcriptional deregulation in aging. Neuroscience 145:13411347 twins. Proc Natl Acad Sci USA 102:1060410609
13. Campisi J (2000) Cancer, aging and cellular senescence. In Vivo 33. Friso S, Choi SW (2002) Genenutrient interactions and DNA
14:183188 methylation. J Nutr 132:2382S2387S
14. Campisi J (2005) Senescent cells, tumor suppression, and 34. Fu VX, Dobosy JR, Desotelle JA, Almassi N, Ewald JA,
organismal aging: good citizens, bad neighbors. Cell 120:513 Srinivasan R, Berres M, Svaren J, Weindruch R, Jarrard DF
522 (2008) Aging and cancer-related loss of insulin-like growth
15. Campisi J, d'Adda di Fagagna F (2007) Cellular senescence: when factor 2 imprinting in the mouse and human prostate. Cancer Res
bad things happen to good cells. Nat Rev Mol Cell Biol 8:729 68:67976802
740 35. Fuks F, Burgers WA, Godin N, Kasai M, Kouzarides T (2001)
16. Campisi J, Kim SH, Lim CS, Rubio M (2001) Cellular Dnmt3a binds deacetylases and is recruited by a sequence-
senescence, cancer and aging: the telomere connection. Exp specific repressor to silence transcription. EMBO J 20:2536
Gerontol 36:16191637 2544
17. Campisi J, Vijg J (2009) Does damage to DNA and other 36. Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J,
macromolecules play a role in aging? If so, how? J Gerontol A Gray JW, Leonhardt H, Jaenisch R (2003) Induction of tumors in
Biol Sci Med Sci 64:175178 mice by genomic hypomethylation. Science 300:489492
18. Chandler VL, Stam M (2004) Chromatin conversations: mecha- 37. Giese H, Snyder WK, van Oostrom C, van Steeg H, Doll ME,
nisms and implications of paramutation. Nat Rev Genet 5:532544 Vijg J (2002) Age-related mutation accumulation at a lacZ
19. Clayton AL, Hazzalin CA, Mahadevan LC (2006) Enhanced reporter locus in normal and tumor tissues of Trp53-deficient
histone acetylation and transcription: a dynamic perspective. Mol mice. Mutat Res 514:153163
Cell 23:289296 38. Glasspool RM, Teodoridis JM, Brown R (2006) Epigenetics as a
20. Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, mechanism driving polygenic clinical drug resistance. Br J
Nelson PS, Desprez PY, Campisi J (2008) Senescence-associated Cancer 94:10871092
secretory phenotypes reveal cell-nonautonomous functions of 39. Gray MD, Jesch SA, Stein GH (1991) 5-Azacytidine-induced
oncogenic RAS and the p53 tumor suppressor. PLoS Biol demethylation of DNA to senescent level does not block
6:28532868 proliferation of human fibroblasts. J Cell Physiol 149:477484
21. DeBusk FL (1972) The Hutchinson-Gilford progeria syndrome. 40. Grewal SI, Elgin SC (2002) Heterochromatin: new possibilities for
Report of 4 cases and review of the literature. J Pediatr 80:697724 the inheritance of structure. Curr Opin Genet Dev 12:178187
22. Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, 41. Grewal SI, Klar AJ (1996) Chromosomal inheritance of
Luise C, Schurra C, Garre M, Nuciforo PG, Bensimon A, epigenetic states in fission yeast during mitosis and meiosis.
Maestro R, Pelicci PG, d'Adda di Fagagna F (2006) Oncogene- Cell 86:95101
induced senescence is a DNA damage response triggered by 42. Guarente L (1999) Diverse and dynamic functions of the Sir
DNA hyper-replication. Nature 444:638642 silencing complex. Nat Genet 23:281285
23. Dillon N, Festenstein R (2002) Unravelling heterochromatin: 43. Haigis MC, Guarente LP (2006) Mammalian sirtuinsemerging
competition between positive and negative factors regulates roles in physiology, aging, and calorie restriction. Genes Dev
accessibility. Trends Genet 18:252258 20:29132921
24. Dolinoy DC, Weidman JR, Waterland RA, Jirtle RL (2006) 44. Harley CB, Futcher AB, Greider CW (1990) Telomeres shorten
Maternal genistein alters coat color and protects Avy mouse during ageing of human fibroblasts. Nature 345:458460
offspring from obesity by modifying the fetal epigenome. 45. Hayflick L (1965) The limited in vitro lifetime of human diploid
Environ Health Perspect 114:567572 cell strains. Exp Cell Res 37:614636
Pflugers Arch - Eur J Physiol (2010) 459:247258 257

46. Hegele RA (2003) Drawing the line in progeria syndromes. 67. Martin SL, Hopkins CL, Naumer A, Dolle ME, Vijg J (2001)
Lancet 362:416417 Mutation frequency and type during ageing in mouse seminif-
47. Herbig U, Jobling WA, Chen BP, Chen DJ, Sedivy JM (2004) erous tubules. Mech Ageing Dev 122:13211331
Telomere shortening triggers senescence of human cells through 68. Menendez L, Benigno BB, McDonald JF (2004) L1 and HERV-
a pathway involving ATM, p53, and p21(CIP1), but not p16 W retrotransposons are hypomethylated in human ovarian
(INK4a). Mol Cell 14:501513 carcinomas. Mol Cancer 3:12
48. Holliday R (1987) The inheritance of epigenetic defects. Science 69. Minamino T, Komuro I (2007) Vascular cell senescence:
238:163170 contribution to atherosclerosis. Circ Res 100:1526
49. Issa JP (1999) Aging, DNA methylation and cancer. Crit Rev 70. Morgan HD, Sutherland HG, Martin DI, Whitelaw E (1999)
Oncol Hematol 32:3143 Epigenetic inheritance at the agouti locus in the mouse. Nat
50. Itahana K, Campisi J, Dimri GP (2007) Methods to detect Genet 23:314318
biomarkers of cellular senescence: the senescence-associated 71. Morley R (2006) Fetal origins of adult disease. Semin Fetal
beta-galactosidase assay. Methods Mol Biol 371:2131 Neonatal Med 11:7378
51. Jablonka E, Lamb MJ (1989) The inheritance of acquired 72. Muller HJ, Altenburg E (1930) The frequency of translocations
epigenetic variations. J Theor Biol 139:6983 produced by x-rays in drosophila. Genetics 15:283311
52. Jacob RA (1999) Evidence that diet modification reduces in vivo 73. Nan X, Campoy FJ, Bird A (1997) MeCP2 is a transcriptional
oxidant damage. Nutr Rev 57:255258 repressor with abundant binding sites in genomic chromatin. Cell
53. Jones PA (2002) DNA methylation and cancer. Oncogene 88:471481
21:53585360 74. Narita M, Nunez S, Heard E, Lin AW, Hearn SA, Spector DL,
54. Jones PA, Laird PW (1999) Cancer epigenetics comes of age. Hannon GJ, Lowe SW (2003) Rb-mediated heterochromatin
Nat Genet 21:163167 formation and silencing of E2F target genes during cellular
55. Kaeberlein M, McVey M, Guarente L (1999) The SIR2/3/4 senescence. Cell 113:703716
complex and SIR2 alone promote longevity in Saccharomy- 75. Oberdoerffer P, Michan S, McVay M, Mostoslavsky R, Vann J,
ces cerevisiae by two different mechanisms. Genes Dev 13: Park SK, Hartlerode A, Stegmuller J, Hafner A, Loerch P,
25702580 Wright SM, Mills KD, Bonni A, Yankner BA, Scully R, Prolla
56. Kanungo A, Chandi SM (1994) Melanotic neuroectodermal tumor TA, Alt FW, Sinclair DA (2008) SIRT1 redistribution on
of the epididymis. A case report. Indian J Cancer 31:138140 chromatin promotes genomic stability but alters gene expression
57. Kator K, Cristofalo V, Charpentier R, Cutler RG (1985) during aging. Cell 135:907918
Dysdifferentiative nature of aging: passage number dependency 76. Okamoto I, Otte AP, Allis CD, Reinberg D, Heard E (2004)
of globin gene expression in normal human diploid cells grown Epigenetic dynamics of imprinted X inactivation during early
in tissue culture. Gerontology 31:355361 mouse development. Science 303:644649
58. Khulan B, Thompson RF, Ye K, Fazzari MJ, Suzuki M, Stasiek 77. Okano M, Takebayashi S, Okumura K, Li E (1999) Assignment
E, Figueroa ME, Glass JL, Chen Q, Montagna C, Hatchwell E, of cytosine-5 DNA methyltransferases Dnmt3a and Dnmt3b to
Selzer RR, Richmond TA, Green RD, Melnick A, Greally JM mouse chromosome bands 12A2A3 and 2H1 by in situ
(2006) Comparative isoschizomer profiling of cytosine methyl- hybridization. Cytogenet Cell Genet 86:333334
ation: the HELP assay. Genome Res 16:10461055 78. Ono T, Dean RG, Chattopadhyay SK, Cutler RG (1985)
59. Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J Dysdifferentiative nature of aging: age-dependent expression of
(2001) Senescent fibroblasts promote epithelial cell growth and MuLV and globin genes in thymus, liver and brain in the AKR
tumorigenesis: a link between cancer and aging. Proc Natl Acad mouse strain. Gerontology 31:362372
Sci USA 98:1207212077 79. Park PJ (2008) Epigenetics meets next-generation sequencing.
60. Kudlow BA, Kennedy BK, Monnat RJ Jr (2007) Werner Epigenetics 3:318321
and Hutchinson-Gilford progeria syndromes: mechanistic 80. Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K,
basis of human progeroid diseases. Nat Rev Mol Cell Biol Sjostrom M, Golding J (2006) Sex-specific, male-line trans-
8:394404 generational responses in humans. Eur J Hum Genet 14:159166
61. Laird CD, Pleasant ND, Clark AD, Sneeden JL, Hassan KM, 81. Rakyan VK, Chong S, Champ ME, Cuthbert PC, Morgan HD, Luu
Manley NC, Vary JC Jr, Morgan T, Hansen RS, Stoger R (2004) KV, Whitelaw E (2003) Transgenerational inheritance of epigenetic
Hairpin-bisulfite PCR: assessing epigenetic methylation patterns states at the murine Axin(Fu) allele occurs after maternal and
on complementary strands of individual DNA molecules. Proc paternal transmission. Proc Natl Acad Sci USA 100:25382543
Natl Acad Sci USA 101:204209 82. Reik W, Dean W, Walter J (2001) Epigenetic reprogramming in
62. Lane N, Dean W, Erhardt S, Hajkova P, Surani A, Walter J, Reik W mammalian development. Science 293:10891093
(2003) Resistance of IAPs to methylation reprogramming may 83. Robertson KD (2001) DNA methylation, methyltransferases, and
provide a mechanism for epigenetic inheritance in the mouse. cancer. Oncogene 20:31393155
Genesis 35:8893 84. Rodenhiser D, Mann M (2006) Epigenetics and human disease:
63. Liu B, Wang J, Chan KM, Tjia WM, Deng W, Guan X, Huang translating basic biology into clinical applications. CMAJ
JD, Li KM, Chau PY, Chen DJ, Pei D, Pendas AM, Cadinanos J, 174:341348
Lopez-Otin C, Tse HF, Hutchison C, Chen J, Cao Y, Cheah KS, 85. Rogina B, Helfand SL (2004) Sir2 mediates longevity in the fly
Tryggvason K, Zhou Z (2005) Genomic instability in through a pathway related to calorie restriction. Proc Natl Acad
laminopathy-based premature aging. Nat Med 11:780785 Sci USA 101:1599816003
64. Lumey LH, Stein AD (1997) In utero exposure to famine and 86. Romanov GA, Vaniushin BF (1980) Intragenomic specificity of
subsequent fertility: The Dutch Famine Birth Cohort Study. Am DNA methylation in animals. Qualitative differences in tissues
J Public Health 87:19621966 and changes in methylation of repeating sequences during aging,
65. Marks PA, Breslow R (2007) Dimethyl sulfoxide to vorinostat: carcinogenesis and hormonal induction. Mol Biol (Mosk)
development of this histone deacetylase inhibitor as an antican- 14:357368
cer drug. Nat Biotechnol 25:8490 87. Rountree MR, Bachman KE, Baylin SB (2000) DNMT1 binds
66. Martin GM (2005) Epigenetic drift in aging identical twins. Proc HDAC2 and a new co-repressor, DMAP1, to form a complex at
Natl Acad Sci USA 102:1041310414 replication foci. Nat Genet 25:269277
258 Pflugers Arch - Eur J Physiol (2010) 459:247258

88. Santos F, Hendrich B, Reik W, Dean W (2002) Dynamic 101. Vidal DO, Paixao VA, Brait M, Souto EX, Caballero OL, Lopes
reprogramming of DNA methylation in the early mouse embryo. LF, Vettore AL (2007) Aberrant methylation in pediatric
Dev Biol 241:172182 myelodysplastic syndrome. Leuk Res 31:175181
89. Scaffidi P, Misteli T (2005) Reversal of the cellular phenotype in 102. Vijg J, Doll ME (2002) Large genome rearrangements as a
the premature aging disease Hutchinson-Gilford progeria syn- primary cause of aging. Mech Ageing Dev 123:907915
drome. Nat Med 11:440445 103. Vogelstein B, Kinzler KW (1993) The multistep nature of cancer.
90. Scaffidi P, Misteli T (2006) Lamin A-dependent nuclear defects Trends Genet 9:138141
in human aging. Science 312:10591063 104. Wang H, Cao R, Xia L, Erdjument-Bromage H, Borchers C,
91. Schmid M, Haaf T, Grunert D (1984) 5-Azacytidine-induced under- Tempst P, Zhang Y (2001) Purification and functional charac-
condensations in human chromosomes. Hum Genet 67:257263 terization of a histone H3-lysine 4-specific methyltransferase.
92. Schumacher A, Petronis A (2006) Epigenetics of complex Mol Cell 8:12071217
diseases: from general theory to laboratory experiments. Curr 105. Wang SC, Oelze B, Schumacher A (2008) Age-specific epigenetic
Top Microbiol Immunol 310:81115 drift in late-onset Alzheimer's disease. PLoS ONE 3:e2698
93. Shay JW, Roninson IB (2004) Hallmarks of senescence in 106. Wareham KA, Lyon MF, Glenister PH, Williams ED (1987) Age
carcinogenesis and cancer therapy. Oncogene 23:29192933 related reactivation of an X-linked gene. Nature 327:725727
94. Shumaker DK, Dechat T, Kohlmaier A, Adam SA, Bozovsky 107. Waterland RA, Lin JR, Smith CA, Jirtle RL (2006) Post-weaning
MR, Erdos MR, Eriksson M, Goldman AE, Khuon S, Collins diet affects genomic imprinting at the insulin-like growth factor 2
FS, Jenuwein T, Goldman RD (2006) Mutant nuclear lamin A (Igf2) locus. Hum Mol Genet 15:705716
leads to progressive alterations of epigenetic control in premature 108. Watt F, Molloy PL (1988) Cytosine methylation prevents binding
aging. Proc Natl Acad Sci USA 103:87038708 to DNA of a HeLa cell transcription factor required for optimal
95. Siegmund KD, Connor CM, Campan M, Long TI, Weisenberger expression of the adenovirus major late promoter. Genes Dev
DJ, Biniszkiewicz D, Jaenisch R, Laird PW, Akbarian S (2007) 2:11361143
DNA methylation in the human cerebral cortex is dynamically 109. Weaver IC, Diorio J, Seckl JR, Szyf M, Meaney MJ (2004) Early
regulated throughout the life span and involves differentiated environmental regulation of hippocampal glucocorticoid receptor
neurons. PLoS ONE 2:e895 gene expression: characterization of intracellular mediators and
96. Sims RJ 3rd, Reinberg D (2009) Processing the H3K36me3 potential genomic target sites. Ann N Y Acad Sci 1024:182212
signature. Nat Genet 41:270271 110. Wiemann SU, Satyanarayana A, Tsahuridu M, Tillmann HL,
97. Sinclair DA, Guarente L (1997) Extrachromosomal rDNA circles Zender L, Klempnauer J, Flemming P, Franco S, Blasco MA,
a cause of aging in yeast. Cell 91:10331042 Manns MP, Rudolph KL (2002) Hepatocyte telomere shortening
98. Sommer M, Poliak N, Upadhyay S, Ratovitski E, Nelkin BD, and senescence are general markers of human liver cirrhosis.
Donehower LA, Sidransky D (2006) DeltaNp63alpha over- FASEB J 16:935942
expression induces downregulation of Sirt1 and an accelerated 111. Wilson VL, Smith RA, Ma S, Cutler RG (1987) Genomic 5-
aging phenotype in the mouse. Cell Cycle 5:20052011 methyldeoxycytidine decreases with age. J Biol Chem 262:9948
99. Thompson RF, Suzuki M, Lau KW, Greally JM (2009) A 9951
pipeline for the quantitative analysis of CG dinucleotide 112. Wright WE, Shay JW (1992) Telomere positional effects and the
methylation using mass spectrometry. Bioinformatics Resource regulation of cellular senescence. Trends Genet 8:193197
Number 113. Young J, Smith JR (2000) Epigenetic aspects of cellular
100. Tra J, Kondo T, Lu Q, Kuick R, Hanash S, Richardson B (2002) senescence. Exp Gerontol 35:2332
Infrequent occurrence of age-dependent changes in CpG island 114. Young JI, Smith JR (2001) DNA methyltransferase inhibition in
methylation as detected by restriction landmark genome scan- normal human fibroblasts induces a p21-dependent cell cycle
ning. Mech Ageing Dev 123:14871503 withdrawal. J Biol Chem 276:1961019616

Anda mungkin juga menyukai