Sciadv 1600584
Sciadv 1600584
Sciadv 1600584
Over the past decade, a growing number of studies have revealed that progressive changes to epigenetic information
accompany aging in both dividing and nondividing cells. Functional studies in model organisms and humans indicate
that epigenetic changes have a huge influence on the aging process. These epigenetic changes occur at various levels,
including reduced bulk levels of the core histones, altered patterns of histone posttranslational modifications and DNA
methylation, replacement of canonical histones with histone variants, and altered noncoding RNA expression, during
both organismal aging and replicative senescence. The end result of epigenetic changes during aging is altered local
accessibility to the genetic material, leading to aberrant gene expression, reactivation of transposable elements, and
genomic instability. Strikingly, certain types of epigenetic information can function in a transgenerational manner to
influence the life span of the offspring. Several important conclusions emerge from these studies: rather than being
genetically predetermined, our life span is largely epigenetically determined; diet and other environmental influences
can influence our life span by changing the epigenetic information; and inhibitors of epigenetic enzymes can influence
life span of model organisms. These new findings provide better understanding of the mechanisms involved in aging.
Given the reversible nature of epigenetic information, these studies highlight exciting avenues for therapeutic inter-
vention in aging and age-associated diseases, including cancer.
INTRODUCTION
Aging is a complex multifactorial biological process shared by all living as queen bees and worker bees (1, 12, 13). Although longevity studies on
organisms. It is manifested by a gradual decline of normal physiological the human population have shown that genetic factors could explain a
functions in a time-dependent manner. Organismal aging holds signif- fraction (20 to 30%) of the differences observed in life spans of mono-
icant importance for human health because it increases susceptibility to zygotic twins, the majority of the remainder of variation is thought to
many diseases, including cancer, metabolic disorders, such as diabetes, have arisen through epigenetic drift during their lifetime (9, 10, 12, 14, 15).
The addition of linker histones, such as histone H1, and other non- complexity of the process of aging and the influence of overlapping
histone proteins, such as heterochromatin protein 1 (HP1), facilitates pathways on the outcome (22, 23).
the formation of higher-order repressive chromatin structures, such Hutchinson-Gilford progeria syndrome (HGPS) and Werner syn-
as heterochromatin (19). Packaging of the genomic DNA into the high- drome are rare human genetic disorders characterized by premature
ly organized chromatin structure regulates all genomic processes in the aging phenotypes with a shortened life span. This group of diseases re-
nucleus, including DNA replication, transcription, recombination, and sembles physiological aging to a certain extent, serving as excellent
DNA repair, by controlling access to the DNA (2). models to gain insight into the biology of aging in humans (24, 25). These
Studies on the aging of mammals are rather limited by the long life diseases are due to either a mutation in genes encoding the DNA repair
span of the commonly used model organisms. Thus, both nonverte- machinery or the A-type lamin, leading to disorganized chromatin
brate and invertebrate organisms, with their shorter life span and ease structures. The causative mutations behind these progeria syndromes in-
of genetic and environmental manipulations, gained popularity among dicate that genomic instability and chromatin deterioration are causes of
researchers in the aging field as experimental models for aging studies. human aging. Furthermore, the knowledge we gain from understanding
Among them, budding yeast or Saccharomyces cerevisiae is a highly in- the molecular pathology of these human premature aging diseases pro-
formative organismal model for aging studies with its genetic tools, vides us with useful information to understand the complex aging pro-
short life span, and fully sequenced genome (20, 21). Despite being uni- cess. Individuals with HGPS do not recapitulate all aging phenotypes
cellular, yeast has been an excellent model to identify and characterize because they usually show segmental progeria affecting multiple tis-
conserved basic biological processes, including aging. Yeast has been sues. By recapitulating some molecular and cellular changes that are
extensively used to identify genes and interventions responsible for life characteristics of the natural aging process, these models provide us
span extension and to gain insights into the aging processes of all eu- with a unique opportunity to understand the aging process in a human
karyotic organisms. In parallel, over the years, studies on invertebrate model (24, 25).
organisms, such as Drosophila melanogaster (flies) and Caenorhabditis Studies in humans and these powerful models of aging reveal that,
elegans (worms), and certain vertebrate models, such as mice, zebrafish, similar to all other biological structures inside the cells, the epigenome
naked mole rats, and, most recently, African turquoise killifish, have suffers from a progressive loss in its configuration during aging. This
also provided invaluable information to help us understand the results in a profound change in the chromosomal architecture, genomic
Chromosome mRNA
Transcriptional drift
DNA mutation
New transposable
element insertions
DNA methylation
Me Histone variant
Me Ac Me
Me Ac
Me Me
Ac Me Me Altered patterns of
Chromatin DNA mutation
histone modifications
modifiers
Fig. 1. Overview of epigenetic changes during aging. In young individuals, the cells within each cell type have a similar pattern of gene expression,
determined in large part by each cell having similar epigenetic information. During aging, the epigenetic information changes sporadically in response to
exogenous and endogenous factors. The resulting abnormal chromatin state is characterized by different histone variants being incorporated, altered DNA
methylation patterns, and altered histone modification patterns, resulting in the recruitment of different chromatin modifiers. The abnormal chromatin state
in old cells includes altered transcription patterns and transcriptional drift within the population. The abnormal chromatin state in old cells also leads to new
transposable elements being inserted into the genome and genomic instability, including DNA mutations.
integrity, and gene expression patterns (1, 10). Where examined, these A rather important unresolved paradox for the heterochromatin loss
effects are mostly conserved all the way from single-celled organisms, model of aging was the observation of loss of heterochromatin during
such as budding yeast, to complex multicellular eukaryotes. These con- aging at the same time as the formation of senescence-associated het-
served mechanisms help us gain a clearer picture of the aging process. erochromatin foci (SAHF), which are domains of heterochromatin over
Here, we will discuss the epigenetic changes that have emerged in recent proliferation-promoting genes in senescent cells (42). For a long time,
years to significantly influence the aging process, from studies on single- SAHF have been perceived as another difference between the aging pro-
celled model organisms to human models of aging. We will synthesize cess and replicative senescence (43). However, very recently, global re-
the recent developments in this area and highlight potential future organization of genome architecture has been reported in senescent
directions. cells using Hi-C (43) and FAIRE (44) analyses, leading to a proposed
two-step mechanism for the formation of SAHF. This two-step mech-
anism consists of heterochromatin decondensation in senescent cells, as
THE HETEROCHROMATIN LOSS MODEL OF AGING seen during organismal aging, followed by the heterochromatinization
of formerly euchromatin regions (Fig. 2) that spatially cluster to form
One of the earlier proposed models of aging was the “heterochromatin the compact SAHF structures. This process has been referred to as het-
loss model of aging” (26, 27). This model suggests that the loss of het- erochromatin redistribution (45) and provides a unifying theme to re-
erochromatin that accompanies aging leads to changes in global nuclear solve the paradox.
architecture and the expression of genes residing in those regions, di-
rectly or indirectly causing aging and cellular senescence. As with any
other model of aging, the heterochromatin loss model is supported by GLOBAL HISTONE PROTEIN REDUCTION DURING AGING
experimental data, but there are also confounding observations. Loss of
transcriptional silencing due to decay of the heterochromatin occurs Recently, the aging research field has experienced a leap from the earlier
during aging in all eukaryotes examined from yeast to humans (26–30), paradigm of the heterochromatin loss model. Not only is the hetero-
and there is evidence that accelerating or reversing this process can chromatin reorganized during aging, but a global loss of core histone
either shorten or lengthen life span, respectively (Fig. 2). Gene silencing proteins from the genome during aging has also been observed in
requires the absence of histone acetylation within heterochromatin re- multiple scenarios, and this has been shown to be a cause of aging
gions. Accordingly, treatment with histone deacetylase (HDAC) inhi- in yeast.
bitors or deletion of genes encoding HDACs, such as yeast SIR2 or its In budding yeast, replicative aging is accompanied by loss of approx-
sirtuin counterparts in metazoan species, shortens life span, whereas imately half of the core histone proteins (46, 47). The extensive nucleo-
during replicative aging is not restricted to budding yeast and has also rejuvenated population (48). Most of the nucleosomal positions are
been observed during aging in worms (55), during replicative aging of maintained in aged cells, whereas nucleosome occupancy is reduced
human diploid primary fibroblasts (56), and in senescent human cells to half the normal proportion and nucleosome locations on particular
(57). Notably, global histone loss has not yet been reported in mitotically DNA sequences become less stringent or fuzzier. Additionally, partial
dividing mammalian tissues in vivo. In human primary fibroblasts, the overexpression of histone proteins H3 and H4 partially reversed the
reduced synthesis of new histones during replicative senescence was a transcriptional defects observed during aging. It will be intriguing to in-
consequence of the shortened telomeres that activate the DNA damage vestigate in the future whether the longevity effect conferred by supply-
response (56), potentially explaining the mechanism by which telomere ing extra histones is equally pertinent in multicellular organisms.
shortening limits the number of cell divisions. Hence, loss of core histones
may be a more generalized phenomenon observed with aging in many
organisms. GENOMIC INSTABILITY RESULTING FROM CHROMATIN
Decreased levels of core histone proteins in aged cells would likely RELAXATION DURING AGING AND THE “AGING BY
cause a marked effect on the chromatin landscape by providing in- TRANSPOSITION” MODEL
appropriate access to the genetic material. The consequences of having
limiting amounts of histone proteins in the DNA during age progres- A more relaxed chromatin structure, due to heterochromatin loss or
sion have been elaborately examined in a recent global analysis of chro- histone loss, is predictive of not only transcriptional deregulation (as
matin from aged cells in budding yeast, in comparison to the young discussed above) but also genome instability. Indeed, the reduced
Extra histones
Integration
Yeast replicative aging cDNA
Reverse transcription
Integration
Human cell cDNA
Reverse transcription
X replicative senescence RNA
Calorie restriction
Integration
Mouse organismal aging cDNA
X Reverse transcription
RNA
Fig. 2. Alterations in chromatin structure during aging leads to activation of retrotransposons. The schematic at the top depicts the reduction in the
level of bulk histone proteins during aging (for example, as seen in budding yeast), leading to a more open chromatin structure and subsequent transcrip-
tional activation of the normally silenced Ty elements. The resulting transcripts from the retrotransposable elements are reverse-transcribed into cDNAs that
reinsert elsewhere into the genome of old cells. Overexpression of histones reverses the loss of histones, reduces Ty retrotransposition, and extends repli-
cative life span. The schematic in the middle depicts the heterochromatin reorganization that occurs during aging, as seen, for example, in tissue culture cells
during replicative senescence. The normally heterochromatinized retrotransposable elements become euchromatinized, leading to their transcription and
transposition elsewhere into the genome. Knockdown of the Alu element transcripts in human adult stem cells leads to escape from senescence and reduced
genomic instability. The schematic at the bottom depicts the heterochromatin reorganization that occurs in mouse tissues during aging and is accompanied
by activation of retrotransposable elements (RTEs). Calorie restriction reduces the level of retrotransposition in the aged mice genome.
nucleosome occupancy in old yeast led to increased genomic perturba- Ty transcription and retrotransposition in old yeast by overexpressing
tions, including higher levels of DNA breaks, damaged foci formation, histones (Fig. 2) (48). Life span–extending interventions, such as CR,
inter- and intrachromosomal translocations, insertion of mitochondrial have also been shown to counteract the increased expression of retro-
DNA into the nuclear genome, and high levels of retrotransposition transposons in aged mice (Fig. 2) (66).
(48). Notably, partial overexpression of histone proteins H3 and H4 The increased mobility of retrotransposons, observed in the gen-
partially reversed the transcriptional defects observed during aging omes of aged cells and tissues from multiples species, provides strong
and reduced retrotransposition, indicating that the increased retrotrans- evidence in favor of a recently hypothesized model of aging: aging by
position in old yeast was a consequence of histone loss during aging transposition (73). According to this model, these mobile elements and
(48). Increased levels of DNA breaks or unrepaired DNA damage, as their transposases are the driving force to cause structural dysregulation
illustrated by the formation of g-H2AX foci (a hallmark of DNA dam- of the genome to manifest aging phenotypes. Indeed, in an adult stem
age), have been observed in aged cells from multiple species, including cell model of ex vivo aging, entry into senescence was accompanied by
aged mice, senescent human cells, and cells derived from patients with increased transcription from the SINE/Alu retrotransposable elements
premature aging disorders (58–61). This suggests that DNA damage and persistent DNA damage foci (70). Experimental suppression of the
may be a driving force behind organismal aging, although the full ex- transcripts from Alu elements reversed the arrested phenotype and
tent of the role of DNA damage during age progression still needs to eradicated the DNA damage foci, directly indicating that retrotranspo-
be explored. son transcription was driving the entry into senescence (Fig. 2). Given
It is a long-standing belief that accumulation of DNA mutations the many recent links between activation of retrotransposable elements
over a period of time affects organismal aging because mutation accu- and aging, it is interesting to note that transposition also becomes more
mulation happens as a function of age in different organisms, such as frequent during cancer development (74, 75), wherein aging is the high-
flies or mice and, in some cases, also correlates with life span (62, 63). est risk factor for most cancers. Neurodegeneration, another disease of
However, a recent study indicated that accumulation of somatic DNA aging, is also characterized by increased retrotransposition (76, 77). Fu-
mutations, at least in budding yeast, does not have a causal role in aging ture studies will hopefully illuminate the full influence of transposition
(64). Therefore, it is yet to be conclusively determined whether the per- on the process of aging and the contribution of the age-dependent in-
sistent DNA damage observed in aging cells is the result of the increased crease in transposition to these other human diseases.
susceptibility of the accessible genome to accrue DNA damage with time Notably, the processes described in this section are unlikely to
or the inability of aged cells to efficiently repair the damage, or a combi- function independently from each other, and one event may lead to an-
nation of both. Notably, another recent analysis of the DNA repair tran- other during the aging process. For example, heterochromatin decay
scriptome of liver in species with significant life span differences revealed and histone loss lead to retrotransposition and changes in gene expres-
of senescence was most robust for the cleaved variant H3.3cs1, revealing gene transcription, DNA repair, DNA replication, effective condensa-
the importance of proteolytic processing of H3.3 during senescence. tion of chromatin, and many others, which affect almost all biological
The deposition of H3.3cs1 onto DNA was mainly attributed to the his- processes in eukaryotes, including aging. The histone posttranslational
tone chaperones ASF1a and UBN1 and putatively to HIRA in senescent modifications are added and removed by enzymes, usually in a tightly
cells (81). Notably, deletion of the yeast counterpart of HIRA, the Hir regulated manner. However, during aging, the abundance, activity, and
complex, extended replicative life span of yeast, but this is more likely function of some of these enzymes change, leading to alterations in the
due to the resulting increase in histone gene expression that counters epigenome.
the loss of core histones during aging (47). A similar correlation of Among the histone modifications that are known to affect the lon-
increased H3.3 incorporation during aging has also been observed in gevity process, the most prominent ones are acetylation and methyla-
the aged mouse brain, where H3.1 levels gradually decline with age tion of lysine residues. In budding yeast, acetylation present on the
and H3.3 gradually accumulates (82). The incorporation of H3.3 into globular domain of histone H3 on lysine 56 (H3 K56Ac) and acetylation
actively transcribed DNA regions in neuronal cells and its efficient at the N-terminal tail of histone H4 on lysine 16 (H4 K16Ac) both in-
turnover in neuronal cells are crucial to maintain cell type–specific gene fluence replicative aging, although through distinct mechanisms. It
expression as well as neuronal plasticity and cognition (82). It appears that seems that either the dynamic nature or a balanced level of each acety-
the histone variant H3.3 and its cleavage should be considered as impor- lation mark is crucial for the cell because either inability to acetylate
tant new players in maintaining functional chromatin structure during these residues or the presence of mutations that mimic permanent acet-
senescence. ylation reduces replicative life span of budding yeast (46, 47). Levels of
There are multiple different variants of histone H2A, and one in par- H3 K56Ac decrease during yeast aging (46, 47). Meanwhile, deletion
ticular has been implicated in aging. The H2A variant macroH2A is a of the genes encoding the relevant HDACs that remove H3 K56Ac,
prominent feature of the SAHF (83, 84). MacroH2A’s function is gen- Hst3, and Hst4 leads to a shortened yeast life span (46, 93) and ge-
erally assumed to be transcriptional repression (85, 86). Although nomic instability (46), including a high frequency of loss of hetero-
macroH2A has not been presumed to be important for the formation zygosity (94). The notion that there is a precise level of H3 K56Ac for
of SAHFs, it was suggested that macroH2A is crucial to maintain the optimal life span is borne out by the fact that overexpression of Hst3
transcriptional silencing at those sites (83, 84). The level of macroH2A shortens life span, whereas addition of one extra gene copy of HST3 or
increases in an age-dependent manner during replicative senescence in HST4 extends life span (95). Similarly, deletion of the yeast histone acetyl-
cultured human fibroblast cells and also in several tissues of aged mice transferase (HAT) that acetylates H3 K56Ac, Rtt109, or the histone
and primates (87). More recently, enrichment of macroH2A1 at sites chaperone Asf1, which assists in the acetylation, causes a shortened life
encoding the senescence-associated secretory phenotype (SASP) genes span (47). Exactly how a slightly elevated level of H3 K56Ac promotes
Other HATs and HDACs have also been implicated in aging. Dele- modification levels from cell populations or tissues misses important
tion of a component of the NuA4 HAT extends yeast replicative life additional information on gene-specific effects that would hopefully
span (52). NuA4 acetylates histone H4 but also has several nonhistone be available soon from profiling histone modifications by ChIP-seq,
substrates that may be relevant to its role in limiting longevity, such as eventually in individual cells, during aging.
Pck1p, which encodes a phosphoenolpyruvate carboxykinase enzyme, Evidence of histone lysine methylation playing a causal role during
Tap42p, a putative downstream member in the mammalian target of aging comes from studies modulating the levels of histone methyltrans-
rapamycin (mTOR) pathway, and/or Nnt1p, a putative nicotinamide ferases and demethylases. RNA interference (RNAi) screens in C. elegans
N-methyltransferase (102). Deletion of genes expressing components identified several histone methyltransferases, including ASH-2, SET-2,
of the histone H3 deacetylase complex Rpd3 extends replicative life span and SET-9, and several demethylases, including UTX-1, as potential
of yeast (103) and organismal life span of flies (104). Rpd3-mediated factors influencing the outcome of aging (55, 115). Reduction in the lev-
deacetylation appears to limit longevity through multiple pathways el of H3 K4me3 methylation, which serves as a signature of active chro-
(105), highlighting the pleiotropic effects of altering levels of histone matin, by knockdown of the H3 K4 methyltransferase SET-2 or the
acetylation. Further studies are required to fully understand the molec- components of the regulatory complex of H3 K4me3, ASH-2 and
ular mechanisms used by these HATs and HDACs to influence life span. WDR-5, caused life span extension in fertile worms. In contrast,
Histone acetylation also appears to play an important role in the knockdown of the H3 K4me3 demethylase protein RBR-2 shortened life
aging brain. Global histone hypoacetylation occurs in the repeated span of worms. On the other hand, overexpression of RBR-2 has also been
DNA elements in aged mice brain, which suggests a loss of chromatin shown to be sufficient for extending life span of worms (116). It is unclear
integrity with aging (106). Aged mice brains also show memory impair- how reduced levels of H3 K4me3 increase longevity, but it is evident that a
ment that has been proposed to be due to failure to up-regulate acety- limiting amount of this modification is favorable for optimal C. elegans
lation at histone H4 K12, which is a histone mark promoting transcription life span (117). Consistent with the findings in C. elegans, flies deficient
elongation, and due to the resulting altered gene expression pattern. in Lid, the RBR-2 ortholog in D. melanogaster, have shortened life span
Restoration of H4 K12 acetylation recovered both gene expression (117, 118). However, this is only evident in male flies, implying that
and learning behavior in aged mice, signifying the critical role of this the effect of this demethylase may be sex-specific in some organisms.
histone acetylation in aged mice brains (107). Alteration in the level of H3 K27me3 also influences life span of both
The trends in changed levels of histone methylation during aging worms and flies. Contrary to H3 K4me3, H3 K27me3 is a prominent mark
have also been extensively examined recently. Significant changes dur- of repressed chromatin. Attenuation of the H3 K27me3 demethylase
ing aging have been seen for trimethylation marks on histone H3 lysines UTX-1 in C. elegans extends life span through an insulin-dependent
4, 9, 27, and 36 (H3 K4me3, H3 K9me3, H3 K27me3, and H3 K36me3, pathway (119, 120). The opposing effects of H3 K4me3 and H3 K27me3
transcripts may limit life span. Deletion of the relevant demethylase ablation of ATP-dependent nucleosome remodeler function have been
generates a more compact chromatin conformation, resulting in life linked to several disease syndromes and cancers (130, 131). Recently,
span extension (124). Although this study also identified increased cryp- some members of the ATP-dependent nucleosome remodeler families
tic transcripts during C. elegans aging, it remains to be validated whether have also been linked to the aging process.
a similar mechanism also applies in mammals. An interesting correla- Both physiological aging and premature aging syndromes reveal
tion exists between the gene expression change during aging and the signs of extensive alteration of chromatin structure, followed by
level of H3 K36me3 in gene bodies in C. elegans and D. melanogaster increased susceptibility to persistent DNA damage (132). Although sev-
(125). The gene bodies of genes with a robust expression change during eral studies suggest that altered chromatin conformations occur with
aging are marked with almost undetectable levels of H3 K36me3 in both age (43, 44), almost no direct connections established the involvement
organisms, suggesting a conserved mechanism for this modification in of ATP-dependent nucleosome remodelers until recently. The C. elegans
promoting longevity by preventing age-dependent changes in mRNA homolog of the catalytic subunit of the NURD chromatin remodeling
expression. Inactivation or depletion of the methyltransferase met-1 re- complex, LET-418/Mi2, has recently been identified as a longevity-
sulted in a global reduction of H3 K36me3 level, increased changes in determining factor because let-418 deficiency leads to prolonged life span
gene expression during aging, and shortening of life span in C. elegans and increased environmental stress resistance. This phenotype is partially
(125). Together, these studies suggest that loss of histone H3 K36 meth- dependent on the transcription factor DAF-16/FOXO. Genetic interaction
ylation during aging leads to aberrant gene expression, reminiscent of a analysis further suggests that let-418 may serve as a life span determinant
transcriptional drift-like effect, which presumably limits life span. In this by acting through the germ cell loss pathway. This protein is highly evo-
light, it is of interest that a recent report showed that pharmaceutical lutionarily conserved, and indeed, it has been found that these increased
suppression of transcriptional drift extends C. elegans life span by longevity and enhanced stress resistance functions are also conserved
delaying the onset of mortality (126). It is tempting to speculate that in fruit flies and plants (133). One study that aims to identify the mo-
deterioration of the epigenome during aging is responsible for the tran- lecular basis for the age-related defect in chromatin structure used cells
scriptional drift that causes the onset of mortality (Fig. 1). from HGPS patients and noted that components of the NURD ATP-
Histone ubiquitination is another modification that has been impli- dependent nucleosome remodeler complex, including RBBP4 and
cated in yeast replicative life span. A genome-wide screen for long-lived RBBP7, were substantially reduced in HGPS cells (132). Conversely, ex-
strains in yeast identified components of the histone deubiquinase perimental depletion of RBBP4 and RBBP7 in cell culture caused an in-
module (DUBm) of the SAGA (Spt-Ada-Gcn5-acetyltransferase) crease in endogenous DNA damage similar to what has been observed in
complex (127), including SCF73, the yeast ortholog of human Ataxin- HGPS cells and cells from aged individuals. Furthermore, it appeared
7. This protein serves as an adapter protein in the complex, which links likely that the defect in chromatin structure precedes the increased en-
nucleosome remodeler SWI/SNF as a crucial cofactor in transcriptional methylation becomes more and more divergent because of epigenetic
activation of DAF-16 target genes. The well-known prolongevity effect drift caused by environmental factors or spontaneous stochastic errors
of DAF-16/FOXO is dependent on SWI/SNF because RNAi inactivation in the process of transmission of DNA methylation (12). Epigenetic
of core subunits of the SWI/SNF complex abolished DAF-16/FOXO– drift leads to unpredictable differences in the methylome among aging
mediated life span extension (136). Deletion of the catalytic subunit of individuals (Fig. 3). However, some of the methylation changes that oc-
the ATP-dependent nucleosome remodeler Chd1 has also been shown cur with age are directional and involve specific regions of the genome.
to extend yeast replicative life span in a comprehensive analysis of repli- This fact indicates that at least part of the DNA methylation changes
cation life span in almost 4700 deletion mutants (52). Given the con- during aging are not stochastic but could be associated with biological
served roles of the ATP-dependent nucleosome remodelers from yeast mechanisms involved in the aging process. DNA methylation, as well as
to humans, it will be intriguing to explore the potential roles of these re- life span, also differs between organisms of identical genetic makeup,
modelers to modulate life span in mammals. such as queen and worker honeybees, when subjected to different en-
vironmental stimuli, such as diet, which results in an altered gene expres-
sion pattern. Strikingly, similar methylome profiles were experimentally
DNA METHYLATION CHANGES DURING AGING found following RNAi-mediated silencing of a DNA methyltransferase
enzyme, Dnmt3, in laboratory-bred bees (16). Although it is yet to be
DNA methylation is one of the most extensively studied and best- determined whether the diet-induced DNA methylation changes are
characterized epigenetic modifications during aging (Fig. 3) (137). In indeed responsible for causing the transcriptional shift observed in these
young cells, the majority of CpGs within the genome have cytosine cases, it provides great promise for unraveling a key concept of epige-
methylation. CpG methylation within promoters leads to transcription- netic reprogramming.
al repression, through the formation of compact chromatin structures, With a few exceptions, mammalian aging is more commonly associated
such as heterochromatin. Conversely, promoters of genes that are high- with CpG hypomethylation, especially at repetitive DNA sequences
ly expressed are devoid of DNA methylation, hence their name—CpG (Fig. 3) (138–144). This is likely to be at least partly responsible for
islands. DNA methylation is particularly important during develop- the loss of heterochromatin during aging. Loss of CpG methylation
ment, when it is used to silence genes in tissues in which their expression at repetitive sequences will heighten the risk of retrotransposition
is never going to be needed, or at developmental stages in which they no events and, hence, genomic instability during aging, given that the retro-
longer need to be expressed. As identical twins age, the pattern of DNA transposons comprise much of the repetitive DNA. The global decrease
LINE Non–CpG island gene CpG island gene SINE CpG island gene LTR
LINE Non–CpG island gene CpG island gene SINE CpG island gene LTR
CpG poor
DNA repeats promoter CpG island DNA repeats CpG island DNA repeats
Key:
Unmethylated CpG Methylated CpG Transposable elements
Fig. 3. Summary of DNA methylation changes during aging. Young mammalian cells are characterized by DNA hypermethylation over the genome, with
the exception of CpG islands within the promoters of expressed genes. In particular, DNA repeats, such as LINE, SINE, and long terminal repeat (LTR) trans-
posable elements, are heavily DNA-methylated, helping to maintain them in a constitutive heterochromatin state. During aging, there is general DNA hy-
pomethylation over the genome, which mostly occurs in a stochastic manner within the cell population. Loss of DNA methylation leads to activation of
normally silenced DNA sequences like the transposable elements. However, DNA methylation also increases in a nonstochastic manner over the CpG islands
of certain genes, correlating with their heterochromatinization and silencing.
in DNA methylation upon aging may be attributed to the progressive de- mechanistic details of this phenomenon are unclear (162). One of the
cline in levels of the DNA methyltransferase DNMT1 (137). The contri- remaining challenges in the analyses of DNA methylation during aging
bution of DNA methylation to aging is further suggested by a Dnmt1+/− is to identify the causal pathways that contribute to the functional de-
mouse model that showed reduced age-dependent impaired learning cline of the DNA methylome during aging.
and memory function (145). In addition to the general DNA hypo- The age-dependent changes in the DNA methylome are reminiscent
methylation that occurs during aging, progressive age-dependent loss of those occurring in cancer. Global hypomethylation at the repetitive
of DNA methylation also occurs at specific gene promoters, including regions and site-specific hypermethylation at certain promoters have
ITGAL and IL17RC, leading to their transcriptional induction and sub- also been reported during cancer, suggesting a potential connection be-
sequent autoimmune responses (146, 147). Simultaneous with the general tween age-dependent DNA methylation changes and increased cancer
and localized DNA hypomethylation during aging, hypermethylation risk observed in the elderly population (163, 164). Notably, local hyper-
occurs at specific CpG sites of the genome, presumably to repress expres- methylation events during aging sometimes occur at the promoters of
sion of specific genes (Fig. 3) (137, 142, 148, 149). A recent analysis also tumor-suppressor genes, potentially preceding cell transformation
observed loss of DNA methylation from cis regulatory elements, such as events. The analogy between cancer and aging also extends to the
enhancer regions in pancreatic b cells, and suggested the possibility that PcG target genes because both aging and cancer are accompanied by
transcription factor binding may hinder the ability of DNA methyltrans- DNA hypermethylation of these genes. Hence, a better understanding
ferases to eventually recognize those sites, resulting in aberrant gene ex- of the reasons behind the changes in the DNA methylome during aging
pression in aged cells (150). The hypothesis seems promising but is yet to may help us to also understand the causes of cancer.
be addressed during aging in different cells, tissues, or organisms. How-
ever, our current knowledge about the limited tissue types tested so far did
not show a striking global correlation between DNA methylation and al- ncRNAs AND AGING
terations in gene expression profile during hematopoietic stem cell aging
or in whole blood during aging (151, 152). ncRNAs are the most recent players in the epigenetics field, influencing
The development of novel next-generation sequencing technologies seemingly all biological processes in virtually all organisms. The advent
for genome-wide assessment of DNA methylation levels has permitted and widespread use of deep sequencing has provided unbiased insights
the confirmation and extension of earlier studies. A comparison be- into the eukaryotic genome, including the existence and functional roles
tween the methylome of CD4+ T cells from newborn and centenarian of ncRNAs. In contrast to earlier beliefs, it is now widely accepted that
individuals further solidifies the notion of global decreases in DNA approximately 60 to 90% of the human genome is transcribed, with
methylation with aging, accompanied by heterogeneous DNA methyl- some variability observed in other organisms (165, 166), giving rise to
miRNAs are sncRNAs that negatively control their target gene ex- network in mice (190). Loss of imprinting from the IGF2/H19 locus
pression posttranscriptionally and have been implicated in aging. Al- has been shown during aging in both mice and human prostate as-
though miRNAs do not alter the chromatin structure, they are sociated with reexpression of certain inactive genes and loss of binding
considered mediators of epigenetics because they lead to heritable of chromatin-associated protein CTCF (191). This may serve as a po-
changes in gene expression that do not involve a change in DNA tential reason for increased prostate cancer occurrence in aging men,
sequence. The best-characterized examples of roles of miRNAs during further supporting the connection between aging and cancer develop-
aging come from studies in C. elegans. Several miRNAs have been ment. Other lncRNAs that are implicated in major senescence-associated
shown to be involved in modulating life span and in controlling tissue pathways, such as p53/p21 pathways, are differentially expressed in pro-
aging (178, 179). One of the most prominent examples includes the reg- liferating early passage compared to senescent late-passage fibroblast
ulation of aging by the miRNA lin-4 and its pro-aging target miRNA cells (192, 193). Similarly, differential expression patterns of various
lin-14 (178). Loss of function of lin-4 shortens life span, whereas over- lncRNAs have been implicated in the pathophysiology of another
expression of lin-4 extends life span. In contrast, knocking down lin-14, age-onset neurological disorder, Huntington’s disease, wherein some
even in adult animals, extends life span (178). The key question is what of these lncRNAs have been postulated to modulate chromatin archi-
are the target genes of lin-14 that are regulating aging? Of all the miRNAs tecture and/or transcription (194, 195). Other ncRNAs, mostly products
reported in C. elegans, more than a quarter are differentially expressed of the RNAi pathway, are involved in heterochromatin assembly in re-
during aging, including some of the miRNA families conserved with petitive DNA elements in diverse organisms (196). Here, they function
humans (176, 177, 179). Whereas some miRNAs have an antiaging effect to target histone-modifying activities in DNA repeats through reader
by promoting longevity in C. elegans, others show a pro-aging effect by proteins. Other ncRNAs act to organize the three-dimensional orga-
antagonizing longevity (177, 179, 180). Even the expression pattern of nization of chromosomes or as boundaries or insulators to prevent the
some miRNAs serves as a predictor of longevity. However, the evolution- spreading of heterochromatin (197). Whether the changes in chromatin
arily conserved nature of some of these miRNAs indicates that their role structure that occur during aging are due to changes in these particular
in life span regulation likely extends beyond this organism to larger eu- ncRNAs is yet to be examined.
karyotes, including humans. Differential expression of miRNAs is also
evident in mice and humans during aging, although the differences are
not always present in all organs (181–184). TRANSGENERATIONAL EPIGENETIC CHANGES THAT
One of the most notable features of human aging is neurodegenera- AFFECT AGING
tion and reduced brain function. Premature death of neurons is
considered to be a major feature of neurodegenerative diseases. Studies According to biological dogma, genetics governs all the inherited traits
could have contributed to evolution, whereby the ancestors’ environmental cancer agents to treat age-related increases in carcinogenesis (201). Clear-
exposure determined the fate of the descendants. Given the intriguing ly, some of the antiaging effects of CR are through the down-regulation of
nature of the subject, more studies will undoubtedly further explore this the nutrient-sensing mTOR pathway (Fig. 4), and this has been discussed
exciting direction in the near future. elsewhere (204).
Because of the challenges for humans in adherence to a CR regimen,
significant research emphasis has been placed on finding effective “CR
LIFE SPAN–EXTENDING REGIMENS THAT FUNCTION, AT mimetics.” These are compounds or drugs that can essentially mimic
LEAST IN PART, THROUGH EPIGENETIC CHANGES the prolongevity effects of CR in humans by using the same pathways
as CR, without actual restriction of calorie intake. In this context, several
There is considerable interest in extending health span as well as life new, as well as already existing marketed, drugs are being assessed
span. Accordingly, improvements in our knowledge of the basic mech- (205, 206). Given that the sirtuins promote longevity in diverse species,
anisms that cause aging have led to the field that identifies different they have attracted considerable interest as drug targets. The first sirtuin-
ways to promote longevity. Experimental manipulations involving ge- activating compounds (STACs) to be identified included plant-derived
netic, epigenetic, environmental, or even pharmacological interventions metabolites, such as flavones, stilbenes, anthocyanidins, and chalcones
in different model organisms have been extremely informative for dem- (207). The best known of these is resveratrol. Resveratrol and synthetic
onstrating the potential of manipulating the processes under study, to STACs have been shown to induce physiological changes and gene
delay aging. Of all the hallmarks of aging, epigenetic alterations have expression changes that are similar to those that are induced by CR
attracted attention as a promising avenue for life span extension be- and to extend life span in many model organisms (207). The ability
cause, unlike most of other age-promoting processes, epigenetic altera- of resveratrol to extend life span in mice depends on them being fed a
tions can be modulated by inhibition or stimulation of the relevant high-fat diet because there was no extended life span when mice were
enzymes. Thus, interventions targeting epigenetic information hold im- given resveratrol on a standard diet (208). Although resveratrol stimu-
mense potential to extend life span and to counteract age-associated dis- lates Sir2/SIRT1 activity in vitro (209), its stimulatory interaction with
eases, including cancer and neurodegenerative disorders. In this section, SIRT1 depended on binding to the 7-amino-4-methylcoumarin (AMC)
we will discuss the relationship of well-known life span–extending in- fluorophore within the histone peptide substrate used to measure deace-
terventions with epigenetics. tylase activity (210). The fluorophore used in the in vitro HDAC assay
Some of the most effective life span extension regimens function at mimics bulky hydrophobic amino acids, and resveratrol only stimulates
least in part through epigenetic pathways (Fig. 4). Of all the experimen- SIRT1-mediated deacetylation of natural substrates that have large hy-
tal manipulations known to date, by far the most accessible means drophobic residues C-terminal to the acetyl lysine (211–214). The recent
during chronological aging in yeast and human cells and extend the tophagy, contributing to the aging process and diseases of aging. Hence,
life span of flies, worms, and human cells. In agreement, administration dietary and pharmacological manipulations that decrease acetyl-CoA
of spermidine through diet in aging mice also reduced age-related ox- levels might extend life span by stimulating autophagy. Notably, SIRT1
idative stress (222). Another potential way to reduce histone acetylation also stimulates autophagy, wherein SIRT1 induces FOXO-mediated ex-
globally is to feed a diet that depletes acetyl–coenzyme A (CoA), the pression of autophagy pathway components and deacetylates major
donor for acetylation reactions. But how does reducing acetylation ex- players of the autophagy induction pathway (Fig. 4) (228). Indeed, the
tend life span? Depletion of acetyl-CoA by starvation of mammalian longevity-inducing role of CR appears to be through the SIRT1-dependent
cells in culture and fasting in mice has been recently shown to induce induction of autophagy (229). Hence, it is tempting to speculate that
autophagy (223). In agreement, spermidine also induced autophagy, multiple different manipulations to extend life span, such as CR, acetyl-
extending chronological life span in yeast (223, 224). Autophagy may CoA depletion, polyamines, and sirtuin activation, all function through
be regarded as a mechanism of cellular “cleansing,” and its up-regulation a common pathway to stimulate autophagy, potentially through an ef-
has clear cellular benefits that would promote longevity (225). According- fect on histone acetylation (Fig. 4).
ly, note that stimulating autophagy is a common theme among multiple An exciting new way to improve health span and, subsequently, life
interventions that extend life span (Fig. 4) (226). Recently, histone acet- span in mice is to specifically kill off senescent cells with senolytic agents
ylation has also been implicated in the mechanism whereby autophagy is (230). Given the epigenetic changes that are required for the maintenance
activated in metazoans. Fly life span is extended and autophagy is induced of senescence, another potential avenue to extend life span may be to de-
when acetyl-CoA levels were reduced in fly brains (224). The mechanism velop senolytic drugs that target specific epigenetic enzymes.
wherein high acetyl-CoA levels block autophagy is not totally clear, but
it likely involves histone acetylation. In one study, the HAT p300, whose
substrates include histone H3, was required for high levels of acetyl-CoA CONCLUSION AND FUTURE DIRECTIONS (RESETTING THE
to block autophagy (223). In another study, high levels of H4 K16Ac (the AGING CLOCK)
substrate of Sir2/SIRT1) down-regulated autophagy, and low levels of H4
K16Ac induced autophagy (227). The natural increase in H4 K16Ac that The aging process is unquestionably complex. During an organism’s
occurs during aging, because of Sir2 loss, could lead to the decline of au- lifetime, aging cells undergo a plethora of changes and accumulate
Longevity
Nutrients Ac
SIRT1 Autophagy
Ins
uli
n
1
SIRT1
F-
IG
Me ATG genes
Cy cleus
Nu
Heterochromatin
top
reorganization
Intervention
DNMTs
during aging
lasm
Spermidine
Ac
HATs
Ac SIRT1
Acetyl CoA STACs
depletion
Ac Ac Ac
RNA
Ac
X
Ac
ATG genes
Fig. 4. Schematic showing how some external interventions trigger longevity, often at least partly through stimulating autophagy. The pink
writing refers to dietary, chemical, or therapeutic interventions that can extend life span, in at least some organisms (described in the text). Arrows indicate
stimulating effects, and blocked lines indicate inhibitory effects. This schematic is not meant to be exhaustive but highlights the pathways that alter the
epigenetic information and autophagy.
macromolecular damage. The aging phenotypes are manifested by the interventions on the epigenome in an aging individual, given the in-
summation of alterations of many different signaling cascades. Here, we terconnectivity of the epigenetic mechanisms.
attempted to provide an overview of some of the major changes in aging
cells, tissues, and organisms that have come to light in recent years
REFERENCES AND NOTES
through the lens of chromatin and epigenetic regulation. Genetic and
environmental manipulations are unequivocally important to decipher 1. A. Brunet, S. L. Berger, Epigenetics of aging and aging-related disease. J. Gerontol. A Biol.
Sci. Med. Sci. 69 (Suppl. 1), S17–S20 (2014).
the effect of any specific factor over the longevity process. It is becoming 2. J. Feser, J. Tyler, Chromatin structure as a mediator of aging. FEBS Lett. 585, 2041–2048
apparent mechanistically that many of those factors that do affect lon- (2011).
gevity act primarily through the modification of the epigenome. Un- 3. B. K. Kennedy, S. L. Berger, A. Brunet, J. Campisi, A. M. Cuervo, E. S. Epel, C. Franceschi,
doubtedly, epigenetic influences over the aging process need to be G. J. Lithgow, R. I. Morimoto, J. E. Pessin, T. A. Rando, A. Richardson, E. E. Schadt,
T. Wyss-Coray, F. Sierra, Geroscience: Linking aging to chronic disease. Cell 159,
incorporated into our current understanding of aging. Almost all classes
709–713 (2014).
of epigenetic alterations reported so far influence longevity pathways, 4. A. Moskalev, A. Aliper, Z. Smit-McBride, A. Buzdin, A. Zhavoronkov, Genetics and epige-
adding further complexity to understanding the aging process. In sum- netics of aging and longevity. Cell Cycle 13, 1063–1077 (2014).
mary, young healthy cells maintain an epigenetic state that promotes 5. J. Campisi, Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75, 685–705 (2013).
6. D. Muñoz-Espín, M. Serrano, Cellular senescence: From physiology to pathology. Nat. Rev.
the formation of a compact chromatin structure and precise regulation
Mol. Cell Biol. 15, 482–496 (2014).
of all the basic biological processes. However, aging cells experience al- 7. C. López-Otín, M. A. Blasco, L. Partridge, M. Serrano, G. Kroemer, The hallmarks of aging.
terations in all aspects of the chromatin landscape, DNA accessibility, Cell 153, 1194–1217 (2013).
and ncRNA production, until a threshold of altered gene expression and 8. A. D. Goldberg, C. D. Allis, E. Bernstein, Epigenetics: A landscape takes shape. Cell 128,
compromised genomic integrity is crossed, and the cells finally succumb 635–638 (2007).
9. U. Muñoz-Najar, J. M. Sedivy, Epigenetic control of aging. Antioxid. Redox Signal. 14,
to a permanent halt in progression through the cell cycle. The reversible 241–259 (2011).
nature of epigenetic mechanisms makes it possible to restore or reverse 10. R. J. O’Sullivan, J. Karlseder, The great unravelling: Chromatin as a modulator of the aging
some of these phenotypes to attain more youthful cells. process. Trends Biochem. Sci. 37, 466–476 (2012).
Whereas some of the molecular changes during aging can be cate- 11. L. Zane, V. Sharma, T. Misteli, Common features of chromatin in aging and cancer: Cause
or coincidence? Trends Cell Biol. 24, 686–694 (2014).
gorized as causal to aging, other changes simply accompany the aging
12. M. F. Fraga, E. Ballestar, M. F. Paz, S. Ropero, F. Setien, M. L. Ballestar, D. Heine-Suñer,
process. However, when characterizing the causes or consequences of J. C. Cigudosa, M. Urioste, J. Benitez, M. Boix-Chornet, A. Sanchez-Aguilera, C. Ling,
aging, one has to carefully dissect the experimental findings because E. Carlsson, P. Poulsen, A. Vaag, Z. Stephan, T. D. Spector, Y.-Z. Wu, C. Plass, M. Esteller,
most of the relevant pathways are interconnected, making it difficult Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad.
to separate the effects of the pro-aging pathways from bystander path- Sci. U.S.A. 102, 10604–10609 (2005).
13. M. Sargent, Why twins age differently. Nature 464, 1130–1131 (2010).
30. T. Smeal, J. Claus, B. Kennedy, F. Cole, L. Guarente, Loss of transcriptional silencing causes A. A. Rodriguez, B. Ros, M. Sage, M. K. Singh, E. D. Smith, K. Snead, A. Solanky, B. L. Spector,
sterility in old mother cells of S. cerevisiae. Cell 84, 633–642 (1996). K. K. Steffen, B. Nga Tchao, M. K. Ting, H. Vander Wende, D. Wang, K. Linnea Welton,
31. L. Guarente, Franklin H. Epstein Lecture: Sirtuins, aging, and medicine. N. Engl. J. Med. E. A. Westman, R. B. Brem, X.-. Liu, Y. Suh, Z. Zhou, M. Kaeberlein, B. K. Kennedy, A compre-
364, 2235–2244 (2011). hensive analysis of replicative lifespan in 4,698 single-gene deletion strains uncovers con-
32. M. C. Haigis, D. A. Sinclair, Mammalian sirtuins: Biological insights and disease relevance. served mechanisms of aging. Cell Metab. 22, 895–906 (2015).
Annu. Rev. Pathol. 5, 253–295 (2010). 53. C. Lee, V. Longo, Dietary restriction with and without caloric restriction for healthy aging.
33. P. Oberdoerffer, S. Michan, M. McVay, R. Mostoslavsky, J. Vann, S.-K. Park, A. Hartlerode, F1000Res. 5, pii: F1000 (2016).
J. Stegmuller, A. Hafner, P. Loerch, S. M. Wright, K. D. Mills, A. Bonni, B. A. Yankner, 54. G. McColl; Supported by the American Federation for Aging Research, D. W. Killilea,
R. Scully, T. A. Prolla, F. W. Alt, D. A. Sinclair, SIRT1 redistribution on chromatin promotes A. E. Hubbard, M. C. Vantipalli, S. Melov, G. J. Lithgow, Pharmacogenetic analysis of lithium-
genomic stability but alters gene expression during aging. Cell 135, 907–918 (2008). induced delayed aging in Caenorhabditis elegans. J. Biol. Chem. 283, 350–357 (2008).
34. B. K. Kennedy, M. Gotta, D. A. Sinclair, K. Mills, D. S. McNabb, M. Murthy, S. M. Pak, 55. Z. Ni, A. Ebata, E. Alipanahiramandi, S. S. Lee, Two SET domain containing genes link
T. Laroche, S. M. Gasser, L. Guarente, Redistribution of silencing proteins from telomeres to epigenetic changes and aging in Caenorhabditis elegans. Aging Cell 11, 315–325 (2012).
the nucleolus is associated with extension of life span in S. cerevisiae. Cell 89, 381–391 (1997). 56. R. J. O’Sullivan, S. Kubicek, S. L. Schreiber, J. Karlseder, Reduced histone biosynthesis and
35. P. Oberdoerffer, D. A. Sinclair, The role of nuclear architecture in genomic instability and chromatin changes arising from a damage signal at telomeres. Nat. Struct. Mol. Biol. 17,
ageing. Nat. Rev. Mol. Cell Biol. 8, 692–702 (2007). 1218–1225 (2010).
36. T. Kobayashi, Regulation of ribosomal RNA gene copy number and its role in modulating 57. A. Ivanov, J. Pawlikowski, I. Manoharan, J. van Tuyn, D. M. Nelson, T. S. Rai, P. P. Shah,
genome integrity and evolutionary adaptability in yeast. Cell. Mol. Life Sci. 68, 1395–1403 G. Hewitt, V. I. Korolchuk, J. F. Passos, H. Wu, S. L. Berger, P. D. Adams, Lysosome-
(2011). mediated processing of chromatin in senescence. J. Cell Biol. 202, 129–143 (2013).
37. D. A. Sinclair, L. Guarente, Extrachromosomal rDNA circles—A cause of aging in yeast. Cell 58. B. Liu, R. Yip, Z. Zhou, Chromatin remodeling, DNA damage repair and aging. Curr. Genomics
91, 1033–1042 (1997). 13, 533–547 (2012).
38. R. D. Goldman, D. K. Shumaker, M. R. Erdos, M. Eriksson, A. E. Goldman, L. B. Gordon, 59. L.-J. Mah, A. El-Osta, T. C. Karagiannis, gH2AX: A sensitive molecular marker of DNA dam-
Y. Gruenbaum, S. Khuon, M. Mendez, R. Varga, F. S. Collins, Accumulation of mutant lamin age and repair. Leukemia 24, 679–686 (2010).
A causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syn- 60. O. A. Sedelnikova, I. Horikawa, C. Redon, A. Nakamura, D. B. Zimonjic, N. C. Popescu,
drome. Proc. Natl. Acad. Sci. U.S.A. 101, 8963–8968 (2004). W. M. Bonner, Delayed kinetics of DNA double-strand break processing in normal and
39. P. Scaffidi, T. Misteli, Lamin A-dependent nuclear defects in human aging. Science 312, pathological aging. Aging Cell 7, 89–100 (2008).
1059–1063 (2006). 61. O. A. Sedelnikova, I. Horikawa, D. B. Zimonjic, N. C. Popescu, W. M. Bonner, J. C. Barrett,
40. D. K. Shumaker, T. Dechat, A. Kohlmaier, S. A. Adam, M. R. Bozovsky, M. R. Erdos, Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-
M. Eriksson, A. E. Goldman, S. Khuon, F. S. Collins, T. Jenuwein, R. D. Goldman, Mutant strand breaks. Nat. Cell Biol. 6, 168–170 (2004).
nuclear lamin A leads to progressive alterations of epigenetic control in premature aging. 62. A. M. Garcia, R. B. Calder, M. E. T. Dollé, M. Lundell, P. Kapahi, J. Vijg, Age- and temperature-
Proc. Natl. Acad. Sci. U.S.A. 103, 8703–8708 (2006). dependent somatic mutation accumulation in Drosophila melanogaster. PLOS Genet. 6,
41. W. Zhang, J. Li, K. Suzuki, J. Qu, P. Wang, J. Zhou, X. Liu, R. Ren, X. Xu, A. Ocampo, e1000950 (2010).
T. Yuan, J. Yang, Y. Li, L. Shi, D. Guan, H. Pan, S. Duan, Z. Ding, M. Li, F. Yi, R. Bai, 63. J. Vijg, M. E. T. Dollé, Large genome rearrangements as a primary cause of aging.
Y. Wang, C. Chen, F. Yang, X. Li, Z. Wang, E. Aizawa, A. Goebl, R. Devi Soligalla, Mech. Ageing Dev. 123, 907–915 (2002).
P. Reddy, C. Rodriguez Esteban, F. Tang, G.-H. Liu, J. Carlos Izpisua Belmonte, Aging 64. A. Kaya, A. V. Lobanov, V. N. Gladyshev, Evidence that mutation accumulation does not
stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations cause aging in Saccharomyces cerevisiae. Aging Cell 14, 366–371 (2015).
77. H. Tan, A. Qurashi, M. Poidevin, D. L. Nelson, H. Li, P. Jin, Retrotransposon activation 103. S. Kim, A. Benguria, C.-Y. Lai, S. M. Jazwinski, Modulation of life-span by histone deace-
contributes to fragile X premutation rCGG-mediated neurodegeneration. Hum. Mol. Genet. tylase genes in Saccharomyces cerevisiae. Mol. Biol. Cell 10, 3125–3136 (1999).
21, 57–65 (2012). 104. B. Rogina, S. L. Helfand, S. Frankel, Longevity regulation by Drosophila Rpd3 deacetylase
78. R. T. Kamakaka, S. Biggins, Histone variants: Deviants? Genes Dev. 19, 295–310 (2005). and caloric restriction. Science 298, 1745 (2002).
79. P. J. Skene, S. Henikoff, Histone variants in pluripotency and disease. Development 140, 105. S. Frankel, J. Woods, T. Ziafazeli, B. Rogina, RPD3 histone deacetylase and nutrition have
2513–2524 (2013). distinct but interacting effects on Drosophila longevity. Aging 7, 1112–1129 (2015).
80. D. Filipescu, S. Muller, G. Almouzni, Histone H3 variants and their chaperones during 106. S. H. Ryu, K. Kang, T. Yoo, C. O. Joe, J. H. Chung, Transcriptional repression of repeat-
development and disease: Contributing to epigenetic control. Annu. Rev. Cell Dev. Biol. derived transcripts correlates with histone hypoacetylation at repetitive DNA elements
30, 615–646 (2014). in aged mice brain. Exp. Gerontol. 46, 811–818 (2011).
81. L. F. Duarte, A. R. J. Young, Z. Wang, H.-A. Wu, T. Panda, Y. Kou, A. Kapoor, D. Hasson, 107. S. Peleg, F. Sananbenesi, A. Zovoilis, S. Burkhardt, S. Bahari-Javan, R. C. Agis-Balboa,
N. R. Mills, A. Ma’ayan, M. Narita, E. Bernstein, Histone H3.3 and its proteolytically pro- P. Cota, J. L. Wittnam, A. Gogol-Doering, L. Opitz, G. Salinas-Riester, M. Dettenhofer,
cessed form drive a cellular senescence programme. Nat. Commun. 5, 5210 (2014). H. Kang, L. Farinelli, W. Chen, A. Fischer, Altered histone acetylation is associated with
82. I. Maze, W. Wenderski, K. M. Noh, R. C. Bagot, N. Tzavaras, I. Purushothaman, S. J. Elsasser, age-dependent memory impairment in mice. Science 328, 753–756 (2010).
Y. Guo, C. Ionete, Y. L. Hurd, C. A. Tamminga, T. Halene, L. Farrelly, A. A. Soshnev, D. Wen, 108. J. G. Wood, S. Hillenmeyer, C. Lawrence, C. Chang, S. Hosier, W. Lightfoot, E. Mukherjee,
S. Rafii, M. R. Birtwistle, S. Akbarian, B. A. Buchholz, R. D. Blitzer, E. J. Nestler, Z.-F. Yuan, N. Jiang, C. Schorl, A. S. Brodsky, N. Neretti, S. L. Helfand, Chromatin remodeling in the
B. A. Garcia, L. Shen, H. Molina, C. David Allis, Critical role of histone turnover in neuronal aging genome of Drosophila. Aging Cell 9, 971–978 (2010).
transcription and plasticity. Neuron 87, 77–94 (2015). 109. C. M. Wang, S. N. Tsai, T. W. Yew, Y. W. Kwan, S. M. Ngai, Identification of histone meth-
83. P. D. Adams, Remodeling of chromatin structure in senescent cells and its potential ylation multiplicities patterns in the brain of senescence-accelerated prone mouse 8.
impact on tumor suppression and aging. Gene 397, 84–93 (2007). Biogerontology 11, 87–102 (2010).
84. R. Zhang, M. V. Poustovoitov, X. Ye, H. A. Santos, W. Chen, S. M. Daganzo, 110. B. S. McCauley, W. Dang, Histone methylation and aging: Lessons learned from model
J. P. Erzberger, I. G. Serebriiskii, A. A. Canutescu, R. L. Dunbrack, J. R. Pehrson, systems. Biochim. Biophys. Acta 1839, 1454–1462 (2014).
J. M. Berger, P. D. Kaufman, P. D. Adams, Formation of macroH2A-containing senescence- 111. I. M. Bochkis, D. Przybylski, J. Chen, A. Regev, Changes in nucleosome occupancy asso-
associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev. Cell 8, ciated with metabolic alterations in aged mammalian liver. Cell Rep. 9, 996–1006 (2014).
19–30 (2005). 112. I. Cheung, H. P. Shulha, Y. Jiang, A. Matevossian, J. Wang, Z. Weng, S. Akbarian, Develop-
85. A. Gaspar-Maia, Z. A. Qadeer, D. Hasson, K. Ratnakumar, N. A. Leu, G. Leroy, S. Liu, mental regulation and individual differences of neuronal H3K4me3 epigenomes in the
C. Costanzi, D. Valle-Garcia, C. Schaniel, I. Lemischka, B. Garcia, J. R. Pehrson, prefrontal cortex. Proc. Natl. Acad. Sci. U.S.A. 107, 8824–8829 (2010).
E. Bernstein, MacroH2A histone variants act as a barrier upon reprogramming towards 113. H. P. Shulha, I. Cheung, Y. Guo, S. Akbarian, Z. Weng, Coordinated cell type–specific ep-
pluripotency. Nat. Commun. 4, 1565 (2013). igenetic remodeling in prefrontal cortex begins before birth and continues into early
86. V. Pasque, R. P. Halley-Stott, A. Gillich, N. Garrett, J. B. Gurdon, Epigenetic stability of adulthood. PLOS Genet. 9, e1003433 (2013).
repressed states involving the histone variant macroH2A revealed by nuclear transfer 114. D. Sun, M. Luo, M. Jeong, B. Rodriguez, Z. Xia, R. Hannah, H. Wang, T. Le, K. F. Faull,
to Xenopus oocytes. Nucleus 2, 533–539 (2011). R. Chen, H. Gu, C. Bock, A. Meissner, B. Göttgens, G. J. Darlington, W. Li, M. A. Goodell,
87. J. A. Kreiling, M. Tamamori-Adachi, A. N. Sexton, J. C. Jeyapalan, U. Munoz-Najar, Epigenomic profiling of young and aged HSCs reveals concerted changes during aging
A. L. Peterson, J. Manivannan, E. S. Rogers, N. A. Pchelintsev, P. D. Adams, J. M. Sedivy, that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).
Age-associated increase in heterochromatic marks in murine and primate tissues. Aging Cell 115. B. Hamilton, Y. Dong, M. Shindo, W. Liu, I.Odell, G. Ruvkun, S. S. Lee, A systematic RNAi
10, 292–304 (2011). screen for longevity genes in C. elegans. Genes Dev. 19, 1544–1555 (2005).
128. J. Cao, Q. Yan, Histone ubiquitination and deubiquitination in transcription, DNA damage I. Iglesias-Platas, F. Formiga, A. F. Fernandez, M. F. Fraga, S. C. Heath, A. Valencia,
response, and cancer. Front. Oncol. 2, 26 (2012). I. G. Gut, J. Wang, M. Esteller, Distinct DNA methylomes of newborns and centenarians.
129. J. D. Aalfs, R. E. Kingston, What does ‘chromatin remodeling’ mean? Trends Biochem. Sci. Proc. Natl. Acad. Sci. U.S.A. 109, 10522–10527 (2012).
25, 548–555 (2000). 154. C. I. Weidner, Q. Lin, C. M. Koch, L. Eisele, F. Beier, P. Ziegler, D. O. Bauerschlag, K. H. Jöckel,
130. C. R. Clapier, B. R. Cairns, The biology of chromatin remodeling complexes. Annu. Rev. Biochem. R. Erbel, T. W. Mühleisen, M. Zenke, T. Henrik Brümmendorf, W. Wagner, Aging of blood can
78, 273–304 (2009). be tracked by DNA methylation changes at just three CpG sites. Genome Biol. 15, R24 (2014).
131. G. J. Narlikar, R. Sundaramoorthy, T. Owen-Hughes, Mechanisms and functions of ATP- 155. M. T. Bocker, I. Hellwig, A. Breiling, V. Eckstein, A. D. Ho, F. Lyko, Genome-wide promoter
dependent chromatin-remodeling enzymes. Cell 154, 490–503 (2013). DNA methylation dynamics of human hematopoietic progenitor cells during differenti-
132. G. Pegoraro, N. Kubben, U. Wickert, H. Göhler, K. Hoffmann, T. Misteli, Ageing-related ation and aging. Blood 117, e182–e189 (2011).
chromatin defects through loss of the NURD complex. Nat. Cell Biol. 11, 1261–1267 (2009). 156. S. Maegawa, G. Hinkal, H. S. Kim, L. Shen, L. Zhang, J. Zhang, N. Zhang, S. Liang,
133. V. De Vaux, C. Pfefferli, M. Passannante, K. Belhaj, A. von Essen, S. G. Sprecher, F. Müller, L. A. Donehower, J.-P. J. Issa, Widespread and tissue specific age-related DNA methyla-
C. Wicky, The Caenorhabditis elegans LET-418/Mi2 plays a conserved role in lifespan reg- tion changes in mice. Genome Res. 20, 332–340 (2010).
ulation. Aging Cell 12, 1012–1020 (2013). 157. A. E. Teschendorff, U. Menon, A. Gentry-Maharaj, S. J. Ramus, D. J. Weisenberger, H. Shen,
134. W. Dang, G. L. Sutphin, J. A. Dorsey, G. L. Otte, K. Cao, R. M. Perry, J. J. Wanat, D. Saviolaki, M. Campan, H. Noushmehr, C. G. Bell, A. P. Maxwell, D. A. Savage, E. Mueller-Holzner,
C. J. Murakami, S. Tsuchiyama, B. Robison, B. D. Gregory, M. Vermeulen, R. Shiekhattar, C. Marth, G. Kocjan, S. A. Gayther, A. Jones, S. Beck, W. Wagner, P. W. Laird, I. J. Jacobs,
F. B. Johnson, B. K. Kennedy, M. Kaeberlein, S. L. Berger, Inactivation of yeast Isw2 chro- M. Widschwendter, Age-dependent DNA methylation of genes that are suppressed in
matin remodeling enzyme mimics longevity effect of calorie restriction via induction of stem cells is a hallmark of cancer. Genome Res. 20, 440–446 (2010).
genotoxic stress response. Cell Metab. 19, 952–966 (2014). 158. J. L. McClay, K. A. Aberg, S. L. Clark, S. Nerella, G. Kumar, L. Y. Xie, A. D. Hudson, A. Harada,
135. M. E. Moynahan, M. Jasin, Mitotic homologous recombination maintains genomic stabil- C. M. Hultman, P. K. Magnusson, P. F. Sullivan, E. J. C. G. Van Den Oord, A methylome-wide
ity and suppresses tumorigenesis. Nat. Rev. Mol. Cell Biol. 11, 196–207 (2010). study of aging using massively parallel sequencing of the methyl-CpG-enriched genomic
136. C. G. Riedel, R. H. Dowen, G. F. Lourenco, N. V. Kirienko, T. Heimbucher, J. A. West, fraction from blood in over 700 subjects. Hum. Mol. Genet. 23, 1175–1185 (2014).
S. K. Bowman, R. E. Kingston, A. Dillin, J. M. Asara, G. Ruvkun, DAF-16 employs the chro- 159. G. Raddatz, S. Hagemann, D. Aran, J. Söhle, P. P. Kulkarni, L. Kaderali, A. Hellman,
matin remodeller SWI/SNF to promote stress resistance and longevity. Nat. Cell Biol. 15, M. Winnefeld, F. Lyko, Aging is associated with highly defined epigenetic changes in
491–501 (2013). the human epidermis. Epigenetics Chromatin 6, 36 (2013).
137. M. Jung, G. P. Pfeifer, Aging and DNA methylation. BMC Biol. 13, 7 (2015). 160. C. M. Koch, S. Joussen, A. Schellenberg, Q. Lin, M. Zenke, W. Wagner, Monitoring of
138. H. T. Bjornsson, M. I. Sigurdsson, M. D. Fallin, R. A. Irizarry, T. Aspelund, H. Cui, W. Yu, cellular senescence by DNA-methylation at specific CpG sites. Aging Cell 11, 366–369
M. A. Rongione, T. J. Ekström, T. B. Harris, L. J. Launer, G. Eiriksdottir, M. F. Leppert, (2012).
C. Sapienza, V. Gudnason, A. P. Feinberg, Intra-individual change over time in DNA meth- 161. L. J. Ions, L. A. Wakeling, H. J. Bosomworth, J. E. J. Hardyman, S. M. Escolme, D. C. Swan,
ylation with familial clustering. JAMA 299, 2877–2883 (2008). R. A. Valentine, J. C. Mathers, D. Ford, Effects of Sirt1 on DNA methylation and expression
139. V. Bollati, J. Schwartz, R. Wright, A. Litonjua, L. Tarantini, H. Suh, D. Sparrow, P. Vokonas, of genes affected by dietary restriction. Age 35, 1835–1849 (2013).
A. Baccarelli, Decline in genomic DNA methylation through aging in a cohort of elderly 162. L. A. Wakeling, L. J. Ions, S. M. Escolme, S. J. Cockell, T. Su, M. Dey, E. V. Hampton,
subjects. Mech. Ageing Dev. 130, 234–239 (2009). G. Jenkins, L. J. Wainwright, J. A. McKay, D. Ford, SIRT1 affects DNA methylation of poly-
140. F. Bormann, M. Rodríguez-Paredes, S. Hagemann, H. Manchanda, B. Kristof, J. Gutekunst, comb group protein target genes, a hotspot of the epigenetic shift observed in ageing.
G. Raddatz, R. Haas, L. Terstegen, H. Wenck, L. Kaderali, M. Winnefeld, F. Lyko, Reduced Hum. Genomics 9, 14 (2015).
DNA methylation patterning and transcriptional connectivity define human skin aging. 163. T. Finkel, M. Serrano, M. A. Blasco, The common biology of cancer and ageing. Nature
Z. Moqtaderi, Z. Zhu, X. Xu, S. Squazzo, M. J. Oberley, D. Inman, M. A. Singer, 185. N. Liu, M. Landreh, K. Cao, M. Abe, G.-J. Hendriks, J. R. Kennerdell, Y. Zhu, L.-S. Wang,
T. A. Richmond, K. J. Munn, A. Rada-Iglesias, O. Wallerman, J. Komorowski, J. C. Fowler, N. M. Bonini, The microRNA miR-34 modulates ageing and neurodegeneration in Drosophila.
P. Couttet, A. W. Bruce, O. M. Dovey, P. D. Ellis, C. F. Langford, D. A. Nix, G. Euskirchen, Nature 482, 519–523 (2012).
S. Hartman, A. E. Urban, P. Kraus, S. Van Calcar, N. Heintzman, T. H. Kim, K. Wang, C. Qu, 186. W.-X. Wang, Q. Huang, Y. Hu, A. J. Stromberg, P. T. Nelson, Patterns of microRNA expres-
G. Hon, R. Luna, C. K. Glass, M. G. Rosenfeld, S. F. Aldred, S. J. Cooper, A. Halees, J. M. Lin, sion in normal and early Alzheimer’s disease human temporal cortex: White matter ver-
H. P. Shulha, X. Zhang, M. Xu, J. N. Haidar, Y. Yu, Y. Ruan, V. R. Iyer, R. D. Green, C. Wadelius, sus gray matter. Acta Neuropathol. 121, 193–205 (2011).
P. J. Farnham, B. Ren, R. A. Harte, A. S. Hinrichs, H. Trumbower, H. Clawson, 187. X. Wang, P. Liu, H. Zhu, Y. Xu, C. Ma, X. Dai, L. Huang, Y. Liu, L. Zhang, C. Qin, miR-34a, a
J. Hillman-Jackson, A. S. Zweig, K. Smith, A. Thakkapallayil, G. Barber, R. M. Kuhn, microRNA up-regulated in a double transgenic mouse model of Alzheimer’s disease, in-
D. Karolchik, L. Armengol, C. P. Bird, P. I. de Bakker, A. D. Kern, N. Lopez-Bigas, hibits bcl2 translation. Brain Res. Bull. 80, 268–273 (2009).
J. D. Martin, B. E. Stranger, A. Woodroffe, E. Davydov, A. Dimas, E. Eyras, 188. J. Lee, A. Padhye, A. Sharma, G. Song, J. Miao, Y.-Y. Mo, L. Wang, J. K. Kemper, A
I. B. Hallgrímsdóttir, J. Huppert, M. C. Zody, G. R. Abecasis, X. Estivill, G. G. Bouffard, pathway involving farnesoid X receptor and small heterodimer partner positively
X. Guan, N. F. Hansen, J. R. Idol, V. V. Maduro, B. Maskeri, J. C. McDowell, M. Park, regulates hepatic sirtuin 1 levels via microRNA-34a inhibition. J. Biol. Chem. 285,
P. J. Thomas, A. C. Young, R. W. Blakesley, D. M. Muzny, E. Sodergren, D. A. Wheeler, 12604–12611 (2010).
K. C. Worley, H. Jiang, G. M. Weinstock, R. A. Gibbs, T. Graves, R. Fulton, E. R. Mardis, 189. I. Meier, L. Fellini, M. Jakovcevski, M. Schachner, F. Morellini, Expression of the snoRNA
R. K. Wilson, M. Clamp, J. Cuff, S. Gnerre, D. B. Jaffe, J. L. Chang, K. Lindblad-Toh, host gene gas5 in the hippocampus is upregulated by age and psychogenic stress and
E. S. Lander, M. Koriabine, M. Nefedov, K. Osoegawa, Y. Yoshinaga, B. Zhu, correlates with reduced novelty-induced behavior in C57BL/6 mice. Hippocampus 20,
P. J. de Jong, Identification and analysis of functional elements in 1% of the human ge- 1027–1036 (2010).
nome by the ENCODE pilot project. Nature 447, 799–816 (2007). 190. P. Monnier, C. Martinet, J. Pontis, I. Stancheva, S. Ait-Si-Ali, L. Dandolo, H19 lncRNA
166. J. E. Wilusz, H. Sunwoo, D. L. Spector, Long noncoding RNAs: Functional surprises from controls gene expression of the imprinted gene network by recruiting MBD1. Proc. Natl.
the RNA world. Genes Dev. 23, 1494–1504 (2009). Acad. Sci. U.S.A. 110, 20693–20698 (2013).
167. S. Djebali, C. A. Davis, A. Merkel, A. Dobin, T. Lassmann, A. Mortazavi, A. Tanzer, J. Lagarde, 191. V. X. Fu, J. R. Dobosy, J. A. Desotelle, N. Almassi, J. A. Ewald, R. Srinivasan, M. Berres,
W. Lin, F. Schlesinger, C. Xue, G. K. Marinov, J. Khatun, B. A. Williams, C. Zaleski, J. Svaren, R. Weindruch, D. F. Jarrard, Aging and cancer-related loss of insulin-like
J. Rozowsky, M. Röder, F. Kokocinski, R. F. Abdelhamid, T. Alioto, I. Antoshechkin, growth factor 2 imprinting in the mouse and human prostate. Cancer Res. 68, 6797–6802
M. T. Baer, N. S. Bar, P. Batut, K. Bell, I. Bell, S. Chakrabortty, X. Chen, J. Chrast, (2008).
J. Curado, T. Derrien, J. Drenkow, E. Dumais, J. Dumais, R. Duttagupta, E. Falconnet, 192. K. Abdelmohsen, A. Panda, M.-J. Kang, J. Xu, R. Selimyan, J.-H. Yoon, J. L. Martindale, S. De,
M. Fastuca, K. Fejes-Toth, P. Ferreira, S. Foissac, M. J. Fullwood, H. Gao, D. Gonzalez, W. H. Wood III, K. G. Becker, M. Gorospe, Senescence-associated lncRNAs: Senescence-
A. Gordon, H. Gunawardena, C. Howald, S. Jha, R. Johnson, P. Kapranov, B. King, associated long noncoding RNAs. Aging Cell 12, 890–900 (2013).
C. Kingswood, O. J. Luo, E. Park, K. Persaud, J. B. Preall, P. Ribeca, B. Risk, D. Robyr, 193. S. Kour, P. C. Rath, Long noncoding RNAs in aging and age-related diseases. Ageing Res. Rev.
M. Sammeth, L. Schaffer, L.-H. See, A. Shahab, J. Skancke, A. Maria Suzuki, H. Takahashi, 26, 1–21 (2015).
H. Tilgner, D. Trout, N. Walters, H. Wang, J. Wrobel, Y. Yu, X. Ruan, Y. Hayashizaki, J. Harrow, 194. R. Johnson, Long non-coding RNAs in Huntington’s disease neurodegeneration. Neurobiol. Dis.
M. Gerstein, T. Hubbard, A. Reymond, S. E. Antonarakis, G. Hannon, M. C. Giddings, 46, 245–254 (2012).
Y. Ruan, B. Wold, P. Carninci, R. Guigó, T. R. Gingeras, Landscape of transcription in human 195. C. Soldati, A. Bithell, C. Johnston, K.-Y. Wong, L. W. Stanton, N. J. Buckley, Dysregulation of
cells. Nature 489, 101–108 (2012). REST-regulated coding and non-coding RNAs in a cellular model of Huntington’s disease.
168. M. J. Hangauer, I. W. Vaughn, M. T. McManus, Pervasive transcription of the human ge- J. Neurochem. 124, 418–430 (2013).
nome produces thousands of previously unidentified long intergenic noncoding RNAs. 196. S. E. Castel, R. A. Martienssen, RNA interference in the nucleus: Roles for small RNAs in
210. D. Cao, M. Wang, X. Qiu, D. Liu, H. Jiang, N. Yang, R.-M. Xu, Structural basis for allosteric, 222. T. Eisenberg, H. Knauer, A. Schauer, S. Buttner, C. Ruckenstuhl, D. Carmona-Gutierrez,
substrate-dependent stimulation of SIRT1 activity by resveratrol. Genes Dev. 29, J. Ring, S. Schroeder, C. Magnes, L. Antonacci, H. Fussi, L. Deszcz, R. Hartl, E. Schraml,
1316–1325 (2015). A. Criollo, E. Megalou, D. Weiskopf, P. Laun, G. Heeren, M. Breitenbach,
211. H. Dai, A. W. Case, T. V. Riera, T. Considine, J. E. Lee, Y. Hamuro, H. Zhao, Y. Jiang, B. Grubeck-Loebenstein, E. Herker, B. Fahrenkrog, K.-U. Fröhlich, F. Sinner,
S. M. Sweitzer, B. Pietrak, B. Schwartz, C. A. Blum, J. S. Disch, R. Caldwell, N. Tavernarakis, N. Minois, G. Kroemer, F. Made, Induction of autophagy by spermidine
B. Szczepankiewicz, C. Oalmann, P. Y. Ng, B. H. White, R. Casaubon, R. Narayan, promotes longevity. Nat. Cell Biol. 11, 1305–1314 (2009).
K. Koppetsch, F. Bourbonais, B. Wu, J. Wang, D. Qian, F. Jiang, C. Mao, M. Wang, E. Hu, 223. G. Mariño, F. Pietrocola, T. Eisenberg, Y. Kong, S. A. Malik, A. Andryushkova, S. Schroeder,
J. C. Wu, R. B. Perni, G. P. Vlasuk, J. L. Ellis, Crystallographic structure of a small molecule T. Pendl, A. Harger, M. Niso-Santano, N. Zamzami, M. Scoazec, S. Durand, D. P. Enot,
SIRT1 activator-enzyme complex. Nat. Commun. 6, 7645 (2015). Á. F. Fernández, I. Martins, O. Kepp, L. Senovilla, C. Bauvy, E. Morselli, E. Vacchelli,
212. M. Gertz, G. T. T. Nguyen, F. Fischer, B. Suenkel, C. Schlicker, B. Fränzel, J. Tomaschewski, M. Bennetzen, C. Magnes, F. Sinner, T. Pieber, C. López-Otín, M. Chiara Maiuri,
F. Aladini, C. Becker, D. Wolters, C. Steegborn, A molecular mechanism for direct sirtuin P. Codogno, J. S. Andersen, J. A. Hill, F. Madeo, G. Kroemer, Regulation of autophagy
activation by resveratrol. PLOS One 7, e49761 (2012). by cytosolic acetyl-coenzyme A. Mol. Cell 53, 710–725 (2014).
213. B. P. Hubbard, A. P. Gomes, H. Dai, J. Li, A. W. Case, T. Considine, T. V. Riera, J. E. Lee, S. Y. E, 224. T. Eisenberg, S. Schroeder, A. Andryushkova, T. Pendl, V. Kuttner, A. Bhukel, G. Marino,
D. W. Lamming, B. L. Pentelute, E. R. Schuman, L. A. Stevens, A. J. Y. Ling, S. M. Armour, F. Pietrocola, A. Harger, A. Zimmermann, T. Moustafa, A. Sprenger, E. Jany, S. Büttner,
S. Michan, H. Zhao, Y. Jiang, S. M. Sweitzer, C. A. Blum, J. S. Disch, P. Yee Ng, K. T. Howitz, D. Carmona-Gutierrez, C. Ruckenstuhl, J. Ring, W. Reichelt, K. Schimmel, T. Leeb,
A. P. Rolo, Y. Hamuro, J. Moss, R. B. Perni, J. L. Ellis, G. P. Vlasuk, D. A. Sinclair, Evidence for C. Moser, S. Schatz, L.-P. Kamolz, C. Magnes, F. Sinner, S. Sedej, K.-U. Fröhlich,
a common mechanism of SIRT1 regulation by allosteric activators. Science 339, G. Juhasz, T. R. Pieber, J. Dengjel, S. J. Sigrist, G. Kroemer, F. Madeo, Nucleocytosolic de-
1216–1219 (2013). pletion of the energy metabolite acetyl-coenzyme a stimulates autophagy and prolongs
214. M. Lakshminarasimhan, D. Rauh, M. Schutkowski, C. Steegborn, Sirt1 activation by resver- lifespan. Cell Metab. 19, 431–444 (2014).
atrol is substrate sequence-selective. Aging 5, 151–154 (2013). 225. S. Gelino, M. Hansen, Autophagy—An emerging anti-aging mechanism. J. Clin. Exp. Pathol.
215. R. K. Minor, J. A. Baur, A. P. Gomes, T. M. Ward, A. Csiszar, E. M. Mercken, K. Abdelmohsen, (Suppl. 4), pii: 006 (2012).
Y. K. Shin, C. Canto, M. Scheibye-Knudsen, M. Krawczyk, P. M. Irusta, A. Martín-Montalvo, 226. F. Madeo, N. Tavernarakis, G. Kroemer, Can autophagy promote longevity? Nat. Cell Biol.
B. P. Hubbard, Y. Zhang, E. Lehrmann, A. A. White, N. L. Price, W. R. Swindell, K. J. Pearson, 12, 842–846 (2010).
K. G. Becker, V. A. Bohr, M. Gorospe, J. M. Egan, M. I. Talan, J. Auwerx, C. H. Westphal, 227. J. Füllgrabe, M. A. Lynch-Day, N. Heldring, W. Li, R. B. Struijk, Q. Ma, O. Hermanson,
J. L. Ellis, Z. Ungvari, G. P. Vlasuk, P. J. Elliott, D. A. Sinclair, R. de Cabo, SRT1720 improves M. G. Rosenfeld, D. J. Klionsky, B. Joseph, The histone H4 lysine 16 acetyltransferase
survival and healthspan of obese mice. Sci. Rep. 1, 70 (2011). hMOF regulates the outcome of autophagy. Nature 500, 468–471 (2013).
216. V. N. Anisimov, Metformin: Do we finally have an anti-aging drug? Cell Cycle 12, 228. F. Ng, B. L. Tang, Sirtuins’ modulation of autophagy. J. Cell. Physiol. 228, 2262–2270 (2013).
3483–3489 (2013). 229. E. Morselli, M. C. Maiuri, M. Markaki, E. Megalou, A. Pasparaki, K. Palikaras, A. Criollo,
217. F. Cabreiro, C. Au, K.-Y. Leung, N. Vergara-Irigaray, H. M. Cochemé, T. Noori, D. Weinkove, L. Galluzzi, S. A. Malik, I. Vitale, M. Michaud, F. Madeo, N. Tavernarakis, G. Kroemer, Caloric
E. Schuster, N. D. E. Greene, D. Gems, Metformin retards aging in C. elegans by altering restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction
microbial folate and methionine metabolism. Cell 153, 228–239 (2013). of autophagy. Cell Death Dis. 1, e10 (2010).
218. B. Onken, M. Driscoll, Metformin induces a dietary restriction–like state and the oxidative 230. C. M. Roos, B. Zhang, A. K. Palmer, M. B. Ogrodnik, T. Pirtskhalava, N. M. Thalji, M. Hagler,
stress response to extend C. elegans healthspan via AMPK, LKB1, and SKN-1. PLOS One 5, D. Jurk, L. A. Smith, G. Casaclang-Verzosa, Y. Zhu, M. J. Schafer, T. Tchkonia, J. L. Kirkland,
e8758 (2010). J. D. Miller, Chronic senolytic treatment alleviates established vasomotor dysfunction in
219. A. Martin-Montalvo, E. M. Mercken, S. J. Mitchell, H. H. Palacios, P. L. Mote, aged or atherosclerotic mice. Aging Cell 10.1111/acel.12458 (2016).
Science Advances (ISSN 2375-2548) is published by the American Association for the Advancement of Science. 1200 New York Avenue
NW, Washington, DC 20005. The title Science Advances is a registered trademark of AAAS.
Copyright © 2016, The Authors