Vous êtes sur la page 1sur 17

REVIEWS

Adult stem cells

Cellular and epigenetic drivers


of stem cell ageing
Maria Ermolaeva1*, Francesco Neri   1*, Alessandro Ori1* and K. Lenhard Rudolph1,2*
Abstract | Adult tissue stem cells have a pivotal role in tissue maintenance and regeneration
throughout the lifespan of multicellular organisms. Loss of tissue homeostasis during post-​
reproductive lifespan is caused, at least in part, by a decline in stem cell function and is associated
with an increased incidence of diseases. Hallmarks of ageing include the accumulation of
molecular damage, failure of quality control systems, metabolic changes and alterations in
epigenome stability. In this Review , we discuss recent evidence in support of a novel concept
whereby cell-​intrinsic damage that accumulates during ageing and cell-​extrinsic changes in
ageing stem cell niches and the blood result in modifications of the stem cell epigenome. These
cumulative epigenetic alterations in stem cells might be the cause of the deregulation of
developmental pathways seen during ageing. In turn, they could confer a selective advantage to
mutant and epigenetically drifted stem cells with altered self-​renewal and functions, which
contribute to the development of ageing-​associated organ dysfunction and disease.

Quiescence The ‘shadowed’ regulation of developmental path- Therefore, they have an intrinsically increased risk of
A reversible state that can ways has been proposed as a new theory of ageing1–3. acquiring DNA mutations and epigenetic alterations,
describe non-​dividing stem According to this theory, developmental pathways impairing the fidelity of gene expression.
cells. Cells are in the G0 phase cooperate to ensure that embryonic and postnatal Stem cells have special mechanisms to minimize
of the cell cycle, characterized
by very low metabolic,
development leads to optimal fitness of the organism at the risk of damage accumulation. These include the
transcriptional and proliferative reproductive age. During post-​reproductive age, there suppression of metabolic activity, to reduce the intra­
activities. is little evolutionary selection for the maintenance of cellular production of toxic metabolites, and the
organisms, as the main function of the individual in maintenance of a state of quiescence to reduce
terms of evolution is fulfilled. According to this theory, replication-​associated DNA damage4. Minimizing the
the same pathways that control organism development risk of DNA damage is important, as DNA repair sys-
to the point of optimal fitness become deregulated at tems generally are not more active in stem cells than in
post-​reproductive age and promote the ageing process. other cells, but by contrast, DNA repair can be even more
Given that adult stem cells (also known as tissue stem error prone in the G0 and G1 phases of the cell cycle in
cells) are the remnants of embryonic development which many stem cells reside5,6. Stem cell quiescence and
in which many of the key developmental pathways the associated reduction in transcription and translation
remain actively in use to ensure postnatal organ homeo­ contribute to the maintenance of a healthy proteome
stasis and regeneration, these cell populations may be (proteostasis), as the folding and refolding and the
particularly prone to losing accuracy in regulating removal of misfolded and damaged proteins are highly
developmental pathways. energy-​consuming processes that limit the energy avail-
1
Leibniz Institute on Aging – Adult stem cells are rare cells that are located in able to other quality control systems7,8. Stem cells employ
Fritz Lipmann Institute (FLI),
Jena, Germany.
specialized niches that contribute to the proper control various damage removal systems, including autophagy
of stem cell self-​renewal and differentiation. Adult stem (which can remove damaged proteins and organelles),
2
Medical Faculty Jena,
University Hospital Jena cells retain the capacity to differentiate into the cell types but it is unclear whether these systems are any more
(UKJ), Jena, Germany. of the organ in which they reside, and they contribute, efficient in stem cells than in somatic cells or whether
*e-​mail: Maria.Ermolaeva@ throughout life, to the regeneration and homeostasis stem cells rely on reducing damage load by maintaining
leibniz-​fli.de; of almost all tissues. During ageing, the functionality a quiescent state, as in the case of DNA damage9. Despite
Francesco.Neri@leibniz-​fli.de; of adult stem cells declines, and various types of cell-​ the protective measures in place, stem cells accumulate
Alessandro.Ori@leibniz-​fli.de;
intrinsic damage and alterations in the niche and the molecular damage and lose functionality during ageing,
Lenhard.Rudolph@
leibniz-​fli.de circulating blood have been demonstrated to contribute which involves cell-​intrinsic processes as well as ageing-​
https://doi.org/10.1038/ to this age-​related decline. Adult stem cells are the long- associated alterations in the stem cell microenvironment
s41580-018-0020-3 est living proliferative cells in multicellular organisms. (niche) and in the blood circulation10,11.

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Progenitor cells In this Review, we first describe the types of intra- a multitude of cellular malfunctions, including genetic
Cells that are constantly cellular damage that accumulate in stem cells during and epigenetic insults (Fig. 1).
generated from stem cells with ageing, as well as extrinsic factors, such as systemic
reduced self-​renewal and high factors, metabolism and changes in the stem cell niche, Metabolic changes in ageing stem cells
proliferative activity; needed
for production of cell types for
that contribute to the functional decline of stem cells Depending on the tissue and the niche they reside in and
tissue regeneration and during ageing. We then discuss mechanisms linking their activation status (for example, quiescence versus
homeostasis. damage accumulation to epigenome stability in ageing proliferation and differentiation), the metabolic state
stem cells and the deregulation of developmental path- of stem cells can be biased towards glycolysis, such as
ways. We propose that this axis represents a major driver in quiescent haematopoietic stem cells (HSCs)12,13, or
of epigenetic drift and the selection of clonal mutations towards oxidative phosphorylation (OXPHOS), as in
in stem and progenitor cells, resulting in ageing-​associated differentiating HSCs14 and intestinal stem cells (ISCs)15
decline in the maintenance of tissue and organ function (Box 1). These specific metabolic states are important
and the onset of diseases, including cancer. for preventing the accumulation of molecular damage
and for the maintenance of stem-​cell-specific epigenetic
Molecular damage and quality control landscapes.
Many types of damage that accumulate in stem cells The regulation of stem cell metabolism is disrupted
during ageing are driven by altered metabolism and a during ageing, causing cellular malfunctions. Even in
decline in quality control pathways, which can result in cells that rely mainly on glycolysis, mitochondria produce

Ageing
↓ Autophagy ↓ Ubiquitin-proteasome system
↓ UPRmt ↓ Asymmetric division
1 Accumulation of protein aggregates
and dysfunctional organelles,
including mitochondria

Toxic protein Mitochondrial


aggregates damage

Increase in mitochondrial damage


and dysfunction disrupts the balance
between glycolysis and OXPHOS, Increased levels
causing changes in the production of ROS and stress
of epigenetic cofactors and regulators signalling disrupt
stem cell function
Glycolysis OXPHOS ROS
2 3
p38 Nuclear DNA
Acetyl-CoA α-KG and SAM FOXO damage

NAD+

Histone DNA and histone Stress Cell Cell


acetylation methylation signalling death senescence

Epigenetic Aberrant proliferation Depletion of


alterations and differentiation stem cell pool

Fig. 1 | Molecular damage in ageing stem cells. Cell-​intrinsic changes that occur in ageing stem cells are highly
interconnected. A decline in protein quality control pathways, which include mechanisms that would normally ensure the
clearance of damaged proteins (autophagy and proteasome-​mediated degradation) and damaged organelles (mitophagy
and mitochondrial unfolded protein response (UPRmt)), as well as their asymmetric distribution during self-​renewing
divisions, leads to the accumulation of toxic protein aggregates and dysfunctional mitochondria (step 1). Damaged
mitochondria produce an excess of reactive oxygen species (ROS), which induce further mitochondrial damage (including
mitochondrial DNA damage) and lead to a metabolic imbalance between glycolysis and oxidative phosphorylation
(OXPHOS) (step 2). The glycolysis–OXPHOS balance is crucial for the homeostasis of stem cells, and its disruption leads to
inadequate levels of epigenetic cofactors and regulators (step 2). ROS accumulation induces nuclear DNA damage,
which is exacerbated by DNA replication errors and faulty DNA repair in aged stem cells, promoting cell senescence and
apoptotic cell death. Although moderate production of ROS and other stresses is required for the control of proliferation
and differentiation of stem cells in normal physiology (at young age), high levels of ROS activate stress mediators (p38 and
forkhead box protein (FOXO)), leading to aberrant stem cell function (step 3). Altogether, these changes result in defective
self-​renewal and the depletion of stem cell reserves. α-​KG, α-​ketoglutarate; SAM, S-​adenosylmethionine.

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | Stem cell metabolism — links to niche and epigenetic state


Depending on the tissue of origin, the niches populated by adult stem differentiation and repopulation capacity14. similar to activated HsCs,
cells vary in their characteristics as does the average number of cell which utilize OXPHOs metabolism for self-​renewal and differentiation,
divisions: intestinal stem cells (isCs) are highly proliferative and exposed isCs rely on OXPHOs supported by carbon substrates provided by niche
to the gut lumen, including commensal microorganisms and food Paneth cells, which perform glycolysis15. Controversial data exist for
metabolites86, whereas quiescent haematopoietic stem cells (HsCs) dormant versus activated and differentiating MusCs: on the one hand,
divide less frequently and reside in a protected and hypoxic bone the prevalence of OXPHOs was observed in activated (versus dormant)
marrow niche13. Muscle stem cells (MusCs, also known as satellite cells) MusCs freshly isolated from the injured muscle235; on the other hand,
are scattered on the surface of myofibres and start dividing in fibre culture experiments associated MusC differentiation with the
response to muscle injury230. Diverse niches and specific proliferation metabolic signature of enhanced glycolysis25. Potential biases related to
requirements probably determine the distinct metabolic signatures of cell culture conditions need to be taken into account in this case. Overall,
adult stem cells. Pluripotent stem cells, mesenchymal stem cells and reactive oxygen species produced via OXPHOs appear to facilitate cell
quiescent HsCs are biased towards glycolysis13,231,232, which is achieved proliferation, while glycolysis renders cells sensitive to systemic nutrient
by inhibiting entry of pyruvate into the tricarboxylic acid (tCa) cycle, cues. the use of glycolysis over OXPHOs also affects the epigenome
a step that is essential for oxidative phosphorylation (OXPHOs)232,233. of stem cells: having pyruvate excluded from mitochondria, the tCa
in quiescent HsCs, the OXPHOs-​to-glycolysis switch is mediated by the cycle consumes glutamate as a carbon source, reducing levels of
hypoxia-​inducible factor 1– pyruvate dehydrogenase kinase pathway α-ketoglutarate (α-​KG), the cofactor of Jumonji C histone demethylases
(HIF1–PKD2/PKD4 pathway), which promotes pyruvate retention in and of methylcytosine dioxygenase tet enzymes (DNa demethylases).
response to low oxygen levels of the niche13,14. HsCs also inhibit Glycolytic bias increases cellular availability of acetyl-​Coa, a cofactor
OXPHOs by removal of active mitochondria through autophagy9. High of histone acetyltransferases236, and promotes biosynthesis of
glycolysis and low OXPHOs are critical for quiescence and self-​renewal S-​adenosylmethionine required for histone and DNa methylation. reduced
of HsCs: mutations that impair HiF1 signalling lead to exhaustion of the OXPHOs (as in quiescent HsCs) has a negative impact on the activity of
stem cell pool by inducing untimely differentiation of HsCs14,233; the NaD-​dependent histone deacetylase sirtuin family by affecting the
mitochondrial changes that promote OXPHOs elicit a comparable NaD+:NaDH ratio237, making chromatin less compact and increasing
outcome234. the ability to switch from glycolysis to OXPHOs is exposure of DNa to modifications and damage. the metabolism of stem
equally critical for the functionality of HsCs; its loss impedes their cells is thus tightly linked to their epigenetic state.

substantial levels of reactive oxygen species (ROS), which cells, had increased levels of α-​ketoglutarate (α-​KG)
damage the mitochondrial genome and cause imbal- and reduced levels of S-​adenosylmethionine (SAM).
ance between glycolysis and OXPHOS16. The elevated Fluctuations of these cofactors are probably responsible
production of ROS by damaged mitochondria initiates for ageing-​associated epigenetic changes, reduced self-​
intracellular processes that lead to nuclear DNA damage17 renewal and possibly myeloid bias in HSCs27. Age-​linked
(Fig. 1). High ROS levels also induce aberrant stem cell epigenetic shifts in stem cells thus seem to be linked to
differentiation by activating stress signalling pathways metabolic alterations that occur during ageing.
such as p38 and forkhead box protein (FOXO)16,18–20. In
the ageing intestine of Drosophila melanogaster, high Ageing-​associated protein damage
stress levels and deregulation of stress responses induce At young age, proteostasis is supported by protein quality
hyperproliferation of ISCs, leading to loss of homeostasis control pathways, such as autophagy, the ubiquitin pro-
HIF1–PKD2/PKD4 pathway
A signalling pathway that and life-​limiting intestinal dysplasia21–23. Accumulation teasome system (UPS) and the unfolded protein response
adjusts cellular metabolism to of mitochondrial DNA mutations leads to the altered in the mitochondria (UPRmt) (reviewed in ref.28) and the
reduced levels of oxygen production of mitochondrial epigenetic cofactors, which endoplasmic reticulum (UPRER). The efficiency of these
(hypoxia), by suppressing the cause epigenome changes in ageing stem cells (Fig. 1). protein quality control systems, which ensure the cor-
entry of glycolytic metabolites
into mitochondria.
For example, levels of mitochondrial NAD+, an essen- rect folding and targeting of proteins and the removal of
tial cofactor of the NAD-​dependent protein deacetylase misfolded or mislocalized proteins, declines with ageing,
p38 sirtuin 1 (SIRT1), are reduced during ageing24. Active compromising cell function29 (Fig. 1). The mechanisms
A MAPK that regulates SIRT1 promotes self-​renewal and inhibits differentiation of this decline are incompletely understood, but reduced
expression of innate immune
of neuronal progenitors and muscle stem cells (MuSCs; activity of regulatory transcription factors such as
molecules and stress effectors
in response to cell-​extrinsic also known as satellite cells)25. Restoring NAD+ levels was FOXO30 and epigenetic silencing of effector genes such
and cell-​intrinsic stimuli. found to improve the functionality of aged MuSCs in a as UPRmt components31 seem to contribute.
SIRT1-dependent manner24.
FOXO A recent study linked defective self-​renewal and UPRmt and autophagy in stem cell ageing. Ageing HSCs
A subgroup of the forkhead
myeloid bias of ageing HSCs to epigenetic changes driven exhibit increased levels of mitochondrial protein folding
family of transcription factors
regulated by the insulin–PI3K– by increased mitochondrial activity9. Elevated OXPHOS stress, which is counteracted by the SIRT7-dependent
AKT pathway and involved in in old HSCs was attributed to the reduced removal of branch of UPRmt (ref.32). SIRT7-deficient animals showed
induction of stress response active mitochondria owing to the autophagy machinery early ageing of HSCs accompanied by myeloid bias
and survival genes.
becoming impaired9,26. Only a fraction (approximately and reduced reconstitution capacity, whereas SIRT7
Myeloid bias 30%) of aged HSCs had adequate levels of autophagy overexpression ameliorated ageing-​associated defects
Myeloid bias designates sufficient to suppress OXPHOS. Importantly, knockout in HSCs. Autophagy was shown to decline with ageing in
preferential differentiation of of the ATG12 autophagy gene in young adult HSCs leads HSCs and MuSCs9,26, leading to impaired self-​renewal
haematopoietic stem cells into to their accelerated ageing, characterized by elevated and senescence. Autophagy defects in both cell types
myeloid cells, such as
monocytes, macrophages,
OXPHOS, reduced self-​renewal capacity and skewed converge at mitochondrial deregulation: in MuSCs,
neutrophils, platelets and differentiation9. Activated HSCs, which contain active impaired autophagy was linked to alterations of mito-
dendritic cells. mitochondria and resemble aged, autophagy-​impaired chondrial membrane potential and enhanced ROS

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

production by mitochondria, whereas in HSCs, it led stem cells44 and neural stem cells45, it was shown that dur-
to elevated OXPHOS and imbalance of mitochondrial ing such asymmetrical divisions, damaged proteins and
epigenetic cofactors (SAM and α-​KG), reshaping the mitochondria are segregated into differentiating cells while
epigenetic landscape of HSCs towards myeloid lineage the stem cell daughter remains damage free43–45. The asym-
(Box 1; Fig. 1). A recent study linked impaired autophagy metrical distribution of damaged cellular components is
in MuSCs to ageing-​associated reprogramming of rhyth- an active process involving sirtuins46, chaperones46 and
mic gene expression during circadian oscillations33. It is the lateral diffusion barrier in the endoplasmic reticu-
possible that epigenetic alterations induced by ageing-​ lum45, ensuring segregation before cell division. During
associated mitochondrial dysfunction contribute to the ageing, the mechanisms and cellular structures that sup-
epigenetic rewiring of circadian regulated genes. port this asymmetrical partitioning decline45,46, resulting
in the accumulation of protein damage43,45 and damaged
Protein synthesis regulation in stem cells. Low rates of mitochondria44 in stem cells. In some cells, the asymmet-
protein translation support cellular proteostasis by redu­ rical distribution of damaged cellular components works
cing pressure on protein folding and sorting machineries. differently: germline stem cells (in D. melanogaster)
At the organism level, reduced protein synthesis (follow- and yeast mother cells retain damaged proteins, ensur-
ing inactivation of protein translation genes) facilitated ing the fitness of progenitors destined to each become a
reduced energy expenditure, enhanced proteostasis and new organism43,46. Loss of asymmetrical division during
extended lifespan in yeast and animal models (reviewed in ageing owing to altered p38 MAPK signalling has been
ref.34). Nucleolar expansion, enhanced ribosomal biogen- described in MuSCs; it remains to be investigated whether
esis and elevated protein synthesis are hallmarks of pre- this failure affects the segregation of damaged proteins in
mature ageing in humans35. In D. melanogaster germline ageing MuSCs47.
stem cells, elevated protein synthesis accompanies the
transition from self-​renewal to differentiation36. Unlike Protein damage and epigenetic modulators. Aside
other haematopoietic cells, HSCs need to maintain tightly from epigenetic changes driven by the accumulation
regulated, low rates of protein synthesis, as increases and of protein and organelle damage during ageing, the
decreases in protein synthesis rates impair HSC self-​ functionality of epigenetic regulators could also directly
renewal and function37. The crosstalk between protein depend on cellular proteostasis. For example, the stabil-
synthesis rates and the epigenetic regulation of stem cell ity and turnover of chromatin modifiers such as histone
self-​renewal and differentiation remains to be elucidated. deacetylase 1 (HDAC1) and DNA (cytosine-5)-methyl-
transferase 1 (DNMT1) are regulated by the UPS, which
UPRER, proteasome and chaperones in stem cell ageing. deteriorates during ageing48. Protein aggregation events
UPRER ensures proper folding and sorting of newly are more frequent in aged cells owing to proteostasis
produced proteins in the endoplasmic reticulum, while failure, and the aggregation of Polycomb group protein
cytosolic chaperones and the UPS support refolding or polyhomeotic was linked to a loss of coordinated repres-
timely degradation of damaged proteins. UPRER regulates sion of genes regulating cell fate during D. melanogaster
the transition of D. melanogaster ISCs from a quiescent development49. The assembly of histones around DNA
to a proliferating state in response to intrinsic and extrin- and the incorporation of specific histones into chroma-
Co-​chaperone
A protein that regulates the sic stimuli38. In old flies, overactive UPRER promotes tin during replication are assisted by specific histone
activity of chaperones, such as aberrant proliferation of ISCs and intestinal dysplasia, chaperones (such as ASF1A/B and chromatin assembly
heat shock protein 70 (HSP70) leading to death, while inhibition of intestinal UPRER at factor 1 (CAF1)), which become less abundant during
and HSP90, during protein
old age confers lifespan extension38. The pluripotency of senescence and ageing, leading to altered chromatin
folding.
embryonic stem cells depends upon tightly controlled structure, loss of core histones from chromatin and gene
Polycomb group protein proteostasis, which requires high levels of chaperones expression changes50,51; consistently, deletion of the ASF1
A chromatin remodeller and the protea­some39–41. A comparable protein quality gene caused accelerated ageing in yeast52.
implicated in the timely control landscape has not yet been reported for adult
silencing of developmental
stem cells. It was nonetheless shown that increased Accumulation of DNA damage
genes.
expression of the co-​chaperone endoplasmic reticu- Here, we focus on links between DNA damage and epi­
Nucleotide excision repair lum DNA J domain-​containing protein 4 (ERDJ4; also genetic changes and refer to other reviews for an extended
(NER). A DNA repair pathway known as DNAJB9) enhances stemness and repopulation discussion of ageing-​associated DNA damage in stem cells
responsible for removal and
capacity of HSCs by supporting protein folding in the (see refs4,53,54). Given their capacity for self-​renewal, stem
repair of DNA helix-​distorting
lesions, such as cyclobutane
endoplasmic reticulum42. The activity of the proteasome cells are at high risk of accumulating replication-​associated
pyrimidine dimers and and the expression of chaperones both decline with age, DNA damage; the accumulation of genotoxic lesions in
pyrimidine (6–4) pyrimidone providing additional evidence for the accumulation of stem cells during ageing has indeed been confirmed5,6,55.
photoproducts. protein damage in stem cell ageing29. DNA replication is believed to be the key source of
genotoxic damage during ageing56,57. In the intestine
Non-​homologous end
joining Segregation of damaged cellular components in ageing and skin, stem cells are exposed to additional external
(NHEJ). A DNA repair pathway stem cells. Owing to their high sensitivity to protein genotoxic insults, such as ROS, microbial products
that repairs double-​strand damage, stem cells developed specific, efficient ways of and ultraviolet light. Analysis of HSCs from mice
DNA breaks by directly ligating
handling damaged proteins. During differentiation, stem deficient in telomere maintenance and DNA repair
the broken ends without the
use of a homologous template
cells divide asymmetrically, giving rise to a stem cell and mechanisms ( nucleotide excision repair (NER) and
(a rapid but error-​prone a differentiating cell. In yeast, in D. melanogaster ISCs non-homologous end joining (NHEJ)) revealed increased
process). and neuroblasts43 and in mammalian mammary gland accumulation of genotoxic damage in HSCs at old age

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

accompanied by impaired self-​renewal and loss of biogenesis have been described in telomerase-​deficient
Heterochronic parabiosis
An experimental procedure in regenerative potential39,58. Unlike differentiated cells with mice with persistent DNA damage at telomeres62 (Box 1).
which two animals of different persistent DNA lesions, genome-​damaged quiescent stem Gene expression marks of elevated DNA damage coincide
age, for example, one young cells avoid apoptosis and cellular senescence by activating with global reprogramming of rhythmic gene expression
and one old, are surgically
rapid but error-​prone NHEJ DNA repair and expression in aged epidermal stem cells, possibly owing to changes
connected so that their blood
circulations are shared.
of pro-​survival factors such as BCL-2 (refs5,6,58,59) and by in metabolic factors and epigenetic cues33. Moreover, per-
attenuating DNA damage responses55 (for review, see sistent DNA damage was linked to reduced expression of
ref.4). These strategies ensure the survival of quiescent core histones and reduced levels of histone chaperones
cells but prob­ably lead to accumulation of mutations in (ASF1A/B and CAF1) in somatic cells63, a phenotype
these cells during ageing, which can cause stem cell dys- comparable to that of aged MuSCs64.
function and promote the death of progenitor cells that
exit quiescence58,59. Chromosomal rearrangements gener- Stem cell-​extrinsic factors
ated by NHEJ errors may result in cancer onset and gene Ageing leads to changes in cell-​extrinsic factors that
mutation-​driven dysfunction of epigenetic regulators affect adult stem cell function. These include changes in
(reviewed in ref.60). systemic, blood-​borne factors in the tissue or organ
Rapid activation of DNA repair in stem cells may have in which they reside, as well as in their microenviron-
a direct impact on their metabolism and epigenetic land- ment (stem cell niche). These external cues can affect
scape: DNA repair processes involve poly(ADP-​ribose) epigenetic reprogramming and gene regulation in ageing
polymerase (PARP), which uses NAD+ as a substrate stem cells (Fig. 2).
to generate ADP-​ribose monomers. When the levels of
DNA damage become high, PARP activity might lead to Systemic factors modify stem cell function
substantial depletion of intracellular NAD+ stores, disrupt- Heterochronic parabiosis experiments provided evidence
ing mitochondrial metabolism and the activity of other that circulating systemic factors, which change during
NAD+-dependent enzymes, such as sirtuins61. Elevated ageing, modify stem cell function in different com-
glucose depletion rates and impaired mitochondrial partments, including muscle and brain65,66. Exposure to

A Ba Haematopoietic stem cells


MPP
Hormones Systemic Diet Host • WNT3A and Vascular
inflammation microbiome WNT5A endothelial cell
• CCL5 Adipocyte
• Osteopontin
• Oestrogens
Cytokine
Systemic Stem cell ageing
factors • Metabolic changes
• Epigenetic alterations HSC MSC
Micro- • Loss of quiescence
environment • Altered developmental
pathways
Osteoblast
Neuron
ECM Macrophage

Crosstalk with Niche structure Local immune Bc Intestinal stem cells • WNT3A
niche cells (ECM) system Proliferating TA cell • cADPR
Stem cell • IL-22
• Butyrate
Bb MuSCs Paneth cell
• FGF1 and FGF2 • Sex hormones
• Fibronectin • Oxytocin CBC stem cell
• GDF11 • ECM stiffness
Repair
PAX7+
Pericryptal cell

Muscle fibre

Fig. 2 | Systemic factors and changes in the local microenvironment affect stem cell function during ageing.
a | A combination of physiological changes in hormone levels, increased inflammation and the interplay between diet
and the host microbiome leads to changes in systemic signals that regulate stem cell function in multiple compartments.
Locally , age-​related changes in cellular and molecular composition of the stem cell niche lead to aberrant stimulation of
stem cells affecting their quiescence, metabolism and differentiation potential. Extrinsic signals are often integrated into
epigenetic changes that can lead to the aberrant reactivation of developmental pathways in stem cells. Ba–Bc | Examples
of extrinsic molecules that regulate stem cell function in different stem cell compartments, the levels or properties of
which are affected by ageing or ageing-​modulating factors, such as diet and the microbiome. cADPR , cyclic ADP-​ribose;
CBC, crypt base columnar ; CCL5, CC-​chemokine ligand 5; ECM, extracellular matrix; FGF, fibroblast growth factor ; GDF11,
growth/differentiation factor 11; HSC, haematopoietic stem cell; IL-22, interleukin-22; MPP, haematopoietic multipotent
progenitor ; MSC, mesenchymal stem cell; PAX7 , paired box protein 7; TA , transit amplifying.

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Gut microbiome
blood from young animals had a rejuvenating effect on epigenetic mechanisms and whether these are reverted
The ensemble of commensal MuSCs and neuronal stem cells, as shown by enhanced by heterochronic parabiosis.
bacterial strains that inhabit cell proliferation, regenerative capacity and increased One of the long recognized but still poorly under-
the digestive tract of an adult neurogenesis. These rejuvenating effects of stood systemic effects of ageing is the increase in
organism.
parabiosis are the result of enhancing developmental inflammatory signalling, which affects virtually all
pathways that become less active in aged stem cells, for organs. Accumulation of tissue damage and senescent
example, Notch signalling65,67 (see below). However, cells (Box 2) and repeated exposure to stress-​inducing
it remains to be investigated whether the modulation events, such as infections, lead to an increase in pro-​
of developmental pathways by exposure to a youthful inflammatory factors in the circulation as well as in the
systemic environment is epigenetically regulated and tissue microenvironment. Infection-​induced cytokines
whether parabiosis reprogrammes epigenetic alterations can impair haematopoiesis by limiting self-​renewal and
that accumulate during ageing. by skewing HSC differentiation towards the myeloid
The results of parabiosis experiments sparked inter- lineage76,77. Inflammatory stimuli can directly influence
est in identifying systemically acting molecules that stem cells by altering epigenetic regulators. In MuSCs,
could potentially improve the function of ageing stem tumour necrosis factor (TNF)-activated p38α kinase
cells and tissues. Although some proteins and hormones induces epigenetic repression of paired box protein PAX7,
were identified (for example, growth/differentiation fac- the key transcription factor responsible for maintain-
tor 11 (GDF11), eotaxin, metallopro­teinase inhibitor 2 ing MuSC identity, reducing stem cell self-​renewal and
(TIMP2) and oxytocin), the direct contribution of each increasing differentiation78. Recent data from epigenetic
factor alone or in combination to the ageing process reprogramming of early stage cancers by inflammatory
remains to be elucidated68–71. Other hormonal changes stimuli suggest that similar mechanisms act in the intes-
are known to influence stem cell function72. In par- tinal epithelium79. The persistent inflammatory state that
ticular, increased oestrogen levels during pregnancy accumulates during ageing could thus cause epigenetic
promote HSC self-​renewal73, and sex hormones drive perturbation, leading to changes in stem cell identity.
the conversion from dividing into quiescent MuSCs Additional work is required to demonstrate whether such
during puberty and regeneration following injury74. mechanisms affect stem cells in other compartments.
As hormones are known to influence the expression of Recent evidence indicates crosstalk between systemic
epigenetic-​modifying genes75, it would be interesting to inflammation and changes in the gut microbiome, driving
investigate whether changes in hormone levels contrib- stem cell ageing. In flies, ageing-​associated imbalances
ute to the decline of stem cell function during ageing via in the composition of the gut microbiome can induce

Box 2 | cellular senescence during ageing


Cellular senescence, which is a permanent state of growth arrest, is a response of cells to various types of stress, including
telomere shortening, DNa replication stress, mitogen stimulation and reactive oxygen species (rOs)4. in recent years,
it has become clear that senescence has a role in ageing and tumour suppression, and during embryonic development.
senescence likely influences stem cell and tissue ageing at multiple levels.
Direct impairment of stem cell function by induction of senescence
there is evidence that cellular senescence contributes to ageing-​associated impairments in the function of muscle stem
cells (also known as satellite cells)214, haematopoietic stem cells216 and intestinal stem cells in old wild-​type mice89 and
telomerase-​deficient mice with dysfunctional telomeres238.
indirect effects of senescence on stem cell function and organ maintenance
senescent cells accumulate in ageing tissues and are a prominent source of pro-​inflammatory signals — also referred to
as the senescence-​associated secretory phenotype (sasP)91,92. studies on mice provided proof of concept that the
removal of senescent cells can improve stem cell function, tissue maintenance, metabolic performance and lifespan239–241.
senolytic treatments were shown to improve stem and progenitor cell function in blood and skeletal muscle240,241.
the studies suggested that the beneficial effects of senolytic therapies involve reductions in rOs and inflammatory
signalling — factors that were previously shown to reduce stem cell function and tissue maintenance in mouse models of
DNa damage accumulation during ageing induced by telomere dysfunction242,243 or DNa repair deficiency58.
effects of SaSP factors on epigenome and cell plasticity
sasP factors can influence stem cell plasticity, which is known to be regulated by the epigenome. interleukin-6 (iL-6)
— one of the prominent pro-​inflammatory cytokines secreted from senescent cells — is upregulated in human ageing
and in mice with shortened telomeres10,242. it was recently shown that iL-6 can promote reprogramming of cells into
pluripotent cells in vivo244, and cytokines derived from tissue-​resident immune cells have important roles in regulating
regenerative processes in various tissues85,90,245. Of note, transient pulses of partial reprogramming were shown to
improve tissue maintenance in progeroid mouse models229. Moreover, there is evidence that transient inflammation
contributes to wound healing245. in contrast to the beneficial effects of a transient induction of inflammatory signals,
chronic inflammation is associated with tissue destruction and the promotion of cancer by inducing cell fate plasticity
and self-renewal capacity in genetically altered progenitor cells246. it is conceivable that the sasP contributes to
misdifferentiation of stem cells in ageing tissues, resulting in defects such as fibrosis in muscle or adipogenesis in
bone marrow. it has been suggested that chronic inflammation (for example, as a consequence of accumulation
of senescent cells in ageing tissues) leads to stem cell exhaustion, tissue-​damagingprocesses and diseases such
as cancer91,238–240.

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

ISC hyperproliferation, leading to accelerated tissue and long-​lasting epigenetic modifications in the resident
organism ageing21. In mice, changes in the gut micro- stem cell population.
biome affect intestinal epithelium permeability and The availability of growth factors and molecules
decrease macrophage function, leading to increased sys- mediating the interaction of stem cells with their niches
temic inflammation and tissue damage11. Importantly, and the biophysical properties of the ECM can influ-
modulating the host microbiome can influence the ence stem cell function100,101. In skeletal muscle, an
lifespan in various species11,80,81. age-​dependent increase in fibrosis alters the cellular
and molecular composition of the stem cell niche. As
Ageing of the stem cell niche a consequence, the mechanical properties of the niche
Stem cells reside in specialized microenvironments microenvironment, such as stiffness, are changed, result-
(known as niches), the property and nature of which vary ing in reduced MuSC self-​renewal and regenerative
depending on tissue. The niche provides specific cues in capacity85. In old muscles, an alteration of the architec-
the form of differentiation and self-​renewal-regulating ture of collagen fibres influences the deposition of ECM
signals, adhesion molecules, spatial organization and by fibroblasts, and consequently, it disrupts lineage
metabolic support to stem cells82–86. As such, the niche specification in MuSCs102. As extracellular cues, includ-
is essential in regulating basic functions of stem cells and ing mechanical forces and stiffness, have been shown
in protecting them from the accumulation of molecular to induce epigenetic alterations in stem cells103, ageing-​
damage and toxins, for example, microorganism-​derived associated alterations in the biophysical properties of the
factors in the intestine15,87. The intimate relationship ECM might contribute to epigenetic changes in adult
between stem cells and their niche can be influenced by stem cells.
multiple factors, including ageing, diet and systemically Ageing-​associated changes in the cellular composition
acting factors, which include metabolites from the host of the niche also occur in other stem cell compartments.
microbiome (Fig. 2). In the small intestine, it was shown that ageing is
Changes in the niche may include cell-​intrinsic accompanied by an increase in the number of secretory
changes in niche cells as well as indirect alterations Paneth cells in the intestinal crypts89. Additionally, injury
in their function, induced by secreted factors, such signals can activate reserve stem cells104, for example, via
as inflammatory cytokines, growth factors, adhesion de-​differentiation of mature enteroendocrine cells in
molecules and/or changes in structural components the intestinal epithelium105. These processes involve
of the extracellular matrix (ECM). An age-​dependent a dynamic reorganization of chromatin architecture
increase in fibroblast growth factor 2 (FGF2) produced of genomic regions controlling the expression of stem
by the muscle fibre has been shown to induce loss of and differentiation factors106. As the combinatorial
quiescence in MuSCs, reducing their regenerative capa­ effect of growth factors secreted by different niche cells
city88. The levels of WNTs, growth factors regulating the determines the quiescence of ISCs by modulating their
homeostatic proliferation and differentiation fate of proliferation and differentiation potential in vitro107
ISCs, decrease during ageing in intestinal crypts and and in vivo108, it would be interesting to investigate
the surrounding mesenchyme, thereby reducing the whether changes in crypt composition in old animals are
regenerative potential of ISCs89. In the intestinal epi- directly related to ageing-​associated epigenetic changes
thelium, the local immune cells can also contribute to in ISCs.
the regulation of stem cell function by secreting inter-
leukins, for example, interleukin-22 (IL-22), that can Influence of diet on stem cell ageing
directly target ISCs90. DNA damage signals in senescent Dietary interventions, such as intermittent fasting
cells with dysfunctional telomeres promote the secretion and dietary restriction, have been shown to improve
of inflammatory cytokines. This senescence-​associated health parameters and promote lifespan in multiple
secretory phenotype 91,92 (SASP; Box  2 ) was shown organisms109. Dietary restriction has been extensively
to contribute to change in bone marrow niches and studied, and its positive effect on regenerative capacity
increases in inflammatory cytokines in blood, inhibit- has been described in multiple stem cell compartments,
Exosomes
Vesicles released by cells ing lymphopoiesis in ageing mice with dysfunctional including MuSCs, HSCs and ISCs110–113. A periodic diet
containing RNAs and proteins, telomeres93. How inflammatory signals might induce designed to mimic the effects of fasting was shown to
which have several functions, cell-​intrinsic defects in niche cells remains to be eluci- restore the expression of MuSC markers, rejuvenate the
including cell-​cell dated. The old bone marrow niche is characterized by haematopoietic system and promote adult neurogenesis
communication.
low levels of osteopontin, a secreted factor that influ- in aged mice114 and promote the reprogramming of
Fibrosis ences the polarity and epigenetic state of HSCs and pancreatic cells towards insulin-​producing β-​cells in
The excessive deposition of thereby their proliferation and differentiation fate94. both mice and humans115.
extracellular matrix proteins Ageing-​associated alterations in ECM components Dietary restriction has been shown to have a positive
caused by aberrant tissue
were shown to impair stem cell function in skeletal effect in preserving the pool of functional stem cells
repair and chronic
inflammation. muscle and in the germ line95–97. Stem cells can con- during ageing in different stem cell compartments in
tribute to the remodelling of their own niche dur- flies and mice110,111,113,116. However, there seems to be a
Reserve stem cells ing ageing. Activated MuSCs secrete exosomes (for trade-​off between the positive effects of dietary restric-
Pools of cells already review, see ref.98) that can modulate the ECM depo- tion on extending stem cell maintenance by reducing
committed to differentiation,
generally quiescent, that upon
sition by niche fibroblasts99. Although experimental stem cell activity and the ability of the organism to
injury can de-​differentiate and evidence is still scarce, the aberrant signals deriving respond to insults and maintain tissue integrity. Indeed,
revert to a stem-​like state. from the ageing niche are likely to be integrated into it was shown that a decrease in intestinal epithelium

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

proliferation (that is, stem cell activity) correlates with Other vitamins, such as vitamin C, can influence stem
extended lifespan in D. melanogaster. Conversely, an cells by altering the activity of epigenetic modifiers124,125
excessive reduction in stem cell proliferation results and niche signalling126. However, a direct link between
in impairment of epithelial barrier function and is age-​related vitamin deficiency and impairment of adult
ultimately detrimental to lifespan22. Similarly, in mice, stem cell function remains to be demonstrated.
long-​term dietary restriction was shown to improve the In summary, changes in the epigenome seem to
maintenance of the haematopoietic capacity of HSCs in integrate multiple environmental signals that influence
middle-​aged mice by favouring a quiescent state, but at stem cell biology127. The mechanisms by which external
the same time, dietary restriction impaired the differen- cues lead to changes in the epigenome, the activity
tiation potential of HSCs, resulting in immune defects of developmental pathways and the functionality of
in the context of prolonged bacterial infection110,117. The stem cells remain to be understood. As niche cells are
outcome of dietary regimes on stem cell function seems among the descendants of stem cells, it is possible that
to depend on the timing and duration of the regimes: niche-​induced changes in stem cells in turn aggravate
subjecting mice to lifelong dietary restriction did not ageing-​associated remodelling of the stem cell niche. It
prevent the ageing-​associated functional decline of could be envisioned that the combination of long-​term
HSCs118, in contrast to when dietary restriction was exposure to aberrant niche signals, incorrect activa-
limited to short periods of time in middle-​aged mice98. tion of developmental pathways and loss of stem cell
Diet can influence stem cell function either directly identity might underlie the loss of regenerative capacity
or indirectly by affecting niche cells, mainly via in ageing.
nutrient-​sensing pathways. Long-​term dietary restric-
tion increases the regenerative capacity of ISCs via Epigenetic drift and clonal expansion
reduction of mTOR complex I (mTORC1) signalling in Throughout the organismal lifespan, adult stem cells
Paneth cells111. ISCs seem to respond to dietary restric- face the challenge of being able to promptly differen-
tion indirectly, but dietary-​restriction-mediated changes tiate into committed progenitor cells in response to the
in paracrine signals from Paneth cells activate mTORC1 in environmental stimuli while maintaining a functional
ISCs, thereby leading to increased protein synthesis undifferentiated stem cell pool. Chromatin architecture
and proliferation119. In skeletal muscle, short-​term diet­ and the epigenome are instrumental in preserving stem
ary restriction increases the number and functionality cell identity and function. As discussed above, the accu-
of MuSCs in both young and old mice113. Interestingly, mulation of molecular damage, changes in metabolism
MuSC transplantation experiments showed that the bene­ and alterations in the local and systemic environment of
fi­cial effects of dietary restriction are not limited to the adult stem cells that occur during ageing can lead to epi-
stem cell donor but can also be observed when the recipi­ genetic changes that could contribute to the appearance
ent animal is subjected to dietary restriction. These data of altered clones, which become dominant, gradually
indicate that dietary restriction induces changes in the replacing normal cells. There is increasing evidence that
stem cell niche that promote the engraftment of MuSCs epigenome alteration and DNA mutations contribute
after transplantation113. to the loss of stem cell function and disease develop-
Diet has profound effects on the whole organism. ment during ageing (Fig. 3). Recent studies have revealed
The way it affects stem cell function might be, at least mutual crosstalk between genetic and epigenetic
in part, induced by systemic factors such as decreased alterations in ageing adult stem cells (Box 3).
level of inflammation109 or changes in the composition
of the host microbiome120. Dietary restriction can influ- Epigenetic landscape of ageing stem cells
ence the epigenome by delaying age-​related changes in The DNA methylome of different organs and tissues
DNA methylation81,121, and nutrient availability alters can be used to precisely predict the biological age of
the expression of chromatin modifiers in neuronal mouse and human organisms121,128–131. In the haemato-
stem cells100. Whether the duration of dietary restric- poietic system, the majority of ageing-​associated DNA
tion (short-​term versus long-​term) and the resulting methylation changes were independent of changes in
epigenetic changes have an effect on HSC maintenance the cellular composition of the tissues132, suggesting
and function (see above) remains to be investigated. that ageing-​associated DNA methylation changes in this
Epigenetic mechanisms are expected to also contribute to compartment originate at the stem or early progenitor
the transgenerational effects of dietary interventions via cell level.
alteration of chromatin structure in the germ line109,122. Whole-​genome DNA methylation analysis in HSCs
How and whether diet regulates the stem cell epigenome from young and old mice showed a slight increase in
in different compartments remain to be explored, as well global DNA methylation levels during ageing133,134. By
as whether specific epigenome alterations underlie the contrast, somatic tissues and other adult stem cells show
positive and negative effects of dietary restriction on a decrease in global DNA methylation levels but hyper-
stem cell ageing and stress resistance. methylation of CpG islands in specific loci81,121,130,135–137.
Dietary vitamins can also influence stem cell An increase in histone repressive marks, such as histone
plasticity. Vitamin A has been shown to promote the H3 lysine 9 trimethylation (H3K9me3) and H3K27me3,
maintenance of the quiescent state in HSCs, and a has been observed in old MuSCs and in HSCs64,138,139,
vitamin A-​deficient diet alters the response of HSCs to suggesting that a progressive intensification of hetero­
inflammatory stress stimuli by impeding the re-​entry chromatin takes place during ageing, which could
into quiescence, leading to stem cell pool exhaustion123. reduce stem cell plasticity. The open (active) chromatin

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Tissue dysfunction

Persistent Deregulation of
epigenome-sensible
mechanisms

Young stem cell Old stem cell


population population
Cancer
Epigenetic modifiers
mutated in HSCs
• DNMT3A Mutation of epigenetic Expansion of a
• TET2 regulator or epimutation clonal stem cell
• ASXL1

Fig. 3 | epigenetic drift and clonal expansion altering adult stem cells during ageing. After reproductive age, the
epigenome of the adult stem cells undergoes an epigenetic drift, possibly driven by molecular damage, changes in
the stem cell niche or aberrant activation of developmental programmes. DNA methylation and histone modification
alterations that accumulate during ageing can be different and have different effects depending on the stem cell
compartment. Recent evidence suggests that epigenetic drift increases self-​renewal and impairs differentiation of adult stem
cells. In some stem cell compartments, persistent epigenetic drift leads to deregulation of epigenome-​sensible gene pathways
that promote stem cell dysfunctions135. In other compartments, mutations in the epigenetic modifiers (for example, DNA
(cytosine-5)-methyltransferase 3A (DNMT3A), methylcytosine dioxygenase TET2 and putative Polycomb group protein ASXL1)
have been frequently found in aged haematopoietic stem cells (HSCs) and can lead to the selection of mutant stem cells (blue)
that gain clonal dominance, leading to impairments in adult stem cell differentiation, tissue dysfunction and cancer153,158.

mark H3K4me3 is enriched in old HSCs, suggesting an age-​dependent change in the CpG methylome, a rein-
epigenetic reinforcement in the transcriptional activa- forcement of open chromatin regions and progressive
tion of stem-​related genes139. By contrast, MuSCs display heterochromatinization — can be profiled, but more
reduced histone acetylation (a mark for open chromatin) studies are required to understand the ageing-​associated
in old mice138. However, in contrast to young cells, old epigenetic landscape. Transcriptomic analyses have
MuSCs show a shift towards a euchromatin state follow- shown that these alterations do not strongly change
ing activation, with increased histone acetylation and the transcriptional programme and identity of adult
decreased H3K27me3 (ref.138). Age-​dependent changes stem cells but can probably affect the differentiation
in DNA methylation and histone modifications have programme and functionality of their terminally com-
previously been reviewed140. The mechanisms of these mitted progeny, causing tissue and organ dysfunction. It
alterations and the functional consequences on ageing has recently been reported that subpopulations of stem
stem cells remain largely unknown. cells in the haematopoietic system undergo ageing-​
Specific DNA methylation and histone modifica- associated changes in the proliferative rate153. Many
tion patterns are associated with the proliferative and epigenetic studies have been performed in bulk stem
differentiation status of ISCs141–143. DNA methylation is cell populations, which could mask cell-​to-cell hetero-
necessary for the maintenance of asymmetrical division geneity and alterations acquired at the single-​cell level
of ISCs, probably by activating specific enhancers144,145. during ageing. By employing single-​cell RNA sequenc-
Impaired absorptive function in the intestine of elderly ing, a recent study showed that whole-​transcriptome
individuals has been associated with a decrease in the cell-​to-cell variability, transcriptional noise and signs of
number of differentiated cells relative to the number of fate drift increase in human pancreas during ageing154.
proliferating, undifferentiated cells146,147. These obser- Recently, single-​cell chromatin modification profiling
vations suggest that ageing-​induced alteration in DNA by mass cytometry showed that human HSC ageing is
methylation alters ISC and progenitor cell differentiation associated with increased heterogeneity between indi-
and thus tissue composition, causing tissue dysfunction. viduals and elevated cell-​to-cell variability in chromatin
A study with a small cohort of patients revealed ageing-​ modifications155. Single-​cell analyses could therefore be
associated gain of DNA methylation in small intesti- crucial to fully understand stem cell epigenetic land-
nal crypts148. Recent work has demonstrated that diet, scapes during ageing. Another important question that
inflammation and the gut microbiome contribute to the remains to be addressed is how the epigenetic integra-
changes in the epi­genetic profile of the intestine, including tion of molecular damage and alteration in the stem
DNA methylation, that could alter the biological function cell environment during ageing enable the selection of
of ISCs149–152. mutant clones and/or population drifts in stem cells dur-
In summary, epigenome changes occurring during ing ageing. Impairment in cell differentiation and slight
ageing seem to be cell dependent and tissue depend- changes in the self-​renewal of adult stem cells could con-
ent and not as drastic and well coordinated as during fer a minimal selective advantage to the aberrant stem
embryonic development, at least in mouse. However, cells and to the appearance of dominant clones in the
an epigenetic drift — which involves a specific somatic stem cell compartment (discussed below).

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 3 | genetics and epigenetics crosstalk in ageing stem cells and exponentially increased during age up to 30–50% in
70-year-​old people162. The presence of these mutations is
genome instability causing epigenome instability associated with a significant increase in the risk of hae-
• Genetic alterations can occur in epigenetic regulators or in non-​coding rNas that in matological cancers, ageing-​associated cardiovascular
turn perturb the epigenome of adult stem cells. this mechanism seems to be involved in diseases and all-​cause mortality. These associations may
the development of many different types of cancer for which stem cells represent the even currently be underestimated owing to technical
cell of origin247. For example, the mixed-​lineage leukaemia 1 gene (MLL1; also known as
limitations. For example, clones bearing mutations
KMT2A), which encodes a histone 3 lysine 4 methyltransferase, and DNa
outside the exome cannot be identified by using exon
methyltransferase genes are frequently translocated and mutated during leukaemia
stem cell development248,249. recent noteworthy studies157,161,165 reported that sequencing. A whole-​genome sequencing approach
epigenetic regulators are also frequently mutated in haematopoietic stem cells of could reveal functional DNA mutations in intergenic or
healthy elderly individuals. intronic regions. Moreover, dominant stem cell clones
• Mutations in genes that regulate metabolism or in mitochondrial DNa lead to changes could arise from the beneficial acquisition of epimutations
in cellular metabolism that alter the availability of essential cofactors of chromatin due to ageing-​related epigenetic drift or sporadic but spe-
modifiers (such as S-​adenosylmethionine and α-​ketoglutarate)250,251. the resulting cific epigenetic events. Most of the identified mutations
impairment in the enzymatic activity of epigenome modifiers may alter the epigenome were dispersed in the genome, but the protein-​coding
during stem cell divisions, leading to aberrant self-​renewal, acquisition of incorrect cell regions of three genes (DNMT3A, TET2 and ASXL1)
identity during differentiation and a functionally altered stem cell pool. were disruptedwith very high recurrence, suggesting
epigenome instability causing genome instability that these genes are functional drivers of such clonal
• several studies report that many genes involved in DNa repair undergo epigenetic haematopoiesis157,161. These three genes all encode master
silencing in different types of cancer252. Promoter hypermethylation of the mismatch epigenetic regulators, and DNMT3A and methylcytosine
repair gene MLH1 leads to microsatellite instability that is responsible for dioxygenase TET2 in particular are the main contrib-
approximately 15–20% of all colorectal cancers253,254. utors to de novo establishment of DNA methylation
• Human mutation rate is associated with DNa replication timing that in turn depends on and removal of DNA methylation, respectively, in adult
the chromatin status of the DNa, suggesting that specific epigenetic modifications can tissues. These findings indicate that alterations in the
promote the frequency of replication-​dependent mutations255,256. epigenome are crucial for the development of clonal
• Most of the mutations accumulated during ageing are C>t transitions, which are dominance in stem cell compartments during ageing.
characteristic of spontaneous deamination of methylated cytosine residues into The most prevalent gene mutations in clonal hae-
thymine at CpG sites257. this mutation spectrum is specifically enriched in highly matopoiesis — DNMT3A and TET2 — are also often
proliferative stem cell compartments (such as the colon and small intestine) and mutated in leukaemia166–169, supporting the hypothesis
suggests an additional function of DNa methylation during ageing257.
that the clonal haematopoiesis originating from recur-
• repetitive elements in our genome are epigenetically repressed. senescent cells rently mutated genes represents a ground stage for the
and aged tissues exhibit an activation of retrotransposons, leading to genome
development of leukaemogenesis. However, these two
instability183,184.
enzymes are also essential regulators of normal HSC
self-​renewal and differentiation170–172, and according to
clinical studies, clonal haematopoiesis is also associated
• Mutation of epigenetic regulators with non-​cancerous diseases157,161. Interestingly, recent
• Mutation of metabolic regulators studies reported that Tet2-deficient haematopoiesis
accelerates atherosclerosis development and cardiovas-
Genome Epigenome cular diseases in mice by enhancing IL-1b inflammatory
instability instability signalling in macrophages173,174. The association of clonal
• Epigenetic silencing of DNA repair genes
• DNA replication-dependent mutations haematopoiesis with cardiovascular diseases in aged
• Deamination of methylated cytosine humans suggests that similar mechanisms are relevant
• Reactivation of retrotransposons
to cardiovascular disease development in humans.
Clonal haematopoiesis may by itself contribute to an
increase in inflammatory signalling in ageing, which
in turn disturbs epigenome regulation, gene regulation
Clonal dominance of aged mutant stem cells and stem cell function in a variety of tissues, including
Recent observations support the hypothesis that clonal non-​mutant stem cells and non-​stem cell compartments.
dominance is a common feature of aged stem cell The latter may also be linked to increases in cell-​to-cell
compartments 156–161. Ageing-​d ependent alterations variation in stochastic alteration in gene expression in
in the genome and epigenome of a single stem cell ageing cardiomyocytes175.
may provide this cell with proliferative and metabolic Ageing-​dependent clonal dominance occurs in adult
advantages, resulting in a strong benefit in cell-​to-cell stem cell compartments other than the blood. A recent
competition and compartment colonization (Fig. 3). This study performed on sun-​exposed human eyelid epider-
mechanism highly depends on the structure and com- mis revealed a high level of somatic mutations (similar
partmentalization of the stem cell niche and, in some to cancer) and a strong positive selection for cancer gene
cases, is believed to generate a precancerous stage162–164. mutations in otherwise physiologically normal cells.
Deep exome-​sequencing analysis of a large human A selective clonal expansion has been found in 18–32% of
cohort showed recurrent somatic mutations in healthy normal skin cells preserving the physiological functions
Epimutations
Alterations of a specific
elderly individuals with clonal haematopoiesis157,161,165. of epidermis176.
chromatin modification in Somatic mutation frequency in blood was extremely rare Even in germ cells, which are meant to have highly
a specific genomic locus. in young individuals (<1% in persons <50 years of age) efficient protection against mutations, mutant stem cell

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

clones emerge in an age-​dependent manner. Mutant activation to generate cryptic transcripts and aberrant
spermatogonial stem cells are positively selected and proteins185. Cryptic promoter regulation was recently
progressively expand clonally in ageing testes. Although found to be a crucial mechanism for driving termi-
this clonal expansion is very common, it rarely gives rise nal differentiation of germ cells in D. melanogaster186.
to testicular tumours. However, mutant spermatogonial Intragenic DNA methylation can also regulate alterna-
stem cells were found to be the cause of a set of genetic tive splicing187–189, which plays an important part during
diseases affecting children of older fathers, also referred cellular differentiation190.
to as paternal age effect mutations158. The epigenome is the major regulator of developmental
In the gut epithelium, all the cells that compose a sin- genes and pathways191. Aberrant chromatin modifications
gle intestinal crypt originate from a single dominant ISC can lead to altered expression of specific differentiation-​
that outcompetes other ISCs to colonize the whole stem associated genes. During development, adult stem cells
cell niche over time177. Although these studies reported are generated and maintained by the activation of spe-
that ISCs follow a stochastic pattern of behaviour that cific epigenetic programmes. Genes responsible for adult
recapitulates a neutral drift dynamic, we cannot exclude stem cell self-​renewal are activated, while genes of the final
that, especially in long-​living organisms such as humans, commitment are kept reversibly switched off. During age-
accumulating ageing-​dependent damage will alter this ing, the epigenetic programme of stem cells may slowly
dynamic process. There is evidence for mutated stem evolve away from the concise regulation of developmen-
cells undergoing clonal crypt dominance increasingly tal pathways during development leading to a ‘shadowed’
during ageing159,160. Moreover, clones can expand by a activation of developmental programmes and epigenetic
crypt fission event159,160. However, additional experi- drift. This may result in hyperactivation of self-​renewal
ments are required to assess the contribution of stem genes and/or skewing in gene expression pathways con-
cell DNA mutations and epigenetic alterations in field trolling lineage differentiation, thus disrupting the balance
colonization of the ageing gut epithelium. between stem cell self-​renewal and differentiation and
leading to the selection of mutant stem clones. Another,
Possible mechanisms promoting clonal dominance non-​mutually exclusive hypothesis could be that increas-
In the haematopoietic system, clonally dominant HSCs ing epigenome instability can increase the cell plasticity to
in ageing humans can be identified by the presence of respond to an aged environment. Under selective pressure,
mutant clones in the peripheral blood. On the basis of the a new stem cell identity with a dominant phenotype can
high recurrence of mutations in the same set of genes in arise in that specific aged niche.
population studies and experimental studies, most of these
mutations can be regarded as drivers of clonal dominance Deregulation of developmental pathways
of HSCs in ageing. Many of the drivers are directly (for The deregulation of developmental pathways is increas-
example, DNMT3A, TET2 and ASXL1) or indirectly ingly being recognized as underlying stem cell ageing in
(for example, IDH2, TP53, JAK2, SRSF2, U2AF1 and different tissues (Figs 4,5). The pathways that are affected
cohesin genes) involved in the epigenome regulation178–181. include Notch192, WNT193,194, FGFs195, TGFβ196, p38
How the epigenome is able to promote clonal dom- (ref.197) and cellular senescence signalling198,199 (Box 2).
inance of mutant stem and/or progenitor cells and why
epigenetic regulator mutations are selected in aged organ- WNT signalling
isms remain poorly understood. One hypothesis suggests Ageing MuSCs have increased activity of canonical WNT
that these epimutations do not strongly affect the molecular signalling, which promotes the erroneous differentiation
and metabolic machine of the adult stem cells but provoke of MuSCs into fibrogenic lineages and muscle fibrosis,
small but persistent epigenome changes that lead to slight as well as a decline in muscle regeneration following
advantages to the stem cells to outcompete non-​mutant injury200. In the haematopoietic system, a switch from
stem cells in colonizing stem cell niches. Alternatively, canonical (WNT3A) to non-​c anonical (WNT5A)
these epimutations may allow mutant adult stem cells to WNT signalling promotes HSC ageing by inducing the
adapt to the accumulation of molecular damage during loss of stem cell polarity — a CDC42-dependent pro-
ageing (see below). Mutant clones that arise from stem cells cess important in regulating the interaction of HSCs
may escape cellular checkpoints and immune response with the niche, self-​renewal and differentiation201,202.
that contribute day by day to the reinforcement of a dom- As WNT3A promotes lymphopoiesis and WNT5A
inant phenotype. It is currently not fully understood how enhances myelopoiesis203, the switch from canonical to
ageing affects the selection of mutant stem cell clones at non-​canonical WNT signalling may contribute to the
both cell-​intrinsic and cell-​extrinsic levels. ageing-​associated skewing of haematopoiesis towards
Epigenetic modifications have been demonstrated to myelopoiesis204. It was also reported that activation of
regulate gene transcription and many other transcription-​ non-​canonical WNT signalling maintains HSC self-​
associated genomic events, such as alternative promoter renewal and repopulation capacity by enhancing stem
Retrotransposon
A class of transposable DNA usage, retrotransposon repression, spurious transcription cell quiescence (which protects long-​term function of
sequences using RNA as a splicing and RNA maturation 182. Recent studies stem cells) through reduction of intracellular levels
mobile intermediate. reported that senescent cells and aged tissues undergo of ROS and suppression of Ca 2+–nuclear factor of
epigenetic changes that result in the reactivation of activated T cells (NFAT)–interferon-​γ (IFNγ) signal-
Spurious transcription
An RNA transcription process
retrotransposons 183,184. Some aged stem cells show ling205–207. It remains to be investigated whether switches
starting from an intragenic or global loss of DNA methylation in intragenic regions in canonical to non-​canonical WNT signalling (WNT3A
intergenic region. that could result in increased spurious transcription to WNT5A) and the selection of subpopulations of stem

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

MuSCs: p16 derepression reduces Notch and


induces CDK1 and TGFβ, reducing MuSC
Reproduction regenerative capacity
Embryogenesis Ageing
HSCs: p16 induction supresses Notch, which
impairs self-renewal of activated HSCs
WNT
MuSCs: FGF1 signalling is reduced and p38–MAPK
signalling is enhanced, impairing asymmetric cell
division of MuSCs; derepression of FGF2 can lead
to MuSC exhaustion
HSCs: administration of FGF7 or FGF21 restores
thymopoiesis
Notch
ISCs: p38–MAPK is induced by PDGF and VEGF,
Master regulators leading to hyperproliferation and
of development misdifferentiation
such as
HOX genes MuSCs: Notch reduction and TGFβ induction
impair regeneration
FGF– HSCs: Notch is suppressed, impairing self-renewal
p38–MAPK of activated HSCs
ISCs: Notch is suppressed or mutated, impairing
proliferation and differentiation
MuSCs: enhanced canonical WNT signalling drives
Senescence fibrosis
HSCs: switch from canonical to non-canonical
WNT signalling promotes myeloid bias
ISCs: DNA damage gives selective advantage to
WNTlow ISCs; low WNT impairs regeneration

Fig. 4 | Shadowed activation of developmental pathways in stem cells during ageing. Developmental pathways are
tightly regulated during embryogenesis. Master regulators of developmental pathways exhibit ageing-​associated
alteration in gene expression, which impairs the function of stem cells during ageing. These findings support the concept
that tightly controlled activity of developmental pathways, which drives embryogenesis and postnatal development until
the point of reproduction, is lost during ageing. After the reproductive age, the evolutionary pressure to maintain the
organism and its functions is reduced, and epigenetic changes that result from age-​associated increases in intracellular
damage and changes in the stem cell microenvironment and macroenvironment contribute to deregulation of these
developmental pathways in stem cells — the remnants of embryonic development in adult organisms. Aberrant expression
of master regulators of development, such as the HOX genes, which are highly responsive to changes in the epigenome,
could play a key part in causing ageing-​associated stem cell dysfunction, loss of organ homeostasis and disease. CDK1,
cyclin-​dependent kinase 1; FGF1, fibroblast growth factor 1; HSC, haematopoietic stem cell; ISC, intestinal stem cell;
MuSC, muscle stem cell; PDGF, platelet-​derived growth factor ; TGFβ, transforming growth factor-​β; VEGF, vascular
endothelial growth factor.

cells with high WNT5A activity are mutually exclusive leads to increased expression of cyclin-​dependent kinase
or parallel processes in HSC ageing. (CDK) inhibitors, including p16 (also known as p16INK4,
In the gut, canonical WNT signalling enhances DNA encoded by CDKN2A) and p21 (encoded by CDKN1A),
damage responses in ISCs208. In telomerase mutant mice which are two well-​established activators of cellular
with critically shortened, dysfunctional telomeres, accu- senescence that also have a role in development67,198,199.
mulation of DNA damage promotes the survival of stem Of note, ectopic expression of Notch rescued regener-
cells in niche positions exposed to low levels of WNT ative defects of aged MuSCs by counteracting ΤGFβ–
ligands208. Similar selection processes may contribute MAD3-driven expression of these CDK inhibitors, p15
to the reduction of canonical WNT signalling in ISCs (encoded by CDKN2B), p16, p21 and p27 (encoded by
in ageing wild-​type mice89 and in hair follicle stem cells CDKN1B)67. It is possible that decreases in Notch and
of progeroid mice209,210. Ageing-​associated decreases in activation of TGFβ signalling contribute to the derepres-
WNT signalling reduce the differentiation and regener- sion of p16 in MuSCs during advanced ageing, which
ative capacity of ISCs, thus affecting organ homeostasis. inhibits cell cycle re-​entry and regenerative capacity of
Progeroid mice
Mice carrying mutations in
Deregulation of Notch and WNT signalling is also seen MuSCs in geriatric mice214.
genes that are mutated in in HSCs and neuronal stem cells of patients with Down In the haematopoietic system, Notch promotes self-​
human progeroid syndromes syndrome211. It is possible that aneuploidy-​induced repli- renewal of HSCs, which is required for WNT-​mediated
that are characterized by cation stress and DNA damage contribute to the selection HSC proliferation215. While Notch activity is dispensable
segmental premature ageing
of stem cells with low WNT activity in these patients212. for the proliferative response of WNT-​activated HSCs,
(affecting specific organs).
the inhibition of Notch results in differentiation rather
Aneuploidy Notch signalling than self-​renewal of WNT-​activated HSCs. During
The irregular deviation of Ageing-​associated suppression of Notch signalling and ageing, the deletion of p16 rescued declines in the repopu­
chromosome numbers from an increase in ΤGFβ/mothers against decapentaplegic lation capacity of HSCs in mice, and this rescue was
the normal situation — not
including doubling of
homologue 3(MAD3 also known as SMAD3) signal- associated with enhanced Notch signalling216, implying
chromosome numbers as ling impair the regenerative capacity of MuSCs213. The that the suppressive regulation of gene expression
in tetraploidy. imbalance between ΤGFβ–SMAD3 and Notch signalling between Notch and p16 is bidirectional.

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Dysbiosis
In the intestine, Notch signalling controls the balance X-​linked in Drosophila, and male flies frequently exhibit
A deviation in the composition between self-​renewal and differentiation of stem and spontaneous somatic mutations of the locus resulting in
of bacterial strains and taxa in progenitor cells into different lineages of the intestinal inactivation of Notch and neoplasia formation in the
the microbiome of an organ or epithelium217. In D. melanogaster, the JUN N-​terminal ageing gut epithelium218. In contrast to the phenotypes
organism characterized by
kinase (Jnk) stress-​signalling pathway is activated by in Drosophila, Notch is important for the maintenance
decreases in overall complexity
and overgrowth of certain dysbiosis of the commensal bacteria, which occurs dur- of self-​renewal of ISCs in mice, and its deletion leads
species. ing ageing. Jnk signalling in turn activates proliferation to loss of proliferative cells in the basal crypts219. In
and aberrant differentiation of ISCs, which leads to the old mice, Notch1 signalling is decreased in ISCs, and
Lateral inhibition formation of misdifferentiated epithelial cell areas, loss there is an increase in the expression of protein atonal
A mechanism by which cells of
one type enforce a difference
of intestinal barrier function and organism death21. homologue 1 (ATOH1) — a regulator of lateral inhibition
in cell fate and/or cell identity Notch signalling in stem cell-​derived progenitor cells through δ-​like canonical Notch ligand (Dll) genes89.
in neighbouring cells. (enteroblasts) and neighbouring cells impinges on this
trajectory in two ways: it inhibits Jnk-​mediated over- FGF–p38–MAPK signalling
proliferation of ISCs but instructs misdifferentiation FGF1 signalling controls asymmetric activation of the
into dysplastic cells23. Interestingly, the Notch gene is p38–MAPK signalling pathway during MuSC divi-
sion, which regulates asymmetric cell division — the
Stem cell-intrinsic Stem cell-extrinsic p38–MAPK high-​expressing daughter cell undergoes
alterations alterations differentiation, and the p38–MAPK low-​expressing
Integrated alteration counterpart undergoes self renewal47. Ageing MuSCs
of the epigenome exhibit reduced responsiveness to FGF1 as well as ele-
vated p38–MAPK signalling. Both processes disturb
Deregulation of developmental pathways FGF1–p38–MAPK-​mediated regulation of asymmetric
• Most sensitive to epigenetic regulation MuSC divisions47,220. Enforced FGF1 signalling and inhib­
• Ground stage activity in adult stem cells
• Naturally regulated by the epigenome ition of p38–MAPK signalling, even if only transiently,
rescue the self-​renewal and regenerative capacity of aged
MuSCs47,220. MuSC niches secrete FGF2, which activates
Stem cell ageing Defects in stem cell function
stage 1 MuSCs to exit quiescence or dormancy88. Interestingly,
• Self-renewal
• Differentiation the expression of protein sprouty homologue 1 — an
Therapeutic inhibitor of FGF signalling — protects MuSC quies-
epigenetic
interventions cence, and imbalances in this regulatory system can lead
to age-​dependent MuSC exhaustion88.
Stem cell ageing In serum-​free cultured HSCs and progenitor cells,
stage 2
supplementation of FGF1 or FGF2 increased cell pro-
Epigenetic drift Mutations with altered liferation and self-​renewal221. Furthermore, in vivo
epigenome plasticity administration of FGF7 or FGF21 reversed ageing-​
Impairments in tissue associated thymic involution and enhanced thymic output
maintenance, systemic side of naive T lymphocytes in ageing mice, which could have
effects, ageing-associated
dysfunction and diseases beneficial effects for adaptive immune functions dur-
ing ageing222. However, the role of FGFs in HSC ageing
Fig. 5 | a model of stem cell ageing by epigenetic integration of damage signals and remains to be investigated. Downstream of growth factor
deregulation of developmental pathways. During ageing, stem cells accumulate stimuli, several mitogenic signalling pathways, such as
several types of molecular damage, and the stem cell environment (the stem cell niche ROS–MAPK and Janus kinase (JAK)–signal transducer
and in the blood system) undergoes several modifications. The epigenome undergoes and activator of transcription (STAT), were shown to
integrated changes in response to cell-​intrinsic and cell-​extrinsic alterations, which aggravate HSC ageing by induction of p53–Cdkn2a/
result in the deregulation of developmental signalling pathways in aged stem cells. In the p19ARF-​dependent DNA damage responses18,153.
adult organism, stem cells retain a low level of activity of signalling pathways that are FGFs contribute to the development of the intestinal
active during development and, therefore, are sensitive to ageing-​associated alterations
epithelium in various species223–226, but their role in the
of the epigenome that can deregulate these pathways. As a consequence, important
processes that are regulated by developmental pathways in stem cells are disturbed, maintenance of adult ISCs and any effects during ageing
including the control of self-​renewal, differentiation and overall stem cell capacity to remain to be investigated. There is experimental evidence
regenerate and maintain tissues. In stem cells, the stochastic integration of cell-​intrinsic that activation of ISC proliferation impairs ISC mainte-
damage and cell-​extrinsic alterations by the epigenome occurs progressively early nance and tissue homeostasis in ageing fly (see above)22.
during ageing (first stage of stem cell ageing) and leads to impairments in stem cell The activation of p38–MAPK signalling downstream of
function. This reduced stem cell function confers a selective advantage on stem cells that PDGF-​related and VEGF-​related factor 2 (PVF2) was
acquire drifts in the epigenome or mutations that increase epigenome plasticity. Both shown to aggravate this phenotype227.
processes could revert growth inhibitory effects that are imposed by the epigenetic
deregulation of developmental pathways in ageing stem cells. We propose that this is Hox genes
the second stage of stem cell ageing and represents a maladaptive change driven by
The functional impact of developmental pathways on
selection. Both stages of stem cell ageing lead to impairments in tissue maintenance,
systemic side effects (for example, increased inflammatory immune cells deriving stem cell ageing indicates that master regulators of
from mutant haematopoietic stem cells) and the development of ageing-​associated embryogenesis, such as Hox genes, could contribute
diseases. Therapeutic interventions that target epigenetic alterations during these two to stem cell ageing. Interestingly, many Hox genes are
stages of stem cell ageing may have the potential to delay the development of expressed in adult stem cells of different tissue com-
age-associated diseases. partments and have been associated with the formation

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Thymic involution
of malignancies, for example, in the haematopoietic sys- for future research. For example, it will be important to
A reduction in the thymus tem; however, functional studies of these genes in stem understand what mechanisms drive epigenome dysreg-
mass and in the output of naive cell ageing are mostly lacking. In skeletal muscle, a recent ulation during ageing, how molecular damage impinges
T lymphocytes. study revealed that the Hoxa9 gene is aberrantly acti- on these processes and how this leads to the aberrant
vated in MuSCs in response to injury. Hoxa9 induction activation of developmental pathways during ageing.
led to deregulation of several developmental pathways In addition to these slowly evolving integrated changes,
that were previously shown to impair MuSC function acute modification of the epigenome in response to
during ageing138 (see above). The role of master regu- tissue injury and regenerative stress seems to be per-
lators of development in the ageing of other stem cell turbed during ageing and lead to impairments in MuSC
compartments remains to be delineated. function and muscle regeneration by activation of
Hox genes138.
Conclusions and future perspectives It remains to be investigated whether there are
During ageing, stem cells that reside in different tis- common mechanisms that lead to changes in the epi­
sues accumulate several defects that can prevent them genome during ageing in quiescent and activated stem
from performing the crucial functions of repairing tis- cells. Delineating acute and chronic alterations in the
sue damage and maintaining tissue homeostasis. These control of epigenetic modification in stem cells during
defects include a decline in the maintenance of a healthy ageing is also important to understand the mechanisms
proteome, metabolic changes, alterations in intrinsic underlying ageing-​associated clonal dominance of stem
and extrinsic signalling pathways, DNA damage and cells carrying mutations in epigenome regulators — a
epigenetic changes. phenomenon that has emerged as a hallmark of the
Developmental pathways that are epigenetically ageing haematopoietic system in humans (Fig. 5). Lastly,
regulated and known to be crucial during embryogen- the concept of epigenetic integration of damage sig-
esis have been recently proposed to contribute to stem nals as a cause of stem cell and organism ageing holds
cell ageing. Epigenetic alterations and dysregulation of new promises for translational approaches, because
these pathways might impair the functionality of adult ­epigenome alterations are in principle reversible, and
stem cells during ageing (Figs 4,5). Multiple regulators there is evidence that epigenome reprogramming can
of the epigenome were shown to have important roles improve tissue maintenance, regenerative capacity and
in controlling self-​renewal, differentiation and regener- health span103,138,229.
ative potential of adult stem cells (for review, see ref.228),
Published online xx xx xxxx
and these observations are raising important questions

1. Martin, N., Beach, D. & Gil, J. Ageing as implications. Proc. Am. Thorac Soc. 9, 64–65 25. Ryall, J. G. et al. The NAD+-dependent SIRT1
developmental decay: insights from p16INK4a. (2012). deacetylase translates a metabolic switch into
Trends Mol. Med. 20, 667–674 (2014). 13. Simsek, T. et al. The distinct metabolic profile of regulatory epigenetics in skeletal muscle stem cells.
2. Henderson, S. T. & Johnson, T. E. daf-16 integrates hematopoietic stem cells reflects their location in a Cell Stem Cell 16, 171–183 (2015).
developmental and environmental inputs to mediate hypoxic niche. Cell Stem Cell 7, 380–390 (2010). 26. Cuervo, A. M. Autophagy and aging: keeping that old
aging in the nematode Caenorhabditis elegans. 14. Takubo, K. et al. Regulation of glycolysis by Pdk broom working. Trends Genet. 24, 604–612 (2008).
Curr. Biol. 11, 1975–1980 (2001). functions as a metabolic checkpoint for cell cycle 27. Geiger, H., de Haan, G. & Florian, M. C. The ageing
3. Blagosklonny, M. V. Aging is not programmed. quiescence in hematopoietic stem cells. Cell Stem Cell haematopoietic stem cell compartment. Nat. Rev.
Cell Cycle 12, 3736–3742 (2013). 12, 49–61 (2013). Immunol. 13, 376–389 (2013).
4. Behrens, A., van Deursen, J. M., Rudolph, K. L. & 15. Rodríguez-​Colman, M. J. et al. Interplay between 28. Shpilka, T. & Haynes, C. M. The mitochondrial UPR:
Schumacher, B. Impact of genomic damage and metabolic identities in the intestinal crypt supports mechanisms, physiological functions and implications in
ageing on stem cell function. Nat. Cell Biol. 16, stem cell function. Nature 543, 424–427 (2017). ageing. Nat. Rev. Mol. Cell. Biol. 19, 109–120 (2017).
201–207 (2014). 16. Park, C. B. & Larsson, N. G. Mitochondrial DNA 29. Douglas, P. M. & Dillin, A. Protein homeostasis and
5. Mohrin, M. et al. Hematopoietic stem cell quiescence mutations in disease and aging. J. Cell Biol. 193, aging in neurodegeneration. J. Cell Biol. 190,
promotes error-​prone DNA repair and mutagenesis. 809–818 (2011). 719–729 (2010).
Cell Stem Cell 7, 174–185 (2010). 17. Garcia-​Prat, L. et al. Autophagy maintains stemness 30. Lapierre, L. R., Kumsta, C., Sandri, M., Ballabio, A. &
6. Sotiropoulou, P. A. et al. Bcl-2 and accelerated DNA by preventing senescence. Nature 529, 37–42 Hansen, M. Transcriptional and epigenetic regulation
repair mediates resistance of hair follicle bulge stem (2016). of autophagy in aging. Autophagy 11, 867–880
cells to DNA-​damage-induced cell death. Nat. Cell 18. Ito, K. et al. Reactive oxygen species act through p38 (2015).
Biol. 12, 572–582 (2010). MAPK to limit the lifespan of hematopoietic stem 31. Merkwirth, C. et al. Two conserved histone
7. Goloubinoff, P., Sassi, A. S., Fauvet, B., Barducci, A. & cells. Nat. Med. 12, 446–451 (2006). demethylases regulate mitochondrial stress-​induced
De Los Rios, P. Chaperones convert the energy from 19. Owusu-​Ansah, E. & Banerjee, U. Reactive oxygen longevity. Cell 165, 1209–1223 (2016).
ATP into the nonequilibrium stabilization of native species prime Drosophila haematopoietic progenitors 32. Mohrin, M. et al. A mitochondrial UPR-​mediated
proteins. Nat. Chem. Biol. https://doi.org/10.1038/ for differentiation. Nature 461, 537–541 (2009). metabolic checkpoint regulates hematopoietic stem
s41589-018-0013-8 (2018). 20. Mantel, C. R. et al. Enhancing hematopoietic stem cell cell aging. Science 347, 1374–1377 (2015).
8. Peth, A., Nathan, J. A. & Goldberg, A. L. The ATP transplantation efficacy by mitigating oxygen shock. 33. Solanas, G. et al. Aged stem cells reprogram their
costs and time required to degrade ubiquitinated Cell 161, 1553–1565 (2015). daily rhythmic functions to adapt to stress. Cell 170,
proteins by the 26 S proteasome. J. Biol. Chem. 288, 21. Guo, L., Karpac, J., Tran, S. L. & Jasper, H. PGRP-​SC2 678–692.e20 (2017).
29215–29222 (2013). promotes gut immune homeostasis to limit 34. Cellerino, A. & Ori, A. What have we learned on aging
9. Ho, T. T. et al. Autophagy maintains the metabolism commensal dysbiosis and extend lifespan. Cell 156, from omics studies? Semin. Cell Dev. Biol. 70,
and function of young and old stem cells. Nature 543, 109–122 (2014). 177–189 (2017).
205–210 (2017). 22. Biteau, B. et al. Lifespan extension by preserving 35. Buchwalter, A. & Hetzer, M. W. Nucleolar expansion
This study reports that subpopulations of HSCs proliferative homeostasis in Drosophila. PLoS Genet. and elevated protein translation in premature aging.
exhibit defects in autophagy during ageing that are 6, e1001159 (2010). Nat. Commun. 8, 328 (2017).
associated with impairments in stem cell function. 23. Biteau, B., Hochmuth, C. E. & Jasper, H. JNK activity in 36. Zhang, W. et al. A Werner syndrome stem cell model
10. Franceschi, C. et al. Chronic inflammation somatic stem cells causes loss of tissue homeostasis unveils heterochromatin alterations as a driver of
(inflammaging) and its potential contribution to in the aging Drosophila gut. Cell Stem Cell 3, 442–455 human aging. Science 348, 1160–1163 (2015).
age-associated diseases. J. Gerontol. A Biol. Sci. (2008). 37. Signer, R. A. J., Magee, J. A., Salic, A. & Morrison, S. J.
Med. Sci. 69 (Suppl. 1), S4–S9 (2014). 24. Zhang, H. et al. NAD+ repletion improves Haematopoietic stem cells require a highly
11. Thevaranjan, N. et al. Age-​associated microbial mitochondrial and stem cell function and enhances life regulated protein synthesis rate. Nature 509,
dysbiosis promotes intestinal permeability, systemic span in mice. Science 352, 1436–1443 (2016). 49–54 (2014).
inflammation, and macrophage dysfunction. Cell Host This study shows that treatment with NAD+ 38. Wang, L., Ryoo, H. D., Qi, Y. & Jasper, H. PERK limits
Microbe 21, 455–466.e4 (2017). precursor nicotinamide riboside improves stem cell Drosophila lifespan by promoting intestinal stem cell
12. Baylin, S. B. The cancer epigenome: its origins, function and prevents their senescence in ageing by proliferation in response to ER stress. PLoS Genet. 11,
contributions to tumorigenesis, and translational enhancing mitochondrial quality. e1005220 (2015).

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

39. Bradley, E., Bieberich, E., Mivechi, N. F., 63. O’Sullivan, R. J., Kubicek, S., Schreiber, S. L. & 92. Herbig, U., Ferreira, M., Condel, L., Carey, D. &
Tangpisuthipongsa, D. & Wang, G. Regulation of Karlseder, J. Reduced histone biosynthesis and Sedivy, J. M. Cellular senescence in aging primates.
embryonic stem cell pluripotency by heat shock chromatin changes arising from a damage signal at Science 311, 1257 (2006).
protein 90. Stem Cells 30, 1624–1633 (2012). telomeres. Nat. Struct. Mol. Biol. 17, 1218–1225 93. Song, Z., Zhang, J., Ju, Z. & Rudolph, K. L. Telomere
40. Noormohammadi, A. et al. Somatic increase of CCT8 (2010). dysfunctional environment induces loss of quiescence
mimics proteostasis of human pluripotent stem cells 64. Liu, L. et al. Chromatin modifications as determinants and inherent impairments of hematopoietic stem cell
and extends C. elegans lifespan. Nat. Commun. 7, of muscle stem cell quiescence and chronological function. Aging Cell 11, 449–455 (2012).
13649 (2016). aging. Cell Rep. 4, 189–204 (2013). 94. Guidi, N. et al. Osteopontin attenuates
41. Vilchez, D. et al. Increased proteasome activity in 65. Conboy, I. M. et al. Rejuvenation of aged progenitor aging-associated phenotypes of hematopoietic stem
human embryonic stem cells is regulated by PSMD11. cells by exposure to a young systemic environment. cells. EMBO J. 36, 840–853 (2017).
Nature 489, 304–308 (2012). Nature 433, 760–764 (2005). 95. Lukjanenko, L. et al. Loss of fibronectin from the
42. van Galen, P. et al. The unfolded protein response 66. Villeda, S. A. et al. The ageing systemic milieu aged stem cell niche affects the regenerative
governs integrity of the haematopoietic stem-​cell pool negatively regulates neurogenesis and cognitive capacity of skeletal muscle in mice. Nat. Med. 22, 897
during stress. Nature 510, 268–272 (2014). function. Nature 477, 90–94 (2011). (2016).
43. Bufalino, M. R., DeVeale, B. & van der Kooy, D. 67. Carlson, M. E., Hsu, M. & Conboy, I. M. Imbalance 96. Rozo, M., Li, L. & Fan, C.-M. Targeting β1-integrin
The asymmetric segregation of damaged proteins is stem between pSmad3 and Notch induces CDK inhibitors in signaling enhances regeneration in aged and dystrophic
cell-​type dependent. J. Cell Biol. 201, 523–530 (2013). old muscle stem cells. Nature 454, 528–532 (2008). muscle in mice. Nat. Med. 22, 889–896 (2016).
44. Katajisto, P. et al. Stem cells. Asymmetric apportioning 68. Sinha, M. et al. Restoring systemic GDF11 levels 97. Boyle, M. et al. Decline in self-​renewal factors
of aged mitochondria between daughter cells is reverses age-​related dysfunction in mouse skeletal contributes to aging of the stem cell niche in the
required for stemness. Science 348, 340–343 (2015). muscle. Science 344, 649–652 (2014). Drosophila testis. Cell Stem Cell 1, 470–478 (2007).
45. Moore, D. L., Pilz, G. A., Arauzo-​Bravo, M. J., Barral, Y. 69. Elabd, C. et al. Oxytocin is an age-​specific circulating 98. van Niel, G., D’Angelo, G. & Raposo, G. Shedding light
& Jessberger, S. A mechanism for the segregation of hormone that is necessary for muscle maintenance on the cell biology of extracellular vesicles. Nat. Rev.
age in mammalian neural stem cells. Science 349, and regeneration. Nat. Commun. 5, 4082 (2014). Mol. Cell Biol. 19, 213–228 (2018).
1334–1338 (2015). 70. Egerman, M. A. et al. GDF11 increases with age and 99. Fry, C. S., Kirby, T. J., Kosmac, K., McCarthy, J. J. &
46. Erjavec, N., Larsson, L., Grantham, J. & Nystrom, T. inhibits skeletal muscle regeneration. Cell Metab. 22, Peterson, C. A. Myogenic progenitor cells control
Accelerated aging and failure to segregate damaged 164–174 (2015). extracellular matrix production by fibroblasts during
proteins in Sir2 mutants can be suppressed by 71. Castellano, J. M. et al. Human umbilical cord plasma skeletal muscle hypertrophy. Cell Stem Cell 20, 56–69
overproducing the protein aggregation-​remodeling proteins revitalize hippocampal function in aged mice. (2017).
factor Hsp104p. Genes Dev. 21, 2410–2421 (2007). Nature 544, 488–492 (2017). 100. Fusco, S. et al. A CREB-​Sirt1-Hes1 circuitry mediates
47. Bernet, J. D. et al. p38 MAPK signaling underlies a 72. Gancz, D. & Gilboa, L. Hormonal control of stem cell neural stem cell response to glucose availability.
cell-​autonomous loss of stem cell self-​renewal in systems. Annu. Rev. Cell Dev. Biol. 29, 137–162 Cell Rep. 14, 1195–1205 (2016).
skeletal muscle of aged mice. Nat. Med. 20, (2013). 101. Gjorevski, N. et al. Designer matrices for intestinal stem
265–271 (2014). 73. Nakada, D. et al. Oestrogen increases haematopoietic cell and organoid culture. Nature 539, 560–564 (2016).
48. Zou, C. & Mallampalli, R. K. Regulation of histone stem-​cell self-​renewal in females and during 102. Stearns-​Reider, K. M. et al. Aging of the skeletal
modifying enzymes by the ubiquitin-​proteasome system. pregnancy. Nature 505, 555–558 (2014). muscle extracellular matrix drives a stem cell
Biochim. Biophys. Acta 1843, 694–702 (2014). 74. Kim, J.-H. et al. Sex hormones establish a reserve pool fibrogenic conversion. Aging Cell 16, 518–528 (2017).
49. Gambetta, M. C. & Müller, J. O-​GlcNAcylation of adult muscle stem cells. Nat. Cell Biol. 18, 103. Crowder, S. W., Leonardo, V., Whittaker, T.,
prevents aggregation of the polycomb group repressor 930–940 (2016). Papathanasiou, P. & Stevens, M. M. Material cues as
polyhomeotic. Dev. Cell 31, 629–639 (2014). 75. Bolstad, B. M., Irizarry, R. A., Astrand, M. & Speed, T. potent regulators of epigenetics and stem cell
50. Hammond, C. M., Strømme, C. B., Huang, H., A comparison of normalization methods for high function. Cell Stem Cell 18, 39–52 (2016).
Patel, D. J. & Groth, A. Histone chaperone networks density oligonucleotide array data based on variance 104. Buczacki, S. J. A. et al. Intestinal label-​retaining cells
shaping chromatin function. Nat. Rev. Mol. Cell Biol. and bias. Bioinformatics 19, 185–193 (2003). are secretory precursors expressing Lgr5. Nature
18, 141–158 (2017). 76. Ergen, A. V., Boles, N. C. & Goodell, M. A. Rantes/Ccl5 495, 65–69 (2013).
51. Das, C. & Tyler, J. K. Histone exchange and histone influences hematopoietic stem cell subtypes and 105. Rosenberger, G. et al. Statistical control of peptide
modifications during transcription and aging. causes myeloid skewing. Blood 119, 2500–2509 and protein error rates in large-​scale targeted data-​
Biochim. Biophys. Acta 1819, 332–342 (2013). (2012). independent acquisition analyses. Nat. Methods 14,
52. Feser, J. et al. Elevated histone expression promotes 77. Mossadegh-​Keller, N. et al. M-​CSF instructs myeloid 921–927 (2017).
life span extension. Mol. Cell 39, 724–735 (2010). lineage fate in single haematopoietic stem cells. 106. Jadhav, U. et al. Dynamic reorganization of chromatin
53. Sperka, T., Wang, J. & Rudolph, K. L. DNA damage Nature 497, 239–243 (2013). accessibility signatures during dedifferentiation of
checkpoints in stem cells, ageing and cancer. Nat. Rev. 78. Palacios, D. et al. TNF/p38?/Polycomb signaling to secretory precursors into Lgr5+ intestinal stem cells.
Mol. Cell Biol. 13, 579–590 (2012). Pax7 locus in satellite cells links inflammation to the Cell Stem Cell 21, 65–77.e5 (2017).
54. Burkhalter, M. D., Rudolph, K. L. & Sperka, T. Genome epigenetic control of muscle regeneration. Cell Stem 107. Basak, O. et al. Induced quiescence of Lgr5+ stem
instability of ageing stem cells-​Induction and defence Cell 7, 455–469 (2010). cells in intestinal organoids enables differentiation of
mechanisms. Ageing Res. Rev. 23, 29–36 (2015). 79. Weir, H. J. et al. Dietary restriction and AMPK hormone-​producing enteroendocrine cells. Cell Stem
55. Beerman, I., Seita, J., Inlay, M. A., Weissman, I. L. & increase lifespan via mitochondrial network and Cell 20, 177–190.e4 (2017).
Rossi, D. J. Quiescent hematopoietic stem cells peroxisome remodeling. Cell Metab. 26, 884–896.e5 108. Yan, K. S. et al. Non-​equivalence of Wnt and R-​spondin
accumulate DNA damage during aging that is repaired (2017). ligands during Lgr5+ intestinal stem-​cell self-​renewal.
upon entry into cell cycle. Cell Stem Cell 15, 37–50 80. Smith, P. et al. Regulation of life span by the gut Nature 545, 238–242 (2017).
(2014). microbiota in the short-​lived African turquoise killifish. 109. Fontana, L. & Partridge, L. Promoting health and
56. Flach, J. et al. Replication stress is a potent driver of eLife 6, e27014 (2017). longevity through diet: from model organisms to
functional decline in ageing haematopoietic stem cells. 81. Hahn, O. et al. Dietary restriction protects from age-​ humans. Cell 161, 106–118 (2015).
Nature 512, 198–202 (2014). associated DNA methylation and induces epigenetic 110. Tang, D. et al. Dietary restriction improves
This study shows that DNA damage increases in reprogramming of lipid metabolism. Genome Biol. 18, repopulation but impairs lymphoid differentiation
ageing HSCs as a consequence of increased 1194 (2017). capacity of hematopoietic stem cells in early aging.
replication stress owing to imbalances in the 82. Mendelson, A. & Frenette, P. S. Hematopoietic stem J. Exp. Med. 213, 535–553 (2016).
expression of components of the DNA replication cell niche maintenance during homeostasis and 111. Yilmaz, Ö. H. et al. mTORC1 in the Paneth cell niche
complex. regeneration. Nat. Med. 20, 833–846 (2014). couples intestinal stem-​cell function to calorie intake.
57. Walter, D. et al. Exit from dormancy provokes DNA-​ 83. Tumbar, T. et al. Defining the epithelial stem cell niche Nature 486, 490–495 (2012).
damage-induced attrition in haematopoietic stem in skin. Science 303, 359–363 (2004). 112. Mair, W., McLeod, C. J., Wang, L. & Jones, D. L.
cells. Nature 520, 549–552 (2015). 84. Morrison, S. J. & Scadden, D. T. The bone marrow Dietary restriction enhances germline stem cell
This study shows that entry into the cell cycle from niche for haematopoietic stem cells. Nature 505, maintenance. Aging Cell 9, 916–918 (2010).
dormancy is a phase of vulnerability in HSCs, 327–334 (2014). 113. Cerletti, M., Jang, Y. C., Finley, L. W. S., Haigis, M. C.
leading to an accumulation of DNA damage and 85. Blau, H. M., Cosgrove, B. D. & Ho, A. T. V. The central & Wagers, A. J. Short-​term calorie restriction
stem cell exhaustion. role of muscle stem cells in regenerative failure with enhances skeletal muscle stem cell function. Cell Stem
58. Rossi, D. J. et al. Deficiencies in DNA damage repair aging. Nat. Med. 21, 854–862 (2015). Cell 10, 515–519 (2012).
limit the function of haematopoietic stem cells with 86. Barker, N. Adult intestinal stem cells: critical drivers of 114. Brandhorst, S. et al. A periodic diet that mimics
age. Nature 447, 725–729 (2007). epithelial homeostasis and regeneration. Nat. Rev. fasting promotes multi-​system regeneration, enhanced
59. Nijnik, A. et al. DNA repair is limiting for Mol. Cell Biol. 15, 19–33 (2013). cognitive performance, and healthspan. Cell Metab.
haematopoietic stem cells during ageing. Nature 447, 87. Kaiko, G. E. et al. The colonic crypt protects stem cells 22, 86–99 (2015).
686–690 (2007). from microbiota-​derived metabolites. Cell 165, 115. Cheng, C.-W. et al. Fasting-​mimicking diet promotes
60. Gorbunova, V. & Seluanov, A. DNA double strand 1708–1720 (2016). Ngn3-driven β-​cell regeneration to reverse diabetes.
break repair, aging and the chromatin connection. 88. Chakkalakal, J. V., Jones, K. M., Basson, M. A. & Cell 168, 775–788.e12 (2017).
Mutat. Res. Mol. Mech. Mutag. 788, 2–6 (2016). Brack, A. S. The aged niche disrupts muscle stem cell 116. Regan, J. C. et al. Sex difference in pathology of the
61. Kauppinen, T. M., Gan, L. & Swanson, R. A. quiescence. Nature 490, 355–360 (2012). ageing gut mediates the greater response of female
Poly(ADP-ribose) polymerase-1-induced NAD(+) 89. Nalapareddy, K. et al. Canonical Wnt signaling lifespan to dietary restriction. eLife 5, e10956 (2016).
depletion promotes nuclear factor-​κB transcriptional ameliorates aging of intestinal stem cells. Cell Rep. 117. Goldberg, E. L. et al. Lifespan-​extending caloric
activity by preventing p65 de-​acetylation. 18, 2608–2621 (2017). restriction or mTOR inhibition impair adaptive
Biochim. Biophys. Acta 1833, 1985–1991 (2013). 90. Lindemans, C. A. et al. Interleukin-22 promotes immunity of old mice by distinct mechanisms.
62. Missios, P. et al. Glucose substitution prolongs intestinal-​stem-cell-​mediated epithelial regeneration. Aging Cell 14, 130–138 (2015).
maintenance of energy homeostasis and lifespan Nature 528, 560–564 (2015). 118. Lazare, S. et al. Lifelong dietary intervention does not
of telomere dysfunctional mice. Nat. Commun. 5, 91. Campisi, J. Aging, Cellular senescence, and cancer. affect hematopoietic stem cell function. Exp. Hematol.
4924 (2014). Annu. Rev. Physiol. 75, 685–705 (2013). 53, 26–30 (2017).

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

119. Igarashi, M. & Guarente, L. mTORC1 and SIRT1 146. Ciccocioppo, R. et al. Small bowel enterocyte of hematopoietic stem cells in mice. Proc. Natl Acad.
cooperate to foster expansion of gut adult stem cells apoptosis and proliferation are increased in the Sci. USA 108, 14566–14571 (2011).
during calorie restriction. Cell 166, 436–450 (2016). elderly. Gerontology 48, 204–208 (2002). 172. Zhang, X. et al. DNMT3A and TET2 compete and
120. Zhang, C. et al. Structural modulation of gut 147. Corazza, G. R. et al. Proliferating cell nuclear cooperate to repress lineage-​specific transcription
microbiota in life-​long calorie-​restricted mice. antigen expression is increased in small bowel factors in hematopoietic stem cells. Nat. Genet. 48,
Nat. Commun. 4, 2163 (2013). epithelium in the elderly. Mech. Ageing Dev. 104, 1014–1023 (2016).
121. Petkovich, D. A. et al. Using DNA methylation profiling 1–9 (1998). 173. Fuster, J. J. et al. Clonal hematopoiesis associated
to evaluate biological age and longevity interventions. 148. Kim, J. Y., Siegmund, K. D., Tavaré, S. & Shibata, D. with Tet2 deficiency accelerates atherosclerosis
Cell Metab. 25, 954–960.e6 (2017). Age-​related human small intestine methylation: development in mice. Science 355, 842–847
122. Öst, A. et al. Paternal diet defines offspring chromatin evidence for stem cell niches. BMC Med. 3, 10 (2005). (2017).
state and intergenerational obesity. Cell 159, 149. Camp, J. G. et al. Microbiota modulate transcription in 174. Jaiswal, S. et al. Clonal hematopoiesis and risk of
1352–1364 (2014). the intestinal epithelium without remodeling the atherosclerotic cardiovascular disease. N. Engl. J.
123. Cabezas-​Wallscheid, N. et al. Vitamin A-​retinoic acid accessible chromatin landscape. Genome Res. 24, Med. 377, 111–121 (2017).
signaling regulates hematopoietic stem cell dormancy. 1504–1516 (2014). References 172 and 173 report that the clonal
Cell 169, 807–823.e19 (2017). 150. Hahn, M. A. et al. Methylation of polycomb target dominance of HSCs with mutations in epigenome
124. Cimmino, L. et al. Restoration of TET2 function blocks genes in intestinal cancer is mediated by regulators contributes to the development of
aberrant self-​renewal and leukemia progression. Cell inflammation. Cancer Res. 68, 10280–10289 ageing-​associated diseases.
170, 1079–1095.e20 (2017). (2008). 175. Bahar, R. et al. Increased cell-​to-cell variation in gene
125. Agathocleous, M. et al. Ascorbate regulates 151. Mugatroyd, C., Wu, Y., Bockmühl, Y. & Spengler, D. expression in ageing mouse heart. Nature 441,
haematopoietic stem cell function and The Janus face of DNA methylation in aging. Aging 2, 1011–1014 (2006).
leukaemogenesis. Nature 549, 476 (2017). 107–110 (2010). 176. Martincorena, I. et al. High burden and pervasive
126. D’Aniello, C., Cermola, F., Patriarca, E. J. & Minchiotti, G. 152. Takahashi, K. et al. Epigenetic control of the host positive selection of somatic mutations in normal
Vitamin C in stem cell biology: impact on extracellular gene by commensal bacteria in large intestinal human skin. Science 348, 880–886 (2015).
matrix homeostasis and epigenetics. Stem Cells Int. epithelial cells. J. Biol. Chem. 286, 35755–35762 177. Snippert, H. J. et al. Intestinal crypt homeostasis
2017, 8936156 (2017). (2011). results from neutral competition between
127. Booth, L. N. & Brunet, A. The aging epigenome. 153. Kirschner, K. et al. Proliferation drives aging-​related symmetrically dividing Lgr5 stem cells. Cell 143,
Mol. Cell 62, 728–744 (2016). functional decline in a subpopulation of the 134–144 (2010).
128. Horvath, S. et al. Aging effects on DNA methylation hematopoietic stem cell compartment. Cell Rep. 19, 178. Figueroa, M. E. et al. Leukemic IDH1 and IDH2
modules in human brain and blood tissue. 1503–1511 (2017). mutations result in a hypermethylation phenotype,
Genome Biol. 13, R97 (2012). 154. Enge, M. et al. Single-​cell analysis of human pancreas disrupt TET2 function, and impair hematopoietic
129. Horvath, S. DNA methylation age of human tissues reveals transcriptional signatures of aging and differentiation. Cancer Cell 18, 553–567 (2010).
and cell types. Genome Biol. 14, R115 (2013). somatic mutation patterns. Cell 171, 321–330.e14 179. He, J. & Zhang, Y. Janus kinase 2: an epigenetic
130. Stubbs, T. M. et al. Multi-​tissue DNA methylation age (2017). ‘writer’ that activates leukemogenic genes. J. Mol.
predictor in mouse. Genome Biol. 18, 68 (2017). 155. Cheung, P. et al. Single-cell chromatin modification Cell. Biol. 2, 231–233 (2010).
131. Weidner, C. et al. Aging of blood can be tracked by profiling reveals increased epigenetic variations with 180. Mian, S. A. et al. Spliceosome mutations exhibit
DNA methylation changes at just three CpG sites. aging. Cell https://doi.org/10.1016/j.cell.2018.03.079 specific associations with epigenetic modifiers and
Genome Biol. 15, R24 (2014). (2018). proto-​oncogenes mutated in myelodysplastic
132. Yuan, T. et al. An integrative multi-​scale analysis of the 156. Alcolea, M. P. et al. Differentiation imbalance in single syndrome. Haematologica 98, 1058–1066 (2013).
dynamic DNA methylation landscape in aging. oesophageal progenitor cells causes clonal 181. Vrba, L., Junk, D. J., Novak, P. & Futscher, B. W. p53
PLoS Genet. 11, e1004996 (2015). immortalization and field change. Nat. Cell Biol. 16, induces distinct epigenetic states at its direct target
133. Beerman, I. et al. Proliferation-​dependent alterations 615–622 (2014). promoters. BMC Genomics 9, 486 (2008).
of the DNA methylation landscape underlie 157. Genovese, G. et al. Clonal hematopoiesis and blood-​ 182. Reddington, J. P., Pennings, S. & Meehan, R. R.
hematopoietic stem cell aging. Cell Stem Cell 12, cancer risk inferred from blood DNA sequence. Non-​canonical functions of the DNA methylome in
413–425 (2013). N. Engl. J. Med. 371, 2477–2487 (2014). gene regulation. Biochem. J. 451, 13–23 (2013).
134. Arai, K. et al. Total synthesis of 6-deoxypladienolide D 158. Goriely, A. & Wilkie, A. O. M. Paternal age effect 183. De Cecco, M. et al. Genomes of replicatively senescent
and assessment of splicing inhibitory activity in a mutations and selfish spermatogonial selection: cells undergo global epigenetic changes leading to
mutant SF3B1 cancer cell line. Org. Lett. 16, causes and consequences for human disease. Am. J. gene silencing and activation of transposable
5560–5563 (2014). Hum. Genet. 90, 175–200 (2012). elements. Aging Cell 12, 247–256 (2013).
135. Fernández, A. F. et al. H3K4me1 marks DNA 159. Greaves, L. C. et al. Mitochondrial DNA mutations are 184. Van Meter, M. et al. SIRT6 represses LINE1
regions hypomethylated during aging in human stem established in human colonic stem cells, and mutated retrotransposons by ribosylating KAP1 but this
and differentiated cells. Genome Res. 25, 27–40 clones expand by crypt fission. Proc. Natl Acad. Sci. repression fails with stress and age. Nat. Commun. 5,
(2014). USA 103, 714–719 (2006). 5011 (2014).
136. Cole, J. J. et al. Diverse interventions that extend 160. Hsieh, J. C. F., Van Den Berg, D., Kang, H., Hsieh, C.-L. 185. Neri, F. et al. Intragenic DNA methylation prevents
mouse lifespan suppress shared age-​associated & Lieber, M. R. Large chromosome deletions, spurious transcription initiation. Nature 543, 72–77
epigenetic changes at critical gene regulatory regions. duplications, and gene conversion events accumulate (2017).
Genome Biol. 18, E503 (2017). with age in normal human colon crypts. Aging Cell 12, 186. Kim, J. et al. Blocking promiscuous activation at
137. Maegawa, S. et al. Caloric restriction delays 269–279 (2013). cryptic promoters directs cell type-​specific gene
age-related methylation drift. Nat. Commun. 8, 539 161. Jaiswal, S. et al. Age-​related clonal hematopoiesis expression. Science 356, 717–721 (2017).
(2017). associated with adverse outcomes. N. Engl. J. Med. 187. Maunakea, A. K., Chepelev, I., Cui, K. & Zhao, K.
138. Schwörer, S. et al. Epigenetic stress responses induce 371, 2488–2498 (2014). Intragenic DNA methylation modulates alternative
muscle stem-​cell ageing by Hoxa9 developmental 162. McKerrell, T. et al. Leukemia-​associated somatic splicing by recruiting MeCP2 to promote exon
signals. Nature 540, 428–432 (2016). mutations drive distinct patterns of age-​related clonal recognition. Cell Res. 23, 1256–1269 (2013).
This study shows that epigenetic alteration in hemopoiesis. Cell Rep. 10, 1239–1245 (2015). 188. Shukla, S. et al. CTCF-​promoted RNA polymerase II
response to activation limits the self-​renewal and 163. Okuchi, Y. et al. Identification of aging-​associated gene pausing links DNA methylation to splicing. Nature
function of aged muscle stem cells by activation of expression signatures that precede intestinal 479, 74–79 (2011).
developmental signals. tumorigenesis. PLoS ONE 11, e0162300 (2016). 189. Yearim, A. et al. HP1 is involved in regulating the
139. Sun, D. et al. Epigenomic profiling of young and aged 164. Shlush, L. I. et al. Identification of pre-​leukaemic global impact of DNA methylation on alternative
HSCs reveals concerted changes during aging that haematopoietic stem cells in acute leukaemia. Nature splicing. Cell Rep. 10, 1122–1134 (2015).
reinforce self-​renewal. Cell Stem Cell 14, 673–688 506, 328–333 (2014). 190. Irimia, M. & Blencowe, B. J. Alternative splicing:
(2014). 165. Busque, L. et al. Recurrent somatic TET2 mutations in decoding an expansive regulatory layer. Curr. Opin.
140. Beerman, I. & Rossi, D. J. Epigenetic control of stem normal elderly individuals with clonal hematopoiesis. Cell Biol. 24, 323–332 (2012).
cell potential during homeostasis, aging, and disease. Nat. Genet. 44, 1179–1181 (2012). 191. Jenuwein, T. & Allis, C. D. Translating the histone code.
Cell Stem Cell 16, 613–625 (2015). 166. Lu, R. et al. Epigenetic perturbations by Arg882- Science 293, 1074–1080 (2001).
141. Kazakevych, J., Sayols, S., Messner, B., Krienke, C. & mutated DNMT3A potentiate aberrant stem cell gene-​ 192. Artavanis-​Tsakonas, S., Rand, M. D. & Lake, R. J.
Soshnikova, N. Dynamic changes in chromatin states expression program and acute leukemia development. Notch signaling: cell fate control and signal
during specification and differentiation of adult Cancer Cell 30, 92–107 (2016). integration in development. Science 284, 770–776
intestinal stem cells. Nucleic Acids Res. 45, 167. Scourzic, L. et al. DNMT3A(R882H) mutant and Tet2 (1999).
5770–5784 (2017). inactivation cooperate in the deregulation of DNA 193. Logan, C. Y. & Nusse, R. The Wnt signaling pathway in
142. Kim, K.-M. & Shibata, D. Methylation reveals a niche: methylation control to induce lymphoid malignancies development and disease. Annu. Rev. Cell Dev. Biol.
stem cell succession in human colon crypts. Oncogene in mice. Leukemia 30, 1388–1398 (2016). 20, 781–810 (2004).
21, 5441–5449 (2002). 168. Tefferi, A. et al. TET2 mutations and their clinical 194. Clevers, H. Wnt/beta-​Catenin signaling in development
143. Ro, S. & Rannala, B. Methylation patterns and correlates in polycythemia vera, essential and disease. Cell 127, 469–480 (2006).
mathematical models reveal dynamics of stem cell thrombocythemia and myelofibrosis. Leukemia 23, 195. Thisse, B. & Thisse, C. Functions and regulations of
turnover in the human colon. Proc. Natl Acad. Sci. 905–911 (2009). fibroblast growth factor signaling during embryonic
USA 98, 10519–10521 (2001). 169. Yang, L. et al. DNMT3A loss drives enhancer development. Dev. Biol. 287, 390–402 (2005).
144. Kaaij, L. T. J. et al. DNA methylation dynamics during hypomethylation in FLT3-ITD-​associated leukemias. 196. Kingsley, D. M. The TGF-​beta superfamily: new members,
intestinal stem cell differentiation reveals enhancers Cancer Cell 29, 922–934 (2016). new receptors, and new genetic tests of function in
driving gene expression in the villus. Genome Biol. 14, 170. Challen, G. A. et al. Dnmt3a is essential for different organisms. Genes Dev. 8, 133–146 (1994).
R50 (2013). hematopoietic stem cell differentiation. Nat. Genet. 197. Keren, A., Tamir, Y. & Bengal, E. The p38 MAPK
145. Sheaffer, K. L. et al. DNA methylation is required for 44, 23–31 (2011). signaling pathway: a major regulator of skeletal
the control of stem cell differentiation in the small 171. Ko, M. et al. Ten-​Eleven-Translocation 2 (TET2) muscle development. Mol. Cell. Endocrinol. 252,
intestine. Genes Dev. 28, 652–664 (2014). negatively regulates homeostasis and differentiation 224–230 (2006).

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

198. Muñoz-​Espín, D. et al. Programmed cell senescence 220. Cosgrove, B. D. et al. Rejuvenation of the muscle stem function and organ maintenance, leading to
during mammalian embryonic development. Cell 155, cell population restores strength to injured aged increased health span.
1104–1118 (2013). muscles. Nat. Med. 20, 255–264 (2014). 241. Berry, D. C. et al. Cellular aging contributes to failure
199. Storer, M. et al. Senescence is a developmental 221. de Haan, G. et al. In vitro generation of long-​term of cold-​induced beige adipocyte formation in old mice
mechanism that contributes to embryonic growth and repopulating hematopoietic stem cells by fibroblast and humans. Cell Metab. 25, 166–181 (2016).
patterning. Cell 155, 1119–1130 (2013). growth factor-1. Dev. Cell 4, 241–251 (2003). 242. Ju, Z. et al. Telomere dysfunction induces environmental
200. Brack, A. S. et al. Increased Wnt signaling during 222. Berent-​Maoz, B., Montecino-​Rodriguez, E., Signer, R. A. J. alterations limiting hematopoietic stem cell function
aging alters muscle stem cell fate and increases & Dorshkind, K. Fibroblast growth factor-7 partially and engraftment. Nat. Med. 13, 742–747 (2007).
fibrosis. Science 317, 807–810 (2007). reverses murine thymocyte progenitor aging by 243. Passos, J. F. et al. Feedback between p21 and reactive
201. Yang, L. et al. Rho GTPase Cdc42 coordinates repression of Ink4a. Blood 119, 5715–5721 (2012). oxygen production is necessary for cell senescence.
hematopoietic stem cell quiescence and niche 223. Al Alam, D. et al. Fibroblast growth factor 10 alters Mol. Syst. Biol. 6, 347 (2010).
interaction in the bone marrow. Proc. Natl Acad. the balance between goblet and Paneth cells in the 244. Mosteiro, L. et al. Tissue damage and senescence
Sci. USA 104, 5091–5096 (2007). adult mouse small intestine. Am. J. Physiol. provide critical signals for cellular reprogramming
202. Florian, M. C. et al. Cdc42 activity regulates Gastrointest. Liver Physiol. 308, G678–G690 (2015). in vivo. Science 354, aaf4445 (2016).
hematopoietic stem cell aging and rejuvenation. 224. Spence, J. R. et al. Directed differentiation of human 245. Demaria, M. et al. An essential role for senescent cells
Cell Stem Cell 10, 520–530 (2012). pluripotent stem cells into intestinal tissue in vitro. in optimal wound healing through secretion of
203. Famili, F. et al. Discrete roles of canonical and Nature 470, 105–109 (2011). PDGF-AA. Dev. Cell 31, 722–733 (2014).
non-canonical Wnt signaling in hematopoiesis and 225. Vidrich, A. et al. Fibroblast growth factor receptor-3 246. Schwitalla, S. et al. Intestinal tumorigenesis initiated
lymphopoiesis. Cell Death Dis. 6, e1981 (2015). regulates Paneth cell lineage allocation and accrual of by dedifferentiation and acquisition of stem-​cell-like
204. Florian, M. C. et al. A canonical to non-​canonical Wnt epithelial stem cells during murine intestinal properties. Cell 152, 25–38 (2013).
signalling switch in haematopoietic stem-​cell ageing. development. Am. J. Physiol. Gastrointest. Liver 247. You, J. S. & Jones, P. A. Cancer genetics and
Nature 503, 392–396 (2013). Physiol. 297, G168–G178 (2009). epigenetics: two sides of the same coin? Cancer Cell
205. Povinelli, B. J. & Nemeth, M. J. Wnt5a regulates 226. Liu, D.-W., Tsai, S.-M., Lin, B.-F., Jiang, Y.-J. & 22, 9–20 (2012).
hematopoietic stem cell proliferation and repopulation Wang, W.-P. Fibroblast growth factor receptor 2c 248. Krivtsov, A. V. & Armstrong, S. A. MLL translocations,
through the Ryk receptor. Stem Cells 32, 105–115 signaling is required for intestinal cell differentiation in histone modifications and leukaemia stem-​cell
(2014). zebrafish. PLoS ONE 8, e58310 (2013). development. Nat. Rev. Cancer 7, 823–833 (2007).
206. Nemeth, M. J., Topol, L., Anderson, S. M., Yang, Y. & 227. Park, J.-S., Kim, Y.-S. & Yoo, M.-A. The role of p38b 249. Yang, L., Rau, R. & Goodell, M. A. DNMT3A in
Bodine, D. M. Wnt5a inhibits canonical Wnt signaling MAPK in age-​related modulation of intestinal stem cell haematological malignancies. Nat. Rev. Cancer 15,
in hematopoietic stem cells and enhances proliferation and differentiation in Drosophila. Aging 152–165 (2015).
repopulation. Proc. Natl Acad. Sci. USA 104, 1, 637–651 (2009). 250. Wallace, D. C. Mitochondrial DNA mutations in
15436–15441 (2007). 228. Avgustinova, A. & Benitah, S. A. Epigenetic control of disease and aging. Environ. Mol. Mutagen. 51,
207. Sugimura, R. et al. Noncanonical Wnt signaling adult stem cell function. Nat. Rev. Mol. Cell. Biol. 17, 440–450 (2010).
maintains hematopoietic stem cells in the niche. Cell 643–658 (2016). 251. Parker, S. J. & Metallo, C. M. Metabolic consequences
150, 351–365 (2012). 229. Ocampo, A. et al. In vivo amelioration of age-​ of oncogenic IDH mutations. Pharmacol. Ther. 152,
208. Tao, S. et al. Wnt activity and basal niche associated hallmarks by partial reprogramming. Cell 54–62 (2015).
position sensitize intestinal stem and progenitor 167, 1719–1733.e12 (2016). 252. Lahtz, C. & Pfeifer, G. P. Epigenetic changes of DNA
cells to DNA damage. EMBO J. 34, 624–640 230. Yin, H., Price, F. & Rudnicki, M. A. Satellite cells and the repair genes in cancer. J. Mol. Cell. Biol. 3, 51–58 (2011).
(2015). muscle stem cell niche. Physiol. Rev. 93, 23–67 (2013). 253. Nazemalhosseini Mojarad, E., Kuppen, P. J., Aghdaei,
209. Scaffidi, P. & Misteli, T. Lamin A-​dependent 231. Nuschke, A., Rodrigues, M., Wells, A. W., Sylakowski, K. H. A. & Zali, M. R. The CpG island methylator
misregulation of adult stem cells associated with & Wells, A. Mesenchymal stem cells/multipotent phenotype (CIMP) in colorectal cancer. Gastroenterol.
accelerated ageing. Nat. Cell Biol. 10, 452–459 stromal cells (MSCs) are glycolytic and thus glucose is Hepatol. Bed Bench 6, 120–128 (2013).
(2008). a limiting factor of in vitro models of MSC starvation. 254. Ogino, S. et al. CpG island methylator phenotype
210. Espada, J. et al. Nuclear envelope defects cause stem Stem Cell Res. Ther. 7, 179 (2016). (CIMP) of colorectal cancer is best characterised by
cell dysfunction in premature-​aging mice. J. Cell Biol. 232. Zhang, J. et al. UCP2 regulates energy metabolism quantitative DNA methylation analysis and prospective
181, 27–35 (2008). and differentiation potential of human pluripotent cohort studies. Gut 55, 1000–1006 (2006).
211. Cairney, C. J. et al. A systems biology approach to stem cells. EMBO J. 30, 4860–4873 (2011). 255. Alabert, C. et al. Nascent chromatin capture
Down syndrome: Identification of Notch/Wnt 233. Takubo, K. et al. Regulation of the HIF-1alpha level is proteomics determines chromatin dynamics during
dysregulation in a model of stem cells aging. essential for hematopoietic stem cells. Cell Stem Cell DNA replication and identifies unknown fork
Biochim. Biophys. Acta 1792, 353–363 (2009). 7, 391–402 (2010). components. Nat. Cell Biol. 16, 281–293 (2014).
212. Meena, J. K. et al. Telomerase abrogates aneuploidy-​ 234. Maryanovich, M. et al. An MTCH2 pathway repressing 256. Stamatoyannopoulos, J. A. et al. Human mutation rate
induced telomere replication stress, senescence and mitochondria metabolism regulates haematopoietic associated with DNA replication timing. Nat. Genet.
cell depletion. EMBO J. 34, 1371–1384 (2015). stem cell fate. Nat. Commun. 6, 7901 (2015). 41, 393–395 (2009).
213. Conboy, I. M., Conboy, M. J., Smythe, G. M. & 235. Rocheteau, P., Gayraud-​Morel, B., Siegl-​Cachedenier, I., 257. Blokzijl, F. et al. Tissue-​specific mutation accumulation
Rando, T. A. Notch-​mediated restoration of Blasco, M. A. & Tajbakhsh, S. A. Subpopulation of in human adult stem cells during life. Nature 538,
regenerative potential to aged muscle. Science 302, adult skeletal muscle stem cells retains all template 260–264 (2016).
1575–1577 (2003). DNA strands after cell division. Cell 148, 112–125
214. Sousa-​Victor, P. et al. Geriatric muscle stem cells (2012). Acknowledgements
switch reversible quiescence into senescence. Nature 236. Moussaieff, A. et al. Glycolysis-​mediated changes in F.N. is supported by the Alexander von Humboldt Foundation
506, 316–321 (2014). acetyl-​CoA and histone acetylation control the early and the German Federal Ministry for Education and Research.
215. Duncan, A. W. et al. Integration of Notch and Wnt differentiation of embryonic stem cells. Cell Metab. K.L.R. is supported by a European Research Council
signaling in hematopoietic stem cell maintenance. 21, 392–402 (2015). Advanced Grant (StemCellGerontoGenes). The Fritz Lipmann
Nat. Immunol. 6, 314–322 (2005). 237. Imai, S. & Guarente, L. NAD+ and sirtuins in aging and Institute is a member of the Leibniz Association and is finan-
216. Janzen, V. et al. Stem-​cell ageing modified by the disease. Trends Cell Biol. 24, 464–471 (2014). cially supported by the federal government of Germany and
cyclin-​dependent kinase inhibitor p16INK4a. Nature 238. Choudhury, A. R. et al. Cdkn1a deletion improves the State of Thuringia.
443, 421–426 (2006). stem cell function and lifespan of mice with
217. Chen, K.-Y. et al. A Notch positive feedback in the dysfunctional telomeres without accelerating cancer Author contributions
intestinal stem cell niche is essential for stem cell formation. Nat. Genet. 39, 99–105 (2007). M.E., F.N., A.O. and K.L.R. contributed equally to writing,
self-renewal. Mol. Syst. Biol. 13, 927 (2017). 239. Baker, D. J. et al. Naturally occurring p16Ink4a-​positive revising, researching and discussing the article.
218. Siudeja, K. et al. Frequent somatic mutation in adult cells shorten healthy lifespan. Nature 530, 184–189
intestinal stem cells drives neoplasia and genetic (2016). Competing interests
mosaicism during aging. Cell Stem Cell 17, 240. Chang, J. et al. Clearance of senescent cells by The authors declare no competing interests.
663–674 (2015). ABT263 rejuvenates aged hematopoietic stem cells in
219. van Es, J. H. et al. Notch/γ-​secretase inhibition turns mice. Nat. Med. 22, 78–83 (2016). Publisher’s note
proliferative cells in intestinal crypts and adenomas References 238 and 239 report that the depletion Springer Nature remains neutral with regard to jurisdictional
into goblet cells. Nature 435, 959–963 (2005). of senescent cells in aged mice improves stem cell claims in published maps and institutional affiliations.

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

Vous aimerez peut-être aussi