You are on page 1of 11

f o c u s o nRa

EVg Iei
EWng
S

stem cells

How stem cells age and why


this makes us grow old
Norman E. Sharpless* and Ronald A. DePinho‡
Abstract | Recent data suggest that we age, in part, because our self-renewing stem cells
grow old as a result of heritable intrinsic events, such as DNA damage, as well as extrinsic
forces, such as changes in their supporting niches. Mechanisms that suppress the
development of cancer, such as senescence and apoptosis, which rely on telomere
shortening and the activities of p53 and p16INK4a, may also induce an unwanted consequence:
a decline in the replicative function of certain stem-cell types with advancing age.
This decreased regenerative capacity appears to contribute to some aspects of mammalian
ageing, with new findings pointing to a ‘stem-cell hypothesis’ for human age-associated
conditions such as frailty, atherosclerosis and type 2 diabetes.

Forkhead transcription Diverse acquired and genetic factors drive the com- Recent evidence supports the model that stem cells
factor class O plex cellular and organismal process of mammalian in several tissues are largely retained in a quiescent state
(FOXO). One of a family of ageing. The process appears to be hastened by envi- but can be coaxed back into the cell cycle in response
evolutionarily conserved
ronmental and behavioural factors including obesity, to extracellular cues, even after prolonged periods
transcription factors that are
linked to lifespan regulation in
diabetes, end-stage renal disease and exposure to of dormancy. Once stimulated to divide, stem cells
lower systems and to stem-cell mutagens such as chemotherapy, ultraviolet light and yield undifferentiated progeny, which in turn produce
maintenance in mice. The tobacco smoke. Likewise, various genetic lesions that differentiated effector cells through subsequent rounds
FOXO proteins are thought to alter DNA metabolism and nuclear architecture have of proliferation (BOX 1). This ‘hierarchical’ differentia-
exert these effects by
been associated with progeroid syndromes in humans tion scheme makes sense from the perspective of organ-
regulating the expression of
genes involved in apoptosis, such as ataxia telangiectasia, Werner syndrome ismal longevity — it permits the production of large
proliferation, glucose and Hutchinson–Gilford syndrome 1. Most comp­ numbers of differentiated cells from a single stem cell
metabolism, DNA repair, elling have been genetic studies in yeast, Drosophila by combining subsequent steps in differentiation with
regulation of reactive oxygen
melanogaster, Caenorhabditis elegans and mice, which proliferation5,6. Therefore, this approach balances the
species and other diverse
cellular processes.
have established crucial roles for the insulin–insulin- high rates of homeostatic proliferation that are required
like growth factor-1 (IGF1)–AKT–forkhead transcription in tissues like the bone marrow and intestine with the
factor class O (FOXO) pathway (see the Review by long-term need to protect stem cells from mutagenic
*Departments of Medicine
and Genetics, The Lineberger Russell and Kahn in this issue) and oxygen-radical insult and carcinogenesis. Indeed, under homeostatic
Comprehensive Cancer Center, regulators (see the Opinion article by Pelicci and conditions, there is limited proliferative demand on
The University of North colleagues in this issue) as determinants of lifespan the self-renewing stem cells themselves and so these
Carolina, Chapel Hill, North in these model systems. Although these disparate cells divide infrequently, sparing stem cells the perils
Carolina 27599‑7295, USA.

Center for Applied Cancer
experimental observations suggest common molecular of DNA replication and mitosis. Additionally, as stem
Science of the Belfer Institute themes, human ageing in physiological terms remains cells appear to be less metabolically active in their qui-
for Innovative Cancer Science, an enigma. escent state, they may be subjected to lower levels of
and Department of Medical In this Review, we suggest that some aspects of DNA-damage-inducing metabolic side products such
Oncology, Dana-Farber
mammalian ageing result from an age-associated as reactive oxygen species (ROS)7.
Cancer Institute; Departments
of Medicine and Genetics, decline in the replicative function of certain tissue- Once believed to be restricted only to ‘high-turnover’
Harvard Medical School, specific regenerative cells — adult stem cells (FIG. 1). organs like the bone marrow and intestine, resident self-
Boston, Massachusetts These rare and specialized cells are required for renewing cells are now thought to have a significant role
02115, USA. tissue replacement throughout the human lifespan, in the homeostatic maintenance of many organs, includ-
e-mails:
ron_depinho@dfci.harvard.
and appear to be characterized by a few specific ing organs with lower turnover rates such as the brain
edu; nes@med.unc.edu physio­logical and biochemical properties (reviewed and pancreatic islets8. Although there is disagreement
doi:10.1038/nrm2241 in Refs 2–4), particularly the capacity for self-renewal. as to the significance of such cells in these low-turnover

nature reviews | molecular cell biology volume 8 | september 2007 | 703


© 2007 Nature Publishing Group
REVIEWS

Stem cell Progenitors Effectors kinase inhibitor p16INK4a and the tumour suppressor
p53 exert their pro-ageing effects in part through their
activation in specific self-renewing compartments such
Young as tissue-specific stem cells. Further, we describe findings
from a series of recently published human association
analyses that suggest a link between the INK4/ARF locus
(also called CDKN2a and CDKN2b) and the onset of
distinct human age-associated phenotypes. These recent
Physiological ageing, mutagen
exposure or forced regeneration observations, together with the findings in genetic model
systems, provide experimental support for the concept
that the activation of tumour-suppressor mechanisms in
self-renewing compartments contributes to the ageing
Old processes in humans.

Do stem cells age?


A decline in replicative function with age is appreciable
in many mammalian tissues. In most mammalian tissues,
however, the inability to purify the resident stem cell to
Figure 1 | How stem cells age. Stem-cell number and self-renewal (curved arrow) does homogeneity, as well as the lack of adequate models
not necessarily decline with ageing, but function —Nature Reviews
the ability | Molecular
to produce Cell Biology
progenitors
to test the function of these cells, has made it difficult to
(blue) and differentiated effector cells (depicted in different colours) — does decline.
Stem-cell ageing can result in some systems from the heritable accumulation of DNA determine if a decline in stem-cell function is indeed a
damage, which can engage tumour-suppressor activation as the stem cell attempts to cause of the degradation of the regenerative capacities
divide asymmetrically. DNA damage can occur stochastically with normal ageing as a that is seen in many organs with ageing.
result of exposure to external mutagens, or from increased proliferation as in forced
regeneration (see main text). Ageing of haematopoietic stem cells. In the haemato­
poietic system, it is possible to purify haematopoietic
stem cells (HSCs) to near-homogeneity and assay their
organs in the adult human, their importance in rodents function using validated assays. Therefore, the ques-
has been demonstrated experimentally. It is therefore tions of whether and how stem cells age are presently
reasonable to surmise that perhaps some characteristics best addressed in this system, with the caveat that stem-
of ageing — once thought to be degenerative — might cell ageing may differ mechanistically in other tissues.
reflect a decline in the regenerative capacity of resident Several effects of ageing on the blood organ are described
stem cells across many different tissues. in humans: decreased immunity11, increased incidence of
Self-renewal comes with some danger for the organ- bone marrow failure and haematological neoplasia12, and
ism; in particular, a risk of malignant transformation4,9,10. moderate anaemia13,14. Older individuals are more likely
Unrepaired genetic lesions in stem cells are passed on to suffer toxicity such as prolonged myelosuppression in
to their self-renewing daughters and accumulate with response to traditional cytotoxic chemotherapy drugs,
ageing in this way. Functional mutations that provide a suggesting a reduced marrow regenerative capacity15–17.
growth or survival advantage in turn produce positive Particularly telling is the clinical observation that
selection for the mutant stem-cell clone, with full-fledged increased donor age in bone marrow transplantation is a
Self-renewal
The capacity of replicating
cancer resulting from the accumulation of multiple predictor of transplant-related mortality18–22, suggesting
stem cells to generate cancer-promoting events. To offset this possibility, stem that the diminished reconstituting ability of HSCs from
daughter cells with the same cells appear to have evolved multiple reinforcing mecha- elderly donors is partly cell-autonomous. These observa-
biological and molecular profile nisms that are aimed at maintaining genomic integrity tions suggest a clinically overt decay in HSC function
that endows continued renewal
beyond that of other proliferating cells (reviewed in with normal human ageing.
potential. This can occur either
asymmetrically when a stem Refs 2–4). When mutations occur despite these error- These correlative findings in humans are buttressed
cell produces another stem cell prevention capacities, potent tumour-suppressor mecha- by related studies in rodents. A surprising finding
and a more differentiated nisms such as senescence and apoptosis exist to sense has been that although HSC function clearly declines
daughter cell, or symmetrically damaged stem-cell genomes with malignant potential with age, the number of HSCs does not necessarily
when stem-cell division gives
rise to two identical stem cells.
and limit replicative expansion or cull such clones. This also decline. In some strains of mice, the HSC number
Importantly, in mature organ relationship between self-renewing cells and cancer actually expands with advancing age7,23–27 and this
systems, most cell-division raises the possibility that — while carrying out a benefi- age-dependent expansion of HSCs is a transplantable,
activity that is responsible for cial, anti-cancer function — these tumour-suppressor cell-autonomous property of HSCs7,25. Moreover, as
tissue maintenance and
mechanisms may inadvertently contribute to ageing by demonstrated by Harrison and colleagues, HSCs can be
expansion is not self-renewing.
causing stem-cell arrest or attrition. serially transplanted into sequential recipients and show
Progeny Here, we discuss recent refinements of this so-called persistent function for >8 years, thus exceeding the life-
Along with progenitor cells, ‘cancer–ageing hypothesis’, according to which cells time of the original donor animal28. These experiments
these are relatively within a tissue are compromised by these anti-cancer have established that cell-autonomous, replicative HSC
undifferentiated cell types that
are derived from asymmetric
mechanisms (see also the Opinion article by Serrano and exhaustion does not necessarily occur during periods of
stem-cell division and lack the Blasco in this issue). Specifically, we reason that growth- normal ageing in some strains of inbred mice that are
capacity to self-renew. inhibitory molecules such as the cyclin-dependent maintained under laboratory conditions.

704 | september 2007 | volume 8 www.nature.com/reviews/molcellbio


© 2007 Nature Publishing Group
f o c u s o nRa
EVg Iei
EWng
S

Box 1 | The hierarchy of haematopoiesis appear to decline with age in other inbred strains of
mice 26,27,29–31, and that several external stimuli (for
Self-renewal example, chemotherapy, ionizing radiation and so on)
LT-HSC hasten stem-cell exhaustion in humans and mice32–38.
Stem cells (LIN– KIT+ SCA1+ CD34Lo FLK2Lo)
Thus, genetically outbred mammals in the wild may
ST-HSC
or may not experience HSC exhaustion with ageing,
(LIN– KIT+ SCA1+ CD34Hi FLK2Lo) and exposure to environmental stresses that are not
Multipotent normally encountered in the laboratory setting may
progenitors further induce HSC exhaustion. Finally, independently
MPP
(LIN– KIT+ SCA1+ CD34Hi FLK2Hi) of replicative function, HSCs exhibit cell-intrinsic,
functional signs of ageing. Numerous studies have
shown that ageing alters HSC function with regard to
mobilization39, homing7,23,24,31,39 and lineage choice7,24,31.
In particular, there is a loss of lymphoid lineage poten-
CMP CLP tial with a skewing toward myeloid lineages in HSCs
Oligopotent from old mice, and old HSCs demonstrate reproduc-
progenitors ible changes in gene expression with age, including
increased expression of myeloid lineage transcripts7.
MEP GMP Therefore, the preponderance of evidence suggests
Lineage- that HSCs undergo cell-intrinsic ageing, although
restricted there is also emerging evidence that the ageing HSC
progenitors micro­environment may influence HSC function in an
extrinsic manner (see below).
Effector
cells
Ageing in other self-renewing compartments. Several
Erythrocytes Platelets Granulocytes Macrophages T cells B cells lines of evidence suggest that stem cells from other
Multipotent tissue-specific stem cells produce differentiated effector cells through a tissues suffer proliferative decline with advancing
Nature Reviews | Molecular Cell Biology
series of increasingly more committed progenitor intermediates. This tissue stem-cell- age (FIG. 1). For example, in rodents, a decline in the
derived differentiation process has been best characterized in the haematopoietic number of new neurons produced by neural stem cells
system (see figure). Here, long-term haematopoietic stem cells (LT-HSCs) represent the (NSCs) with age or telomere dysfunction has been docu-
‘true’ stem cells that self-renew and produce multipotent progenitors (short-term (ST)- mented40,41, as has the in vivo proliferation of NSCs41–43.
HSCs and, subsequently, multipotent progeny (MPP)) with no self-renewal capacity. This decline in the capacity for murine neurogenesis
These in turn give rise to oligopotent progenitors including the common lymphoid has been associated with a progressive Parkinsonian
progenitor (CLP) and common myeloid progenitor (CMP), which then yields the disease41 and with an impairment of olfactory discrimin­
granulocyte–macrophage progenitor (GMP) that differentiates into monocytes,
ation with ageing44. Likewise, hair greying has been
macrophages and granulocytes, and the megakaryocyte–erythrocyte progenitor (MEP)
that differentiates into megakaryocytes, platelets and erythrocytes. Similar hierarchies
linked to decreased melanocyte stem-cell maintenance,
are also thought to exist in other stem-cell-containing tissues (for example, brain, gut, possibly in association with melanoblast senescence45.
liver and lung). The capacity of the insulin-producing β‑cell of the
A principal advantage of studies in the haematopoietic system is that stem and pancreatic islet to replicate in adult rodents throughout
progenitor cells from this organ can be purified to near-homogeneity by surface life has been known for decades, although the importance
markers. For example, LT‑HSCs express low levels of lineage markers (LIN–), high levels of this regenerative capacity with regard to the develop-
of the CD117/c-KIT receptor (KIT+), high levels of a surface marker called SCA1 (SCA1+), ment of type 2 diabetes mellitus (also called adult-onset
and low levels of another surface marker, CD34 (CD34Lo). With each asymmetric diabetes mellitus) has only recently been appreciated.
division of a LIN –KIT+SCA1+CD34Lo LT‑HSC, there is the production of another LT‑HSC The prevailing view had been that type 2 diabetes mell­
and a multipotent daughter cell with limited renewal potential, ST‑HSC, which has a
itus was a metabolic disease resulting largely from an
similar surface immunophenotype to LT‑HSC except that it has higher levels of CD34
(CD34Hi). As ST‑HSC in turn proliferate to form more differentiated MPP, they increase
age-associated decline in the ability of muscle and liver to
expression of another surface marker, FLK2. Development from oligopotent respond to insulin (insulin resistance), but recent careful
progenitors to mature blood cells proceeds through several intermediate progenitors analyses of islet mass have challenged this view. Studies
that are not shown. in the immediate post-mortem period have shown sig-
nificant rates of β‑cell production and apoptosis, even
in aged adults, with an increased β‑cell mass noted in
This is not to say, however, that the replicative func- obese individuals and a relatively reduced β‑cell mass
tion of HSCs is not limited by anti-cancer mechanisms among adults with diabetes46,47. Therefore, the de novo
with ageing. It is entirely possible that senescent HSCs synthesis of β‑cells through self-renewal appears to be
change their surface immunophenotype (and therefore the predominant source of islet mass in adult humans.
are no longer identified as HSCs by flow cytometry) or Islet replication appears to decline with human ageing47,
that the senescence and apoptosis mechanisms are not although β‑cell replication has been reported in human
engaged until old HSCs attempt to divide asymmetri- patients up to 89 years of age48. Consistent with these
cally. In the latter model, senescence or apoptosis could findings in humans, a sharp decline in β‑cell prolifera-
limit HSC function without decreasing the HSC num­ tion with ageing in mice has been recently described49.
ber. Moreover, it is important to note that HSC numbers More than 1% of islet cells from young mice proliferate

nature reviews | molecular cell biology volume 8 | september 2007 | 705


© 2007 Nature Publishing Group
REVIEWS

1.4 satellite-cell proliferation suggests that cell-intrinsic


p16INK4a knockout ageing of some other tissue occurs such that it elabo-

Proliferating islet cells (% of total islet cells)


1.2 rates (or fails to elaborate) the relevant serum factor that
modulates the proliferative capacity of satellite cells. The
1.0 ability to target genes conditionally in a tissue-specific
Senescence and temporal manner will permit in vivo analyses to
A specialized form of growth
0.8 determine which compartments age intrinsically, versus
arrest induced by various Wild-type those that age ‘by proxy’.
stressful stimuli including loss
of telomere function, reactive 0.6
oxygen species, some forms of What causes cells to age?
DNA damage and activation of Although a few mechanisms have been suggested to
0.4
certain oncogenes or explain cellular ageing, independent lines of evidence
reactivation of tumour- suggest that forms of DNA damage lead to the activation
suppressor genes. Senescence p16INK4a transgenic
0.2 of tumour-suppressor mechanisms, such as senescence,
is characterized by several
markers such as senescence- to limit stem-cell function with increasing age.
associated‑β-galactosidase,
20 40 60 80
alterations in chromatin DNA damage results in stem-cell attrition. The accu-
structure (senescence- Mouse age (weeks)
mulation of damage to cellular macromolecules (for
associated heterochromatic Figure 2 | Proliferation of β-cells
foci) and a marked increase in Nature Reviewswith age. ACell
| Molecular sharp
Biology example, proteins and DNA) has been postulated to
decline in pancreatic β‑cell proliferation with ageing
the secretion of several be a cause of cellular attrition with ageing52. DNA is
normally occurs in wild-type mice (blue). This decline is
cytokines and other bioactive subjected to spontaneous and extrinsic mutational
molecules (senescence-
modulated by expression of the p16INK4a tumour
suppressor: increased expression (in a transgenic line; events on a daily basis, and despite a formidable cap­
associated secretory
phenotype). green) correlates with reduced proliferation, whereas acity for repair, some damaged DNA appears to evade
decreased activity (in the p16INK4a knockout line; orange) repair and accumulates over time. Evidence in support
Tissue-specific stem cell affords a resistance to β‑cell ageing. of the notion that DNA damage attenuates stem-cell
A specialized cell found in function with age has been provided by the study of
many tissues of adults. These
HSCs from mice that harbour alterations in the DNA-
cells can replace themselves
through self-renewal and are under steady-state conditions, but this frequency damage response. Significant functional defects are seen
generally multipotent, in that declines by nearly tenfold after a year of ageing (FIG. 2). in HSCs from mice that are deficient in DNA-repair
they can give rise to progeny Therefore, rather than being merely a disease of insulin proteins such as FANCD1(Ref. 53), MSH2 (Ref. 54) or
that can differentiate into resistance, type 2 diabetes mellitus partly results from a ERCC1 (Ref. 55). A mouse strain with a viable, hypo-
multiple different cell types of
the associated organ.
relative failure of islet replication. morphic allele of the DNA-repair protein DNA ligase IV
that was recently identified through a mutagenesis
Multipotency Cell-extrinsic ageing. These observations, however, do screen exhibits a marked, age-induced decline in HSC
The ability to give rise to not make clear which islet-associated compartment is number and function56. Additionally, Morales et al. have
differentiated progeny of
ageing: is it the β‑cells themselves, a putative pancreatic shown that mice with a mutant allele of RAD50, a mem-
different specialized subtypes.
However, some self-renewing stem cell or another tissue that influences β‑cell prolifer­ ber of the MRE11 DNA-repair complex, demonstrate
cells (for example, pancreatic ation in a cell non-autonomous manner? In fact, cell profound bone marrow hypoplasia57. This effect can be
β‑cells) have a narrow potential non-autonomous factors have been shown to have a rescued in the setting of ataxia-telangiectasia mutated
for differentiation, generating major role in the regulation of stem-cell ageing in other (ATM) kinase deficiency, suggesting that the Rad50
progeny similar to the parental
cell. This type of self-renewing
systems. Rando and colleagues have shown that the allele is hypermorphic and that excess DNA-damage
cell is termed a ‘unipotent ageing of a regenerative cell of muscle, the satellite cell, signalling reduces HSC number and/or function.
progenitor’, which can be is influenced by the aged microenvironment50. Satellite These data suggest that several forms of DNA repair
viewed as a special stem-cell cells from aged mice were ‘rejuvenated’ by exposure to are needed to maintain HSC genomic integrity, and that
subtype, at least in terms of
a young blood supply, suggesting that the aged milieu, activation of a response to DNA damage compromises
long-term proliferative
capacity. For convenience, in hormonal or otherwise, influences the replicative func- HSC function.
this Review the term ‘stem cell’ tion of satellite cells. Likewise, while some aspects of the A demonstration of DNA damage in a tissue-
is applied to both types of functional decline that characterizes ageing of HSCs has specific stem-cell compartment with ageing comes from
adult self-renewing cells. been shown to be cell-autonomous, the HSC niche is a recent report by Rossi and colleagues58. Using mice
Telomere
also expected to influence HSC ageing to some degree. with germline deficiencies in DNA repair or telomere
A nucleoprotein complex at For example, in mice with shortened telomeres, an metabolism (Ku80-, XPD- and mTERC-knockout mice),
the end of chromosomes that age-dependent decrease was noted in the ability of the authors demonstrated a marked premature decline
maintains chromosomal recipient animals to support lymphopoiesis of trans- in the regenerative function per HSC in several assays.
integrity. It consists of many
planted HSCs from young mice with normal telomeres, This study further noted the increased expression of
double-stranded 5′-TTAGGG-3′
repeats, a 3′-single-stranded and enhanced myeloid proliferation occurred when markers of the DNA-damage response (for example,
overhang and associated HSCs were transplanted into a microenvironment that histone H2AX foci) in highly purified HSCs with normal
telomere-binding proteins, harboured dysfunctional telomeres51. physio­logical ageing even in wild-type mice. The authors
which together generate a Although this distinction between cell-intrinsic and noted an increase in apoptosis of progeny from old HSCs,
capped structure that is
impervious to the actions of
cell-extrinsic ageing is important, ageing is, ultimately, the but not a decline in the ability of old HSCs to proliferate
complexes that repair DNA summative property of the organism in toto. For exam- in in vitro assays. These observations correspond well
damage. ple, the finding that serum from young mice enhances with a diminished capacity of the haematopoietic system

706 | september 2007 | volume 8 www.nature.com/reviews/molcellbio


© 2007 Nature Publishing Group
f o c u s o nRa
EVg Iei
EWng
S

Therefore, the excess regeneration that was forced to


Cancer occur to reconstitute proliferative tissues after transient
Atr inactivation in some proliferating cells produced
RAS mutation, a durable compromise of stem-cell function in Atr-
p53 loss Transformation
competent cells (FIG. 1), although it is important to
emphasize that the possible adverse effects of ATR
Regenerative failure, deletion on the stem-cell niche may also have contrib-
SA-SP
Telomere uted to the accelerated ageing. This result is consistent
Senescence
dysfunction with the view that such forced regeneration in response
Stem cell
to homeostatic demands, even in the absence of external
Tissue dysfunction DNA-damaging agents, can be toxic to stem cells across
and failure many organ systems.
Unrepaired DSBs
Apoptosis In the ‘DNA-damage accrual’ model of ageing,
unrepaired (or improperly repaired) genomic damage
Tissue dysfunction accumulates with ageing in stem-cell compartments.
and failure
Y-chromosome (for example, MDS)
At some point, accumulated damage perturbs normal
or 5q – loss Dysfunction stem-cell biology, driving stem cells to a few possible
Figure 3 | Fates of damaged stem cells. A limited number of outcomes appear to be fates: transformation, senescence, apoptosis or dysfunc-
possible for stem cells with heritable DNA damage.Nature Reviews
Although | Molecular
it is likely Cell Biology
that many tion; for example, a loss of the ability to robustly pro-
mutational events do not lead to any alteration of stem-cell function, significant duce progeny or an impaired potential for multilineage
damage is expected to induce apoptosis, senescence, transformation or dysfunction. differentiation (FIG. 3). As this process proceeds with time,
Importantly, these outcomes are not necessarily mutually exclusive; for instance, many depleted and/or dysfunctional stem-cell compartments
genetic events associated with stem-cell dysfunction (for example, the oncogenic cannot match the regenerative needs of a given organ
translocation that fuses BCR and ABL (forming the Tyr kinase BCR/ABL) in and homeostatic failure ensues. Likewise, if oncogenic
haematopoietic stem cells (HSCs)) are also associated with subsequent transformation. DNA-damage-induced lesions accumulate, self-renew-
Examples of DNA lesions associated with each outcome are indicated. Mutations in RAS
ing clones that contain such lesions undergo positive
and the tumour-suppressor p53 are associated with transformation, whereas loss of the
long arm of chromosome 5 (5q–) or the Y‑chromosome are associated with dysfunction selection, leading to cancer. Therefore, it is tempting to
of HSCs, manifesting as myelodysplasia (MDS). Unrepaired double-strand DNA breaks speculate that cancer and ageing are related endpoints
(DSBs) and telomere dysfunction induce apoptosis and senescence, respectively. of accumulating DNA damage within self-renewing
SA‑SP, senescence-associated secretory phenotype. compartments.

How telomere shortening could affect stem-cell ageing.


in telomere-dysfunctional mice to recover following There has been significant interest in the possibility that
chemotherapeutic challenge, as well as with a progres- telomere dysfunction, a specialized form of DNA dam-
sive neurodegenerative condition that is associated with age, contributes to the ageing of human stem cells. In the
decreased NSC reserves and function in the telomerase- absence of adequate telomerase activity, telomere short-
knockout model41,59. Together, these results are consist- ening inexorably occurs with proliferation, eventually
ent with the view that DNA damage accumulates with triggering a change in telomere structure that is sensed
ageing in the HSC compartment, as evidenced by the by the cell as a DNA double-strand break. In humans,
accumulation of H2AX foci, and that this damage can robust telomerase activity is predominantly restricted
be physiologically significant if unrepaired. to germ-cell compartments, some somatic early stem-
But what causes DNA damage in stem-cell com- cell or progenitor compartments and proliferating
partments in adult mice? Recent data from Ruzankina lymphocytes.
et al., who used a conditional allele for the DNA-dam- Whereas telomere dysfunction in the setting of an
age response gene Atr (ataxia telangiectasia and Rad3- intact DNA-damage response can serve as a potent
related), are worth considering in this regard60. Loss tumour-suppressor mechanism, the role of telomere-
of Atr is toxic to proliferating cells61 and when Atr was mediated checkpoints in stem-cell ageing remains an
somatically excised in adult mice in a widespread man- area of active investigation. Mice have long telomeres
ner by conditional inactivation, the vast majority of pro- relative to humans and, thus, telomere dysfunction
liferating cells rapidly disappeared, producing marked appears not to be a major cause of stem-cell exhaustion
intestinal atrophy and bone marrow hypoplasia 2 weeks in many strains of Mus musculus. Accordingly, in the
after conditional activation. However, the animals sur- telomerase-knockout mouse62, the lack of telomerase
vived this transient period of cell loss because rare stem activity per se produces a modest phenotypic effect in
cells that had not recombined the Atr allele replaced adult mice59,63. Murine telomere length can be made
Telomerase the lost cells. By 1 month after conditional activ­ation, the more limiting and reduced to a ‘humanized’ length by
A ribonucleoprotein complex mice appeared largely normal, with rapidly prolifer­ serial intercrossing of mice that are deficient in telo­
that extends the ends of ating tissues that had been fully reconstituted by spor­ merase activity. In this experimental setting, telomere
telomeres after replication by adic Atr-competent cells. Surprisingly though, these length becomes shorter with each successive generation
using telomerase reverse
transcriptase (TERT) and an
reconstituted mice then developed a marked progeroid until telomere dysfunction ensues with dramatic pheno-
RNA template (TERC) that is phenotype a few months later, with osteopaenia, greying typic effects, thus providing a model system to address
part of the enzyme complex. and loss of lymphoid and haematopoietic progenitors. the stem-cell issue.

nature reviews | molecular cell biology volume 8 | september 2007 | 707


© 2007 Nature Publishing Group
REVIEWS

a Senescence contributes to ageing. DNA damage and


telomere dysfunction appear to activate the classical
Exon 1 2 1β 1α 2 3
tumour-suppressor mechanisms of senescence and
Chromosome 9p21
apoptosis. Senescence requires activation of the retino­
DNA damage (for example, blastoma (RB) and/or p53 proteins and expression of
telomere dysfunction, ROS, their regulators, most prominently p16INK4a and ARF
stalled replication forks) (Refs 69–71) (FIG. 4a). The notion that senescence pre-
p15INK4b p16INK4a ARF
vents cancer is well-supported and is not controversial
ATM and/or ATR (reviewed in Refs 72,73). The expression of markers of
CDK4/6 MDM2 senescence such as senescence-associated β‑galactosidase
and p16INK4a markedly increases with ageing in many
CHK1 and/or CHK2
tissues from disparate mammalian species (reviewed in
Ref. 73; see also the Review by Campisi and D’Adda di
Fagagna in this issue). Caloric restriction (CR) potently
pRB family p21CIP p53 slows ageing in rodents, and CR and its related dietary
changes retard or even abolish the age-induced increase
in the expression of senescence markers, including the
expression of p16INK4a (Refs 74–76). Provocatively, CR,
Senescence Apoptosis similar to p16INK4a deficiency77, enhances stem-cell func-
b
tion with ageing78, which suggests the possibility that CR
Human 9p21 22,000k 22,100k may slow ageing in mammals by decreasing the activation
** * * * ** * ** of senescence in self-renewing compartments.
A role for ROS in senescence bears particular rel-
p16INK4a evance. ROS induces senescence in certain cell-culture
ARF systems79,80 but may also have an important role in effect-
ing the senescent phenotype81. In ATM-deficient mouse
p15INK4b
HSCs, increased ROS levels appear to compromise HSC
ANRIL function in part through a p38MAPK-dependent activation
of Ink4a/Arf expression34,35, which can be attenuated by
Region of linkage disequilibrium antioxidants. Likewise, HSCs that lack the FOXO trans­
Figure 4 | SNPs and age-related phenotypes at the INK4a/ARF/INK4b locus on cription factors show diminished self-renewal and pre-
human chromosome 9p21. a | Proteins encoded by Nature Reviews | locus
the INK4/ARF Molecular
on Cell Biology mature exhaustion under conditions of increased ROS
chromosome 9p21 regulate the p53 and retinoblastoma protein (pRB) tumour- production82. Therefore, FOXO transcription factors
suppressor pathways to promote senescence (p53 and pRB-family activation) or maintain HSC quiescence and survival, primarily via
apoptosis (p53 activation). Activation of p53 in response to several types of DNA damage
the regulation of physiological levels of ROS. While
also can occur via an ataxia-telangiectasia mutated (ATM)/ataxia telangiectasia and
Rad3-related (ATR)-mediated pathway independent of ARF function. b | Single FOXO-deficient HSCs show ROS-dependent enhanced
nucleotide polymorphisms that are significantly associated with the indicated cell-cycle entry and increased apoptosis, increased ROS
phenotypes are shown (frailty, green; vascular heart disease, red; type 2 diabetes may also result in the accumulation of DNA damage
mellitus, orange). The open reading frames for p16INK4a, ARF and p15INK4b are indicated, and unscheduled activation of senescence mechanisms
as is the ANRIL transcript. The large (>100 kb) region of linkage disequilibrium is also in this stem-cell compartment in the long term.
indicated. CDK4/6, cyclin-dependent kinases-4/-6; CHK1/2, checkpoint kinases-1/-2; Although the expression of senescence markers
MDM2, murine double minute-2, p53-binding protein; ROS, reactive oxygen species. is associated with ageing, this observation does not
establish a causal relationship between senescence and
ageing. Recent studies of self-renewal in HSCs, NSCs
Animals with dysfunctional telomeres develop fea- and pancreatic islet cells from p16INK4a-deficient and
tures of premature ageing caused by the activation of p16INK4a-overexpressing mice have begun to address this
senescence and apoptosis mechanisms in certain self- issue43,49,77,83. These results showed that increasing levels
renewing compartments such as HSCs, the intestinal of p16INK4a are not only associated with ageing, but partly
crypt and the testes59,63–66. Deficiency of p53 rescues many contribute to the age-induced replicative failure of these
of the stem-cell defects in mice that harbour defective tissues. In all three compartments, p16INK4a deficiency
telomeres, but does not extend the lifespan of these mice attenuated the age-induced decline in proliferation
because of increased tumorigenesis67,68. By contrast, loss and function. Likewise, overexpression of p16 INK4a
of p21CIP, a p53 transcriptional target that potently inhib- attenuated HSC function and islet proliferation in an
its the cell cycle (FIG. 4a), partially extends the longevity age-dependent manner (FIG. 2). The effects of p16INK4a
of mice with telomere dysfunction without increased loss were consistent across these disparate self-renewing
tumorigenesis, and attenuates some of the proliferative tissues, suggesting that p16INK4a can promote ageing
defects that are seen in various stem-cell compartments in tissues that are developmentally distinct. However,
of telomere-deficient mice66. Therefore, DNA dam- the loss of p16INK4a did not completely abrogate the
age induced by telomere shortening is partly sensed effects of ageing in any of the organs studied, indicat-
and managed through p53, and p53 exerts important ing that p16INK4a-independent ageing occurs in each of
anti-proliferative effects in stem cells through p21CIP. these compartments.

708 | september 2007 | volume 8 www.nature.com/reviews/molcellbio


© 2007 Nature Publishing Group
f o c u s o nRa
EVg Iei
EWng
S

Related observations have likewise suggested a pro- Importantly, these SNPs did not correlate with lipids,
ageing role for p53 and its effectors in mice66,84,85 and glycaemia or body mass index, implying that they
humans86. The case for p53, however, appears to be more in­fluence ASHD independently of these known meta-
complicated because p53 and its downstream effectors bolic characteristics. That is, the ASHD SNPs do not
such as p21CIP also have important roles in regulating the appear merely to be type 2 diabetes mellitus SNPs despite
DNA-damage response. In fact, an intriguing recent study the strong link between diabetes and vascular disease.
by Serrano and colleagues, who used carefully designed A mechanistic basis for this association has been supplied
transgenic strains, has shown that increased ARF and by several reports that suggest links between senescence
p53 activity can increase the murine lifespan by prevent- and age-related diseases of the cardiovascular system96–98.
ing cancer without an attendant increase in organismal Given these results, it will be interesting to determine the
ageing87. Additionally, HSCs from p21CIP‑deficient mice effects of p15INK4b, p16INK4a and ARF on atherosclerosis
demonstrate premature exhaustion88, consistent with in murine models.
the notion that a p53- and p21CIP-dependent cell-cycle Regulatory polymorphisms that reside >1 Mb from
pause in response to DNA damage may be important the related ORFs are well established99 and, therefore, the
for stem-cell longevity in vivo. These results suggest INK4/ARF locus appears well within the range of such
that p53 activation can be both pro-ageing and anti- regulatory elements. Although there has been a recent
ageing depending on the nature and duration of the stress description of an apparently non-protein-coding tran-
behind its activation. script in this region (ANRIL100; FIG. 4b), there does not
appear to be another protein-coding transcript near
Ageing and the INK4/ARF locus these SNPs. Therefore, in light of the murine genetic
The described links between stem-cell function and studies that link Ink4a/Arf and stem-cell function, pro-
ageing predict that individuals who differentially regu- teins encoded by the locus are the strongest candidates
late senescence-promoting mechanisms might exhibit to mediate the effects of these polymorphisms on the
different predispositions to cancer versus ageing. incidence of these common human diseases that are
Accordingly, hypomorphic alleles that affect p16INK4a or associated with ageing.
p53 function and that are associated with increased types
of cancer are well described (reviewed in Ref. 73). Recent Outstanding questions
data also suggest that non-coding polymorphisms near We believe that the data derived from disparate rodent
the open reading frames (ORFs) of p16INK4a, ARF and and human experimental systems support the view that
p15INK4b modify the onset of age-associated phenotypes a decline in the regenerative function of stem cells with
in humans. In a remarkable series of recent studies, age contributes to mammalian ageing and age-associated
single nucleotide polymorphisms (SNPs) that are very close disease. Given this model, a few crucial questions facing
(<120 kb) to the INK4/ARF locus were associated with the field remain.
frailty89, type 2 diabetes mellitus90–92 and vascular heart
disease93–95 (FIG. 4b). The identified SNPs may not be Does telomere dysfunction affect human ageing? An
the actual variants that convey disease susceptibility or important issue is whether sufficient telomere attrition
Single nucleotide
polymorphism resistance, and several classes of lesions (for example, occurs in human self-renewing compartments during
A common, single-base small insertions and deletions) would not have been physiological ageing to activate a DNA-damage response
difference in a gene among detected by the chosen approach. Different SNPs near and the subsequent compromise of replicative func-
individuals within a species. the locus have been associated with these phenotypes, tion. We feel that the current data do not yet provide
Frailty
and at least a few of the associated SNPs are not in linkage a clear answer. Humans who harbour short telomeres
A clinically validated, functional disequilibrium with each other, which suggests that more because of congenital deficiencies of components of the
measure used in clinical than one polymorphism near the locus influences these telo­merase complex (the RNA component TERC, telom-
geriatrics. It is scored as a ageing phenotypes. erase reverse transcriptase (TERT) or dyskerin) develop
continuous variable using a
Specifically, three research consortia that undertook an age-related failure of bone marrow or lung101–105.
series of routine, easily
measured tests such as gait genome-wide association studies across large, carefully Moreover, telomere shortening in the liver precedes the
speed. Frail individuals are less annotated patient samples have reported an association onset of liver failure (cirrhosis) in patients with chronic
able to live independently, are between type 2 diabetes mellitus and several SNPs near hepatitis106–109, an association that has been validated in
more likely to harbour co- p16INK4a (Refs 90–92). The INK4/ARF-associated SNPs the telomerase-deficient mouse110. A few studies have
morbid illnesses, and exhibit
increased mortality.
were independently identified and have common risk demonstrated a correlation between telomere length
allele frequencies and considerable effect sizes. This in peripheral blood lymphocytes (PBLs) and the onset
Linkage disequilibrium result suggests that expression of p16INK4a, ARF and/or of certain diseases that are associated with ageing. Such
(LD). A measure of genetic p15INK4b may be a significant determinant of suscepti- studies in non-neoplastic diseases have shown that PBL
associations between alleles at
bility for type 2 diabetes mellitus. This model fits with telomere lengths can provide a biomarker that forecasts
different loci, which indicates
whether allelic or marker the newly appreciated importance of islet replication the development of atherosclerosis111,112, cancer risk113,114
associations on the same in human type 2 diabetes mellitus and the effect of and mortality115. These results support a connection
chromosome are more p16 INK4a on islet regeneration in vivo 49. Also using between telomere dysfunction and human ageing.
common than expected. Loci unbiased, genome-wide approaches, three consortia By contrast, arguments can be made against telo­
are generally considered to be
in strong LD if their correlation
recently described SNPs near the INK4/ARF locus that mere-based ageing in humans. Murine ageing appears to
is higher than a pre-defined correlate with myocardial infarction and athero­sclerotic have many similarities to human ageing, yet we believe
cut-off (for example, 0.8). heart disease (ASHD) in large human cohorts93–95. that it occurs largely independently of telomere length.

nature reviews | molecular cell biology volume 8 | september 2007 | 709


© 2007 Nature Publishing Group
REVIEWS

Additionally, although telomere length shortens with To reconcile these observations, we favour the model
human ageing in many tissues, overall telomere length that the activation of p16INK4a and other senescence-pro-
does not linearly correlate with age, with the most rapid moting pathways (for example, ARF–p53, FOXO–ROS
shortening occurring by young adulthood102,105,116. It is and so on) are repressed in true stem-cell compartments,
possible, however, that mean telomere length is a poor but that such pathways may limit the function of stem
indicator of telomere status because the shortest telomere cells as they asymmetrically produce progeny. In accord-
appears to be capable of activating the DNA-damage ance with this hypothesis, diverse stem-cell compartments
response117. require the persistent ability of Polycomb group proteins
Although the observations of bone marrow failure such as BMI1 to repress the senescence-promoting activi-
and pulmonary fibrosis in patients with defective telo­ ties of the Ink4a/Arf locus121–125 (FIG. 4a). Absence of BMI1
merase components establishes that telomere dysfunc- does not lead to an increase in the numbers of hypofunc-
tion can cause human disease, some individuals from tional LT‑HSCs but instead causes a marked depletion
such kindreds who harbour the defective allele do not of LT‑HSCs, which is partly rescued by concomitant
demonstrate any overt phenotype. In fact, heterozygous Ink4a/Arf inactivation126. Therefore, Ink4a/Arf activation
TERT and TERC germline mutations in humans show in Bmi1-null mice does not produce ‘senescent’ LT‑HSCs,
strong ‘anticipation’ 102,104,105, which means that the at least on the basis of surface immunophenotype, but
phenotypic effects of the mutation are more severe instead decreases their frequency. Given this observation,
in subsequent generations; this is analogous to find- it is likewise not surprising that senescent LT‑HSCs do
ings in telomerase-deficient mice. Therefore, TERT or not accumulate with age, even though p16INK4a activation
TERC deficiency is most phenotypically pronounced in has a causal role in the decline of HSC function with
patients that also inherit fore-shortened telomeres from age77. Accordingly, the increase in p16INK4a expression in
their parents, suggesting that telomerase deficiency per se lineage-negative bone marrow cells from old mice prob-
may not be sufficient to induce overt age-associated ably reflects either the accumulation of senescent progeny
pathology. Importantly, however, studies of the unaf- or senescent cells that were formerly LT‑HSCs but have
fected carriers of these kindreds have so far not been changed their surface immunophenotype.
sufficient to exclude subtle, age-associated phenotypes,
and phenotypic consequences of telomerase deficiency Why are 9p21 SNPs linked to age-associated diseases? The
may be noted in such individuals with more careful recent findings from human association studies correlat-
observation. Considered in aggregate, we believe that the ing loci that are near the INK4a/ARF locus on chromo-
data are consistent with both telomere-independent and some 9 to the development of type 2 diabetes mellitus,
telomere-dependent ageing of human stem cells. ASHD and frailty are striking. Some might argue that
type 2 diabetes mellitus and ASHD are ageing-associated
How do senescence factors contribute to ageing? The diseases rather than ageing per se. Although this is an
simplest explanation for how p16INK4a limits replica- important distinction, we feel that any model of human
tive function with age would be that the induction of ageing that does not account for these causes of significant
p16INK4a, in response to senescence-promoting cues, human morbidity is less useful and, therefore, we believe
intrinsically limits replication by inducing senescence their potential association with p16INK4a, p15INK4b and/or
or at least decreasing cell-cycle entry. For example, ARF expression is of relevance in this discussion.
pancreatic β‑cell replication is known to require cyclin- It should be noted that, despite the data that support
dependent kinase-4 (CDK4) activity118,119, the biochemi- links between p16INK4a and age-associated declines in
cal target of p16INK4. Because p16INK4a accumulates with stem-cell function, it remains possible that these SNPs
physiological ageing in human and rodent islets49,120, do not correlate with altered regulation of INK4/ARF
and the loss of p16INK4a augments islet proliferation in products. It could be that there is an unappreciated
an age-dependent way49, p16INK4a might directly limit protein-coding ORF near the locus, that a non-protein
the replication of self-renewing cells with ageing. This coding transcript such as ANRIL (FIG. 4b) encodes regula-
cell-autonomous model is further supported by the fact tory RNAs, or that a polymorphism in this region exerts
that the enhanced function of p16INK4a-deficient HSCs is long-range (>1 Mb) regulation of another protein-coding
seen even when such cells are transplanted into p16INK4a- or microRNA transcript. Studies in humans and mice
competent recipient mice77,83. Importantly, however, to understand whether this correlation reflects an effect
p16INK4a expression increases significantly with ageing of INK4/ARF-based tumour-suppressor mechanisms on
in lineage-negative bone marrow cells (of which <0.1% human ageing are moving at a rapid pace. Such efforts
are true stem cells), but only modestly in highly purified are hampered, however, by a large region of linkage
long-term (LT)-HSCs (BOX 1) from old mice; LT‑HSCs disequilibrium that comprises the INK4a/ARF locus as
from old mice maintain the ability to replicate in vitro, well as almost all of the disease-correlated SNPs (FIG. 4b),
which suggests that they are not senescent24,58. Therefore, which implies that the relevant genetic regulatory events
although genetic approaches using p16INK4a-deficient and could be ‘hiding’ anywhere within a large region around
p21CIP-deficient mice have suggested an important effect p15INK4b. Regardless of the mechanistic basis, however, as
of these proteins in regulating HSC biology with age, the minor allele frequencies of the relevant SNPs in each
experiments that rely on the analysis of highly purified of these reports are large (>10%), these polymorphisms
LT‑HSC are not consistent with the accumulation of appear to have a major role in determining the onset of
senescent LT‑HSCs with age. these highly common, age-induced phenotypes.

710 | september 2007 | volume 8 www.nature.com/reviews/molcellbio


© 2007 Nature Publishing Group
f o c u s o nRa
EVg Iei
EWng
S

Conclusions apoptosis, which rely on telomere shortening and/or


In summary, we believe the data suggest that we grow p53 and p16INK4a activation, appear to promote ageing
old partly because our stem cells grow old as a result of just as their failure is associated with cancer. We believe
mechanisms that suppress the development of cancer that a further, more precise mechanistic understanding
over a lifetime. In this regard, our self-renewing stem of this process will be required before this knowledge
cells appear to grow old because of heritable intrinsic can be translated into human anti-ageing therapies. For
events, such as DNA damage, but also due to cell- the time being, the most prudent, clinically validated
extrinsic events such as alterations in their supporting advice appears still to be: don’t smoke, eat reasonably
niches. Anti-cancer mechanisms such as senescence and and take exercise.

1. Kudlow, B. A., Kennedy, B. K. & Monnat, R. J. Jr. 22. Yakoub-Agha, I. et al. Allogeneic marrow stem-cell proliferative defect in primitive hematopoietic stem
Werner and Hutchinson–Gilford progeria syndromes: transplantation from human leukocyte antigen- cells (PHSC) after chemotherapy. Exp. Hematol. 25,
mechanistic basis of human progeroid diseases. identical siblings versus human leukocyte 495–501 (1997).
Nature Rev. Mol. Cell Biol. 8, 394–404 (2007). antigen‑allelic‑matched unrelated donors (10/10) in 39. Xing, Z. et al. Increased hematopoietic stem cell
2. Bryder, D., Rossi, D. J. & Weissman, I. L. patients with standard-risk hematologic malignancy: mobilization in aged mice. Blood 108, 2190–2197
Hematopoietic stem cells: the paradigmatic tissue- a prospective study from the French Society of Bone (2006).
specific stem cell. Am. J. Pathol. 169, 338–346 Marrow Transplantation and Cell Therapy. J. Clin. 40. Kuhn, H. G., Dickinson-Anson, H. & Gage, F. H.
(2006). Oncol. 24, 5695–5702 (2006). Neurogenesis in the dentate gyrus of the adult rat:
3. Morrison, S. J. & Kimble, J. Asymmetric and 23. Morrison, S. J., Wandycz, A. M., Akashi, K., age-related decrease of neuronal progenitor
symmetric stem-cell divisions in development and Globerson, A. & Weissman, I. L. The aging of proliferation. J. Neurosci. 16, 2027–2033 (1996).
cancer. Nature 441, 1068–1074 (2006). hematopoietic stem cells. Nature Med. 2, 1011–1016 41. Wong, K. K. et al. Telomere dysfunction and ATM
4. Sharpless, N. E. & DePinho, R. A. Telomeres, stem (1996). deficiency compromises organ homeostasis and
cells, senescence, and cancer. J. Clin. Invest. 113, 24. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age- accelerates ageing. Nature 421, 643–648 (2003).
160–168 (2004). associated characteristics of murine hematopoietic An important study showing that the effects of
5. Hodgson, G. S. & Bradley, T. R. In vivo kinetic status of stem cells. J. Exp. Med. 192, 1273–1280 (2000). ATM loss seen in humans can be reproduced in
hematopoietic stem and progenitor cells as inferred 25. Pearce, D. J., Anjos-Afonso, F., Ridler, C. M., mice if ATM deficiency is combined with telomere
from labeling with bromodeoxyuridine. Exp. Hematol. Eddaoudi, A. & Bonnet, D. Age-dependent increase in dysfunction, which implies that the ataxia
12, 683–687 (1984). side population distribution within hematopoiesis: telangiectasia syndrome partly results from
6. Passegue, E., Wagers, A. J., Giuriato, S., implications for our understanding of the mechanism telomere dysfunction in ATM-deficient cells.
Anderson, W. C. & Weissman, I. L. Global analysis of of aging. Stem Cells 25, 828–835 (2006). 42. Maslov, A. Y., Barone, T. A., Plunkett, R. J. &
proliferation and cell cycle gene expression in the 26. de Haan, G., Nijhof, W. & Van Zant, G. Mouse strain- Pruitt, S. C. Neural stem cell detection,
regulation of hematopoietic stem and progenitor cell dependent changes in frequency and proliferation of characterization, and age-related changes in the
fates. J. Exp. Med. 202, 1599–1611 (2005). hematopoietic stem cells during aging: correlation subventricular zone of mice. J. Neurosci. 24,
7. Rossi, D. J. et al. Cell intrinsic alterations underlie between lifespan and cycling activity. Blood 89, 1726–1733 (2004).
hematopoietic stem cell aging. Proc. Natl Acad. 1543–1550 (1997). 43. Molofsky, A. V. et al. Increasing p16INK4a expression
Sci. USA 102, 9194–9199 (2005). 27. de Haan, G. & Van Zant, G. Dynamic changes in mouse decreases forebrain progenitors and neurogenesis
8. Weissman, I. L. Stem cells: units of development, hematopoietic stem cell numbers during aging. Blood during ageing. Nature 443, 448–452 (2006).
units of regeneration, and units in evolution. Cell 100, 93, 3294–3301 (1999). Shows that NSCs demonstrate a decrease in
157–168 (2000). 28. Harrison, D. E. Mouse erythropoietic stem cell lines replicative function with ageing that is decreased in
9. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. function normally 100 months: loss related to number the setting of p16INK4a, implying that p16INK4a
Stem cells, cancer, and cancer stem cells. Nature 414, of transplantations. Mech. Ageing Dev. 9, 427–433 activation contributes to NSC ageing.
105–111 (2001). (1979). 44. Enwere, E. et al. Aging results in reduced epidermal
10. Campisi, J. Cancer and ageing: rival demons? 29. Chen, J., Astle, C. M. & Harrison, D. E. Genetic growth factor receptor signaling, diminished olfactory
Nature Rev. Cancer 3, 339–349 (2003). regulation of primitive hematopoietic stem cell neurogenesis, and deficits in fine olfactory
11. Linton, P. J. & Dorshkind, K. Age-related changes in senescence. Exp. Hematol. 28, 442–450 (2000). discrimination. J. Neurosci. 24, 8354–8365 (2004).
lymphocyte development and function. Nature 30. Kamminga, L. M. et al. Impaired hematopoietic stem A careful, functional description of NSC ageing.
Immunol. 5, 133–139 (2004). cell functioning after serial transplantation and during 45. Nishimura, E. K., Granter, S. R. & Fisher, D. E.
12. Lichtman, M. A. & Rowe, J. M. The relationship of normal aging. Stem Cells 23, 82–92 (2005). Mechanisms of hair graying: incomplete melanocyte
patient age to the pathobiology of the clonal myeloid 31. Liang, Y., Van Zant, G. & Szilvassy, S. J. Effects of aging stem cell maintenance in the niche. Science 307,
diseases. Semin. Oncol. 31, 185–197 (2004). on the homing and engraftment of murine 720–724 (2005).
13. Beghe, C., Wilson, A. & Ershler, W. B. Prevalence and hematopoietic stem and progenitor cells. Blood 106, An excellent study suggesting that greying, a
outcomes of anemia in geriatrics: a systematic review 1479–1487 (2005). paradigmatic ageing phenotype, appears to result
of the literature. Am. J. Med. 116 (Suppl 7A), 3S–10S 32. Wang, Y., Schulte, B. A., Larue, A. C., Ogawa, M. & from a defect in melanocyte stem-cell maintenance
(2004). Zhou, D. Total body irradiation selectively induces with ageing.
14. Guralnik, J. M., Eisenstaedt, R. S., Ferrucci, L., murine hematopoietic stem cell senescence. Blood 46. Yoon, K. H. et al. Selective β-cell loss and α-cell
Klein, H. G. & Woodman, R. C. Prevalence of anemia 107, 358–366 (2006). expansion in patients with type 2 diabetes mellitus in
in persons 65 years and older in the United States: An important study showing persistent proliferative Korea. J. Clin. Endocrinol. Metab. 88, 2300–2308
evidence for a high rate of unexplained anemia. defects and p16INK4a expression in HSCs after (2003).
Blood 104, 2263–2268 (2004). exposure to DNA-damaging agents. 47. Butler, A. E. et al. β-cell deficit and increased β-cell
15. Appelbaum, F. R. et al. Age and acute myeloid 33. Meng, A., Wang, Y., Van Zant, G. & Zhou, D. Ionizing apoptosis in humans with type 2 diabetes. Diabetes
leukemia. Blood 107, 3481–3485 (2006). radiation and busulfan induce premature senescence 52, 102–110 (2003).
16. Brunello, A. et al. Adjuvant chemotherapy for elderly in murine bone marrow hematopoietic cells. Cancer 48. Meier, J. J. et al. Direct evidence of attempted β cell
patients (> or =70 years) with early high-risk breast Res. 63, 5414–5419 (2003). regeneration in an 89‑year‑old patient with recent-
cancer: a retrospective analysis of 260 patients. 34. Ito, K. et al. Regulation of oxidative stress by ATM is onset type 1 diabetes. Diabetologia 49, 1838–1844
Ann. Oncol. 16, 1276–1282 (2005). required for self-renewal of haematopoietic stem cells. (2006).
17. Lenhoff, S. et al. Impact of age on survival after Nature 431, 997–1002 (2004). 49. Krishnamurthy, J. et al. p16INK4a induces an age-
intensive therapy for multiple myeloma: a population- 35. Ito, K. et al. Reactive oxygen species act through p38 dependent decline in islet regenerative potential.
based study by the Nordic Myeloma Study Group. MAPK to limit the lifespan of hematopoietic stem cells. Nature 443, 453–457 (2006).
Br. J. Haematol. 133, 389–396 (2006). Nature Med. 12, 446–451 (2006). This study, similar to reference 43, shows that
18. Kollman, C. et al. Donor characteristics as risk factors References 34 and 35 suggest roles for p16INK4a loss can ameliorate an ageing phenotype,
in recipients after transplantation of bone marrow senescence-promoting molecules in HSC lifespan in in this case in the pancreatic islet.
from unrelated donors: the effect of donor age. Blood the setting of impaired ATM function and increased 50. Conboy, I. M. et al. Rejuvenation of aged progenitor
98, 2043–2051 (2001). ROS production. cells by exposure to a young systemic environment.
19. Ash, R. C. et al. Bone marrow transplantation from 36. Boccadoro, M. et al. Oral melphalan at diagnosis Nature 433, 760–764 (2005).
related donors other than HLA-identical siblings: effect hampers adequate collection of peripheral blood Using parabiosis, this study provides striking
of T cell depletion. Bone Marrow Transplant. 7, progenitor cells in multiple myeloma. Haematologica evidence that muscle satellite-cell ageing is cell
443–452 (1991). 87, 846–850 (2002). non-autonomous.
20. Castro-Malaspina, H. et al. Unrelated donor marrow 37. Knudsen, L. M., Rasmussen, T., Jensen, L. & 51. Ju, Z. et al. Telomere dysfunction induces
transplantation for myelodysplastic syndromes: Johnsen, H. E. Reduced bone marrow stem cell pool environmental alterations limiting hematopoietic stem
outcome analysis in 510 transplants facilitated by the and progenitor mobilisation in multiple myeloma after cell function and engraftment. Nature Med. 13,
National Marrow Donor Program. Blood 99, melphalan treatment. Med. Oncol. 16, 245–254 742–747 (2007).
1943–1951 (2002). (1999). This paper shows that telomere dysfunction limits
21. Buckner, C. D. et al. Marrow harvesting from normal 38. Gardner, R. V., Astle, C. M. & Harrison, D. E. HSC niche function, suggesting a cause of extrinsic
donors. Blood 64, 630–634 (1984). Hematopoietic precursor cell exhaustion is a cause of ageing in HSCs.

nature reviews | molecular cell biology volume 8 | september 2007 | 711


© 2007 Nature Publishing Group
REVIEWS

52. Kirkwood, T. B. Understanding the odd science of 69. Kamijo, T. et al. Tumor suppression at the mouse 91. Saxena, R. et al. Genome-wide association analysis
aging. Cell 120, 437–447 (2005). INK4a locus mediated by the alternative reading identifies loci for type 2 diabetes and triglyceride
53. Navarro, S. et al. Hematopoietic dysfunction in a frame product p19ARF. Cell 91, 649–659 (1997). levels. Science 316, 1331–1336 (2007).
mouse model for Fanconi anemia group D1. 70. Stein, G. H., Drullinger, L. F., Soulard, A. & Dulic, V. 92. Zeggini, E. et al. Replication of genome-wide
Mol. Ther. 14, 525–535 (2006). Differential roles for cyclin-dependent kinase inhibitors association signals in UK samples reveals risk loci for
54. Reese, J. S., Liu, L. & Gerson, S. L. Repopulating p21 and p16 in the mechanisms of senescence and type 2 diabetes. Science 316, 1336–1341 (2007).
defect of mismatch repair-deficient hematopoietic differentiation in human fibroblasts. Mol. Cell. Biol. 93. Helgadottir, A. et al. A common variant on
stem cells. Blood 102, 1626–1633 (2003). 19, 2109–2117 (1999). chromosome 9p21 affects the risk of myocardial
55. Prasher, J. M. et al. Reduced hematopoietic reserves 71. Alcorta, D. A. et al. Involvement of the cyclin- infarction. Science 316, 1491–1493 (2007).
in DNA interstrand crosslink repair-deficient Ercc1–/– dependent kinase inhibitor p16 (INK4a) in replicative 94. McPherson, R. et al. A common allele on chromosome
mice. EMBO J. 24, 861–871 (2005). senescence of normal human fibroblasts. Proc. Natl 9 associated with coronary heart disease. Science
56. Nijnik, A. et al. DNA repair is limiting for Acad. Sci. USA 93, 13742–13747 (1996). 316, 1488–1491 (2007).
haematopoietic stem cells during ageing. Nature 447, 72. Campisi, J. Suppressing cancer: the importance of 95. The Wellcome Trust Case Control Consortium.
686–690 (2007). being senescent. Science 309, 886–887 (2005). Genome-wide association study of 14,000 cases of
57. Morales, M. et al. The Rad50S allele promotes 73. Kim, W. Y. & Sharpless, N. E. The regulation of INK4/ seven common diseases and 3,000 shared controls.
ATM-dependent DNA damage responses and ARF in cancer and aging. Cell 127, 265–275 (2006). Nature 447, 661–678 (2007).
suppresses ATM deficiency: implications for the Mre11 74. Sone, H. & Kagawa, Y. Pancreatic β cell senescence References 89–95 describe the identification of
complex as a DNA damage sensor. Genes Dev. 19, contributes to the pathogenesis of type 2 diabetes in 9p21 SNPs near the INK4/ARF locus as being
3043–3054 (2005). high-fat diet-induced diabetic mice. Diabetologia 48, associated with type 2 diabetes mellitus,
A striking finding showing that an engineered 58–67 (2005). atherosclerotic heart disease and frailty.
Rad50S allele, which is associated with bone 75. Krishnamurthy, J. et al. Ink4a/Arf expression is a 96. Matthews, C. et al. Vascular smooth muscle cells
marrow hypoplasia, can be rescued by ATM biomarker of aging. J. Clin. Invest. 114, 1299–1307 undergo telomere-based senescence in human
deficiency. This result indicates that the Rad50S (2004). atherosclerosis: effects of telomerase and oxidative
allele is hypermorphic, and suggests that activation 76. Edwards, M. G. et al. Gene expression profiling of stress. Circ. Res. 99, 156–164 (2006).
of the DNA-damage response per se can lead to aging reveals activation of a p53-mediated 97. Chimenti, C. et al. Senescence and death of primitive
stem-cell dysfunction. transcriptional program. BMC Genomics 8, e80 cells and myocytes lead to premature cardiac aging
58. Rossi, D. J. et al. Deficiencies in DNA damage repair (2007). and heart failure. Circ. Res. 93, 604–613 (2003).
limit the function of haematopoietic stem cells with 77. Janzen, V. et al. Stem-cell ageing modified by the 98. Urbanek, K. et al. Myocardial regeneration by
age. Nature 447, 725–729 (2007). cyclin-dependent kinase inhibitor p16INK4a. Nature activation of multipotent cardiac stem cells in ischemic
An interesting study showing the accumulation of 443, 421–426 (2006). heart failure. Proc. Natl Acad. Sci. USA 102,
H2AX foci in HSCs with murine ageing, and an 78. Chen, J., Astle, C. M. & Harrison, D. E. Hematopoietic 8692–8697 (2005).
intrinsic role for several DNA-repair mechanisms in senescence is postponed and hematopoietic stem cell 99. Kleinjan, D. A. & van Heyningen, V. Long-range control
HSCs to prevent stem-cell ageing of this function is enhanced by dietary restriction. Exp. of gene expression: emerging mechanisms and
compartment. Hematol. 31, 1097–1103 (2003). disruption in disease. Am. J. Hum. Genet. 76, 8–32
59. Rudolph, K. L. et al. Longevity, stress response, and A provocative description of the effects of caloric (2005).
cancer in aging telomerase-deficient mice. Cell 96, restriction on HSC function with ageing. 100. Pasmant, E. et al. Characterization of a germ-line
701–712 (1999). 79. Chen, Q. & Ames, B. N. Senescence-like growth arrest deletion, including the entire INK4/ARF locus, in a
Classical study that established the importance of induced by hydrogen peroxide in human diploid melanoma-neural system tumor family: identification
telomere maintenance in age-related processes fibroblast F65 cells. Proc. Natl Acad. Sci. USA 91, of ANRIL, an antisense noncoding RNA whose
and lifespan in mammals. 4130–4134 (1994). expression coclusters with ARF. Cancer Res. 67,
60. Ruzankina, Y. et al. Deletion of the developmentally 80. Chen, J. H. et al. Loss of proliferative capacity and 3963–3969 (2007).
essential gene Atr in adult mice leads to age-related induction of senescence in oxidatively stressed human 101. Vulliamy, T. et al. The RNA component of telomerase is
phenotypes and stem cell loss. Cell Stem Cell 1, fibroblasts. J. Biol. Chem. 279, 49439–49446 mutated in autosomal dominant dyskeratosis
113–126 (2007). (2004). congenita. Nature 413, 432–435 (2001).
A recent demonstration that homeostatic 81. Takahashi, A. et al. Mitogenic signalling and the 102. Yamaguchi, H. et al. Mutations in TERT, the gene for
proliferation, in the absence of external p16(INK4a)-Rb pathway cooperate to enforce telomerase reverse transcriptase, in aplastic anemia.
DNA‑damaging agents, can promote stem-cell irreversible cellular senescence. Nature Cell Biol. 8, N. Engl. J. Med. 352, 1413–1424 (2005).
dysfunction and ageing. 1291–1297 (2006). 103. Mitchell, J. R., Wood, E. & Collins, K. A telomerase
61. Brown, E. J. & Baltimore, D. ATR disruption leads to 82. Tothova, Z. et al. FoxOs are critical mediators of component is defective in the human disease
chromosomal fragmentation and early embryonic hematopoietic stem cell resistance to physiologic dyskeratosis congenita. Nature 402, 551–555 (1999).
lethality. Genes Dev. 14, 397–402 (2000). oxidative stress. Cell 128, 325–339 (2007). 104. Tsakiri, K. D. et al. Adult-onset pulmonary fibrosis
62. Blasco, M. A. et al. Telomere shortening and tumor A detailed genetic study that establishes the caused by mutations in telomerase. Proc. Natl Acad.
formation by mouse cells lacking telomerase RNA. importance of FOXO-dependent regulation of Sci. USA 104, 7552–7557 (2007).
Cell 91, 25–34 (1997). intracellular ROS in the maintenance of HSCs. 105. Armanios, M. Y. et al. Telomerase mutations in
63. Lee, H. W. et al. Essential role of mouse telomerase in 83. Stepanova, L. & Sorrentino, B. P. A limited role for families with idiopathic pulmonary fibrosis. N. Engl. J.
highly proliferative organs. Nature 392, 569–574 p16Ink4a and p19Arf in the loss of hematopoietic stem Med. 356, 1317–1326 (2007).
(1998). cells during proliferative stress. Blood 106, 827–832 References 101–105 are important human studies
64. Herrera, E. et al. Disease states associated with (2005). describing the role of telomere dysfunction in
telomerase deficiency appear earlier in mice with Together with references 43, 49 and 77, this study human syndromes such as aplastic anaemia and
short telomeres. EMBO J. 18, 2950–2960 suggests that p16INK4a loss can ameliorate a murine idiopathic pulmonary fibrosis.
(1999). ageing phenotype, in this case in the HSCs. 106. Kitada, T., Seki, S., Kawakita, N., Kuroki, T. & Monna, T.
65. Allsopp, R. C., Morin, G. B., DePinho, R., Harley, C. B. 84. Tyner, S. D. et al. p53 mutant mice that display early Telomere shortening in chronic liver diseases.
& Weissman, I. L. Telomerase is required to slow ageing-associated phenotypes. Nature 415, 45–53 Biochem. Biophys. Res. Commun. 211, 33–39 (1995).
telomere shortening and extend replicative lifespan of (2002). 107. Miura, N. et al. Progressive telomere shortening and
HSCs during serial transplantation. Blood 102, 85. Maier, B. et al. Modulation of mammalian life span by telomerase reactivation during hepatocellular
517–520 (2003). the short isoform of p53. Genes Dev. 18, 306–319 carcinogenesis. Cancer Genet. Cytogenet. 93, 56–62
This paper shows that telomere dysfunction can (2004). (1997).
limit HSC lifespan in serial transplantation. References 84 and 85 demonstrate that excess 108. Urabe, Y. et al. Telomere length in human liver
66. Choudhury, A. R. et al. Cdkn1a deletion improves p53 activation can markedly accelerate the diseases. Liver 16, 293–297 (1996).
stem cell function and lifespan of mice with development of ageing-associated phenotypes in 109. Wiemann, S. U. et al. Hepatocyte telomere shortening
dysfunctional telomeres without accelerating cancer mice. and senescence are general markers of human liver
formation. Nature Genet. 39, 99–105 (2007). 86. Orsted, D. D., Bojesen, S. E., Tybjaerg-Hansen, A. & cirrhosis. FASEB J. 16, 935–942 (2002).
This paper demonstrates that p21CIP is not Nordestgaard, B. G. Tumor suppressor p53 Arg72Pro 110. Rudolph, K. L., Chang, S., Millard, M., Schreiber-Agus,
required for the anti-cancer effects of p53 polymorphism and longevity, cancer survival, and risk N. & DePinho, R. A. Inhibition of experimental liver
mediated in response to telomere dysfunction, but of cancer in the general population. J. Exp. Med. 204, cirrhosis in mice by telomerase gene delivery. Science
is important for the pro-ageing effects of p53 in 1295–1301 (2007). 287, 1253–1258 (2000).
HSCs and other stem cells in response to telomere 87. Matheu, A. et al. Delayed ageing through damage 111. Samani, N. J., Boultby, R., Butler, R., Thompson, J. R.
dysfunction. protection by the Arf/p53 pathway. Nature 448, & Goodall, A. H. Telomere shortening in
67. Chin, L. et al. p53 deficiency rescues the adverse 375–379 (2007). atherosclerosis. Lancet 358, 472–473 (2001).
effects of telomere loss and cooperates with telomere This paper challenges the view that increased p53 112. Obana, N. et al. Telomere shortening of peripheral
dysfunction to accelerate carcinogenesis. Cell 97, activity is necessarily pro-ageing. blood mononuclear cells in coronary disease patients
527–538 (1999). 88. Cheng, T. et al. Hematopoietic stem cell quiescence with metabolic disorders. Intern. Med. 42, 150–153
68. Artandi, S. E. et al. Telomere dysfunction promotes maintained by p21cip1/waf1. Science 287, (2003).
non-reciprocal translocations and epithelial cancers in 1804–1808 (2000). 113. Tabori, U., Nanda, S., Druker, H., Lees, J. & Malkin, D.
mice. Nature 406, 641–645 (2000). 89. Melzer, D. et al. A common variant of the p16(INK4a) Younger age of cancer initiation is associated with
References 67 and 68 provide insights into the genetic region is associated with physical function in shorter telomere length in Li–Fraumeni syndrome.
mechanisms that underlie the intimate link older people. Mech. Ageing Dev. 128, 370–377 Cancer Res. 67, 1415–1418 (2007).
between cancer and ageing, particularly why aged (2007). In accordance with results in mice (see reference
individuals develop epithelial cancers and why such 90. Scott, L. J. et al. A genome-wide association study of 67), this provocative human study suggests that
cancers emerge with radically altered cytogenetic type 2 diabetes in Finns detects multiple susceptibility telomere dysfunction accelerates tumour formation
profiles. variants. Science 316, 1341–1345 (2007). in the setting of p53 insufficiency.

712 | september 2007 | volume 8 www.nature.com/reviews/molcellbio


© 2007 Nature Publishing Group
f o c u s o nRa
EVg Iei
EWng
S

114. Wu, X. et al. Telomere dysfunction: a potential cancer 123. Molofsky, A. V., He, S., Bydon, M., Morrison, S. J. & Cancer Society Research Professor and an Ellison Medical
predisposition factor. J. Natl Cancer Inst. 95, Pardal, R. Bmi‑1 promotes neural stem cell self- Foundation Senior Scholar. This work was supported by grants
1211–1218 (2003). renewal and neural development but not mouse from the Sidney Kimmel Foundation for Cancer Research
115. Cawthon, R. M., Smith, K. R., O’Brien, E., growth and survival by repressing the p16Ink4a and (N.E.S.), the Ellison Medical Foundation (N.E.S. and R.A.D.),
Sivatchenko, A. & Kerber, R. A. Association between p19Arf senescence pathways. Genes Dev. 19, the American Federation of Aging Research (N.E.S.), the
telomere length in blood and mortality in people aged 1432–1437 (2005). Burroughs Wellcome Fund (N.E.S.) and the US National
60 years or older. Lancet 361, 393–395 (2003). References 121–123 are careful characterizations Institutes of Health (R.A.D.). R.A.D. is supported by the LeBow
116. Frenck, R. W. Jr., Blackburn, E. H. & Shannon, K. M. of the effects of BMI1 loss on NSCs, and the role of Fund to Cure Myeloma and the Robert A. and Renee E. Belfer
The rate of telomere sequence loss in human p16INK4a and ARF in NSCs in the setting of BMI1 Foundation Institute for Innovative Cancer Science.
leukocytes varies with age. Proc. Natl Acad. Sci. USA deficiency.
95, 5607–5610 (1998). 124. Jacobs, J. J., Kieboom, K., Marino, S., DePinho, R. A. Competing interests statement
117. Hemann, M. T., Strong, M. A., Hao, L. Y. & & van Lohuizen, M. The oncogene and Polycomb- The authors declare no competing financial interests.
Greider, C. W. The shortest telomere, not average group gene bmi‑1 regulates cell proliferation and
telomere length, is critical for cell viability and senescence through the ink4a locus. Nature 397,
chromosome stability. Cell 107, 67–77 (2001). 164–168 (1999). DATABASES
118. Rane, S. G. et al. Loss of Cdk4 expression causes 125. Park, I. K. et al. Bmi‑1 is required for maintenance of Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.
insulin-deficient diabetes and Cdk4 activation results adult self-renewing haematopoietic stem cells. Nature fcgi?db=gene
in b-islet cell hyperplasia. Nature Genet. 22, 423, 302–305 (2003). Atr
44–52 (1999). 126. Oguro, H. et al. Differential impact of INK4a and ARF OMIM: http://www.ncbi.nlm.nih.gov/entrez/query.
119. Tsutsui, T. et al. Targeted disruption of CDK4 delays on hematopoietic stem cells and their bone marrow fcgi?db=OMIM
cell cycle entry with enhanced p27(Kip1) activity. microenvironment in BMI1-deficient mice. J. Exp. ataxia telangiectasia | Hutchinson–Gilford syndrome |
Mol. Cell. Biol. 19, 7011–7019 (1999). Med. 203, 2247–2253 (2006). type 2 diabetes mellitus | Werner syndrome
120. Nielsen, G. P. et al. Immunohistochemical survey of References 125 and 126 are careful UniProtKB: http://ca.expasy.org/sprot
p16INK4A expression in normal human adult and infant characterizations of the effects of BMI1 loss on ATM | p15INK4b | p16INK4a | p53 | RB
tissues. Lab. Invest. 79, 1137–1143 (1999). HSCs, and the role of p16INK4a and ARF in the
121. Molofsky, A. V. et al. Bmi‑1 dependence distinguishes setting of BMI1 deficiency.
FURTHER INFORMATION
neural stem cell self-renewal from progenitor Ronald A. DePinho’s homepage: http://www.dana-farber.
proliferation. Nature 425, 962–967 (2003). Acknowledgements org/abo/danafarber/detail.asp?PersonID=51&RD=True
122. Bruggeman, S. W. et al. Ink4a and Arf differentially We thank E. Sahin, J.‑H. Paik, T. Letai and K. Mohlke for criti- Norman E. Sharpless’s homepage:
affect cell proliferation and neural stem cell self- cal reading and advice on the manuscript. R.A.D. is a director, http://genetics.unc.edu/faculty/sharpless.htm
renewal in Bmi1-deficient mice. Genes Dev. 19, co-founder and scientific advisor of AVEO Pharmaceuticals, All links are active in the online pdf.
1438–1443 (2005). Inc. in Cambridge, Massachusetts, USA, and is an American

nature reviews | molecular cell biology volume 8 | september 2007 | 713


© 2007 Nature Publishing Group

You might also like