Sie sind auf Seite 1von 18

Downloaded from genesdev.cshlp.

org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence


Thomas Kuilman, Chrysiis Michaloglou, Wolter J. Mooi, et al.

Genes Dev. 2010 24: 2463-2479


Access the most recent version at doi:10.1101/gad.1971610

References This article cites 209 articles, 66 of which can be accessed free at:
http://genesdev.cshlp.org/content/24/22/2463.full.html#ref-list-1
Email alerting Receive free email alerts when new articles cite this article - sign up in the box at the
service top right corner of the article or click here

Topic Articles on similar topics can be found in the following collections


Collections
Cell Cycle and DNA Replication (41 articles)
Cancer and Disease Models (98 articles)

To subscribe to Genes & Development go to:


http://genesdev.cshlp.org/subscriptions

Copyright © 2010 by Cold Spring Harbor Laboratory Press


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

REVIEW

The essence of senescence


Thomas Kuilman,1,3,4 Chrysiis Michaloglou,1,3,5 Wolter J. Mooi,2 and Daniel S. Peeper1,6
1
Division of Molecular Genetics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; 2Department
of Pathology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands

Almost half a century after the first reports describing by replication. Telomeres are subject to attrition due to
the limited replicative potential of primary cells in the fact that DNA polymerase fails to completely repli-
culture, there is now overwhelming evidence for the cate the lagging strands. In the early 1970s, Olovnikov telomeres reach
existence of ‘‘cellular senescence’’ in vivo. It is being (1971) and Watson (1972) independently described this so- critical minimal
recognized as a critical feature of mammalian cells to called ‘‘end replication problem’’, which contributes to
length, at
suppress tumorigenesis, acting alongside cell death pro- telomere shortening. Thus, telomeres act as a molecular
which point
grams. Here, we review the various features of cellular clock, reflecting the replicative history of a primary cell
senescence and discuss their contribution to tumor (Harley et al. 1990). DNA damage
suppression. Additionally, we highlight the power and When telomeres reach a critical minimal length, their response is
limitations of the biomarkers currently used to identify protective structure is disrupted. This triggers a DNA called in to
senescent cells in vitro and in vivo. damage response (DDR), which is associated with the arrest
appearance of foci that stain positive for g-H2AX (a phos- proliferation;
phorylated form of the histone variant H2AX) and the depending on
DDR proteins 53BP1, NBS1, and MDC1. Moreover, the DNA damages,
Replicative cellular senescence DNA damage kinases ATM and ATR are activated in se- the cell lives on
One of the first observations made for primary cells nescent cells (D’Adda di Fagagna et al. 2003). After withOUT
explanted from human tissue was that such cells do not amplification of the DDR signal, these kinases activate
replicative
proliferate indefinitely but instead are ‘‘mortal.’’ In fact, CHK1 and CHK2 kinases. Communication between
potential in
their proliferative capacity upon explantation consis- DDR-associated factors and the cell cycle machinery is
tently displays three phases: phase I, corresponding to brought about by phosphorylation and activation of sev- senescence OR
a period of little proliferation before the first passage, eral cell cycle proteins, including CDC25 (a family of it undergoes
during which the culture establishes; phase II, character- phosphatases) and p53. In addition, differential expression apoptosis;
ized by rapid cell proliferation; and phase III, during of p53 isoforms has been linked to replicative senescence senescence
which proliferation gradually grinds to a complete halt (Fujita et al. 2009). Together, these changes can induce allows cell to
(Hayflick and Moorhead 1961). Commenting on the a transient proliferation arrest, allowing cells to repair live without
possible causes of the transition to phase III, Hayflick their damage. However, if the DNA damage exceeds a passing on
(1965) hypothesized that ‘‘The finite lifetime of diploid certain threshold, cells are destined to undergo either DNA damage
cell strains in vitro may be an expression of aging or apoptosis or senescence. The factors bringing about this to progeny;
senescence at the cellular level.’’ The term cellular se- differential outcome have remained largely elusive, but
apoptosis=no
Senescence= the cell type and the intensity and duration of the signal,
nescence therefore denotes a stable and long-term loss means for cell
absence of of proliferative capacity, despite continued viability and as well as the nature of the damage, are likely to be
to live without
proliferation metabolic activity. important determinants (D’Adda di Fagagna 2008).
Consistent with Hayflick’s proposal, we now know In addition to p53, replicative senescence is linked to proliferation or
that, with the propagation of human cells in culture, the RB tumor suppressor and its signaling partners, in- cell checkpoint
telomeres (the protective chromosomal termini) are pro- cluding p16INK4A (a cyclin-dependent kinase inhibitor arrest or too
gressively shortened, ultimately causing cells to reach acting upstream of RB). Indeed, activation of both the much DNA
their ‘‘Hayflick limit.’’ This barrier has been termed p53 and p16INK4A–RB pathways is essential for induction damage
replicative (cellular) senescence, since it is brought about of senescence in a variety of human cell strains. The first
evidence for this came from experiments employing the
viral oncoprotein SV40 large T antigen (LT) and mutants
[Keywords: Telomeres; oncogenes; tumor suppressor genes; senescence;
in vivo; biomarkers] thereof lacking the ability to inhibit p53 or RB. While
3These authors contributed equally to this work.
4
expression of wild-type LT allowed cellular immortaliza-
Present addresses: Chromosome Segregation Laboratory, Cancer Re-
search UK, London Research Institute, 44 Lincoln’s Inn Fields, WC2A
tion to occur, expression of either of the mutants failed to
3LY London, United Kingdom; 5Novartis Institutes for Biomedical Re- do so (Shay et al. 1991). A variety of experiments sub-
search, Oncology Disease Area, CH-4002 Basel, Switzerland. sequently confirmed the critical roles of the p53 and
6
Corresponding author.
E-MAIL d.peeper@nki.nl; FAX 31-205122011. p16INK4A–RB pathways (Ben-Porath and Weinberg 2005;
Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1971610. Campisi 2005). The relative contribution of these cascades

GENES & DEVELOPMENT 24:2463–2479 ! 2010 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/10; www.genesdev.org 2463
Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

to senescence depends on the cell strain: While some are number of passages in culture, despite their retaining of
significantly delayed in their onset of senescence upon long telomeres. Elongation of their life span can be achieved
inactivation of p16INK4A alone, others require a deficiency by culturing in serum-free medium supplemented with
in p53 or in p53 as well as p16INK4A for the abrogation of a number of defined growth factors (Loo et al. 1987)
senescence. However, such escape from replicative se- or by culturing under physiological oxygen conditions
nescence results in only limited further proliferation: (Parrinello et al. 2003). Consistent with this, oxidative
Ultimately, cells will undergo telomeric crisis, resulting stress induces cessation of replication in cultured human
in chromosomal instability and death (Shay and Wright cells (Packer and Fuehr 1977; Chen et al. 1995; Yuan et al.
2005). 1995), while the replicative potential of human melano-
The dependence of replicative senescence on telomere cytes and epithelial cells depends largely on the compo-
shortening is evident from its bypass by the ectopic ex- sition of the culture medium used, as well as on the use of
pression of the catalytic subunit of the telomerase holo- feeder layers (Ramirez et al. 2001; Bennett and Medrano
enzyme (hTERT), which elongates telomeres, thereby 2002; Bennett 2003). Senescence of MEFs can be bypassed
abrogating the effect of the end replication problem also by inactivation of p53 or simultaneous ablation of RB
(Bodnar et al. 1998; Vaziri and Benchimol 1998). The family genes (Tanaka et al. 1994; Dannenberg et al. 2000;
limited life span of most primary human cells is ex- Sage et al. 2000). Thus, the immortalization of mamma-
plained by the fact that, in contrast to stem cells, tel- lian cells requires not only telomere maintenance, but
omerase is not expressed in human somatic cells, so they also optimal culture conditions (Mathon et al. 2001;
are unable to maintain telomeres at a sufficient length to Ramirez et al. 2001; Tang et al. 2001; Herbert et al.
suppress a DDR (Harley et al. 1990; Wright et al. 1996; 2002; for review, see Wright and Shay 2002).
Masutomi et al. 2003). Therefore, the ectopic expression
of hTERT is a common practice in vitro, allowing for the
Oncogene-induced senescence (OIS) in vitro
immortalization of primary human cells. Likewise, tumor
cells often express telomerase (Shay and Bacchetti 1997), Early studies on mutant HRAS (HRASV12) led to the
or elongate their telomeres through a mechanism termed discovery that, although it can transform most immortal
alternative lengthening of telomeres (ALT) (Muntoni and mammalian cell lines and collaborate with immortaliz-
Reddel 2005). As a result, telomeres of human cancer cells ing genes in oncogenically transforming primary cells, it
are maintained at a length that permits continued pro- induces cell cycle arrest when it is introduced alone into
liferation (Shay and Wright 2006). primary cells (and at least one immortal rat fibroblast
cell line) (Land et al. 1983; Franza et al. 1986; Serrano
et al. 1997). Serrano et al. (1997) noted the striking
Premature cellular senescence phenotypic resemblance of such nonproliferating cells
to those in replicative senescence, and this phenomenon
Senescence can be induced also in the absence of any
has eventually come to be known as OIS. Unlike
detectable telomere loss or dysfunction, by a variety of
replicative senescence, OIS cannot be bypassed by ex-
conditions. This type of senescence has been termed
pression of hTERT, confirming its independence from
premature, since it arises prior to the stage at which it
telomere attrition (Wei and Sedivy 1999).
is induced by telomere shortening. In recent years, ev-
One of the hallmarks shared by cells undergoing rep-
idence for the existence of premature senescence in vivo
licative senescence and OIS is the critical involvement of
has been accumulating rapidly and points to a critical
the p53 and p16INK4A–RB pathways, at least in certain
role in tumor suppression. We now turn to the various
settings. In murine cells, functional inactivation of p53 or
conditions that can induce such premature cellular
its direct upstream regulator, p19ARF, is sufficient to bypass
senescence.
RASV12-induced senescence (Kamijo et al. 1997; Serrano
et al. 1997). In human cells, p16INK4A seems to play a more
Stress-induced senescence in vitro
prominent role than p53, as some cells depend solely on
In vitro, premature senescence can result from inade- p16INK4A for OIS (Ben-Porath and Weinberg 2005). Whereas
quate culturing conditions. When cells are explanted p19ARF is an exquisite sensor that is activated by onco-
from an organism and placed in culture, they have to genic signals and mediates senescence in cultured murine
adapt to an artificial environment, characterized by ab- cells, in human cells it does not seem to play a similarly
normal concentrations of nutrients and growth factors dominant role (Wei et al. 2001; Michaloglou et al. 2005).
and the presence of ambient O2 levels, as well as the Nonetheless, specific mutations affecting human ARF
absence of surrounding cell types and extracellular ma- (i.e., p14ARF) but not p16INK4A have been identified in
trix components. One or more of these conditions can some human melanoma (Freedberg et al. 2008). Indeed,
induce a culture shock, resulting in stress-induced senes- OIS mechanisms do not seem to be universal across cell
cence (Sherr and DePinho 2000). This type of cell cycle types and genetic contexts. This is also exemplified by the
arrest is independent of telomere length. It also occurs in signaling routes relaying OIS by RASV12 versus BRAFE600:
mouse cells, which, in contrast to most human cells, Whereas RASV12-induced senescence can be bypassed by
express telomerase (Prowse and Greider 1995) and have abrogation of the p16INK4A–RB pathway (Serrano et al. 1997),
long telomeres (Kipling and Cooke 1990). Mouse embry- BRAFE600-triggered senescence cannot be bypassed by func-
onic fibroblasts (MEFs) undergo senescence after a limited tional inactivation of p16INK4A, be it alone (Michaloglou

2464 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

et al. 2005) or in combination with silencing of p14ARF senescent cells by definition withdraw from the cell
(Haferkamp et al. 2009). cycle, their inability to replicate is far from unique.
Recent evidence suggests the relevance of OIS also in Terminal differentiation, for example, similarly results
the context of induced pluripotency in vitro. At least two in stable replicative arrest. An essential difference is the
oncoproteins, c-MYC and KLF4, are required for the gener- fact that differentiation is typically triggered by physio-
ation of induced pluripotent stem (iPS) cells. As the logical cues and does not commonly involve the activa-
INK4A/ARF proteins and p53 limit the efficiency of iPS tion of tumor suppressor networks. Thus, although cell
cell formation, it has been suggested that cellular se- cycle exit constitutes an indispensable biomarker for
nescence counteracts the induced conversion of primary senescence, it can by no means be used in isolation to
cells into pluripotent stem cells (Banito et al. 2009; Hong identify senescent cells.
et al. 2009; Kawamura et al. 2009; Li et al. 2009; Marión Is cellular senescence strictly irreversible? Early in
et al. 2009; Utikal et al. 2009). Alternatively, increased vitro studies—for example, on p16INK4A and RAF1—sug-
proliferation rates associated with p53 loss may result in gested that it essentially is (Kato et al. 1998; Zhu et al.
accelerated kinetics of iPS formation (Hanna et al. 2009). 1998), but subsequent studies have argued that, in fact,
To the extent that this can be extrapolated to an in vivo escape scenarios exist. In spite of the observation that
setting, one could imagine that cancer stem cells arise stable RB-dependent heterochromatin structures and
from a similar reprogramming process (Krizhanovsky and several other factors contribute to lock cells in their
Lowe 2009). Thus, cellular senescence might suppress senescent state (see below), there are multiple ways to
tumor formation not only by inducing a persistent cell reverse the arrest, allowing cells to re-enter the cell cycle.
cycle arrest, but also by limiting the generation of cancer For example, inactivation of the p53 pathway permits
stem cells. senescence reversal, while inactivation of some interleu-
kins also abrogates the arrest (Beauséjour et al. 2003;
Tumor suppressor loss-induced senescence in vitro Dirac and Bernards 2003; Coppé et al. 2008; Kuilman
et al. 2008; see below). In some settings, the reversibility
Similar to oncogene mutation or overexpression, loss of
depends on the expression of p16INK4A prior to entering
a tumor suppressor can also trigger senescence in mouse
senescence (Beauséjour et al. 2003). As discussed in more
and human cells. This was first illustrated for PTEN and
detail below, there is also the possibility that, in addition
NF1. As elaborated further below, for PTEN it was shown
to the levels of certain tumor suppressor proteins, the
that fully deficient MEFs undergo senescence, which is
strength of the oncogenic signal contributes to the re-
accompanied by induction of p53. Concomitant loss of
versibility of senescence.
p53 allows cells to override the cytostatic effects of Pten
It should be noted here that strong selection exists for
deletions (Chen et al. 2005). Similarly, depletion of NF1
the small subset of cells that are endowed with the ability
causes senescence in vitro, which is eventually accom-
to exit the senescent state. While this may be a rare event
panied by decreases in ERK and AKT activities (Courtois-
in cultured cells, it is highly relevant in in vivo settings,
Cox et al. 2006). An elegant model was proposed in which
where clonal expansion of such reverted cells likely
the increase in RAS activity following NF1 loss is
represents a rate-limiting event on the path toward tumor
dampened by a negative feedback loop. Of note, although
progression. Indeed, activation of senescence-abrogating
loss of NF1 triggers senescence in human diploid fibro-
pathways can be seen in melanomas clonally emerging
blasts (HDFs), it immortalizes MEFs. Another example
from melanocytic nevi, which carry the same driver
within this theme is VHL, loss of which triggers senescence
mutation (LCW Vredeveld and DS Peeper, unpubl.).
in an RB- and p400-dependent manner (Young et al. 2008).
Notably, wound healing can create a microenvironment
that induces regrowth in nevi (Schoenfeld and Pinkus
Biomarkers and mechanisms of cellular senescence 1958; Park et al. 1987). It will be of interest to identify the
signaling pathways that apparently relax the cytostatic
While cellular senescence is induced by a wide variety of
constraints. A variation on this theme is preventing se-
conditions, senescent cells display a number of charac-
nescence, rather than reversing it. One could imagine
teristics that allow their identification both in vitro and
that this can be achieved by either a failure to engage
in vivo. Some of these biomarkers reflect the activation of
senescence pathways or a lack of fundamental senescence
mechanisms (such as the induction of tumor suppressor
mediators. An example for the first scenario would be
networks) that contribute to the senescence program. For
the inability of mutant KRAS to induce a senescence re-
others that accompany the execution of the senescence
sponse in the small intestine of mice that display evi-
program (like the increase in senescence-associated b-ga-
dence of senescence in the colon. In the same study, the
lactosidase [SA-b-GAL] activity), it is as yet unclear to
rapid development of colon adenocarcinomas in the ab-
what extent they contribute mechanistically.
sence of INK4a/Arf is an example of the second scenario
(Bennecke et al. 2010; see below).
Cell cycle arrest
Long-term exit from the cell cycle is the central and, in
Morphological transformation
our view, only indispensable marker for the identification
of all types of cellular senescence both in vitro and in Cell senescence is generally accompanied by morpholog-
vivo. However, it is important to realize that, although ical changes, which may be quite striking. Depending

GENES & DEVELOPMENT 2465


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

on the senescence trigger, cells can become large, flat, ing melanoma (Gruis et al. 1995; Krimpenfort et al. 2001;
and multinucleated, or rather refractile. A flat cell phe- Sharpless et al. 2001). This notwithstanding, at least in
notype is commonly seen in cells undergoing H-RASV12- melanomagenesis, genetic and immunohistochemical
induced senescence (Serrano et al. 1997; Denoyelle et al. evidence in mice and humans suggests that p16INK4A
2006), stress-induced senescence (Parrinello et al. 2003), plays a redundant role in senescence (Gruis et al. 1995;
or DNA damage-induced senescence (Chen and Ames Michaloglou et al. 2005; Dhomen et al. 2009). This may
1994; Chen et al. 2001). Cells senescing due to BRAFE600 be different in other settings, like human colon ade-
expression or the silencing of p400, however, acquire noma, in which a strict correlation is observed between
a more spindle-shaped morphology (Chan et al. 2005; p16INK4A expression and cell cycle arrest (Dai et al.
Michaloglou et al. 2005). Melanocytes undergoing 2000; Kuilman et al. 2008). In murine cells, its neighbor,
RASV12-induced senescence display extensive vacuoliza- p19ARF, appears to take on this dominant role as a com-
tion as a result of endoplasmic reticulum stress caused by mon senescence marker. Concordantly, Arf acts as
the unfolded protein response (Denoyelle et al. 2006). a potent tumor suppressor gene in mice (Kamijo et al.
1997) and is induced in response to oncogenic signals in
vivo (Zindy et al. 2003).
Activation of tumor suppressor networks
As mentioned above, the p53 and p16INK4A–RB signal
transduction cascades commonly mediate the activation Induction of SA-b-GAL activity
of the senescence program (Lowe et al. 2004). Conse- SA-b-GAL is a commonly used senescence biomarker
quently, components thereof have been used as biomarkers (Dimri et al. 1995; Debacq-Chainiaux et al. 2009). Its
to identify senescent cells. In human fibroblasts undergo- increased activity in senescent cells derives from lyso-
ing replicative or premature senescence, RB accumulates somal b-D-galactosidase, which is encoded by the GLB1
in its active, hypophosphorylated form (Stein et al. 1990; gene. Its detection requires tissues to be snap-frozen to
Serrano et al. 1997; Lin et al. 1998), and p53 displays preserve enzymatic activity (Debacq-Chainiaux et al.
increased activity and/or levels (Atadja et al. 1995; Vaziri 2009). Also, nonsenescent cells display b-galactosidase
et al. 1997; Bunz et al. 1998; Wei et al. 2001). The p53 activity in the lysosomes that functions most optimally
protein can also be phosphorylated on Ser 15 by ATM as at pH 4 (Lee et al. 2006). Accordingly, the increase in SA-
part of the senescence response (Calabrese et al. 2009). p53 b-GAL activity in senescent cells is likely due to an
serves as a node, mediating prosenescence signals emerg- expansion of the lysosomal compartment, giving rise to
ing from unscheduled oncogene activation, telomere dys- an increase in b-galactosidase activity that can be
function, DNA damage, and reactive oxygen species (ROS). measured also at suboptimal pH 6 (hence, SA-b-GAL)
Among the pocket proteins, RB has a unique role in (Kurz et al. 2000; Yang and Hu 2005; Lee et al. 2006).
mediating senescence in human cells (Chicas et al. 2010). There is, however, as yet no evidence pointing to an
One of its primary activators, p16INK4A, is commonly actual involvement of this enzyme in the senescence
induced in senescent cells in many contexts in vitro response (Lee et al. 2006).
(Serrano et al. 1997; Campisi 2005). Other proteins in
the p16INK4A–RB and p53 pathways, notably p21CIP1 and
Senescence-associated heterochromatic foci (SAHF)
p15INK4B, also often accumulate in senescent cells, and
have been used as markers reflecting the activation of Cellular senescence can be associated with an altered
these pathways in senescence (Alcorta et al. 1996; Hara chromatin structure, at least in vitro. While DNA dyes
et al. 1996; Reznikoff et al. 1996; Serrano et al. 1997; display overall homogenous staining patterns in cycling
Erickson et al. 1998; Lin et al. 1998; Robles and Adami or quiescent human cells, senescent cells often show
1998; Zhu et al. 1998; Hitomi et al. 2007). Induction of strikingly different punctate staining patterns. These
p16INK4A is commonly seen in senescent mouse and hu- DNA SAHF (Narita et al. 2003) are specifically enriched
man lesions in vivo also (te Poele et al. 2002; Collado et al. in methylated Lys 9 of histone H3 (a modification cat-
2005; Michaloglou et al. 2005; Gray-Schopfer et al. 2006; alyzed by the histone methyltransferase Suv39h1), while
Dhomen et al. 2009; Goel et al. 2009), while p53 and histone H3-Lys 9 acetylation and Lys 4 methylation (both
p21CIP1 induction can be evident, too; for example, in euchromatin markers) are excluded from SAHF. Whereas
senescent mouse prostate tumors (Chen et al. 2005). polycomb group proteins have been shown to repress the
Disruption of the p16INK4A–RB pathway collaborates INK4A/ARF locus (Jacobs et al. 1999; Gil et al. 2004;
with ectopic hTERT expression in the immortalization Bracken et al. 2007), the histone H3-Lys 27 demethylase
of both primary human epithelial cells (Kiyono et al. JMJD3 contributes to its transcriptional activation (Agger
1998) and melanocytes (Gray-Schopfer et al. 2006). At et al. 2009; Barradas et al. 2009), thus regulating senes-
least in some in vitro settings, this can be bypassed by cence. Senescent cells display increased binding of het-
altering the cell culture conditions (Ramirez et al. 2001), erochromatin-associated proteins in the promoters of sev-
consistent with the view discussed above that inadequate eral E2F target genes. SAHF formation is circumvented by
circumstances can launch the senescence program (Sherr interference with p16INK4A–RB pathway signaling, corre-
and DePinho 2000). lating with bypass of senescence. Depending on the
Genetic disruption of the gene encoding p16INK4A experimental system, interference with expression of
predisposes mice and humans to tumorigenesis, includ- other genes—including those encoding p53, C/EBPb, or

2466 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

interleukin-6 (IL-6) (see below)—also reduces SAHF-pos- Intriguingly, in addition to influencing their micro-
itivity, which is associated with abrogation of senescence environment, some inflammatory cytokines have an
(Chan et al. 2005; Ye et al. 2007; Zhang et al. 2007; indispensable role in the establishment and maintenance
Kuilman et al. 2008). of the senescence arrest. For example, signaling through
An essential step in the initiation phase of chromosome the IL-6 and IL-8 (CXCR2) receptors is essential, in a cell-
condensation appears to be the translocation of HIRA (a autonomous fashion, for cells to enter senescence in re-
histone chaperone) to PML (promyelocytic leukemia nu- sponse to oncogenic BRAF or replicative exhaustion, re-
clear) bodies (Zhang et al. 2005; Adams 2007, 2009). These spectively (Acosta et al. 2008; Kuilman et al. 2008). This
subnuclear organelles are thought to serve as a staging is relayed by the C/EBPb and NF-kB transcription factors
ground for the formation of HIRA/ASF1A-containing com- and is associated with the activation of an inflammatory
plexes, which are subsequently incorporated into chroma- transcriptome. Correspondingly, elevated levels of cyto-
tin and play an essential role in instigating SAHF forma- kines are found in senescent human neoplasms. Like-
tion. RNAi depletion of either ASF1A or HMGA1 (a SAHF wise, the ECM regulator PAI-1 has been shown to me-
component) leads to a partial bypass of senescence (Zhang diate senescence (Kortlever et al. 2006), although clinical
et al. 2005; Narita et al. 2006). data seem to point to a more complex picture (Kuilman
and Peeper 2009).
It thus appears that, while senescent cells can promote
Secreted factors in senescence
cancer by stimulating the proliferation of incipient tumor
Cells undergoing senescence—whether in response to cells that reside in their microenvironment, senescence
telomere malfunction, DNA damage, or oncogenic alter- also acts as a cell-autonomous tumor suppression mech-
ations—exhibit profound changes in their transcrip- anism limiting the expansion of early malignant cells.
tomes. A major consequence of this is the secretion of Therefore, the name ‘‘senescence-messaging secretome’’
many dozens of factors, including cytokines and chemo- (SMS) has been proposed in order to emphasize that, next
kines (Campisi 2005). The first indication of changes in to their requirement for the senescence response
the secretome of human cells accompanying senescence (whether or not involving a DDR), many of these fac-
was reported for fibroblasts undergoing replicative senes- tors are also endowed with communicative functions
cence. Microarray analysis revealed a strong inflamma- (Kuilman and Peeper 2009). The SMS creates a complex
tory response, as seen in wound healing (Shelton et al. signaling network in which secreted factors affect not
1999). Subsequent work from various laboratories has re- only the cells producing them (autocrine effects), but
vealed that cells undergoing either replicative or pre- also the microenvironment and hence neighboring cells
mature senescence display profound changes in their (paracrine effects). In some cases, SMS factors can stim-
secretome, termed the senescence-associated secretory ulate the innate immune system, triggering tumor clear-
phenotype (SASP) (Coppé et al. 2008; Rodier et al. 2009). ance (Xue et al. 2007; see below). The ultimate effect on
A recent study indicates that, for the induction of several target cells may depend largely on their genetic consti-
of these SASP factors, persistent DNA damage is re- tution. A classic example is the pro- or anti-proliferative
quired. Because DNA damage accompanies some but effect of TGFb on tumor cells as a function of the stage of
not all senescence settings (see below), the SASP is not tumorigenesis (Massagué 2008). Also, the effect of IL-6 on
strictly coupled to senescence per se (Rodier et al. 2009). melanomagenesis depends on tumor progression (Lu et al.
Proinflammatory cytokines—when secreted by senes- 1992, 1996). Although in these examples it is as yet
cent cells—trigger a variety of cellular responses. In cer- unclear to what extent this differential response is linked
tain settings, pro-oncogenic effects result: The prolifera- to the ability of cells to undergo senescence, it is conceiv-
tive rate, migration, and invasion of premalignant cells able that the effect of SMS factors on their target cells is
are enhanced when they are cocultured with, or grown in influenced by the oncogenic lesions that are carried by
medium conditioned by, senescent fibroblasts (Krtolica the latter.
et al. 2001; Dilley et al. 2003; Parrinello et al. 2005; Yang
et al. 2006). For example, a study on ovarian tumorigen-
ROS
esis has revealed that preneoplastic epithelial ovarian
cells expressing oncogenic RAS induce expression of As already alluded to, the replicative potential of primary
GRO1 (a cytokine recognized by CXCR2) (Yang et al. cells is influenced by the oxygen levels in which they are
2006). Once secreted, GRO1 has the potential to induce cultured. In fact, lowering the oxygen tension can prevent
senescence in ovarian fibroblasts in vitro. In turn, these OIS in HDF (Lee et al. 1999) and tissue culture stress-
fibroblasts promote tumor growth when coinjected with induced senescence in MEFs (Parrinello et al. 2003). It is
preneoplastic epithelial cells into mice. Interestingly, fi- widely assumed that not oxygen itself, but rather ROS
broblasts surrounding human ovarian tumors express produced by mitochondria are involved in senescence
several senescence markers, suggesting that this two- (Lu and Finkel 2008; Moiseeva et al. 2009). While ROS
way communication exists in vivo as well. Similar levels increase in both replicative senescence and OIS
findings have been reported for several additional weakly (Furumoto et al. 1998; Lee et al. 1999), treatment of HDF
oncogenic epithelial cell lines that show increased tu- with a sublethal concentration of H2O2 induces senescence
morigenic potential upon exposure to senescent fibro- (Chen and Ames 1994; Chen et al. 1998). ROS contribute
blasts (Krtolica et al. 2001). also to induction of replicative senescence, as evidenced

GENES & DEVELOPMENT 2467


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

by its delayed or premature onset upon treatment with Lenain and DS Peeper, unpubl.). In line with this, senes-
antioxidants or inhibitors of cellular oxidant scavengers, cence induced upon genetic loss of Skp2 in the context of
respectively (Chen et al. 1995; Yuan et al. 1995). A recent Pten heterozygosity is not associated with the emergence
systems biology study has suggested that ROS mediates of DDR markers and does not depend on p53 signaling
senescence through induction of DNA damage foci, (Lin et al. 2010). Shedding some light on a possible link
with a contribution of p21CIP1 (Passos et al. 2010). between the DDR and senescence, an elegant study
How do ROS relate to p53 and RB? A study on the showed that, in the context of Rb loss-driven murine
maintenance of a senescence-like arrest upon temporary adenomas, the DDR preceded cellular senescence
expression of SV40 LT revealed that ROS and PKCd func- (Shamma et al. 2009; see also below). While early and
tion in a positive feedback loop to maintain this pro- late adenomas similarly expressed DDR markers, senes-
liferative halt (Takahashi et al. 2006). Furthermore, ROS cence was apparent only in late (arrested) lesions, sug-
have been suggested to impinge either directly or in- gesting that, at least in this experimental system, it is
directly on the p53 and p16INK4A–RB signal transduction primarily senescence and not DNA damage signaling that
cascades (Itahana et al. 2003; Iwasa et al. 2003). For acts cytostatically (Peeper 2009). It thus seems that DDR
instance, the MINK–p38–PRAK pathway is activated in activation is involved in certain, but not all, OIS settings.
senescence and controls the activation of p16INK4A and
p53, as well as expression of p21CIP1, in a p53-independent Autophagy
manner (Wang et al. 2002; Bulavin et al. 2003; Iwasa et al.
Increasing evidence points to a link between cellular
2003; Nicke et al. 2005; Sun et al. 2007). These results not
senescence and autophagy. This catabolic process is me-
withstanding, although ROS undoubtedly plays an im-
diated by the formation of double-membraned vesicles
portant role in senescence, the nature and mechanism of
(autophagosomes) engulfing cytoplasmic content that
this contribution remains largely unclear (Lu and Finkel
fuse with lysosomes. Ultimately, this leads to the break-
2008). Major questions include how the increased levels
down of the vesicles and their constituents (Levine and
of ROS are generated, and what the cellular targets for
Kroemer 2008). Early studies have demonstrated that
ROS in senescence are.
chaperone-mediated autophagy is decreased in senescent
human cells (Cuervo and Dice 2000). Treatment of
DNA damage-induced senescence human umbilical vein endothelial cells (HUVECs) with
glycated collagen I (GC) results in an increase of SA-
The involvement of DNA damage in the induction of
b-GAL activity (Patschan et al. 2008). This seems to be
replicative senescence by telomere attrition has been
correlated with a transient induction of the formation of
established (D’Adda di Fagagna 2008). Also DNA-damag-
autophagosomes, as evidenced by the up-regulation of
ing agents such as ionizing radiation can induce cell cycle
the LC3-II protein. The fusion of autophagosomes with
arrest with characteristics of senescence in HDFs (Di
lysosomes, as measured by colocalization of LC3 and
Leonardo et al. 1994; Robles and Adami 1998). Further-
LAMP1 (a lysosomal marker), correlates with expression
more, DNA damage-inducing drugs can cause senescence
of LC3-II. Thus, both autophagosome formation and its
of tumor cells in vitro and in vivo, a phenomenon termed
fusion with lysosomes are eventually decreased com-
drug-induced senescence, which may be exploited for the
pared with nonsenescent cells. Inhibition of autophago-
treatment of cancer (Wang et al. 1998; Chang et al. 1999;
some formation by treatment with the pharmacological
Berns 2002; Schmitt et al. 2002; te Poele et al. 2002;
inhibitor 3-methyladenine decreases the number of SA-
Roninson 2003).
b-GAL-positive cells upon treatment with GC (Patschan
Activation of a DDR has also been shown to contribute
et al. 2008). This might indicate a decrease in the amount
to OIS in several settings in vitro; among others, in the
of senescent cells, although one cannot exclude that it
context of oncogene-induced DNA hyperreplication
reflects a drop in the number of lysosomes due to a de-
(D’Adda di Fagagna 2008). The DDR and p53 often func-
creased demand for lysosome–autophagosome fusion.
tion in a common signal transduction cascade in which
However, senescence and autophagy have recently been
interference with specific DDR components can sub-
connected more tightly. It was shown that autophagic
stitute for the loss of p53 function (Bartkova et al. 2006;
activity increases in OIS, as monitored by the activation
Di Micco et al. 2006; Moiseeva et al. 2006; Mallette et al.
of LC3-II, through a delayed activation of the mTORC1
2007; Kim et al. 2009; Santoriello et al. 2009). For ex-
pathway. Furthermore, autophagy contributes to cell
ample, a shRNA targeting ATM can serve as a proxy for
cycle arrest and production of senescence-associated in-
HPV E6 (which inactivates p53), contributing to bypass of
terleukins (Young et al. 2009).
senescence by STAT5 (Mallette et al. 2007).
However, DDR activation is not a universal feature of
Identifying senescent cells
OIS. For example, in mice, in the context of several types
of senescent adenomas, no DDR is observed, nor is there As described above, the execution of senescence pro-
a requirement for ATM in (oncogene-induced) senescence grams is associated with the appearance of several
(Efeyan et al. 2009). Furthermore, senescence triggered by markers, which are commonly used in order to identify
RAF1 or BRAFE600 and, in some cases, RASV12 does not senescent cells in vitro and in vivo. They include the loss
require p53 (Olsen et al. 2002; Nicke et al. 2005; Kuilman of proliferation, morphological changes, increased SA-
et al. 2008) and fails to mount an appreciable DDR (C b-GAL activity, the production of SAHF, and a rise in

2468 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

the levels of DDR markers as well as the cell cycle in- Cellular senescence as a pathophysiological
hibitors p16INK4A, p15INK4B, p21CIP1, and p53. DEC1, phenomenon
DCR2 (Collado et al. 2005), and PAI1 (Goldstein et al.
1994) have also been used as senescence biomarkers Ever since the discovery of cellular senescence in vitro,
(Collado and Serrano 2010). Furthermore, a number of researchers have wondered whether there are in vivo
inflammation-linked factors have been associated with correlates of this program. Hayflick (1965) described
senescence. However, it should be noted that different possible implications of cellular senescence for carcino-
senescent lesions may be characterized by different bio- genesis and aging: ‘‘Thus the phenomenon of the alter-
markers. ation of a cell strain to a cell line is important because, in
It should be emphasized that, in isolation, none of its simplest terms, it can be regarded as oncogenesis in
these markers can indisputably identify senescent cells, vitro.’’ Indeed, the conversion from a mortal to an im-
whether in vitro or in vivo. Indeed, several of the markers mortal cell reflects a rate-limiting step in tumorigenesis.
used in vitro await in vivo confirmation. For example, The identification of OIS in cultured cells opened a debate
due to the lack of appropriate antibodies, it has remained on the possible role of this type of proliferation arrest in
unclear whether the senescence-associated cell cycle vivo. Many were quick to realize that, in principle, OIS
protein p15INK4B can be used to specifically identify se- could serve as a powerful line of defense against tumor-
nescent human cells in vivo. Moreover, although recently igenesis. It would allow an incipient neoplastic cell har-
identified markers encoded by the inflammatory tran- boring an oncogenic lesion to withdraw itself from the
scriptome—particularly IL-6, IL-8, and CXCR2 (Acosta proliferative pool, so that tumor formation is prevented.
et al. 2008; Coppé et al. 2008; Kuilman et al. 2008)—have This is consistent with the fact that p16INK4A and p53,
been demonstrated to identify senescent cells in some two factors often associated with senescence, are among
early murine and human tumors, it remains to be de- the most commonly mutated proteins in cancer (The
termined to what extent they can be used across different Cancer Genome Atlas Gene Ranker GBM 6000, http://
tissues and genetic contexts. cbio.mskcc.org/tcga-generanker/index.jsp).
In spite of its popularity, SA-b-GAL should not be Early in vitro experiments on senescence commonly
regarded as a unique identifier for senescent cells. In used supraphysiological expression levels of RASV12.
addition to senescent cells, contact-inhibited or serum- Subsequently, several groups reported that RASV12 and
starved cells in tissue culture, as well as proliferating mutant RAF proteins act mitogenically or, conversely,
dysplastic epithelium in the gastrointestinal tract, can be drive cells into senescence as a function of increasing
positive for SA-b-GAL (Severino et al. 2000; Going et al. expression levels (Woods et al. 1997; Guerra et al. 2003;
2002; Yang and Hu 2005). Furthermore, one would expect Deng et al. 2004; Tuveson et al. 2004; Sarkisian et al.
that increases in the number or activity of lysosomes 2007; Chen et al. 2009). Some of these observations, and
unrelated to senescence would also result in elevated the fact that commonly used culturing conditions differ
lysosomal b-galactosidase protein levels, and thus in in- markedly from those in intact tissue and presumably act
creased SA-b-GAL activity. Thus, employing SA-b-GAL as a further stressor, initially caused some skepticism
in the absence of additional senescence markers may about a physiological role for senescence. In the last 5
inappropriately classify cells as being senescent. For these years, however, a plethora of mouse models as well as
reasons, SA-b-GAL can serve as senescence marker only human lesions have provided convincing and indepen-
in conjunction with additional markers. dent arguments in favor of OIS serving as a bona fide and
A major challenge to the field, therefore, is to increase effective tumor suppressor mechanism.
the number as well as the reliability of markers to Although some in vitro senescence models may have
identify senescent cells, especially in vivo. Ideally, one been artificial, they did not at all rule out a possible role
needs to uncover biomarkers that are causally involved in for senescence in vivo. For instance, increased expression
this unique type of proliferative arrest. For the time of activated RAS genes is a common feature of human tu-
being, in the absence of unambiguous single markers, we mors (Elenbaas et al. 2001; Johnson et al. 2005; Okamura
would recommend that, for the identification of senes- et al. 2006; Chen et al. 2009) that can be associated
cent cells, one uses, in addition to a proliferation marker with the conversion from benign to malignant tumors
such as the Ki-67 antigen (by immunostaining) or the (Quintanilla et al. 1986; Finney and Bishop 1993; Rodriguez-
thymidine analog bromodeoxyuridine (BrdU) (by in vivo Puebla et al. 1999). Amplification of HRASV12 is common
labeling), at least two additional senescence markers, in human Spitz nevi, which rarely progress to malignancy
the choice of which may vary for different settings. For (Maldonado et al. 2004), probably representing the best
example, p16INK4A often correlates with senescence available evidence of high levels of a mutant RAS gene in
both in vitro and in vivo, and as such it is a common a growth-arrested benign neoplastic lesion.
(but not universal) senescence marker. Also, tissue As already alluded to above, the propensity to launch
sections for immunostaining should be analyzed with a senescence program in response to an oncogenic alter-
multiple senescence markers. Ideally, this is done on ation differs among tissue and cell types. In murine
consecutive sections or, even better, on single sections pulmonary tumors, for example, p19ARF is induced to
by double staining, if feasible. Only careful and multi- relatively low levels upon expression of KRASG12D, while
parameter analyses will allow the appropriate detection it is highly elevated in sarcomas (Young and Jacks 2010).
of presumed senescent lesions. Along these lines, KRAS-driven senescence is observed in

GENES & DEVELOPMENT 2469


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

several mouse tissues, including lung adenomas, pancre- BCL2-driven lymphomagenesis (Feldser and Greider 2007).
atic intraductal neoplasia, and colonic epithelium, but The lack of telomerase activity had been shown previously
not small intestinal epithelial cells (Collado et al. 2005; to result in an induction of apoptosis, but, in these two
Bennecke et al. 2010). Similarly, reactivation of p53 in models, this response was abrogated owing to expression
established murine tumors causes senescence in sarco- of the p53 mutant or BCL2, respectively. These studies
mas but apoptosis in lymphomas (Ventura et al. 2007; see show that cells display extensive induction of senescence
below). Thus, while senescence can be considered as in intestine, kidney, and spleen (Cosme-Blanco et al. 2007),
a pathophysiological phenomenon, several physiological or in microlymphomas (Feldser and Greider 2007). This
factors influence its onset. has provided the first direct evidence of replicative senes-
cence preventing tumorigenesis, at least in the context of
an apoptosis block.
Replicative senescence in vivo
Consequently, a colorful picture is emerging for the
A number of studies have suggested that senescent cells role of telomeres in senescence and cancer biology.
accumulate in aging individuals. An increase in SA- Telomere shortening or dysfunction initially triggers
b-GAL activity can be detected in the skin of elderly a robust stress response that, through the induction of cell
people (Dimri et al. 1995). In addition, a small but sig- cycle inhibitory proteins, limits the replicative potential of
nificant decrease in telomere length has been detected in cells. As such, this aspect of telomere biology is likely to
aged tissue, although this is limited to certain cell types act as a potent anti-cancer mechanism. This is consistent
(Cristofalo et al. 2004). These studies, which relied on with a significant drop in the tumor take of telomerase-
single senescence markers, were later supported by the deficient (mTR!/!) mice in the context of INK4a/Arf loss
finding that several DNA damage and heterochromatini- (Greenberg et al. 1999). In the absence of p53, however,
zation markers—including 53BP1, g-H2AX, phospho-Ser telomere dysfunction instead promotes tumorigenesis
1981-ATM, HP-1b, and HIRA—are increased in dermal (Chin et al. 1999). This could occur through the inability
fibroblasts from aging baboons compared with young of cells to engage a full DDR, and through the perturba-
ones. DNA damage markers were shown to colocalize tion of the apoptosis and senescence responses (Artandi
with telomeres, and this correlated with increased and DePinho 2010).
p16INK4A expression (Herbig et al. 2006), suggesting that
dysfunctional telomeres can create DNA damage signals
OIS in vivo
leading to the onset of senescence also in vivo. In line
with the idea that replicative senescence results from The past 5 years have witnessed a boost in the number of
telomeric attrition upon prolonged proliferation, senes- studies demonstrating senescence in vivo in both murine
cence is detected mainly in tissues experiencing sub- and human lesions. Senescence markers have been dem-
stantial cellular turnover (Jeyapalan et al. 2007). onstrated in several contexts in which oncogenes or tu-
Telomere dysfunction has been suggested to be con- mor suppressor genes were perturbed (Fig. 1). Four studies
nected to age-related pathology, and this warrants further that were published in concert provided the first evidence
study. For example, atherosclerosis is accompanied by an for OIS in vivo. Schmitt and colleagues (Braig et al. 2005)
induction of SA-b-GAL activity (Minamino et al. 2002) reported on the tumor-suppressive role of the chromatin
and telomere shortening (Foreman and Tang 2003). Se- remodeling enzyme Suv39h1 in Em-NRAS-driven lympho-
nescent cells have also been observed in osteoarthritis magenesis, which correlates with bypass of OIS in vitro in
(Price et al. 2002). Since these studies used single senes- Suv39h1!/! splenocytes. Serrano and coworkers (Collado
cence markers, they await confirmation. This notwith- et al. 2005) showed that conditional expression of KRASV12
standing, the observations are in agreement with the from its endogenous locus in mice results in the emer-
proposed deleterious effects of senescent cells on their gence of lung adenomas, as well as premalignant pancre-
microenvironment, possibly contributing to age-related atic intraductal neoplasia, most of which fail to progress to
disease (Campisi 2005). malignancy. The adenomatous stage is specifically char-
Conversely, beneficial aspects of replicative senescence acterized by a low proliferative index and the appearance of
have also been uncovered, including some that are un- several senescence markers, including SA-b-GAL, p15INK4B,
related to cancer. In a mouse model of liver fibrosis, se- p16INK4A, and several new markers. Peeper and coworkers
nescent cells were detected, as evidenced by positivity for (Michaloglou et al. 2005) showed that congenital human
SA-b-GAL, p16INK4A, p53, p21CIP1, and HMGA1. These melanocytic nevi, which frequently harbor activating
senescent cells derive from activated hepatocellular stem BRAF or NRAS mutations, display several characteris-
cells and limit the progression of fibrosis (Krizhanovsky tics of senescence (see below). The first demonstration of
et al. 2008). In addition, two reports (Cosme-Blanco et al. tumor suppressor (PTEN) loss-induced senescence in vivo
2007; Feldser and Greider 2007) have provided evidence (Chen et al. 2005) is discussed below.
for the proposal by Hayflick (1965) that replicative se- As a sequel to this first series of observations, several
nescence limits tumorigenesis. Both studies report that other groups have further supported the view that OIS
mice lacking the RNA component of telomerase (mTR) inhibits tumor formation. For example, both established
display decreased tumor formation in the context of and newly identified chemokine and cytokine markers
either one or two copies of mutant p53R172P replacing suggest the presence of senescent cells in human colon
endogenous p53 (Cosme-Blanco et al. 2007) or Em-MYC/ adenoma and prostatic intraepithelial neoplasia (PIN)

2470 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

Figure 1. The essence of senescence: biomarkers in vivo. Several markers of cellular senescence have been identified in early
neoplastic lesions in both mice and humans. Representative examples are shown in the various columns, with the type of genetic
lesions or other types of stress (stress categories, color-coded) that instigate senescence at the very top and very bottom, and from
thereon inward: the tissue types, specific stress signals, biomarkers, and symbols legends, respectively. For instance, in BRAF mutant
nevi, the stress category is oncogene activation (blue), the tissue type is nevus, the specific stress signal is BRAF, and the biomarkers
reported are cell cycle arrest, increase in SA-b-GAL activity, and tumor suppressor activation. (CTX) Cyclophosphamide; (bCAT)
b-catenin; (GL) gland; (N.D.) not determined.

lesions (Acosta et al. 2008; Kuilman et al. 2008). In Collado et al. 2005; Lazzerini Denchi et al. 2005; Acosta
addition, murine papillomas driven by HRASV12 expres- et al. 2008). However, particularly for cell cycle-regulat-
sion or induced upon DMBA/TPA treatment express ing genes like p16INK4A, abundant expression may, in fact,
several senescence markers (Collado et al. 2005; Acosta be maintained during malignancy, which is explained by
et al. 2008; Chen et al. 2009; Yamakoshi et al. 2009). the occurrence of other (epi)genetic aberrations (includ-
Recent evidence indicates that Myc, known mainly for its ing events occurring in parallel and downstream) that
proapoptotic function, drives a subset of murine lympho- neutralize their functions. As such cases are expected to
mas into senescence through stromal secretion of TGFb coincide with immunopositivity for proliferation mark-
(Reimann et al. 2010), providing support for the premise ers, this reinstates the need for careful, multiparameter
that senescence is not solely implemented in a cell- analysis of senescence in vivo.
autonomous manner. In line with the idea that OIS
represents a barrier for cancer development, a senescence
OIS in models of nevus and melanoma formation
index of tumor cells has been associated with better
treatment outcome in high-grade colorectal cancer Melanocytic nevi, benign tumors that have a low pro-
(Haugstetter et al. 2010). Moreover, expression of several pensity to progress toward melanoma, are a well-studied
senescence-associated genes is reduced when tumors system for OIS in vivo in humans, mice, and fish. In 2002,
progress beyond the senescent stage (Chen et al. 2005; BRAFE600 was identified as a frequent mutation in human

GENES & DEVELOPMENT 2471


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

cancer, predominantly melanoma (Davies et al. 2002). At cooperation denotes an essential role of PTEN in
least as remarkable was the finding that the same mu- BRAFE600-induced senescence remains to be established.
tation is present in the large majority of nevi (Pollock
et al. 2003). In spite of the presence of an oncogenic BRAF Tumor suppressor loss-induced senescence in vivo
(or, in some cases, NRAS) allele, an important and com-
Pandolfi and colleagues (Chen et al. 2005) were the first to
mon feature of nevi is their exceedingly low proliferative
show that senescence in vivo can also be triggered by the
activity. This characteristic is typically maintained for
loss of a tumor suppressor gene. They found that condi-
decades until the lesion gradually disappears. Nevi ex-
tional Pten deletion in murine prostate cells results in the
press elevated levels of p16INK4A and display increased
formation of high-grade PIN, which display characteris-
SA-b-GAL activity (Michaloglou et al. 2005). Arguing
tics of senescence. In conjunction with p53 loss, these
against a role for replicative senescence, it was found that
lesions progress to malignancy. Senescence induced by
telomere length in nevi is indistinguishable from that in
Pten loss can be enhanced by inactivation of the E3
normal skin (and longer than in melanoma cells). This
ubiquitin ligase Skp2 (Lin et al. 2010). Similarly, Cichowski
strongly suggests that nevi undergo OIS in vivo. These
and coworkers (Courtois-Cox et al. 2006) showed that, in
findings have been confirmed by Bennett and coworkers
benign dermal neurofibromas carrying NF1 mutations,
(Gray-Schopfer et al. 2006). Secreted IGFBP7 has been
several senescence markers are up-regulated, while phos-
reported to play a central role in the initiation and main-
pho-ERK and phospho-AKT are present at low levels.
tenance of the senescent state of nevus cells (Wajapeyee
Finally, kidneys from Vhl knockout mice display an in-
et al. 2008), but these findings were challenged recently
crease in SA-b-GAL activity as well as induced immu-
(Schrama et al. 2010; Scurr et al. 2010; Wajapeyee et al.
nostaining for p27Kip1 and the senescence marker DcR2
2010).
(Young et al. 2008).
The studies on senescent nevi are consistent with
The Takahashi laboratory (Shamma et al. 2009) used
earlier observations that a mutant BRAF transgene in
a model of senescence in vivo in which genetic loss of one
the melanocytic compartment induces the formation of
allele of Rb drives thyroid adenoma formation. These
nevus-like lesions in zebrafish (Patton et al. 2005). Al-
lesions display enhanced isoprenylation and activation of
though senescence markers were not assessed in this
N-Ras and express several senescence markers, as dis-
study, Patton et al. (2005) describe that, in combination
cussed above. Homozygous loss of NRas in the context of
with loss of p53, the fish develop invasive melanomas.
Rb heterozygosity allowed for progression to carcinomas
In a similar transgenic mouse model, expression of
that was accompanied by the loss of senescence markers.
BRAFE600 in the melanocytic lineage drives the formation
Together, these examples provide the first pieces of evi-
of nevi with several signs of senescence (Goel et al. 2009).
dence that, similar to activated oncogenes, loss of expres-
These animals develop melanoma with a long latency,
sion of tumor suppressors can also cause incipient cancer
depending on the expression of the transgene. Melanoma
lesions to activate features of senescence, serving to limit
formation was strongly accelerated in Cdkn2a or p53-
tumorigenesis in vivo.
deficient backgrounds.
Recently, two advanced mouse models have been
Restoration of cellular senescence in vivo
created that more closely resemble spontaneous mu-
tation of the oncogene by conditionally expressing In light of the steadily increasing evidence that senes-
BRAFE600 from its endogenous promoter (Dankort et al. cence protects humans and other organisms against can-
2009; Dhomen et al. 2009). In these models, specific cer, ideas are being launched to restore this process in
expression of mutant BRAF in the melanocytic compart- tumor cells in which it has become deficient. Induction of
ment results in the formation of nevus-like benign le- apoptosis in tumor cells, whether by chemotherapeutics
sions, which, depending on the model used, remain stable or signaling molecules, is successfully used therapeuti-
for several months to more than a year. Importantly, cally. Would reactivation of senescence be similarly re-
these melanocytic cell groups express several senescence warding clinically? In particular, for those tumors that
markers, corroborating earlier observations on RASV12 are highly resistant to drug- or radiotherapy-induced cell
that physiological expression levels of an activated onco- death, the concept of activating senescence in (early) tu-
gene can also give rise to senescence in neoplasms. In line mors may sound appealing. But is it feasible? To explore
with the idea that p16INK4A is not always strictly required the viability of such an approach, the Jacks (Ventura et al.
for OIS in vivo (Michaloglou et al. 2008), nevus formation 2007) and Lowe (Xue et al. 2007) laboratories have em-
was unimpaired in a p16INK4A-deficient background, al- ployed conditional mouse models. In the system used by
though increased melanoma penetrance and reduced la- Ventura et al. (2007) a transcription–translation stop site
tency were observed (Dhomen et al. 2009). In contrast, in flanked by loxP sites within the p53 gene could be excised
combination with a mutant Braf knock-in allele, loss of upon expression of Cre recombinase. Upon activation of
Pten produced aggressive tumors much resembling human p53 in already established lymphomas and sarcomas,
metastatic melanoma (Dankort et al. 2009). These elegant dramatic tumor regression was observed. While this was
models have convincingly shown that mutation of BRAF associated with induction of apoptosis in lymphomas,
can drive nevus formation, and that a specific secondary sarcomas failed to undergo apoptosis, but instead displayed
lesion (e.g., Pten loss) collaborates to drive melanoma- several characteristics of senescence. In contrast to tumor
genesis. Whether this classical genetic oncogenic cells, restoration of p53 function in normal tissue did not

2472 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

result in detectable expression of the tumor suppressor. research on this topic, four decades have passed before
This suggests that restoration of p53-dependent senes- solid evidence was first presented that senescence occurs
cence occurs only in the context of stress signals (Ventura not only under artificial conditions in vitro, but also
et al. 2007). In the model by Xue et al. (2007), liver under pathophysiological conditions and in several ge-
progenitor cells were infected ex vivo with retrovirus netic settings in vivo. Attempts to increase our under-
encoding HRASV12 and an inducible shRNA targeting standing of this process were boosted by the discovery of
p53. Upon transplantation into the spleen of recipient OIS, which strengthened the link with cancer research. In
mice, the hepatoblasts seeded in the liver and produced spite of this important progress, however, many chal-
invasive hepatocarcinomas. Reactivation of p53 caused lenges remain. Probably most importantly, there is an
these tumors to undergo senescence. Remarkably, an in- urgent need for better senescence biomarkers. It is clear
nate immune response was subsequently elicited, eventu- that, although a plethora of markers has been proposed
ally resulting in the clearance of the tumors (Xue et al. and in fact are commonly used, no single one can reliably
2007). Thus, these models have established that, at least identify senescent cells in vitro or in vivo. Results from
under conditions in which widespread and acute activa- genome-wide expression analyses of senescent cells could
tion of senescence can be achieved, this can culminate in serve as a convenient starting point for the identification
tumor regression. Conversely, as has been shown for Myc, of new and better senescence markers. Integrating these
oncogene inactivation can also set off a senescence pro- data with systematic RNAi approaches is likely to yield
gram in several tumors (Wu et al. 2007) in a TGFb-de- biomarkers with causal relationships to senescence. The
pendent manner (van Riggelen et al. 2010). These studies contribution to senescence in vitro and in vivo of several
may therefore serve as proof of principle that reactivation cellular processes—including activation of DDR, SAHF
of senescence is a realistic means to intervene with cancer. formation, and induction of ROS and autophagy—
In several examples above, a tumor suppressor (p53) also requires further elucidation. In addition, the ques-
that contributed to the tumor initiation and maintenance tion remains of which factors determine whether tissues
was restored, resulting in senescence. Since inactivation, primarily undergo senescence or other stress-induced
rather than restoration, of a tumor suppressor can also programs like apoptosis. This may be determined by the
induce senescence in vivo (as discussed above for PTEN abundance of cell-intrinsic survival proteins, but other
and NF1), it thus appears that tumor suppressors can factors, perhaps of stromal origin, are likely to add to the
differentially influence senescence. These effects can be complexity of this choice. Several of these issues can be
seen even for a single tumor suppressor. For example, as effectively investigated in mouse models; for instance, in
ectopic expression of PTEN or suppression of PI3K also animals conditionally expressing activated oncogenes
causes senescence (Courtois-Cox et al. 2006), together from their endogenous promoters, or conditional tumor
these observations suggest that major pathways like suppressor genes. It is conceivable that these as well as
PI3K/PTEN require fine-tuning to maintain cell prolifer- newly generated models will prove instrumental in vali-
ation: Too little or too much activity may act cytostati- dating results obtained in vitro. As therapeutic exploita-
cally. Recently, the window in which reduction in PTEN tion of enforced cellular senescence may become a realistic
levels induces senescence (PICS) has been proposed as goal, answers to these questions may eventually provide
a means to establish a prosenescence therapy (Alimonti us with new tools to effectively treat cancer patients.
et al. 2010). It was shown that pharmacologic inhibition
of PTEN caused increased SA-b-GAL activity along with Acknowledgments
a reduction in cell proliferation both in vitro and in tumor
xenografts. Along these lines, Lin et al. (2010) reported T.K., C.M., W.J.M, and D.S.P. were supported by grants from the
that a Skp2–SCF inhibitor triggers senescence and in- Dutch Cancer Society, including a Queen Wilhelmina Research
Award, and a Vici grant from the Netherlands Organization for
hibition of tumor growth of PTEN-deficient cancer cells.
Scientific Research (NWO).
In further support of this emerging theme, Amati and
colleagues (Campaner et al. 2010) observed that onco-
genic stress caused by the Myc oncoprotein sensitizes References
Cdk2-deficient cells to undergo senescence in vitro and Acosta JC, O’Loghlen A, Banito A, Guijarro MV, Augert A,
in vivo. Similarly, in advanced non-small-cell lung carci- Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, et al.
noma driven by activation of oncogenic KRas, tumor 2008. Chemokine signaling via the CXCR2 receptor rein-
progression is halted and senescence is induced upon forces senescence. Cell 133: 1006–1018.
genetic ablation of Cdk4 (Puyol et al. 2010). The rising Adams PD. 2007. Remodeling of chromatin structure in sen-
number of examples of enforced senescence in vivo escent cells and its potential impact on tumor suppression
justifies further exploration of this possibility by address- and aging. Gene 397: 84–93.
Adams PD. 2009. Healing and hurting: Molecular mechanisms,
ing several more clinically oriented questions.
functions, and pathologies of cellular senescence. Mol Cell
36: 2–14.
Concluding remarks Agger K, Cloos PA, Rudkjaer L, Williams K, Andersen G,
Christensen J, Helin K. 2009. The H3K27me3 demethylase
The discovery of cellular senescence in cultured cells JMJD3 contributes to the activation of the INK4A–ARF locus
came with exciting hypotheses about the potential roles in response to oncogene- and stress-induced senescence.
it may have in tumorigenesis and aging. Despite intensive Genes Dev 23: 1171–1176.

GENES & DEVELOPMENT 2473


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

Alcorta DA, Xiong Y, Phelps D, Hannon G, Beach D, Barrett JC. p53 and p21 to sustain G2 arrest after DNA damage. Science
1996. Involvement of the cyclin-dependent kinase inhibitor 282: 1497–1501.
p16 (INK4a) in replicative senescence of normal human Calabrese V, Mallette FA, Deschênes-Simard X, Ramanathan S,
fibroblasts. Proc Natl Acad Sci 93: 13742–13747. Gagnon J, Moores A, Ilangumaran S, Ferbeyre G. 2009.
Alimonti A, Nardella C, Chen Z, Clohessy JG, Carracedo A, SOCS1 links cytokine signaling to p53 and senescence.
Trotman LC, Cheng K, Varmeh S, Kozma SC, Thomas G, Mol Cell 36: 754–767.
et al. 2010. A novel type of cellular senescence that can be Campaner S, Doni M, Hydbring P, Verrecchia A, Bianchi L,
enhanced in mouse models and human tumor xenografts to Sardella D, Schleker T, Perna D, Tronnersjö S, Murga M,
suppress prostate tumorigenesis. J Clin Invest 120: 681–693. et al. 2010. Cdk2 suppresses cellular senescence induced
Artandi SE, DePinho RA. 2010. Telomeres and telomerase in by the c-myc oncogene. Nat Cell Biol 12: 54–59; sup pp 51–
cancer. Carcinogenesis 31: 9–18. 14.
Atadja P, Wong H, Garkavtsev I, Veillette C, Riabowol K. 1995. Campisi J. 2005. Senescent cells, tumor suppression, and or-
Increased activity of p53 in senescing fibroblasts. Proc Natl ganismal aging: Good citizens, bad neighbors. Cell 120: 513–
Acad Sci 92: 8348–8352. 522.
Banito A, Rashid ST, Acosta JC, Li S, Pereira CF, Geti I, Pinho S, Chan HM, Narita M, Lowe SW, Livingston DM. 2005. The p400
Silva JC, Azuara V, Walsh M, et al. 2009. Senescence impairs E1A-associated protein is a novel component of the p53 /
successful reprogramming to pluripotent stem cells. Genes p21 senescence pathway. Genes Dev 19: 196–201.
Dev 23: 2134–2139. Chang BD, Xuan Y, Broude EV, Zhu H, Schott B, Fang J,
Barradas M, Anderton E, Acosta JC, Li S, Banito A, Rodriguez- Roninson IB. 1999. Role of p53 and p21waf1/cip1 in senes-
Niedenfuhr M, Maertens G, Banck M, Zhou MM, Walsh MJ, cence-like terminal proliferation arrest induced in human
et al. 2009. Histone demethylase JMJD3 contributes to tumor cells by chemotherapeutic drugs. Oncogene 18: 4808–
epigenetic control of INK4a/ARF by oncogenic RAS. Genes 4818.
Dev 23: 1177–1182. Chen Q, Ames BN. 1994. Senescence-like growth arrest induced
Bartkova J, Rezaei N, Liontos M, Karakaidos P, Kletsas D, Issaeva by hydrogen peroxide in human diploid fibroblast F65 cells.
N, Vassiliou L-VF, Kolettas E, Niforou K, Zoumpourlis VC, Proc Natl Acad Sci 91: 4130–4134.
et al. 2006. Oncogene-induced senescence is part of the Chen Q, Fischer A, Reagan JD, Yan LJ, Ames BN. 1995.
tumorigenesis barrier imposed by DNA damage checkpoints. Oxidative DNA damage and senescence of human diploid
Nature 444: 633–637. fibroblast cells. Proc Natl Acad Sci 92: 4337–4341.
Beauséjour CM, Krtolica A, Galimi F, Narita M, Lowe SW, Chen QM, Bartholomew JC, Campisi J, Acosta M, Reagan JD,
Yaswen P, Campisi J. 2003. Reversal of human cellular Ames BN. 1998. Molecular analysis of H2O2-induced sen-
senescence: Roles of the p53 and p16 pathways. EMBO escent-like growth arrest in normal human fibroblasts: p53
J 22: 4212–4222. and Rb control G1 arrest but not cell replication. Biochem
Bennecke M, Kriegl L, Bajbouj M, Retzlaff K, Robine S, Jung A, J 332: 43–50.
Arkan MC, Kirchner T, Greten FR. 2010. Ink4a/Arf and Chen QM, Prowse KR, Tu VC, Purdom S, Linskens MH. 2001.
oncogene-induced senescence prevent tumor progression Uncoupling the senescent phenotype from telomere short-
during alternative colorectal tumorigenesis. Cancer Cell ening in hydrogen peroxide-treated fibroblasts. Exp Cell Res
18: 135–146. 265: 294–303.
Bennett DC. 2003. Human melanocyte senescence and mela- Chen Z, Trotman LC, Shaffer D, Lin H-K, Dotan ZA, Niki M,
noma susceptibility genes. Oncogene 22: 3063–3069. Koutcher JA, Scher HI, Ludwig T, Gerald W, et al. 2005.
Bennett DC, Medrano EE. 2002. Molecular regulation of mela- Crucial role of p53-dependent cellular senescence in sup-
nocyte senescence. Pigment Cell Res 15: 242–250. pression of Pten-deficient tumorigenesis. Nature 436: 725–
Ben-Porath I, Weinberg RA. 2005. The signals and pathways 730.
activating cellular senescence. Int J Biochem Cell Biol 37: Chen X, Mitsutake N, LaPerle K, Akeno N, Zanzonico P, Longo
961–976. VA, Mitsutake S, Kimura ET, Geiger H, Santos E, et al. 2009.
Berns A. 2002. Senescence: A companion in chemotherapy? Endogenous expression of Hras(G12V) induces developmen-
Cancer Cell 1: 309–311. tal defects and neoplasms with copy number imbalances of
Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin the oncogene. Proc Natl Acad Sci 106: 7979–7984.
GB, Harley CB, Shay JW, Lichtsteiner S, Wright WE. 1998. Chicas A, Wang X, Zhang C, McCurrach M, Zhao Z, Mert O,
Extension of life-span by introduction of telomerase into Dickins RA, Narita M, Zhang M, Lowe SW. 2010. Dissecting
normal human cells. Science 279: 349–352. the unique role of the retinoblastoma tumor suppressor
Bracken AP, Kleine-Kohlbrecher D, Dietrich N, Pasini D, during cellular senescence. Cancer Cell 17: 376–387.
Gargiulo G, Beekman C, Theilgaard-Monch K, Minucci S, Chin L, Artandi SE, Shen Q, Tam A, Lee SL, Gottlieb GJ, Greider
Porse BT, Marine JC, et al. 2007. The Polycomb group CW, DePinho RA. 1999. p53 deficiency rescues the adverse
proteins bind throughout the INK4A-ARF locus and are effects of telomere loss and cooperates with telomere dys-
disassociated in senescent cells. Genes Dev 21: 525– function to accelerate carcinogenesis. Cell 97: 527–538.
530. Collado M, Serrano M. 2010. Senescence in tumours: Evidence
Braig M, Lee S, Loddenkemper C, Rudolph C, Peters AHFM, from mice and humans. Nat Rev Cancer 10: 51–57.
Schlegelberger B, Stein H, Dörken B, Jenuwein T, Schmitt Collado M, Gil J, Efeyan A, Guerra C, Schuhmacher AJ, Barradas
CA. 2005. Oncogene-induced senescence as an initial barrier M, Bengurı́a A, Zaballos A, Flores JM, Barbacid M, et al.
in lymphoma development. Nature 436: 660–665. 2005. Tumour biology: Senescence in premalignant tumours.
Bulavin DV, Kovalsky O, Hollander MC, Fornace AJ. 2003. Loss Nature 436: 642.
of oncogenic H-ras-induced cell cycle arrest and p38 mito- Coppé J-P, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J,
gen-activated protein kinase activation by disruption of Nelson PS, Desprez P-Y, Campisi J. 2008. Senescence-asso-
Gadd45a. Mol Cell Biol 23: 3859–3871. ciated secretory phenotypes reveal cell-nonautonomous
Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, functions of oncogenic RAS and the p53 tumor suppressor.
Sedivy JM, Kinzler KW, Vogelstein B. 1998. Requirement for PLoS Biol 6: 2853–2868.

2474 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

Cosme-Blanco W, Shen M-F, Lazar AJF, Pathak S, Lozano G, response triggered by DNA hyper-replication. Nature 444:
Multani AS, Chang S. 2007. Telomere dysfunction sup- 638–642.
presses spontaneous tumorigenesis in vivo by initiating Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C,
p53-dependent cellular senescence. EMBO Rep 8: 497–503. Medrano EE, Linskens M, Rubelj I, Pereira-Smith OM.
Courtois-Cox S, Genther Williams SM, Reczek EE, Johnson BW, 1995. A biomarker that identifies senescent human cells
McGillicuddy LT, Johannessen CM, Hollstein PE, MacCollin in culture and in aging skin in vivo. Proc Natl Acad Sci 92:
M, Cichowski K. 2006. A negative feedback signaling network 9363–9367.
underlies oncogene-induced senescence. Cancer Cell 10: 459– Dirac AMG, Bernards R. 2003. Reversal of senescence in mouse
472. fibroblasts through lentiviral suppression of p53. J Biol Chem
Cristofalo VJ, Lorenzini A, Allen RG, Torres C, Tresini M. 2004. 278: 11731–11734.
Replicative senescence: A critical review. Mech Ageing Dev Efeyan A, Murga M, Martinez-Pastor B, Ortega-Molina A, Soria
125: 827–848. R, Collado M, Fernandez-Capetillo O, Serrano M. 2009.
Cuervo AM, Dice JF. 2000. Age-related decline in chaperone- Limited role of murine ATM in oncogene-induced senes-
mediated autophagy. J Biol Chem 275: 31505–31513. cence and p53-dependent tumor suppression. PLoS ONE
D’Adda di Fagagna F. 2008. Living on a break: Cellular senes- 4: e5475. doi: 10.1371/journal.pone.0005475.
cence as a DNA-damage response. Nat Rev Cancer 8: 512– Elenbaas B, Spirio L, Koerner F, Fleming MD, Zimonjic DB,
522. Donaher JL, Popescu NC, Hahn WC, Weinberg RA. 2001.
D’Adda di Fagagna F, Reaper PM, Clay-Farrace L, Fiegler H, Carr Human breast cancer cells generated by oncogenic trans-
P, Von Zglinicki T, Saretzki G, Carter NP, Jackson SP. 2003. formation of primary mammary epithelial cells. Genes Dev
A DNA damage checkpoint response in telomere-initiated 15: 50–65.
senescence. Nature 426: 194–198. Erickson S, Sangfelt O, Heyman M, Castro J, Einhorn S, Grandér
Dai CY, Furth EE, Mick R, Koh J, Takayama T, Niitsu Y, Enders D. 1998. Involvement of the Ink4 proteins p16 and p15 in
GH. 2000. p16(INK4a) expression begins early in human T-lymphocyte senescence. Oncogene 17: 595–602.
colon neoplasia and correlates inversely with markers of cell Feldser DM, Greider CW. 2007. Short telomeres limit tumor
proliferation. Gastroenterology 119: 929–942. progression in vivo by inducing senescence. Cancer Cell 11:
Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, 461–469.
Damsky WE, You MJ, Depinho RA, McMahon M, Bosenberg Finney RE, Bishop JM. 1993. Predisposition to neoplastic trans-
M. 2009. Braf(V600E) cooperates with Pten loss to induce formation caused by gene replacement of H-ras1. Science
metastatic melanoma. Nat Genet 41: 544–552. 260: 1524–1527.
Dannenberg JH, van Rossum A, Schuijff L, te Riele H. 2000. Foreman KE, Tang J. 2003. Molecular mechanisms of replica-
Ablation of the retinoblastoma gene family deregulates G(1) tive senescence in endothelial cells. Exp Gerontol 38: 1251–
control causing immortalization and increased cell turnover 1257.
under growth-restricting conditions. Genes Dev 14: 3051– Franza BR, Maruyama K, Garrels JI, Ruley HE. 1986. In vitro
3064. establishment is not a sufficient prerequisite for transforma-
Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, tion by activated ras oncogenes. Cell 44: 409–418.
Teague J, Woffendin H, Garnett MJ, Bottomley W, et al. 2002. Freedberg DE, Rigas SH, Russak J, Gai W, Kaplow M, Osman I,
Mutations of the BRAF gene in human cancer. Nature 417: Turner F, Randerson-Moor JA, Houghton A, Busam K, et al.
949–954. 2008. Frequent p16-independent inactivation of p14ARF in
Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. human melanoma. J Natl Cancer Inst 100: 784–795.
2009. Protocols to detect senescence-associated b-galactosi- Fujita K, Mondal AM, Horikawa I, Nguyen GH, Kumamoto K,
dase (SA-bgal) activity, a biomarker of senescent cells in Sohn JJ, Bowman ED, Mathe EA, Schetter AJ, Pine SR, et al.
culture and in vivo. Nat Protoc 4: 1798–1806. 2009. p53 isoforms D133p53 and p53b are endogenous
Deng Q, Liao R, Wu B-L, Sun P. 2004. High intensity ras regulators of replicative cellular senescence. Nat Cell Biol
signaling induces premature senescence by activating p38 11: 1135–1142.
pathway in primary human fibroblasts. J Biol Chem 279: Furumoto K, Inoue E, Nagao N, Hiyama E, Miwa N. 1998. Age-
1050–1059. dependent telomere shortening is slowed down by enrich-
Denoyelle C, Abou-Rjaily G, Bezrookove V, Verhaegen M, ment of intracellular vitamin C via suppression of oxidative
Johnson TM, Fullen DR, Pointer JN, Gruber SB, Su LD, stress. Life Sci 63: 935–948.
Nikiforov MA, et al. 2006. Anti-oncogenic role of the Gil J, Bernard D, Martinez D, Beach D. 2004. Polycomb CBX7
endoplasmic reticulum differentially activated by mutations has a unifying role in cellular lifespan. Nat Cell Biol 6: 67–
in the MAPK pathway. Nat Cell Biol 8: 1053–1063. 72.
Dhomen N, Reis-Filho JS, Da Rocha Dias S, Hayward R, Savage Goel VK, Ibrahim N, Jiang G, Singhal M, Fee S, Flotte T,
K, Delmas V, Larue L, Pritchard C, Marais R. 2009. Onco- Westmoreland S, Haluska FS, Hinds PW, Haluska FG. 2009.
genic Braf induces melanocyte senescence and melanoma in Melanocytic nevus-like hyperplasia and melanoma in trans-
mice. Cancer Cell 15: 294–303. genic BRAFV600E mice. Oncogene 28: 2289–2298.
Di Leonardo A, Linke SP, Clarkin K, Wahl GM. 1994. DNA Going JJ, Stuart RC, Downie M, Fletcher-Monaghan AJ, Keith
damage triggers a prolonged p53-dependent G1 arrest and WN. 2002. ‘Senescence-associated’ b-galactosidase activity
long-term induction of Cip1 in normal human fibroblasts. in the upper gastrointestinal tract. J Pathol 196: 394–400.
Genes Dev 8: 2540–2551. Goldstein S, Moerman EJ, Fujii S, Sobel BE. 1994. Overexpres-
Dilley TK, Bowden GT, Chen QM. 2003. Novel mechanisms of sion of plasminogen activator inhibitor type-1 in senescent
sublethal oxidant toxicity: Induction of premature senes- fibroblasts from normal subjects and those with Werner
cence in human fibroblasts confers tumor promoter activity. syndrome. J Cell Physiol 161: 571–579.
Exp Cell Res 290: 38–48. Gray-Schopfer VC, Cheong SC, Chong H, Chow J, Moss T,
Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, Abdel-Malek ZA, Marais R, Wynford-Thomas D, Bennett
Luise C, Schurra C, Garré M, Nuciforo PG, Bensimon A, DC. 2006. Cellular senescence in naevi and immortalisation
et al. 2006. Oncogene-induced senescence is a DNA damage in melanoma: A role for p16? Br J Cancer 95: 496–505.

GENES & DEVELOPMENT 2475


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

Greenberg RA, Chin L, Femino A, Lee KH, Gottlieb GJ, Singer Kamijo T, Zindy F, Roussel MF, Quelle DE, Downing JR,
RH, Greider CW, DePinho RA. 1999. Short dysfunctional Ashmun RA, Grosveld G, Sherr CJ. 1997. Tumor suppression
telomeres impair tumorigenesis in the INK4a(D2/3) cancer- at the mouse INK4a locus mediated by the alternative
prone mouse. Cell 97: 515–525. reading frame product p19ARF. Cell 91: 649–659.
Gruis NA, van der Velden PA, Sandkuijl LA, Prins DE, Weaver- Kato D, Miyazawa K, Ruas M, Starborg M, Wada I, Oka T, Sakai
Feldhaus J, Kamb A, Bergman W, Frants RR. 1995. Homozy- T, Peters G, Hara E. 1998. Features of replicative senescence
gotes for CDKN2 (p16) germline mutation in Dutch familial induced by direct addition of antennapedia-p16INK4A fusion
melanoma kindreds. Nat Genet 10: 351–353. protein to human diploid fibroblasts. FEBS Lett 427: 203–
Guerra C, Mijimolle N, Dhawahir A, Dubus P, Barradas M, 208.
Serrano M, Campuzano V, Barbacid M. 2003. Tumor in- Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya
duction by an endogenous K-ras oncogene is highly depen- A, Wahl GM, Belmonte JCI. 2009. Linking the p53 tumour
dent on cellular context. Cancer Cell 4: 111–120. suppressor pathway to somatic cell reprogramming. Nature
Haferkamp S, Scurr LL, Becker TM, Frausto M, Kefford RF, 460: 1140–1144.
Rizos H. 2009. Oncogene-induced senescence does not re- Kim K, Kang K, Seu Y, Baek S, Kim J. 2009. Interferon-g induces
quire the p16(INK4a) or p14ARF melanoma tumor suppres- cellular senescence through p53-dependent DNA damage
sors. J Invest Dermatol 129: 1983–1991. signaling in human endothelial cells. Mech Ageing Dev
Hanna J, Saha K, Pando B, van Zon J, Lengner CJ, Creyghton MP, 130: 179–188.
van Oudenaarden A, Jaenisch R. 2009. Direct cell reprogram- Kipling D, Cooke HJ. 1990. Hypervariable ultra-long telomeres
ming is a stochastic process amenable to acceleration. in mice. Nature 347: 400–402.
Nature 462: 595–601. Kiyono T, Foster SA, Koop JI, McDougall JK, Galloway DA,
Hara E, Smith R, Parry D, Tahara H, Stone S, Peters G. 1996. Klingelhutz AJ. 1998. Both Rb/p16INK4a inactivation and
Regulation of p16CDKN2 expression and its implications for telomerase activity are required to immortalize human
cell immortalization and senescence. Mol Cell Biol 16: 859– epithelial cells. Nature 396: 84–88.
867. Kortlever RM, Higgins PJ, Bernards R. 2006. Plasminogen
Harley CB, Futcher AB, Greider CW. 1990. Telomeres shorten activator inhibitor-1 is a critical downstream target of p53
during ageing of human fibroblasts. Nature 345: 458–460. in the induction of replicative senescence. Nat Cell Biol 8:
Haugstetter AM, Loddenkemper C, Lenze D, Grone J, Standfuss 877–884.
C, Petersen I, Dorken B, Schmitt CA. 2010. Cellular senes- Krimpenfort P, Quon KC, Mooi WJ, Loonstra A, Berns A. 2001.
cence predicts treatment outcome in metastasised colorectal Loss of p16Ink4a confers susceptibility to metastatic mela-
cancer. Br J Cancer 103: 505–509. noma in mice. Nature 413: 83–86.
Hayflick L. 1965. The limited in vitro lifetime of human diploid Krizhanovsky V, Lowe SW. 2009. Stem cells: The promises and
cell strains. Exp Cell Res 37: 614–636. perils of p53. Nature 460: 1085–1086.
Hayflick L, Moorhead PS. 1961. The serial cultivation of human Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething
diploid cell strains. Exp Cell Res 25: 585–621. C, Yee H, Zender L, Lowe SW. 2008. Senescence of acti-
Herbert B-S, Wright WE, Shay JW. 2002. p16(INK4a) inactivation vated stellate cells limits liver fibrosis. Cell 134: 657–
is not required to immortalize human mammary epithelial 667.
cells. Oncogene 21: 7897–7900. Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J. 2001.
Herbig U, Ferreira M, Condel L, Carey D, Sedivy JM. 2006. Senescent fibroblasts promote epithelial cell growth and
Cellular senescence in aging primates. Science 311: 1257. tumorigenesis: A link between cancer and aging. Proc Natl
Hitomi T, Matsuzaki Y, Yasuda S, Kawanaka M, Yogosawa S, Acad Sci 98: 12072–12077.
Koyama M, Tantin D, Sakai T. 2007. Oct-1 is involved in the Kuilman T, Peeper DS. 2009. Senescence-messaging secretome:
transcriptional repression of the p15(INK4b) gene. FEBS Lett SMS-ing cellular stress. Nat Rev Cancer 9: 81–94.
581: 1087–1092. Kuilman T, Michaloglou C, Vredeveld LCW, Douma S, van
Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa Doorn R, Desmet CJ, Aarden LA, Mooi WJ, Peeper DS.
M, Okita K, Yamanaka S. 2009. Suppression of induced 2008. Oncogene-induced senescence relayed by an interleu-
pluripotent stem cell generation by the p53–p21 pathway. kin-dependent inflammatory network. Cell 133: 1019–
Nature 460: 1132–1135. 1031.
Itahana K, Zou Y, Itahana Y, Martinez J-L, Beausejour C, Jacobs Kurz DJ, Decary S, Hong Y, Erusalimsky JD. 2000. Senescence-
JJL, van Lohuizen M, Band V, Campisi J, Dimri GP. 2003. associated b-galactosidase reflects an increase in lysosomal
Control of the replicative life span of human fibroblasts by mass during replicative ageing of human endothelial cells.
p16 and the polycomb protein Bmi-1. Mol Cell Biol 23: 389– J Cell Sci 113: 3613–3622.
401. Land H, Parada LF, Weinberg RA. 1983. Tumorigenic conversion
Iwasa H, Han J, Ishikawa F. 2003. Mitogen-activated protein of primary embryo fibroblasts requires at least two cooperat-
kinase p38 defines the common senescence-signalling path- ing oncogenes. Nature 304: 596–602.
way. Genes Cells 8: 131–144. Lazzerini Denchi E, Attwooll C, Pasini D, Helin K. 2005.
Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M. Deregulated E2F activity induces hyperplasia and senes-
1999. The oncogene and Polycomb-group gene bmi-1 regu- cence-like features in the mouse pituitary gland. Mol Cell
lates cell proliferation and senescence through the ink4a Biol 25: 2660–2672.
locus. Nature 397: 164–168. Lee AC, Fenster BE, Ito H, Takeda K, Bae NS, Hirai T, Yu ZX,
Jeyapalan JC, Ferreira M, Sedivy JM, Herbig U. 2007. Accumu- Ferrans VJ, Howard BH, Finkel T. 1999. Ras proteins induce
lation of senescent cells in mitotic tissue of aging primates. senescence by altering the intracellular levels of reactive
Mech Ageing Dev 128: 36–44. oxygen species. J Biol Chem 274: 7936–7940.
Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng Lee BY, Han JA, Im JS, Morrone A, Johung K, Goodwin EC,
A, Labourier E, Reinert KL, Brown D, Slack FJ. 2005. RAS is Kleijer WJ, DiMaio D, Hwang ES. 2006. Senescence-associ-
regulated by the let-7 microRNA family. Cell 120: 635– ated b-galactosidase is lysosomal b-galactosidase. Aging Cell
647. 5: 187–195.

2476 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

Levine B, Kroemer G. 2008. Autophagy in the pathogenesis of Muntoni, A., Reddel, R.R. 2005. The first molecular details of
disease. Cell 132: 27–42. ALT in human tumor cells. Hum Mol Genet 14: R191–R196.
Li H, Collado M, Villasante A, Strati K, Ortega S, Cañamero M, doi: 10.1093/hmg/ddi266.
Blasco MA, Serrano M. 2009. The Ink4/Arf locus is a barrier Narita M, Nũnez S, Heard E, Narita M, Lin AW, Hearn SA,
for iPS cell reprogramming. Nature 460: 1136–1139. Spector DL, Hannon GJ, Lowe SW. 2003. Rb-mediated
Lin AW, Barradas M, Stone JC, van Aelst L, Serrano M, Lowe SW. heterochromatin formation and silencing of E2F target genes
1998. Premature senescence involving p53 and p16 is acti- during cellular senescence. Cell 113: 703–716.
vated in response to constitutive MEK/MAPK mitogenic Narita M, Narita M, Krizhanovsky V, Nuñez S, Chicas A, Hearn
signaling. Genes Dev 12: 3008–3019. SA, Myers MP, Lowe SW. 2006. A novel role for high-mobility
Lin H-K, Chen Z, Wang G, Nardella C, Lee S-W, Chan C-H, Yang group a proteins in cellular senescence and heterochromatin
W-L, Wang J, Egia A, Nakayama KI, et al. 2010. Skp2 formation. Cell 126: 503–514.
targeting suppresses tumorigenesis by Arf-p53-independent Nicke B, Bastien J, Khanna SJ, Warne PH, Cowling V, Cook SJ,
cellular senescence. Nature 464: 374–379. Peters G, Delpuech O, Schulze A, Berns K, et al. 2005.
Loo DT, Fuquay JI, Rawson CL, Barnes DW. 1987. Extended Involvement of MINK, a Ste20 family kinase, in Ras onco-
culture of mouse embryo cells without senescence: Inhibi- gene-induced growth arrest in human ovarian surface epi-
tion by serum. Science 236: 200–202. thelial cells. Mol Cell 20: 673–685.
Lowe SW, Cepero E, Evan GI. 2004. Intrinsic tumour suppres- Okamura M, Unami A, Matsumoto M, Oishi Y, Kashida Y,
sion. Nature 432: 307–315. Mitsumori K. 2006. Gene expression analysis of urethane-
Lu T, Finkel T. 2008. Free radicals and senescence. Exp Cell Res induced lung tumors in ras H2 mice. Toxicology 217: 129–138.
314: 1918–1922. Olovnikov AM. 1971. Principle of marginotomy in template
Lu C, Vickers MF, Kerbel RS. 1992. Interleukin 6: A fibroblast- synthesis of polynucleotides. Dokl Akad Nauk SSSR 201:
derived growth inhibitor of human melanoma cells from 1496–1499.
early but not advanced stages of tumor progression. Proc Olsen CL, Gardie B, Yaswen P, Stampfer MR. 2002. Raf-1-
Natl Acad Sci 89: 9215–9219. induced growth arrest in human mammary epithelial cells
Lu C, Sheehan C, Rak JW, Chambers CA, Hozumi N, Kerbel RS. is p16-independent and is overcome in immortal cells during
1996. Endogenous interleukin 6 can function as an in vivo conversion. Oncogene 21: 6328–6339.
growth- stimulatory factor for advanced-stage human mela- Packer L, Fuehr K. 1977. Low oxygen concentration extends the
noma cells. Clin Cancer Res 2: 1417–1425. lifespan of cultured human diploid cells. Nature 267: 423–
Maldonado JL, Timmerman L, Fridlyand J, Bastian BC. 2004. 425.
Mechanisms of cell-cycle arrest in Spitz nevi with constitu- Park HK, Leonard DD, Arrington JH, Lund HZ. 1987. Recurrent
tive activation of the MAP-kinase pathway. Am J Pathol 164: melanocytic nevi: Clinical and histologic review of 175
1783–1787. cases. J Am Acad Dermatol 17: 285–292.
Mallette FA, Gaumont-Leclerc M-F, Ferbeyre G. 2007. The Parrinello S, Samper E, Krtolica A, Goldstein J, Melov S,
DNA damage signaling pathway is a critical mediator of Campisi J. 2003. Oxygen sensitivity severely limits the
oncogene-induced senescence. Genes Dev 21: 43–48. replicative lifespan of murine fibroblasts. Nat Cell Biol 5:
Marión RM, Strati K, Li H, Murga M, Blanco R, Ortega S, 741–747.
Fernandez-Capetillo O, Serrano M, Blasco MA. 2009. A p53- Parrinello S, Coppe J-P, Krtolica A, Campisi J. 2005. Stromal–
mediated DNA damage response limits reprogramming to epithelial interactions in aging and cancer: Senescent fibro-
ensure iPS cell genomic integrity. Nature 460: 1149–1153. blasts alter epithelial cell differentiation. J Cell Sci 118: 485–
Massagué J. 2008. TGFb in cancer. Cell 134: 215–230. 496.
Masutomi K, Yu EY, Khurts S, Ben-Porath I, Currier JL, Metz Passos JF, Nelson G, Wang C, Richter T, Simillion C, Proctor CJ,
GB, Brooks MW, Kaneko S, Murakami S, DeCaprio JA, et al. Miwa S, Olijslagers S, Hallinan J, Wipat A, et al. 2010.
2003. Telomerase maintains telomere structure in normal Feedback between p21 and reactive oxygen production is
human cells. Cell 114: 241–253. necessary for cell senescence. Mol Syst Biol 6: 347.
Mathon NF, Malcolm DS, Harrisingh MC, Cheng L, Lloyd AC. Patschan S, Chen J, Polotskaia A, Mendelev N, Cheng J,
2001. Lack of replicative senescence in normal rodent glia. Patschan D, Goligorsky MS. 2008. Lipid mediators of
Science 291: 872–875. autophagy in stress-induced premature senescence of endo-
Michaloglou C, Vredeveld LCW, Soengas MS, Denoyelle C, thelial cells. Am J Physiol Heart Circ Physiol 294: H1119–
Kuilman T, van der Horst CMAM, Majoor DM, Shay JW, H1129. doi: 10.1152/ajpheart.00713.2007.
Mooi WJ, Peeper DS. 2005. BRAFE600-associated senes- Patton EE, Widlund HR, Kutok JL, Kopani KR, Amatruda JF,
cence-like cell cycle arrest of human naevi. Nature 436: Murphey RD, Berghmans S, Mayhall EA, Traver D, Fletcher
720–724. CDM, et al. 2005. BRAF mutations are sufficient to promote
Michaloglou C, Vredeveld LCW, Mooi WJ, Peeper DS. 2008. nevi formation and cooperate with p53 in the genesis of
BRAF(E600) in benign and malignant human tumours. On- melanoma. Curr Biol 15: 249–254.
cogene 27: 877–895. Peeper DS. 2009. Ras and pRb: The relationship gets yet more
Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, intimate. Cancer Cell 15: 243–245.
Komuro I. 2002. Endothelial cell senescence in human Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins
atherosclerosis: Role of telomere in endothelial dysfunction. CM, Moses TY, Hostetter G, Wagner U, Kakareka J, et al.
Circulation 105: 1541–1544. 2003. High frequency of BRAF mutations in nevi. Nat Genet
Moiseeva O, Mallette FA, Mukhopadhyay UK, Moores A, 33: 19–20.
Ferbeyre G. 2006. DNA damage signaling and p53-dependent Price JS, Waters JG, Darrah C, Pennington C, Edwards DR,
senescence after prolonged b-interferon stimulation. Mol Donell ST, Clark IM. 2002. The role of chondrocyte senes-
Biol Cell 17: 1583–1592. cence in osteoarthritis. Aging Cell 1: 57–65.
Moiseeva O, Bourdeau V, Roux A, Deschênes-Simard X, Ferbeyre Prowse KR, Greider CW. 1995. Developmental and tissue-
G. 2009. Mitochondrial dysfunction contributes to oncogene- specific regulation of mouse telomerase and telomere length.
induced senescence. Mol Cell Biol 29: 4495–4507. Proc Natl Acad Sci 92: 4818–4822.

GENES & DEVELOPMENT 2477


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

Kuilman et al.

Puyol M, Martin A, Dubus P, Mulero F, Pizcueta P, Khan G, ated with accumulation of p53 and p16INK4a. Cell 88: 593–
Guerra C, Santamaria D, Barbacid M. 2010. A synthetic 602.
lethal interaction between K-Ras oncogenes and Cdk4 un- Severino J, Allen RG, Balin S, Balin A, Cristofalo VJ. 2000. Is
veils a therapeutic strategy for non-small cell lung carci- b-galactosidase staining a marker of senescence in vitro and
noma. Cancer Cell 18: 63–73. in vivo? Exp Cell Res 257: 162–171.
Quintanilla M, Brown K, Ramsden M, Balmain A. 1986. Shamma A, Takegami Y, Miki T, Kitajima S, Noda M, Obara T,
Carcinogen-specific mutation and amplification of Ha-ras Okamoto T, Takahashi C. 2009. Rb Regulates DNA damage
during mouse skin carcinogenesis. Nature 322: 78–80. response and cellular senescence through E2F-dependent
Ramirez RD, Morales CP, Herbert BS, Rohde JM, Passons C, suppression of N-ras isoprenylation. Cancer Cell 15: 255–269.
Shay JW, Wright WE. 2001. Putative telomere-independent Sharpless NE, Bardeesy N, Lee KH, Carrasco D, Castrillon DH,
mechanisms of replicative aging reflect inadequate growth Aguirre AJ, Wu EA, Horner JW, DePinho RA. 2001. Loss of
conditions. Genes Dev 15: 398–403. p16Ink4a with retention of p19Arf predisposes mice to
Reimann M, Lee S, Loddenkemper C, Dörr JR, Tabor V, Aichele tumorigenesis. Nature 413: 86–91.
P, Stein H, Dörken B, Jenuwein T, Schmitt CA. 2010. Tumor Shay JW, Bacchetti S. 1997. A survey of telomerase activity in
stroma-derived TGF-b limits myc-driven lymphomagenesis human cancer. Eur J Cancer 33: 787–791.
via Suv39h1-dependent senescence. Cancer Cell 17: 262– Shay JW, Wright WE. 2005. Senescence and immortalization:
272. Role of telomeres and telomerase. Carcinogenesis 26: 867–
Reznikoff CA, Yeager TR, Belair CD, Savelieva E, Puthenveettil 874.
JA, Stadler WM. 1996. Elevated p16 at senescence and loss of Shay JW, Wright WE. 2006. Telomerase therapeutics for cancer:
p16 at immortalization in human papillomavirus 16 E6, but Challenges and new directions. Nat Rev Drug Discov 5: 577–
not E7, transformed human uroepithelial cells. Cancer Res 584.
56: 2886–2890. Shay JW, Pereira-Smith OM, Wright WE. 1991. A role for both
Robles SJ, Adami GR. 1998. Agents that cause DNA double RB and p53 in the regulation of human cellular senescence.
strand breaks lead to p16INK4a enrichment and the pre- Exp Cell Res 196: 33–39.
mature senescence of normal fibroblasts. Oncogene 16: Shelton DN, Chang E, Whittier PS, Choi D, Funk WD. 1999.
1113–1123. Microarray analysis of replicative senescence. Curr Biol 9:
Rodier F, Coppé J-P, Patil CK, Hoeijmakers WAM, Muñoz DP, 939–945.
Raza SR, Freund A, Campeau E, Davalos AR, Campisi J. Sherr CJ, DePinho RA. 2000. Cellular senescence: Mitotic clock
2009. Persistent DNA damage signalling triggers senescence- or culture shock? Cell 102: 407–410.
associated inflammatory cytokine secretion. Nat Cell Biol Stein GH, Beeson M, Gordon L. 1990. Failure to phosphorylate
11: 973–979. the retinoblastoma gene product in senescent human fibro-
Rodriguez-Puebla ML, LaCava M, Bolontrade MF, Russell J, blasts. Science 249: 666–669.
Conti CJ. 1999. Increased expression of mutated Ha-ras Sun P, Yoshizuka N, New L, Moser BA, Li Y, Liao R, Xie C, Chen
during premalignant progression in SENCAR mouse skin. J, Deng Q, Yamout M, et al. 2007. PRAK is essential for ras-
Mol Carcinog 26: 150–156. induced senescence and tumor suppression. Cell 128: 295–
Roninson IB. 2003. Tumor cell senescence in cancer treatment. 308.
Cancer Res 63: 2705–2715. Takahashi A, Ohtani N, Yamakoshi K, Iida S-i, Tahara H,
Sage J, Mulligan GJ, Attardi LD, Miller A, Chen S, Williams B, Nakayama K, Nakayama KI, Ide T, Saya H, Hara E. 2006.
Theodorou E, Jacks T. 2000. Targeted disruption of the three Mitogenic signalling and the p16INK4a–Rb pathway cooper-
Rb-related genes leads to loss of G(1) control and immortal- ate to enforce irreversible cellular senescence. Nat Cell Biol
ization. Genes Dev 14: 3037–3050. 8: 1291–1297.
Santoriello C, Deflorian G, Pezzimenti F, Kawakami K, Lanfrancone Tanaka N, Ishihara M, Kitagawa M, Harada H, Kimura T,
L, D’Adda di Fagagna F, Mione M. 2009. Expression of Matsuyama T, Lamphier MS, Aizawa S, Mak TW, Taniguchi
H-RASV12 in a zebrafish model of Costello syndrome causes T. 1994. Cellular commitment to oncogene-induced trans-
cellular senescence in adult proliferating cells. Dis Model formation or apoptosis is dependent on the transcription
Mech 2: 56–67. factor IRF-1. Cell 77: 829–839.
Sarkisian CJ, Keister BA, Stairs DB, Boxer RB, Moody SE, Tang DG, Tokumoto YM, Apperly JA, Lloyd AC, Raff MC. 2001.
Chodosh LA. 2007. Dose-dependent oncogene-induced se- Lack of replicative senescence in cultured rat oligodendro-
nescence in vivo and its evasion during mammary tumori- cyte precursor cells. Science 291: 868–871.
genesis. Nat Cell Biol 9: 493–505. te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP.
Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman 2002. DNA damage is able to induce senescence in tumor
RM, Lowe SW. 2002. A senescence program controlled by cells in vitro and in vivo. Cancer Res 62: 1876–1883.
p53 and p16INK4a contributes to the outcome of cancer Tuveson DA, Shaw AT, Willis NA, Silver DP, Jackson EL, Chang
therapy. Cell 109: 335–346. S, Mercer KL, Grochow R, Hock H, Crowley D, et al. 2004.
Schoenfeld R, Pinkus H. 1958. The recurrence of nevi after Endogenous oncogenic K-ras(G12D) stimulates proliferation
incomplete removal. AMA Arch Derm 78: 30–35. and widespread neoplastic and developmental defects. Can-
Schrama D, Kneitz H, Willmes C, Adam C, Houben R, Becker cer Cell 5: 375–387.
JC. 2010. Lack of correlation between IGFBP7 expression and Utikal J, Polo JM, Stadtfeld M, Maherali N, Kulalert W, Walsh
BRAF mutational status in melanoma. J Invest Dermatol RM, Khalil A, Rheinwald JG, Hochedlinger K. 2009. Immor-
130: 897–898. talization eliminates a roadblock during cellular reprogram-
Scurr LL, Pupo GM, Becker TM, Lai K, Schrama D, Haferkamp ming into iPS cells. Nature 460: 1145–1148.
S, Irvine M, Scolyer RA, Mann GJ, Becker JC, et al. 2010. van Riggelen J, Muller J, Otto T, Beuger V, Yetil A, Choi PS,
IGFBP7 is not required for B-RAF-induced melanocyte se- Kosan C, Moroy T, Felsher DW, Eilers M. 2010. The in-
nescence. Cell 141: 717–727. teraction between Myc and Miz1 is required to antagonize
Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. 1997. TGFb-dependent autocrine signaling during lymphoma for-
Oncogenic ras provokes premature cell senescence associ- mation and maintenance. Genes Dev 24: 1281–1294.

2478 GENES & DEVELOPMENT


Downloaded from genesdev.cshlp.org on January 20, 2011 - Published by Cold Spring Harbor Laboratory Press

The essence of senescence

Vaziri H, Benchimol S. 1998. Reconstitution of telomerase lated oncogene 1 (Gro-1) links RAS signaling to the senes-
activity in normal human cells leads to elongation of telo- cence of stromal fibroblasts and ovarian tumorigenesis. Proc
meres and extended replicative life span. Curr Biol 8: 279–282. Natl Acad Sci 103: 16472–16477.
Vaziri H, West MD, Allsopp RC, Davison TS, Wu YS, Arrowsmith Ye X, Zerlanko B, Kennedy A, Banumathy G, Zhang R,
CH, Poirier GG, Benchimol S. 1997. ATM-dependent telo- Adams PD. 2007. Downregulation of Wnt signaling is
mere loss in aging human diploid fibroblasts and DNA a trigger for formation of facultative heterochromatin and
damage lead to the post-translational activation of p53 pro- onset of cell senescence in primary human cells. Mol Cell
tein involving poly(ADP-ribose) polymerase. EMBO J 16: 27: 183–196.
6018–6033. Young NP, Jacks T. 2010. Tissue-specific p19Arf regulation
Ventura A, Kirsch DG, Mclaughlin ME, Tuveson DA, Grimm J, dictates the response to oncogenic K-ras. Proc Natl Acad
Lintault L, Newman J, Reczek EE, Weissleder R, Jacks T. Sci 107: 10184–10189.
2007. Restoration of p53 function leads to tumour regression Young AP, Schlisio S, Minamishima YA, Zhang Q, Li L,
in vivo. Nature 445: 661–665. Grisanzio C, Signoretti S, Kaelin WG. 2008. VHL loss
Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. actuates a HIF-independent senescence programme medi-
2008. Oncogenic BRAF induces senescence and apoptosis ated by Rb and p400. Nat Cell Biol 10: 361–369.
through pathways mediated by the secreted protein IGFBP7. Young ARJ, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot
Cell 132: 363–374. JFJ, Tavaré S, Arakawa S, Shimizu S, Watt FM, et al. 2009.
Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. Autophagy mediates the mitotic senescence transition.
2010. Role for IGFBP7 in senescence induction by BRAF. Cell Genes Dev 23: 798–803.
141: 746–747. Yuan H, Kaneko T, Matsuo M. 1995. Relevance of oxidative
Wang X, Wong SC, Pan J, Tsao SW, Fung KH, Kwong DL, Sham stress to the limited replicative capacity of cultured human
JS, Nicholls JM. 1998. Evidence of cisplatin-induced senes- diploid cells: The limit of cumulative population doublings
cent-like growth arrest in nasopharyngeal carcinoma cells. increases under low concentrations of oxygen and decreases
Cancer Res 58: 5019–5022. in response to aminotriazole. Mech Ageing Dev 81: 159–168.
Wang W, Chen JX, Liao R, Deng Q, Zhou JJ, Huang S, Sun P. Zhang R, Poustovoitov MV, Ye X, Santos HA, Chen W, Daganzo
2002. Sequential activation of the MEK-extracellular signal- SM, Erzberger JP, Serebriiskii IG, Canutescu AA, Dunbrack
regulated kinase and MKK3/6-p38 mitogen-activated protein RL, et al. 2005. Formation of MacroH2A-containing senes-
kinase pathways mediates oncogenic ras-induced premature cence-associated heterochromatin foci and senescence
senescence. Mol Cell Biol 22: 3389–3403. driven by ASF1a and HIRA. Dev Cell 8: 19–30.
Watson JD. 1972. Origin of concatemeric T7 DNA. Nat New Zhang W, Chan HM, Gao Y, Poon R, Wu Z. 2007. BS69 is
Biol 239: 197–201. involved in cellular senescence through the p53-p21Cip1
Wei S, Sedivy JM. 1999. Expression of catalytically active pathway. EMBO Rep 8: 952–958.
telomerase does not prevent premature senescence caused Zhu J, Woods D, McMahon M, Bishop JM. 1998. Senescence of
by overexpression of oncogenic Ha-Ras in normal human human fibroblasts induced by oncogenic Raf. Genes Dev 12:
fibroblasts. Cancer Res 59: 1539–1543. 2997–3007.
Wei W, Hemmer RM, Sedivy JM. 2001. Role of p14(ARF) in Zindy F, Williams RT, Baudino TA, Rehg JE, Skapek SX,
replicative and induced senescence of human fibroblasts. Cleveland JL, Roussel MF, Sherr CJ. 2003. Arf tumor sup-
Mol Cell Biol 21: 6748–6757. pressor promoter monitors latent oncogenic signals in vivo.
Woods D, Parry D, Cherwinski H, Bosch E, Lees E, McMahon Proc Natl Acad Sci 100: 15930–15935.
M. 1997. Raf-induced proliferation or cell cycle arrest is
determined by the level of Raf activity with arrest mediated
by p21Cip1. Mol Cell Biol 17: 5598–5611.
Wright WE, Shay JW. 2002. Historical claims and current
interpretations of replicative aging. Nat Biotechnol 20:
682–688.
Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW. 1996.
Telomerase activity in human germline and embryonic
tissues and cells. Dev Genet 18: 173–179.
Wu C-H, van Riggelen J, Yetil A, Fan AC, Bachireddy P, Felsher
DW. 2007. Cellular senescence is an important mechanism
of tumor regression upon c-Myc inactivation. Proc Natl
Acad Sci 104: 13028–13033.
Xue W, Zender L, Miething C, Dickins RA, Hernando E,
Krizhanovsky V, Cordon-Cardo C, Lowe SW. 2007. Senes-
cence and tumour clearance is triggered by p53 restoration in
murine liver carcinomas. Nature 445: 656–660.
Yamakoshi K, Takahashi A, Hirota F, Nakayama R, Ishimaru N,
Kubo Y, Mann DJ, Ohmura M, Hirao A, Saya H, et al. 2009.
Real-time in vivo imaging of p16Ink4a reveals cross talk
with p53. J Cell Biol 186: 393–407.
Yang N-C, Hu M-L. 2005. The limitations and validities of
senescence associated-b-galactosidase activity as an aging
marker for human foreskin fibroblast Hs68 cells. Exp Gerontol
40: 813–819.
Yang G, Rosen DG, Zhang Z, Bast RC, Mills GB, Colacino JA,
Mercado-Uribe I, Liu J. 2006. The chemokine growth-regu-

GENES & DEVELOPMENT 2479

Das könnte Ihnen auch gefallen