Vous êtes sur la page 1sur 22

AREV403-PM05-05 ARI 10 December 2009 16:41

ANNUAL
REVIEWS Further The Senescence-Associated
Click here for quick links to
Annual Reviews content online,
including:
Secretory Phenotype:
Other articles in this volume
Top cited articles The Dark Side of Tumor
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

Top downloaded articles


Our comprehensive search
Suppression
Jean-Philippe Coppe,1 Pierre-Yves Desprez,2,3
by University of Sussex on 10/10/12. For personal use only.

Ana Krtolica,1 and Judith Campisi1,2


1
Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
2
Buck Institute for Age Research, Novato, California 94945
3
California Pacic Medical Center, Cancer Research Institute, San Francisco,
California 94107; email: pydesprez@cpmcri.org

Annu. Rev. Pathol. Mech. Dis. 2010. 5:99118 Key Words


First published online as a Review in Advance on aging, cancer, inammation, proliferation, invasion
September 28, 2009

The Annual Review of Pathology: Mechanisms of Abstract


Disease is online at pathmechdis.annualreviews.org
Cellular senescence is a tumor-suppressive mechanism that perma-
This articles doi: nently arrests cells at risk for malignant transformation. However, ac-
10.1146/annurev-pathol-121808-102144
cumulating evidence shows that senescent cells can have deleterious
Copyright  c 2010 by Annual Reviews. effects on the tissue microenvironment. The most signicant of these
All rights reserved
effects is the acquisition of a senescence-associated secretory phenotype
1553-4006/10/0228-0099$20.00 (SASP) that turns senescent broblasts into proinammatory cells that
have the ability to promote tumor progression.

99
AREV403-PM05-05 ARI 10 December 2009 16:41

INTRODUCTION response and is the focus of this review. We par-


ticularly emphasize the potential effects of the
The tissue microenvironment is dened by the
SASP (20) on cell behavior in the context of
phenotypes of the cells in the immediate area
tumor progression.
and by the physical and chemical properties
of the soluble and insoluble factors surround-
ing cells within a given tissue. These proper-
ties include temperature and oxygen tension, CELLULAR SENESCENCE
as well as various molecules that may be pro- Cellular senescence occurs in culture and in
duced locallyfor example, growth factors and vivo as a response to excessive extracellular or
cytokines. Further, cells within tissues form a intracellular stress. The senescence program
dynamic network that contributes to their mi- locks the cells into a cell-cycle arrest that pre-
croenvironment. At the same time, the tissue vents the spread of damage to the next cell
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

microenvironment regulates cell behavior. This generation and precludes potential malignant
reciprocal relationship determines tissue func- transformation (19). Senescent cells have been
tion and repair and is also central to a number shown to accumulate over the life span of ro-
of pathologies, including cancer. dents, nonhuman primates, and humans (21).
by University of Sussex on 10/10/12. For personal use only.

A permissive microenvironment supports These cells are found primarily in renewable


and promotes tumor growth and cancer cell tissues and in tissues that experience prolonged
aggressiveness (14). Alterations in the cellu- inammation.
lar and molecular composition of the connec- A plethora of stresses can provoke cellu-
tive tissues surrounding carcinomas allow tu- lar senescence (22, 23). These stresses include
mors to evade detection by the immune sys- telomeric dysfunction (telomere uncapping) re-
tem as well as to proliferate inappropriately, sulting from repeated cell division (termed
invade the surrounding tissue structure, and replicative senescence), mitochondrial deteri-
eventually metastasize. The synergy between oration, oxidative stress, severe or irrepara-
an altered microenvironment and the genetic ble DNA damage and chromatin disruption
alterations acquired by tumor cells allows these (genotoxic stress), and the expression of certain
cells to evade preventive mechanisms and be- oncogenes (oncogene-induced senescence) (see
come fully malignant. Cellular senescence is Figure 1) (2431). Stresses that cause cellular
now recognized as a potent tumor-suppressive senescence can be induced by external or inter-
mechanism that arrests the growth of cells at nal chemical and physical insults encountered
risk for malignant transformation (512). How- during the course of the life span, during thera-
ever, recent studies show that senescent cells peutic interventions (for example, X-irradiation
develop altered secretory activities that may in- or chemotherapy), or as a consequence of en-
duce changes in the tissue microenvironment, dogenous processes such as oxidative respira-
relaxing its control over cell behavior and pro- tion and mitogenic signals. External mitogenic
moting tumorigenesis (1318). signals, for example growth-related oncogene
How can the senescence response be both alpha (GRO) secretion by tumor cells in close
tumor suppressive and procarcinogenic? It is proximity to normal cells (32) or circulating an-
important to consider that a biological process giotensin II (33, 34), have also been shown to
such as cellular senescence can be both bene- induce cellular senescence. All somatic cells that
cial and deleterious. The idea that processes have the ability to divide can undergo senes-
can have such dual effects is consistent with cence. Regardless of the disparate mechanisms
a major evolutionary theory of aging termed of senescence-inducing stresses, the senescence
antagonistic pleiotropy (19). The senescence- program is activated once a cell has sensed a
associated secretory phenotype (SASP) repre- critical level of damage or dysfunction. So far,
sents one of the darkest sides of the senescence the senescence growth arrest has been shown

100 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

to depend on the activities of the major tumor-


Chromatin Overexpressed
suppressor pathways controlled by p16INK4a and instability cell-cycle inhibitors
pRB (retinoblastoma protein), as well as by p53. Irreversible
(p16, p21)
Some of the molecules involved in pathways cell-cycle
upstream and downstream of the senescence- arrest Strong mitogenic/
associated phenotype have been used as mark- Nontelomeric stress signals
DNA damage
ers to detect senescent cells in culture and
in vivo.

Short/dysfunctional Oncogenes/oncogenic
THE SECRETORY PHENOTYPE telomeres mutations
OF SENESCENT CELLS
DNA damage = SASP
The senescent phenotype is not limited to an ar-
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

No DNA damage = No SASP


rest of cell proliferation. In fact, a senescent cell
Figure 1
is a potentially persisting cell that is metabol-
Multiple types of stimuli can provoke cellular senescence and a senescence-
ically active and has undergone widespread associated secretory phenotype (SASP). When irreversible cell-cycle arrest is
changes in protein expression and secretion,
by University of Sussex on 10/10/12. For personal use only.

triggered by severe DNA damage (i.e., dysfunctional telomeres or oncogenic


ultimately developing the SASP. This pheno- stress), the SASP occurs in senescent cells. However, when a senescent-like
type has also been termed the senescence- phenotype is triggered in cells that overexpress cell-cycle inhibitors such as p16
messaging secretome (35). We recently pro- or p21, cells undergo a growth arrest with many characteristics of senescent
cells, but not a SASP.
vided a large-scale characterization of the SASP,
using antibody arrays to quantitatively mea-
may lead to multiple pathologies, including
sure factors secreted by human broblasts and
cancer. SASP factors can be globally divided
epithelial cells (18), as well as mouse brob-
into the following major categories: soluble sig-
lasts ( J.P. Coppe & J. Campisi, unpublished
naling factors (interleukins, chemokines, and
data). The potential existence of the SASP was
growth factors), secreted proteases, and se-
already suggested by large-scale comparative
creted insoluble proteins/extracellular matrix
gene (mRNA) expression studies performed on
(ECM) components. SASP proteases can have
broblasts from different-aged donors and dif-
three major effects: (a) shedding of membrane-
ferent tissues of origin (3646). Among the cells
associated proteins, resulting in soluble versions
that have been shown to senesce and secrete bi-
of membrane-bound receptors, (b) cleavage/
ologically active molecules are liver stellate cells
degradation of signaling molecules, and/or
(47), endothelial cells (36, 4851), and epithelial
(c) degradation or processing of the ECM.
cells of the retinal pigment, mammary gland,
These activities provide potent mechanisms by
colon, lung, pancreas, and prostate (8, 18, 36,
which senescent cells can modify the tissue mi-
41, 5256).
croenvironment. In the following sections, we
Senescence-associated changes in gene ex-
discuss these SASP subsets and some of their
pression are specic and mostly conserved
known paracrine effects on nearby cells, with
within individual cell types. Most differences
an emphasis on their ability to facilitate cancer
between the molecular signatures of presenes-
progression.
cent and senescent cells entail cell-cycle- and
metabolism-related genes, as well as genes en-
coding the secretory proteins that constitute Soluble Signaling Factors as Major
the SASP. The SASP includes several families Components of the Senescence-
of soluble and insoluble factors (see Table 1). Associated Secretory Phenotype
These factors can affect surrounding cells by ac- Senescent cells secrete interleukins, inamma-
tivating various cell-surface receptors and cor- tory cytokines, and growth factors that can af-
responding signal transduction pathways that fect surrounding cells.

www.annualreviews.org SASP and Cancer 101


AREV403-PM05-05 ARI 10 December 2009 16:41

Table 1 The senescence-associated secretory phenotype (SASP). Factors significantly


altered between presenescent and senescent states are listed
Secretory profile of Changes in the SASP due to the loss
SASP factorsa senescent cellsb of p53 and/or gain of oncogenic RAS
Soluble factors
Interleukins (IL)
IL-6
IL-7
IL-1a, -1b
IL-13
IL-15
Chemokines (CXCL, CCL)
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

IL-8
GRO-a,-b,-gc
MCP-2
MCP-4
by University of Sussex on 10/10/12. For personal use only.

MIP-1a
MIP-3a
HCC-4
Eotaxin
Eotaxin-3
TECK
ENA-78
I-309
I-TAC
Other inammatory factors
GM-CSF
G-CSF
IFN-
BLC
MIF
Growth factors and regulators
Amphiregulin
Epiregulin
Heregulin
EGF or
bFGF
HGF
KGF (FGF7)
VEGF
Angiogenin
SCF
SDF-1 or
PIGF
NGF
(Continued )

102 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

Table 1 (Continued )
Secretory profile of Changes in the SASP due to the loss
SASP factorsa senescent cellsb of p53 and/or gain of oncogenic RAS
IGFBP-2, -3, -4, -6, -7 or
Proteases and regulators
MMP-1, -3, -10, -12, -13, -14 or
TIMP-1 or
TIMP-2
PAI-1, -2; tPA; uPA
Cathepsin B
Soluble or shed receptors or ligands
ICAM-1, -3
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

OPG
sTNFRI
TRAIL-R3, Fas, sTNFRII
Fas
by University of Sussex on 10/10/12. For personal use only.

uPAR
SGP130
EGF-R
Nonprotein soluble factors
PGE2
Nitric oxide
Reactive oxygen species Altered
Insoluble factors (ECM)
Fibronectin
Collagens Altered
Laminin Altered

a
Factors are arranged by family.
b
The secretory changes that occur at senescence are indicated by upward arrows (increase), crosses (no change),
and downward arrows (decrease). Loss of p53 or gain of oncogenic RAS increases (upward arrows) or decreases
(downward arrows) the secretion of several SASP factors.
c
Abbreviations: bFGF, basic broblast growth factor; ECM, extracellular matrix; EGF, endothelial growth factor;
GRO, growth-related oncogene; HGF, hepatocyte growth factor; ICAM, intercellular adhesion molecule;
IGFBP, insulin-like growth factorbinding protein; MCP, membrane cofactor protein; MMP, matrix
metalloproteinase; NGF, nerve growth factor; OPG, osteoprotegerin; PAI, plasminogen activator inhibitor;
PGE2, prostaglandin E2; PIGF, placental growth factor; SCF, stem cell factor; SDF, stromal cellderived factor;
sTNFR, soluble tumor necrosis factor receptor; t-PA, tissue-type plasminogen activator; TIMP, tissue inhibitor of
metalloproteinases; TRAIL, tumor necrosis factorrelated apoptosis-inducing ligand; u-PA, urokinase-type
plasminogen activator; uPAR, u-PA receptor; VEGF, vascular endothelial growth factor.

IL-6. The most prominent cytokine of the epithelial cells (16, 18, 57, 58). Further, IL-6
SASP is interleukin-6 (IL-6), a pleiotropic secretion appears to be directly controlled
proinammatory cytokine (see Figure 2). by persistent DNA-damage signaling (ATM
IL-6 has been shown to be associated with and CHK2), independent of the p53 pathway
DNA damage and oncogenic stressinduced (59). Through IL-6 expression, senescent
senescence of mouse and human keratinocytes, cells can directly affect neighboring cells that
melanocytes, monocytes, broblasts, and express the IL-6R (gp80) and gp130 signaling

www.annualreviews.org SASP and Cancer 103


AREV403-PM05-05 ARI 10 December 2009 16:41

IL-6 IL-8 p16 and macrophage inammatory protein (MIP)-


3 and -1 (CCL-20, -3). MCP-3 (CCL-7) is
overexpressed by senescent liver stellate cells
PRE
and by prostate and skin broblasts. Fibroblasts
induced to senesce by oncogenic RAS secrete
high levels of MCP-3 as well as I-309 (CCL-1).
In addition, both broblasts induced to senesce
REP SEN
by RAS and stellate cells induced to senesce by
liver damage secrete high levels of another two
members of the CXCL family, GCP-2 (CXCL-
6) and ENA-78 (CXCL-5). Overexpression of
IR SEN PF-4 (CXCL-4) and SDF-1 (CXCL-12) was
observed in senescent prostate broblasts (46,
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

68). Recently, it was shown that cells undergo-


ing oncogene-induced senescence secrete mul-
RAS SEN tiple CXCR-2 (IL-8RB)-binding chemokines
(15). It was proposed that senescent cells ac-
by University of Sussex on 10/10/12. For personal use only.

tivate a self-amplifying secretory network in


Figure 2 which CXCR-2-binding chemokines reinforce
Human broblasts, either presenescent (PRE) or senescent (SEN), were growth arrest.
immunostained for the inammatory cytokines interleukin (IL)-6 and IL-8, as
well as the senescence marker p16. Cells were made senescent either by IGF pathway. The insulin-like growth fac-
replicative exhaustion (REP) or ionizing radiation (IR) or by expression of
tor (IGF)/IGF receptor network may also con-
oncogenic RAS (RAS).
tribute to the effect senescent cells exert on
complex at their surface, such as epithelial their microenvironment. Senescent endothe-
and endothelial cells of various functions and lial, epithelial, and broblast cells express high
origins. levels of almost all the IGF-binding proteins
(IGFBPs), including IGFBP-2, -3, -4, -5, and
IL-1. Another interleukin signaling pathway -6 (18, 69, 70) and their regulators, IGFBP-
demonstrated to be upregulated by senescent rP1 and -rP2 [also known as connective tis-
cells is that of IL-1 (60, 61). Both IL-1 and sue growth factor (CTGF)] (44, 71). Recently,
- are overexpressed and secreted by senes- activation of the BRAF oncogene in primary
cent endothelial cells (62), broblasts (63, 64), broblasts was shown to lead to the secre-
and chemotherapy-induced senescent epithe- tion of IGFBP-7, which acts through au-
lial cells (53). These cytokines can affect neigh- tocrine/paracrine pathways to induce senes-
boring cells through the cell-surface receptors cence and apoptosis in neighboring cells (72).
(IL-1 receptor/Toll-like receptor superfamily),
which act primarily to trigger the nuclear factor Other soluble factors. There are additional
kappa B and activating protein 1 pathways (65). soluble factors associated with the SASP. For
example, inammatory cytokines such as the
Chemokines (CXCL and CCL). Most senes- colony-stimulating factors (CSFs, including
cent cells overexpress IL-8 (CXCL-8) (see GM-CSF and G-CSF) are secreted at high
Figure 2), along with GRO and GRO levels by senescent broblasts (18). In addi-
(CXCL-1 and -2; the murine CXCL-1 is named tion, osteoprotegerin, a secreted decoy recep-
KC) (58, 66, 67). CCL family members that tor for tumor necrosis factor alpha, is present
are generally upregulated in senescent cells in- at high levels in the extracellular milieu of
clude MCP-2, -4, and -1 (CCL-8, -13, and senescent broblasts. Other molecules upreg-
-2); HCC-4 (CCL-16); eotaxin-3 (CCL-26); ulated at senescence include prostaglandin E2

104 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

(PGE2) (57, 73) and Cox-2, the enzyme re- Extracellular Insoluble Molecules
sponsible for the production of PGE2 and other
Fibronectin is a large multidomain glycopro-
prostaglandins.
tein found in connective tissue, on cell sur-
faces, and in plasma and other body uids. It
Extracellular Proteases interacts with a variety of macromolecules, in-
as an Important Subset cluding cell-surface receptors, components of
of the Senescence-Associated the cytoskeleton, and other ECM molecules.
Secretory Phenotype Through its interactions with cell-surface re-
ceptors, primarily integrins, bronectin can af-
In addition to secreting soluble signaling cy-
fect cell adhesion, survival, growth, and migra-
tokines and growth factors, senescent cells also
tion. Fibronectin production is upregulated in
secrete proteases such as matrix metallopro-
premature aging Werner syndrome broblasts
teinases (MMPs).
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

(89). Moreover, cells undergoing senescence in


culture and in vivo (90) increase bronectin
MMP family. The MMP family members
expression.
that are consistently upregulated in human
and mouse broblasts undergoing replicative
by University of Sussex on 10/10/12. For personal use only.

or stress-induced senescence are stromelysin- Nonprotein Secretions


1 and -2 (MMP-3 and -10, respectively) and
As a result of senescence-induced changes in
collagenase-1 (MMP-1) (7478). In some in-
cellular metabolism, senescent cells may ex-
stances, the MMP-1 and -3 produced by senes-
ert inuences on tissue microenvironments due
cent cells (79) can also regulate the activity of
to the secretion of molecules other than pro-
the soluble factors present in the SASP. For ex-
teins. These molecules include reactive oxygen
ample, these MMPs can cleave MCP-1, -2, and
species and transported ions. For example,
-4 and IL-8 (80). A variety of other CXCL/
senescent cells have been shown to release
CCL family members that constitute the SASP
nitric oxide and reactive oxygen species due
can also be cleaved by MMP-9, -2, or -7. These
to alterations in inducible nitric oxide syn-
CXCL and CCL cytokines can originate from
thase, endothelial nitric oxide synthase, and
neighboring cells, such as leukocytes or tumor
superoxide-dismutase activities (9195). These
cells (81).
reactive molecules are known modulators of
cellular phenotype, such as the differentiation
Serine proteases and their inhibitors.
of monocytes. In addition, these molecules
Another family of proteases involved in car-
can enhance cancer cell aggressiveness and can
cinogenesis and present in the SASP comprises
promote aging and age-related degeneration
serine proteases and regulators of the plasmino-
(96, 97).
gen activation pathway. Members of this family
include urokinase- or tissue-type plasminogen
activators (uPA or tPA, respectively), the uPA
receptor (uPAR), and inhibitors of these serine SPECIFICITY OF THE
proteases (PAI-1 and -2) (82). Indeed, a >50- SENESCENCE-ASSOCIATED
fold increase in plasminogen activator activity SECRETORY PHENOTYPE
has been reported in senescent endothelial Despite the fact that a signicant number of fac-
cells and lung and skin broblasts (83, 84). tors increase their secretion upon senescence,
PAI-1 is also upregulated in broblasts and the SASP is not a general or nonspecic upreg-
endothelial cells from aged donors (8587). ulator of secretion. The levels of expression of
Like the CXCR-2 cytokines, PAI-1 also seems many secreted factors do not change when cells
to reinforce the senescence growth arrest senesce. Interestingly, among these unchanged
(88). secreted molecules are anti-inammatory

www.annualreviews.org SASP and Cancer 105


AREV403-PM05-05 ARI 10 December 2009 16:41

soluble factors such as IL-2, -4, -10, -11, and the SASP, suggesting that the SASP is a specic
-12 (18). Fractalkine (CX3 CL-1), GCP-2, program triggered by genotoxic stress.
GITR, PDGF-BB, and LIGHT (all essential Finally, mouse senescent broblasts also dis-
to leukocyte differentiation or proliferation) play a SASP. Under standard cell-culture con-
also remain unchanged when broblasts are ditions, which include 20% oxygen, mouse
induced to senesce by X-irradiation, RAS cells undergo an arrest that has been termed
overexpression, or replicative exhaustion. senescence but that does not include a SASP.
Intriguingly, no factor was signicantly By contrast, under physiological 3% oxygen,
downregulated in different senescent states the mouse SASP closely resembles the hu-
(18). man SASP. These ndings suggest that the
Despite a specic, conserved core of up- senescent cell secretome is specic and evolu-
regulated and unchanged secreted molecules, tionarily conserved ( J.P. Coppe & J. Campisi,
different senescent states appear to display unpublished data).
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

some unique features. Whereas cells induced


to senesce by replicative exhaustion, telomere
disruption, X-irradiation, or chromatin disrup- REGULATORY MECHANISMS OF
tion seem to express closely related SASPs (18, THE SENESCENCE-ASSOCIATED
by University of Sussex on 10/10/12. For personal use only.

59), broblasts induced to senesce by oncogenic SECRETORY PHENOTYPE


RAS oversecrete more GM-CSF, IL-6, -7, -8,
-1, -13, and GRO than do cells induced to
Intracellular Signaling, Transcription,
senesce by other means. Moreover, such cells
and Chromatin: Locking in the
secrete high levels of factors such as ENA-78,
Senescence-Associated
I-309, G-CSF, and interferon (IFN)- that are
Secretory Phenotype
not secreted by other senescent cells. By con- Overall, the gene (mRNA) expression proles
trast, cells induced to senesce by overexpres- of senescent cells determined by microarrays
sion of the p16INK4a tumor-suppressor protein resemble the proles of secreted proteins de-
do not express a SASP despite other hallmarks termined by antibody arrays (18; J.P. Coppe &
of senescence ( J.P. Coppe, F. Rodier & J. Camp- J. Campisi, unpublished data). This nding sug-
isi, unpublished data). Cells that senesce with gests that the secretory phenotype of senescent
dysfunctional p53 develop a SASP that resem- cells is at least partly regulated at the transcrip-
bles the SASP caused by oncogenic RAS (18). tional level. However, because the changes in
Thus, although a core of SASP factors is a fea- gene expression that occur at senescence are
ture of all senescent cells (with the exception of so widespread, the transcriptional control may
p16INK4a -induced senescence), there are varia- well be at the level of chromatin organization,
tions in the quantity and quality of the SASP rather than due to changes in specic transcrip-
that depend on the cell type and senescence tion factors. In fact, dramatic chromatin alter-
inducer. ations are known to occur at senescence (98
Another feature of the SASP is its dynamic 101). Further support for the idea that gene
development over time (18). In culture, cells expression specic to the senescence program
develop a full SASP >5 days after senescence may be partially attributed to larger changes
induction, and the cells growth arrests within in chromatin conformation is suggested by the
24 h of damage. Not all SASP factors begin to be physical clustering of genes that constitute the
secreted at the same time. This gradual pheno- SASP ( J.P. Coppe & J. Campisi, unpublished
typic transition is a feature conserved between data).
cell types and senescence inducers. Genetic al- Our data suggest that a large propor-
terations, such as loss of p53 or gain of onco- tion of the SASP of senescent broblasts
genic RAS, lead to a more rapid acquisition of is irreversible once established (18). Indeed,

106 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

senescent human broblasts that express low without a SASP) of p16INK4a . That is, p16INK4a
levels of p16INK4a can revert and resume pro- induction/delivery could actively suppress cell
liferation upon p53 inactivation (102). These proliferation without triggering the proinam-
reverted cells, however, retain the SASP (18). matory SASP.
This may imply that, once senescence is es- The p53 tumor suppressor can also pro-
tablished, unknown mechanismspotentially mote aging (111, 112) and senescence (7, 11,
related to chromatin alterationspermanently 12) in mice (see Figure 3). Along with mu-
lock the SASP in an irreversible open chro- tations in p16INK4a , mutations leading to p53
matin conrmation, analogous to the way the inactivation occur very frequently in cancer
p16INK4a /pRB pathway is proposed to lock cells; that is, p53 is well known to act as
growth-promoting genes into a heterochro- a cell-autonomous tumor suppressor by con-
matic state (103). Another implication of these trolling apoptosis and cell-cycle arrest, both
ndings is that the senescence-associated cell- in culture and in vivo. However, p53 mu-
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

cycle arrest and the SASP can be uncoupled (see tations have also been found in the stromal
Figure 1). Further, the SASP is a more per- vicinity of carcinomas, and this p53-decient
manent characteristic of senescence than is the stroma was shown to promote tumorigenesis
growth-arrested state. (32). These data suggest that p53 may have
by University of Sussex on 10/10/12. For personal use only.

benecial cell-nonautonomous effects in pre-


venting cancer development (32, 113). Strik-
Cell-Nonautonomous Tumor ingly, we found that p53 actively restrains the
Suppressors: The Guardians SASP, suggesting a potential mechanism by
of the Senescence-Associated which p53 may suppress tumorigenesisthat
Secretory Phenotype is, by restraining the development of a pro-
The p16INK4a tumor suppressor is a posi- tumorigenic/proinammatory tissue microen-
tive regulator of the pRB tumor-suppressor vironment (18). Thus, loss of p53 activity by
pathway. High levels of ectopic expression of senescing or damaged broblasts greatly en-
p16INK4a induce senescence. Induction of en- hances the SASP and the stimulatory effects of
dogenous p16INK4a is associated with tumor the SASP on malignant epithelial cells (as dis-
prevention and the general age-associated de- cussed further below) (18).
cline in stem cell and tissue function (104109).
Although p16INK4a is a very efcient inducer
of cell-cycle arrest (including the senescence- EFFECTS ON CELL BEHAVIOR
associated arrest), p16INK4a does not seem to Factors secreted by senescent cells can pro-
play a major role in the development of the mote tumor development in vivo and malignant
SASP ( J.P. Coppe, F. Rodier & J. Campisi, un- phenotypes such as proliferation and invasive-
published data). Cells induced to senesce by ec- ness in cell-culture models. These effects have
topic p16INK4a expression secrete signicantly been observed in a number of tissues, including
lower levels of SASP factors compared to cells breast (13, 18, 77, 78, 114), skin (115), prostate
induced to senesce by most other senescence in- (18, 116), pancreas (117), and oropharyngial
ducers. Senescence induced by p16INK4a has po- mucosa (14). The effects of the complex SASP
tential therapeutic applications. As an example, are, of course, dependent on the tissue context.
p16INK4a gene therapy for rheumatoid arthritis Thus, different models show different effects of
was demonstrated to efciently stop the disease the SASP. In the following sections, we discuss
evolution and decrease the inammatory state in greater detail the various behavioral changes
(110). In cancer, such an approach could take cells can undergo when residing in the prox-
advantage of both the cell-autonomous prop- imity of senescent cells and how the senescent
erties (inhibition of cell growth) and the cell- tissue microenvironment can facilitate tumor
nonautonomous properties (senescence arrest initiation and progression.

www.annualreviews.org SASP and Cancer 107


AREV403-PM05-05 ARI 10 December 2009 16:41

Intrinsic or extrinsic
genotoxic stress

Damage sensors
and transducers

Transient arrest and


faithful repair p53
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org
by University of Sussex on 10/10/12. For personal use only.

Cancer, Senescence
SASP
aging, pathologies arrest

Early Later
(?) (?)

Alarm signals and


tissue repair

Chronic SASP
and persistent
inflammation

Figure 3
The DNA damage signaling pathway leads to the activation of the p53 tumor suppressor. Activated p53
triggers cell fate decisions, such as senescence or apoptosis. Depending on the cell context, p53 can suppress
cancer through transient cell-cycle arrest and activation of the DNA-repair machinery. Additionally, p53
restrains the senescence-associated secretory phenotype (SASP). Regulation of the SASP by p53 suggests a
cell-nonautonomous function of this tumor suppressor. In the short term, the SASP may promote tissue
repair. In the long term, it may promote chronic inammation, which in turn can drive cancer and aging.

The Senescence-Associated component ( J.P. Coppe & J. Campisi, unpub-


Secretory Phenotype Promotes lished data). Irradiated stromal cells, which are
Cell Proliferation presumed to be senescent, have been shown
to perturb mammary epithelial microenviron-
One of the most protumorigenic effects of the
ment and to fuel inappropriate epithelial cell
SASP is to promote the proliferation of epithe-
growth in the mammary gland (114, 118). Fur-
lial cells.
thermore, MMPs secreted by senescent bro-
blasts have been shown to be responsible for
Breast cancer. In the case of breast epithe- the higher tumorigenicity of breast epithelial
lial cells, senescent human broblasts can stim- cell xenografts in mice, most likely by allowing
ulate the growth of premalignant and malig- mitogenic and chemotactic signals greater ac-
nant mammary epithelial cells (13, 18, 77). This cess to breast cancer cells (78, 114). In addition
stimulation may be due in large measure to se- to secreted soluble factors, there is evidence
cretion of GRO, which is a prominent SASP that the matrix laid down by senescent cells can

108 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

also stimulate mammary epithelial cell growth variants in nevi can evolve into melanoma.
(13). Malignant melanocytes express high levels of
the CXCR-2 receptor (122) and can be stim-
Prostate cancer. Fibroblasts from the human ulated to grow by its ligands GRO (123)
prostate gland that undergo senescence in cul- and IL-8 (124). Given that both GRO and
ture have been shown to create a local tissue IL-8 form part of the core SASP, the senes-
environment that favors prostate epithelial cell cent microenvironment may therefore stimu-
hyperproliferation, in part owing to amphireg- late the proliferation of rare premalignant cells
ulin secretion (46). Furthermore, CTGF (or in nevi, thereby leading to the development of
IGFBP-rP2) is upregulated in senescent bro- melanoma.
blasts (44), and this protein was shown to reg-
ulate prostate tumor progression in xenografts Other tumor-associated cells. During an-
and to be expressed by the cancer-associated re- giogenesis, endothelial cells can undergo pro-
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

active stroma (119). Whereas upregulation of liferation, which is stimulated by vascular en-
CXCR-4 is observed in most cancer cells, only dothelial growth factor (VEGF), IL-8, I-309,
senescent stromal cells of the prostate display and eotaxin (125127). The proangiogenic ef-
high levels of expression of its ligand SDF- fects of the SASP were shown in vivo in mouse
by University of Sussex on 10/10/12. For personal use only.

1 (CXCL-12). Thus, SDF-1 secretion by xenografts of breast cancer cells. The blood ves-
senescent broblasts may therefore play a se- sel density was signicantly higher when the tu-
lective role in fueling prostate cancer. It has mors developed in the presence of senescent but
recently been determined that senescence in- not presenescent broblasts (127). RAS-driven
duced by irradiation in prostate cancer patients tumors are also known to contain signicant
is associated with a signicantly increased re- numbers of senescent cells (8), and these tu-
lease of exosome-like microvesicles (120). This mors are also highly vascularized (128). Many
novel secretory phenotype depends on the acti- of the SASP factors can also affect leukocyte
vation of p53. Finally, the propensity of prostate proliferation during the course of cancer devel-
cancer patients to relapse after chemotherapy opment. For example, IL-7 directly promotes
may be due to the accumulation of senescent lymphocyte proliferation in peripheral tissues,
tumor cells with inammatory characteristics and GM-CSF stimulates myeloid suppressor
(18). cells, which are known to have important im-
munosuppressive functions that affect cancer
Other carcinomas. In the skin, unidentied progression (129).
factors secreted by human broblasts were
shown to be capable of inducing clonal ex-
pansion of keratinocytes (121). In addition, The Senescence-Associated Secretory
senescent endometrial broblasts promoted Phenotype Stimulates Cell Motility
anchorage-independent epithelial cell growth, (Invasion, Migration, Metastasis)
primarily because of IL-1 oversecretion (64). In Senescent cells secrete an array of factors that
the orobucal cavity, tobacco-driven senescence can create a gradient to promote cell migration
of supportive stromal cells was shown to stim- and invasion.
ulate the hyperplastic growth of epithelial cells
and was associated with the loss of tight junc- Epithelial cells. In pancreatic cancer, hepato-
tions and epithelial integrity (14). cyte growth factor (HGF), and to a lesser de-
gree basic broblast growth factor (bFGF), pro-
Melanoma. Melanocytic nevi (moles) are of- motes cancer cell invasion in culture and can
ten composed of senescent melanocytes that potentially drive cancer dissemination in vivo
are induced owing to oncogenic mutations in (117, 130). In breast cancer, high levels of IL-
BRAF (V600E mutations) (9). Only rare cell 6 and -8 secreted by senescent broblasts are

www.annualreviews.org SASP and Cancer 109


AREV403-PM05-05 ARI 10 December 2009 16:41

responsible for enhancing the invasiveness of a p53-decient RAS-driven tumors induced to


panel of cancer cell lines in cell-culture mod- senesce through reestablishment of p53 func-
els (18). Moreover, the secretion of MMP-2 tion (12), innate immune cells were shown to
and -3 by senescent cells can also promote migrate into the vicinity of the senescent tumor
the invasion of multiple epithelial cell types area. CSF-1, CXCL-1, or MCP-1 and ICAM-
(77, 78, 114, 131, 132). Other proteases, such 1 transcripts were found to be higher in these
as uPA and its regulator (PAI-1), are likewise senescent tumor masses, and they may be re-
implicated in cancer cell invasion. Senescent sponsible for the immune response. For exam-
stromal cells may promote an epithelial-to- ple, neutrophils express CXCR-1, -2, and -4 to
mesenchymal transition (18), which is an im- sense their microenvironment and to invade tis-
portant phenotypic switch that enables cancer sues; eosinophils use the broad-spectrum recep-
cells to migrate and invade. Through use of co- tor CCR-3 to fulll their function; monocytes
cultures of smokeless tobacco extractexposed use CCR-1, -2, and -5, CXCR-4, and CX3 CR1
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

broblasts and human epidermal keratinocytes, to extravasate and enter peripheral sites, where
factors secreted by extract-modied broblasts they differentiate; natural killer cells express
increased the invasiveness of partially trans- CCR -2 and -5, CXCR-4, CX3 CR1, and XCR1;
formed epithelial cells in conjunction with a and immature myeloid dendritic cells display
by University of Sussex on 10/10/12. For personal use only.

loss of E-cadherin, zonula occludens 1, and in- CCR-1, -2, -5, and -6 and CXCR-4, which fa-
volucrin expression (14). Thus, senescent cells cilitate their transport, migration, and function
and the SASP can induce phenotypes in nearby (136139).
human epithelial cells that are common during
cancer progression.
The Senescence-Associated
Endothelial cells. In cell-culture models, en- Secretory Phenotype Regulates
dothelial cells are induced to migrate by fac- Cell Differentiation
tors secreted by senescent broblasts (127). The factors secreted by senescent cells can alter
This is in part due to VEGF secretion and the differentiation status of neighboring cells.
chemokine gradients set up by senescent cells
(133). Neoangiogenesis, which is dependent on Epithelial cells. Senescent human and mouse
endothelial cell motility and invasion, is en- broblasts disrupt the differentiation of mam-
hanced in xenograft models containing senes- mary epithelial cells and inhibit the expression
cent broblasts (127). Further, it is known of differentiation markers (77, 114). This activ-
that IL-1, a SASP component, activates the ity is due in large measure to the secretion of
endothelium and consequently increases the MMP-3 by the senescent cells. Furthermore,
adhesive potential of cancer cells to vessel weakly tumorigenic pancreatic (117) and mam-
walls (134). Thus, senescent cells may pro- mary (18) epithelial cells undergo morphologic
mote extravasation of cancer cells to secondary changes in culture that resemble an epithelial-
metastatic sites. However, the effects of senes- to-mesenchymal transition in the presence of a
cent cells on angiogenesis may depend on senescent conditioned medium. The effect on
cell type. For example, senescent keratinocytes mammary epithelial cells is attributable to IL-
oversecrete maspin, which displays paracrine 6 and -8 (18), as well as to HGF, uPAR, and
antiangiogenic activity and acts as a domi- MMPs (77), all of which can disrupt epithelial
nant inhibitor of endothelial cell migration cell clusters and stimulate dedifferentiation in
(135). culture and in vivo (140142).

Leukocytes. Senescent broblasts may pro- Endothelial cells. Strikingly, no angiostatic


mote leukocyte recruitment because they factors have been reported among SASP con-
chronically release chemokines (136). In stituents (e.g., IFN-, TSP-1, MIG, PF4,

110 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

IP-10, IL-4 and -12, and endostatin). This con- CONCLUSIONS AND FUTURE
trasts with the largely proangiogenic prole of DIRECTIONS
the SASP (which includes IL-8, MCP-1 and
Most insoluble components of the ECM
-2, GROs, PGE2, VEGF, EGF, CSFs, u-/tPA,
are enzymatic targets of secreted proteases.
MMPs, bronectin, and laminin) (143). Fur-
Therefore, the senescence-associated changes
thermore, there may be an amplifying activa-
in proteolytic activities could affect the phys-
tion loop because senescent stromal cells se-
ical properties of the tissue structure around
crete MCPs, CSFs, MIPs, GROs, and CXCLs,
cells. In particular, the accumulation of senes-
which in turn recruit inammatory and immune
cent cells could lessen the supportive role of the
cells that also secrete proangiogenic factors
ECM, globally diminishing tissue tension and
(VEGFs, IL-8, and MMPs). Thus, senescent
elasticity. In addition, the relaxed tissue struc-
cells are poised to support the differentiation
ture and higher levels of MMPs may help tumor
of a new vasculature around and within a pro-
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

cells migrate and invade through the ECM, thus


gressing tumor.
enabling metastasis. The panel of proteases se-
creted by senescent cells extensively overlaps
with those found in malignant tumors. Further,
The Senescence-Associated Secretory
by University of Sussex on 10/10/12. For personal use only.

senescence-induced alterations in the secretion


Phenotype Affects Leukocyte of interleukins, chemokines, growth factors,
Infiltration and Tumor Immunology proteases, and associated processing activities
No anti-inammatory factors (e.g., IFN-, tend to establish the SASP as protumorigenic.
IFN-, IL-3, and IL-5) are signicantly se- Overall, senescence is a molecular program
creted by senescent broblasts, and some of with a unique phenotypic outcome. How its ex-
these factors are even downregulated upon tracellular molecular signature is activated and
senescence (e.g., IL-2 and -12). Nonetheless, maintained and the extent to which it inuences
some reports show that massive amounts of the tissue milieu in healthy tissues, aged tissues,
either MCP-1 or IL-8, which are prominent and diseased tissues are some of the many ques-
components of the SASP, lead to tumor de- tions that remain unanswered. However, even
struction (144, 145). Senescent broblasts may with our currently limited knowledge of the
inuence the macrophage balance in the tumor SASP and its potential effects on carcinogen-
environment. Molecules that are implicated in esis, promising new strategies for cancer ther-
the recruitment and differentiation of circulat- apies are possible. For example, restoring the
ing monocytes to tumor sites also happen to activity of tumor-suppressor proteins is an
be overexpressed by senescent broblasts (146). attractive, potentially powerful therapeutic
These molecules can lead to an inadequate im- approach. Taking into account our present un-
mune response within the close proximity of derstanding of the cell-nonautonomous effects
senescent cells. Senescent broblasts may affect of tumor-suppressor genes such as p53, small
lymphocytic populations inltrating the tumor. chemicals that can pharmacologically restore
Specic T cell populations associated with tu- their normal function would help reestablish
mor progression (i.e., Th2 and regulatory T the proper tissue and cell signals, thereby
cells) respond to inammatory cytokines that stimulating cancer regression (147150). Such
are commonly present in the broblast SASP. approaches could stimulate cancer elimination
Other cells of the specic and innate immune for two reasons: First, they would limit inam-
system, such as natural killer cells, neutrophils, mation and thus possibly allow proper tissue
eosinophils, dendritic cells, and B cells, are also repair; second, they would directly promote
subject to regulation by cytokines that are pro- the immune-mediated clearance of cells that
duced by senescent broblasts. drive cancer progression.

www.annualreviews.org SASP and Cancer 111


AREV403-PM05-05 ARI 10 December 2009 16:41

DISCLOSURE STATEMENT
A.K. is CEO and CSO of StemLifeLine, Inc., a biotech company that uses stem cells for drug
screening and therapy. The other authors are not aware of any afliations, memberships, funding,
or nancial holdings that might be perceived as affecting the objectivity of this review.

LITERATURE CITED
1. Coussens LM, Werb Z. 2002. Inammation and cancer. Nature 420:86067
2. Vakkila J, Lotze MT. 2004. Inammation and necrosis promote tumor growth. Nat. Rev. Immunol.
4:64148
3. Joyce JA. 2005. Therapeutic targeting of the tumor microenvironment. Cancer Cell 7:51320
4. de Visser KE, Eichten A, Coussens LM. 2006. Paradoxical roles of the immune system during cancer
development. Nat. Rev. Cancer 6:2437
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

5. Narita M, Lowe SW. 2005. Senescence comes of age. Nat. Med. 11:92022
6. Braig M, Lee S, Loddenkemper C, Rudolph C, Peters AH, et al. 2005. Oncogene-induced senescence
as an initial barrier in lymphoma development. Nature 436:66065
7. Chen Z, Trotman LC, Shaffer D, Lin H, Dotan ZA, et al. 2005. Critical role of p53 dependent cellular
by University of Sussex on 10/10/12. For personal use only.

senescence in suppression of Pten decient tumourigenesis. Nature 436:72530


8. Collado M, Gil J, Efeyan A, Guerra C, Schumacher AJ, et al. 2005. Tumor biology: senescence in
premalignant tumours. Nature 436:642
9. Michaloglou C, Vredeveld LCW, Soengas MS, Denoyelle C, Kuilman T, et al. 2005. BRAFE600 -
associated senescence-like cell cycle arrest of human nevi. Nature 436:72024
10. Courtois-Cox S, Genther Williams SM, Reczek EE, Johnson BW, McGillicuddy LT, et al. 2006. A
negative feedback signaling network underlies oncogene-induced senescence. Cancer Cell 10:45972
11. Ventura A, Kirsch DG, McLaughlin ME, Tuveson DA, Grimm J, et al. 2007. Restoration of p53 function
leads to tumor regression in vivo. Nature 445:66165
12. Xue W, Zender L, Miething C, Dickins RA, Hernando E, et al. 2007. Senescence and tumor clearance
is triggered by p53 restoration in murine liver carcinomas. Nature 445:65660
13. Krtolica A, Parrinello S, Lockett S, Desprez P, Campisi J. 2001. Senescent broblasts promote epithelial
cell growth and tumorigenesis: a link between cancer and aging. Proc. Natl. Acad. Sci. USA 98:1207277
14. Coppe JP, Boysen M, Sun CH, Wong BJ, Kang MK, et al. 2008. A role for broblasts in mediating the
effects of tobacco-induced epithelial cell growth and invasion. Mol. Cancer Res. 6:108598
15. Acosta JC, OLoghlen A, Banito A, Guijarro MV, Augert A, et al. 2008. Chemokine signaling via the
CXCR2 receptor reinforces senescence. Cell 133:100618
16. Kuilman T, Michaloglou C, Vredeveld LCW, Douma S, van Doorn R, et al. 2008. Oncogene-induced
senescence relayed by an interleukin-dependent inammatory network. Cell 133:101931
17. Green MR. 2008. Senescence: not just for tumor suppression. Cell 134:56264
18. Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, et al. 2008. Senescence-associated secretory phenotypes
reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6:2853
68
19. Campisi J, dAdda di Fagagna F. 2007. Cellular senescence: when bad things happen to good cells. Nat.
Rev. Mol. Cell. Biol. 8:72940
20. Young AR, Narita M. 2009. SASP reects senescence. EMBO Rep. 10:22830
21. Dimri GP, Lee X, Basile G, Acosta M, Scott G, et al. 1995. A novel biomarker identies senescent human
cells in culture and in aging skin in vivo. Proc. Natl Acad. Sci. USA 92:936367
22. Ben-Porath I, Weinberg RA. 2004. When cells get stressed: an integrative view of cellular senescence.
J. Clin. Investig. 113:813
23. Ben-Porath I, Weinberg RA. 2005. The signals and pathways activating cellular senescence. Int. J.
Biochem. Cell Biol. 37:96176
24. Schmitt CA. 2003. Senescence, apoptosis and therapycutting the lifelines of cancer. Nat. Rev. Cancer
3:28695

112 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

25. Campisi J. 2005. Senescent cells, tumor suppression and organismal aging: good citizens, bad neighbors.
Cell 120:51322
26. Martin GM. 2005. Genetic modulation of senescent phenotypes in Homo sapiens. Cell 120:52332
27. Chien KR, Karsenty G. 2005. Longevity and lineages: toward the integrative biology of degenerative
diseases in heart, muscle, and bone. Cell 120:53344
28. Lombard DB, Chua KF, Mostoslavsky R, Franco S, Gostissa M, Alt FW. 2005. DNA repair, genome
stability, and aging. Cell 120:497512
29. Balaban RS, Nemoto S, Finkel T. 2005. Mitochondria, oxidants, and aging. Cell 120:48395
30. Braig M, Schmitt CA. 2006. Oncogene-induced senescence: putting the brakes on tumor development.
Cancer Res. 66:288184
31. Collado M, Serrano M. 2006. The power and the promise of oncogene-induced senescence markers.
Nat. Rev. Cancer 6:47276
32. Hill R, Song Y, Cardiff RD, Van Dyke T. 2005. Selective evolution of stromal mesenchyme with p53
loss in response to epithelial tumorigenesis. Cell 123:100111
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

33. Najjar SS, Scuteri A, Lakatta EG. 2005. Arterial aging: Is it an immutable cardiovascular risk factor?
Hypertension 46:45462
34. Kunieda T, Minamino T, Nishi J, Tateno K, Oyama T, et al. 2006. Angiotensin II induces premature
senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a
by University of Sussex on 10/10/12. For personal use only.

p21-dependent pathway. Circulation 114:95360


35. Kuilman T, Peeper DS. 2009. Senescence-messaging secretome: SMS-ing cellular stress. Nat. Rev. Cancer
9:8194
36. Shelton DN, Chang E, Whittier PS, Choi D, Funk WD. 1999. Microarray analysis of replicative senes-
cence. Curr. Biol. 9:93945
37. Ly DH, Lockhart DJ, Lerner RA, Schultz PG. 2000. Mitotic misregulation and human aging. Science
287:248692
38. Park WY, Hwang CI, Kang MJ, Seo JY, Chung JH, et al. 2001. Gene prole of replicative senescence
is different from progeria or elderly donor. Biochem. Biophys. Res. Commun. 282:93439
39. Kyng KJ, May A, Brosh RM Jr, Cheng WH, Chen C, et al. 2003. The transcriptional response after
oxidative stress is defective in Cockayne syndrome group B cells. Oncogene 22:113549
40. Kulaeva OI, Draghici S, Tang L, Kraniak JM, Land SJ, Tainsky MA. 2003. Epigenetic silencing of
multiple interferon pathway genes after cellular immortalization. Oncogene 22:411827
41. Zhang H, Pan KH, Cohen SN. 2003. Senescence-specic gene expression ngerprints reveal cell-type-
dependent physical clustering of up-regulated chromosomal loci. Proc. Natl. Acad. Sci. USA 100:325156
42. Csoka AB, English SB, Simkevich CP, Ginzinger DG, Butte AJ, et al. 2004. Genome-scale expression
proling of Hutchinson-Gilford progeria syndrome reveals widespread transcriptional misregulation
leading to mesodermal/mesenchymal defects and accelerated atherosclerosis. Aging Cell 3:23543
43. Yoon IK, Kim HK, Kim YK, Song IH, Kim W, et al. 2004. Exploration of replicative senescence-
associated genes in human dermal broblasts by cDNA microarray technology. Exp. Gerontol. 39:1369
78
44. Kim KH, Park GT, Lim YB, Rue SW, Jung JC, et al. 2004. Expression of connective tissue growth
factor, a biomarker in senescence of human diploid broblasts, is up-regulated by a transforming growth
factor--mediated signaling pathway. Biochem. Biophys. Res. Commun. 318:81925
45. Kyng KJ, May A, Stevnsner T, Becker KG, Kolvra S, Bohr VA. 2005. Gene expression responses to
DNA damage are altered in human aging and in Werner syndrome. Oncogene 24:502642
46. Bavik C, Coleman I, Dean JP, Knudsen B, Plymate S, Nelson PS. 2006. The gene expression program
of prostate broblast senescence modulates neoplastic epithelial cell proliferation through paracrine
mechanisms. Cancer Res. 66:794802
47. Schnabl B, Purbeck CA, Choi YH, Hagedorn CH, Brenner D. 2003. Replicative senescence of activated
human hepatic stellate cells is accompanied by a pronounced inammatory but less brogenic phenotype.
Hepatology 37:65364
48. Chen J, Brodsky SV, Goligorsky DM, Hampel DJ, Li H, et al. 2002. Glycated collagen I induces
premature senescence-like phenotypic changes in endothelial cells. Circ. Res. 90:129098

www.annualreviews.org SASP and Cancer 113


AREV403-PM05-05 ARI 10 December 2009 16:41

49. Csiszar A, Ungvari Z, Koller A, Edwards JG, Kaley G. 2003. Aging-induced proinammatory shift in
cytokine expression prole in coronary arteries. FASEB J. 17:118385
50. Kamino H, Hiratsuka M, Toda T, Nishigaki R, Osaki M, et al. 2003. Searching for genes involved
in arteriosclerosis: proteomic analysis of cultured human umbilical vein endothelial cells undergoing
replicative senescence. Cell Struct. Funct. 28:495503
51. Eman MR, Regan-Klapisz E, Pinkse MW, Koop IM, Haverkamp J, et al. 2006. Protein expression dy-
namics during replicative senescence of endothelial cells studied by 2-D difference in gel electrophoresis.
Electrophoresis 27:166982
52. Schwarze SR, DePrimo SE, Grabert LM, Fu VX, Brooks JD, Jarrard DF. 2002. Novel pathways associ-
ated with bypassing cellular senescence in human prostate epithelial cells. J. Biol. Chem. 277:1487783
53. Chang BD, Swift ME, Shen M, Fang J, Broude EV, Roninson IB. 2002. Molecular determinants of
terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proc. Natl Acad. Sci. USA
99:38994
54. Untergasser G, Koch HB, Menssen A, Hermeking H. 2002. Characterization of epithelial senescence by
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

serial analysis of gene expression: identication of genes potentially involved in prostate cancer. Cancer
Res. 62:625562
55. Zhang H, Herbert BS, Pan KH, Shay JW, Cohen SN. 2004. Disparate effects of telomere attrition
on gene expression during replicative senescence of human mammary epithelial cells cultured under
by University of Sussex on 10/10/12. For personal use only.

different conditions. Oncogene 19:619398


56. Schwarze SR, Fu VX, Desotelle JA, Kenowski ML, Jarrard DF. 2005. The identication of senescence-
specic genes during the induction of senescence in prostate cancer cells. Neoplasia 7:81623
57. Lu SY, Chang KW, Liu CJ, Tseng YH, Lu HH, et al. 2006. Ripe areca nut extract induces G1 phase arrests
and senescence-associated phenotypes in normal human oral keratinocyte. Carcinogenesis 27:127384
58. Sarkar D, Lebedeva IV, Emdad L, Kang DC, Baldwin AS Jr, Fisher PB. 2004. Human polynucleotide
phosphorylase (hPNPaseold-35): a potential link between aging and inammation. Cancer Res. 64:7473
78
59. Rodier F, Coppe JP, Patil CK, Hoeijmakers WAM, Munoz D, et al. 2009. Persistent DNA damage
signaling triggers senescence-associated inammatory cytokine secretion. Nat. Cell Biol. 11:97379
60. Garnkel S, Brown S, Wessendorf JH, Maciag T. 1994. Post-transcriptional regulation of interleukin
1 in various strains of young and senescent human umbilical vein endothelial cells. Proc. Natl. Acad. Sci.
USA 91:155963
61. McLachlan JA, Serkin CD, Morrey-Clark KM, Bakouche O. 1995. Immunological functions of aged
human monocytes. Pathobiology 63:14859
62. Maier JAM, Voulalas P, Roeder D, Maciag T. 1990. Extension of the life-span of human endothelial
cells by an interleukin-1 antisense oligomer. Science 249:157074
63. Kumar S, Millis AJ, Baglioni C. 1992. Expression of interleukin 1inducible genes and production of
interleukin 1 by aging human broblasts. Proc. Natl. Acad. Sci. USA 89:468387
64. Palmieri D, Watson JM, Rinehart CA. 1999. Age-related expression of PEDF/EPC-1 in human en-
dometrial stromal broblasts: implications for interactive senescence. Exp. Cell Res. 247:14247
65. Mantovani A, Locati M, Vecchi A, Sozzani S, Allavena P. 2001. Decoy receptors: a strategy to regulate
inammatory cytokines and chemokines. Trends Immunol. 22:32836
66. Castro P, Giri D, Lamb D, Ittmann M. 2003. Cellular senescence in the pathogenesis of benign prostatic
hyperplasia. Prostate 55:3038
67. Bode-Boger SM, Scalera F, Martens-Lobenhoffer J. 2005. Asymmetric dimethylarginine (ADMA) ac-
celerates cell senescence. Vasc. Med. 10(Suppl. 1):S6571
68. Begley L, Monteleon C, Shah RB, Macdonald JW, Macoska JA. 2005. CXCL12 overexpression and
secretion by aging broblasts enhance human prostate epithelial proliferation in vitro. Aging Cell 4:291
98
69. Wang S, Moerman EJ, Jones RA, Thweatt R, Goldstein S. 1996. Characterization of IGFBP-3, PAI-1
and SPARC mRNA expression in senescent broblasts. Mech. Ageing Dev. 92:12132
70. Grillari J, Hohenwarter O, Grabherr RM, Katinger H. 2000. Subtractive hybridization of mRNA from
early passage and senescent endothelial cells. Exp. Gerontol. 35:18797

114 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

71. Lopez-Bermejo A, Buckway CK, Devi GR, Hwa V, Plymate SR, et al. 2000. Characterization of insulin-
like growth factor-binding protein-related proteins (IGFBP-rPs) 1, 2, and 3 in human prostate epithe-
lial cells: potential roles for IGFBP-rP1 and 2 in senescence of the prostatic epithelium. Endocrinology
141:407280
72. Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. 2008. Oncogenic BRAF induces senescence
and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell 132:36374
73. Huang NN, Wang DJ, Heppel LA. 1993. Stimulation of aged human lung broblasts by extracellular
ATP via suppression of arachidonate metabolism. J. Biol. Chem. 268:1078995
74. West MD, Pereira-Smith OM, Smith JR. 1989. Replicative senescence of human skin broblasts corre-
lates with a loss of regulation and overexpression of collagenase activity. Exp. Cell Res. 184:13847
75. Millis AJT, Hoyle M, McCue HM, Martini H. 1992. Differential expression of metalloproteinase and
tissue inhibitor of metalloproteinase genes in diploid human broblasts. Exp. Cell Res. 201:37379
76. Zeng G, Millis AJ. 1996. Differential regulation of collagenase and stromelysin mRNA in late passage
cultures of human broblasts. Exp. Cell Res. 222:15056
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

77. Parrinello S, Coppe JP, Krtolica A, Campisi J. 2005. Stromal-epithelial interactions in aging and cancer:
senescent broblasts alter epithelial cell differentiation. J. Cell Sci. 118:48596
78. Liu D, Hornsby PJ. 2007. Senescent human broblasts increase the early growth of xenograft tumors
via matrix metalloproteinase secretion. Cancer Res. 67:311726
by University of Sussex on 10/10/12. For personal use only.

79. Hornebeck W, Maquart FX. 2003. Proteolyzed matrix as a template for the regulation of tumor pro-
gression. Biomed. Pharmacother. 57:22330
80. McQuibban GA, Gong JH, Wong JP, Wallace JL, Clark-Lewis I, Overall CM. 2002. Matrix metallopro-
teinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists
with anti-inammatory properties in vivo. Blood 100:116067
81. Van Den Steen PE, Wuyts A, Husson SJ, Proost P, Van Damme J, Opdenakker G. 2003. Gelati-
nase B/MMP-9 and neutrophil collagenase/MMP-8 process the chemokines human GCP-2/CXCL6,
ENA-78/CXCL5 and mouse GCP-2/LIX and modulate their physiological activities. Eur. J. Biochem.
270:373949
82. Blasi F, Carmeliet P. 2002. uPAR: a versatile signaling orchestrator. Nat. Rev. Mol. Cell Biol. 3:93243
83. Comi P, Chiaramonte R, Maier JA. 1995. Senescence-dependent regulation of type 1 plasminogen
activator inhibitor in human vascular endothelial cells. Exp. Cell Res. 219:3048
84. West MD, Shay JW, Wright WE, Linskens MH. 1996. Altered expression of plasminogen activator and
plasminogen activator inhibitor during cellular senescence. Exp. Gerontol. 31:17593
85. Mu XC, Higgins PJ. 1995. Differential growth statedependent regulation of plasminogen activator
inhibitor type-1 expression in senescent IMR-90 human diploid broblasts. J. Cell Physiol. 165:64757
86. Mu XC, Staiano-Coico L, Higgins PJ. 1998. Increased transcription and modied growth state
dependent expression of the plasminogen activator inhibitor type-1 gene characterize the senescent
phenotype in human diploid broblasts. J. Cell Physiol. 174:9098
87. Martens JW, Sieuwerts AM, Vries JB, Bosma PT, Swiggers SJ, et al. 2003. Aging of stromal-derived
human breast broblasts might contribute to breast cancer progression. Thromb. Haemost. 89:393404
88. Kortlever RM, Higgins PJ, Bernards R. 2006. Plasminogen activator inhibitor-1 is a critical downstream
target of p53 in the induction of replicative senescence. Nat. Cell Biol. 8:87784
89. Rasoamanantena P, Thweatt R, Labat-Robert J, Goldstein S. 1994. Altered regulation of bronectin
gene expression in Werner syndrome broblasts. Exp. Cell Res. 213:12127
90. Kumazaki T, Kobayashi M, Mitsui Y. 1993. Enhanced expression of bronectin during in vivo cellular
aging of human vascular endothelial cells and skin broblasts. Exp. Cell Res. 205:396402
91. Sato I, Morita I, Kaji K, Ikeda M, Nagao M, Murota S. 1993. Reduction of nitric oxide producing activity
associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochem. Biophys. Res.
Commun. 195:107076
92. Lee AC, Fenster BE, Ito H, Takeda K, Bae NS, et al. 1999. Ras proteins induce senescence by altering
the intracellular levels of reactive oxygen species. J. Biol. Chem. 274:793640
93. Van Der Loo B, Labugger R, Skepper JN, Bachschmid M, Kilo J, et al. 2000. Enhanced peroxynitrite
formation is associated with vascular aging. J. Exp. Med. 192:173144

www.annualreviews.org SASP and Cancer 115


AREV403-PM05-05 ARI 10 December 2009 16:41

94. Macip S, Igarashi M, Fang L, Chen A, Pan ZQ, et al. 2002. Inhibition of p21-mediated ROS accumulation
can rescue p21-induced senescence. EMBO J. 21:218088
95. Xin MG, Zhang J, Block ER, Patel JM. 2003. Senescence-enhanced oxidative stress is associated with
deciency of mitochondrial cytochrome c oxidase in vascular endothelial cells. Mech. Ageing Dev. 124:911
19
96. Finkel T, Holbrook NJ. 2000. Oxidants, oxidative stress and the biology of ageing. Nature 408:23947
97. Finkel T, Serrano M, Blasco MA. 2007. The common biology of cancer and ageing. Nature 448:76774
98. Funayama R, Ishikawa F. 2007. Cellular senescence and chromatin structure. Chromosoma 116:43140
99. Mehta IS, Figgitt M, Clements CS, Kill IR, Bridger JM. 2007. Alterations to nuclear architecture and
genome behavior in senescent cells. Ann. N.Y. Acad. Sci. 1100:25063
100. Narita M. 2007. Cellular senescence and chromatin organisation. Br. J. Cancer 96:68691
101. Adams PD. 2007. Remodeling chromatin for senescence. Aging Cell 6:42527
102. Beausejour CM, Krtolica A, Galimi F, Narita M, Lowe SW, et al. 2003. Reversal of human cellular
senescence: roles of the p53 and p16 pathways. EMBO J. 22:421222
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

103. Narita M, Nunez S, Heard E, Narita M, Lin AW, et al. 2003. Rb-mediated heterochromatin formation
and silencing of E2F target genes during cellular senescence. Cell 113:70316
104. Itahana K, Zou Y, Itahana Y, Martinez JL, Beausejour CM, et al. 2003. Control of the replicative life
span of human broblasts by p16 and the polycomb protein Bmi-1. Mol. Cell. Biol. 23:389401
by University of Sussex on 10/10/12. For personal use only.

105. Herbig U, Jobling WA, Chen BP, Chen DJ, Sedivy JM. 2004. Telomere shortening triggers senescence of
human cells through a pathway involving ATM, p53, and p21CIP1 , but not p16INK4a . Mol. Cell 14:50113
106. Benanti JA, Galloway DA. 2004. Normal human broblasts are resistant to RAS-induced senescence.
Mol. Cell. Biol. 24:284252
107. Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, et al. 2006. p16INK4a induces an age-
dependent decline in islet regenerative potential. Nature 443:45357
108. Janzen V, Forkert R, Fleming H, Saito Y, Waring MT, et al. 2006. Stem cell aging modied by the
cyclin-dependent kinase inhibitor, p16INK4a . Nature 443:42126
109. Molofsky AV, Slutsky SG, Joseph NM, He S, Pardal R, et al. 2006. Increasing Ink4a expression decreases
forebrain progenitors and neurogenesis during ageing. Nature 443:44852
110. Taniguchi K, Kohsaka H, Inoue N, Terada Y, Ito H, et al. 1999. Induction of the p16INK4a senescence
gene as a new therapeutic strategy for the treatment of rheumatoid arthritis. Nat. Med. 5:76067
111. Tyner SD, Venkatachalam S, Choi J, Jones S, Ghebranious N, et al. 2002. p53 mutant mice that display
early aging-associated phenotypes. Nature 415:4553
112. Maier B, Gluba W, Bernier B, Turner T, Mohammad K, et al. 2004. Modulation of mammalian life span
by the short isoform of p53. Genes Dev. 18:30619
113. Kiaris H, Chatzistamou I, Trimis G, Frangou-Plemmenou M, Pati-Kondi A, Kalofoutis A. 2005. Evi-
dence for nonautonomous effect of p53 tumor suppressor in carcinogenesis. Cancer Res. 65:162730
114. Tsai KK, Chuang EY, Little JB, Yuan ZM. 2005. Cellular mechanisms for low-dose ionizing radiation-
induced perturbation of the breast tissue microenvironment. Cancer Res. 65:673444
115. Sun P, Yoshizuka N, New L, Moser BA, Li Y, et al. 2007. PRAK is essential for ras-induced senescence
and tumor suppression. Cell 128:295308
116. Choi J, Shendrik I, Peacocke M, Peehl D, Buttyan R, et al. 2000. Expression of senescence-associated
-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology 56:16066
117. Ohuchida K, Mizumoto K, Murakami M, Qian LW, Sato N, et al. 2004. Radiation to stromal broblasts
increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 64:3215
22
118. Barcellos-Hoff MH, Ravani SA. 2000. Irradiated mammary gland stroma promotes the expression of
tumorigenic potential by unirradiated epithelial cells. Cancer Res. 60:125460
119. Yang F, Tuxhorn JA, Ressler SJ, McAlhany SJ, Dang TD, Rowley DR. 2005. Stromal expression of
connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Res.
65:888795
120. Lehmann BD, Paine MS, Brooks AM, McCubrey JA, Renegar RH, et al. 2008. Senescence-associated
exosome release from human prostate cancer cells. Cancer Res. 68:786471

116 Coppe et al.


AREV403-PM05-05 ARI 10 December 2009 16:41

121. Dilley TK, Bowden GT, Chen QM. 2003. Novel mechanisms of sublethal oxidant toxicity: induction
of premature senescence in human broblasts confers tumor promoter activity. Exp. Cell Res. 290:3848
122. Dhawan P, Richmond A. 2002. Role of CXCL1 in tumorigenesis of melanoma. J. Leukoc. Biol. 72:918
123. Balentien E, Mufson BE, Shattuck RL, Derynck R, Richmond A. 1991. Effects of MGSA/GRO on
melanocyte transformation. Oncogene 6:111524
124. Schadendorf D, Moller A, Algermissen B, Worm M, Sticherling M, Czarnetzki BM. 1993. IL-8 produced
by human malignant melanoma cells in vitro is an essential autocrine growth factor. J. Immunol. 151:2667
75
125. Bernardini G, Spinetti G, Ribatti D, Camarda G, Morbidelli L, et al. 2000. I-309 binds to and activates
endothelial cell functions and acts as an angiogenic molecule in vivo. Blood 96:403945
126. Salcedo R, Young HA, Ponce ML, Ward JM, Kleinman HK, et al. 2001. Eotaxin (CCL11) induces in
vivo angiogenic responses by human CCR3+ endothelial cells. J. Immunol. 166:757178
127. Coppe JP, Kauser K, Campisi J, Beausejour CM. 2006. Secretion of vascular endothelial growth factor
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

by primary human broblasts at senescence. J. Biol. Chem. 281:2956874


128. Sparmann A, Bar-Sagi D. 2004. Ras-induced interleukin-8 expression plays a critical role in tumor growth
and angiogenesis. Cancer Cell 6:44758
129. Frey AB. 2006. Myeloid suppressor cells regulate the adaptive immune response to cancer. J. Clin.
Investig. 116:258790
by University of Sussex on 10/10/12. For personal use only.

130. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. 2003. Met, metastasis, motility and more.
Nat. Rev. Mol. Cell Biol. 4:91525
131. Camphausen K, Moses MA, Beecken WD, Khan MK, Folkman J, OReilly MS. 2001. Radiation therapy
to a primary tumor accelerates metastatic growth in mice. Cancer Res. 61:220711
132. Qian LW, Mizumoto K, Urashima T, Nagai E, Maehara N, et al. 2002. Radiation-induced increase
in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase
inhibitor, CGS27023. Clin. Cancer Res. 8:122327
133. Strieter RM, Burdick MD, Mestas J, Gomperts B, Keane MP, Belperio JA. 2006. Cancer CXC chemokine
networks and tumor angiogenesis. Eur. J. Cancer 42:76878
134. Orr FW, Wang HH. 2001. Tumor cell interactions with the microvasculature: a rate-limiting step in
metastasis. Surg. Oncol. Clin. N. Am. 10:35781
135. Nickoloff BJ, Lingen MW, Chang BD, Shen M, Swift M, et al. 2004. Tumor suppressor maspin is
up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Res.
64:295661
136. Mantovani A. 2004. Chemokines in neoplastic progression. Semin. Cancer Biol. 14:14748
137. Homey B, Muller A, Zlotnik A. 2002. Chemokines: agents for the immunotherapy of cancer? Nat. Rev.
Immunol. 2:17584
138. Balkwill F. 2004. Cancer and the chemokine network. Nat. Rev. Cancer 4:54050
139. Ben-Baruch A. 2006. Inammation-associated immune suppression in cancer: the roles played by cy-
tokines, chemokines and additional mediators. Semin. Cancer Biol. 16:3852
140. Potempa S, Ridley AJ. 1998. Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is
required for hepatocyte growth factor/scatter factorinduced adherens junction disassembly. Mol. Biol.
Cell 9:2185200
141. Paumelle R, Tulasne D, Kherrouche Z, Plaza S, Leroy C, et al. 2002. Hepatocyte growth factor/scatter
factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene
21:230919
142. Thiery JP. 2002. Epithelial-mesenchymal transitions in tumor progression. Nat. Rev. Cancer 2:44254
143. Tonini T, Rossi F, Claudio PP. 2003. Molecular basis of angiogenesis and cancer. Oncogene 22:654956
144. Nesbit M, Schaider H, Miller TH, Herlyn M. 2001. Low-level monocyte chemoattractant protein-1
stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. J. Immunol.
166:648390
145. Schaider H, Oka M, Bogenrieder T, Nesbit M, Satyamoorthy K, et al. 2003. Differential response of
primary and metastatic melanomas to neutrophils attracted by IL-8. Int. J. Cancer 103:33543

www.annualreviews.org SASP and Cancer 117


AREV403-PM05-05 ARI 10 December 2009 16:41

146. Sica A, Schioppa T, Mantovani A, Allavena P. 2006. Tumor-associated macrophages are a distinct M2
polarised population promoting tumor progression: potential targets of anticancer therapy. Eur. J. Cancer
42:71727
147. Selivanova G, Iotsova V, Okan I, Fritsche M, Strom M, et al. 1997. Restoration of the growth suppression
function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nat. Med. 3:632
38
148. Foster BA, Coffey HA, Morin MJ, Rastinejad F. 1999. Pharmacological rescue of mutant p53 confor-
mation and function. Science 286:250710
149. Chene P. 2003. Inhibiting the p53-MDM2 interaction: an important target for cancer therapy. Nat. Rev.
Cancer 3:1029
150. Selivanova G, Wiman KG. 2007. Reactivation of mutant p53: molecular mechanisms and therapeutic
potential. Oncogene 26:224354
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org
by University of Sussex on 10/10/12. For personal use only.

118 Coppe et al.


AR403-FM ARI 9 December 2009 12:46

Annual Review of
Pathology:
Mechanisms of

Contents Disease

Volume 5, 2010
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

Molecular Pathogenesis of Necrotizing Fasciitis


Randall J. Olsen and James M. Musser p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1
The Pathobiology of Glioma Tumors
by University of Sussex on 10/10/12. For personal use only.

Candece L. Gladson, Richard A. Prayson, and Wei Michael Liu p p p p p p p p p p p p p p p p p p p p p p p p p p p33


Mutational Heterogeneity in Human Cancers: Origin and
Consequences
Jesse J. Salk, Edward J. Fox, and Lawrence A. Loeb p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p51
Fibrogenic Reactions in Lung Disease
Jun Araya and Stephen L. Nishimura p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p77
The Senescence-Associated Secretory Phenotype: The Dark Side
of Tumor Suppression
Jean-Philippe Coppe, Pierre-Yves Desprez, Ana Krtolica, and Judith Campisi p p p p p p p p p p p99
Epithelial Barriers in Homeostasis and Disease
Amanda M. Marchiando, W. Vallen Graham, and Jerrold R. Turner p p p p p p p p p p p p p p p p p p 119
Nonalcoholic Fatty Liver Disease: Pathology and Pathogenesis
Dina G. Tiniakos, Miriam B. Vos, and Elizabeth M. Brunt p p p p p p p p p p p p p p p p p p p p p p p p p p p p 145
Pathogenesis of Preeclampsia
Brett C. Young, Richard J. Levine, and S. Ananth Karumanchi p p p p p p p p p p p p p p p p p p p p p p p p 173
Preinvasive Breast Cancer
Dennis C. Sgroi p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 193
The Pathogenesis of Acute Pulmonary Viral and Bacterial Infections:
Investigations in Animal Models
Mary F. Lipscomb, Julie Hutt, Julie Lovchik, Terry Wu, and C. Rick Lyons p p p p p p p p p p p 223
Mammalian Sirtuins: Biological Insights and Disease Relevance
Marcia C. Haigis and David A. Sinclair p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 253
Mitochondrial Energetics and Therapeutics
Douglas C. Wallace, Weiwei Fan, and Vincent Procaccio p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 297

v
AR403-FM ARI 9 December 2009 12:46

p63 in Epithelial Survival, Germ Cell Surveillance, and Neoplasia


Christopher P. Crum and Frank D. McKeon p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 349

Indexes

Cumulative Index of Contributing Authors, Volumes 15 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 373


Cumulative Index of Chapter Titles, Volumes 15 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 376

Errata
Annu. Rev. Pathol. Mech. Dis. 2010.5:99-118. Downloaded from www.annualreviews.org

An online log of corrections to Annual Review of Pathology, Mechanisms of Disease articles


may be found at http://pathol.annualreviews.org
by University of Sussex on 10/10/12. For personal use only.

vi Contents

Vous aimerez peut-être aussi