Skip to main content
Aging Cell logoLink to Aging Cell
. 2016 Sep 14;15(6):999–1017. doi: 10.1111/acel.12518

Small molecule compounds that induce cellular senescence

Nadezhda V Petrova 1, Artem K Velichko 1, Sergey V Razin 1,2,3,, Omar L Kantidze 1,3,
PMCID: PMC6398529  PMID: 27628712

Summary

To date, dozens of stress‐induced cellular senescence phenotypes have been reported. These cellular senescence states may differ substantially from each other, as well as from replicative senescence through the presence of specific senescence features. Here, we attempted to catalog virtually all of the cellular senescence‐like states that can be induced by low molecular weight compounds. We summarized biological markers, molecular pathways involved in senescence establishment, and specific traits of cellular senescence states induced by more than fifty small molecule compounds.

Keywords: cellular senescence, cell stress, DNA damage, DNA replication stress, epigenetic modifiers, aging


Cellular senescence is a stable arrest of the cell cycle and is characterized by complex phenotypic changes. It was first described in studies of human fibroblasts that ceased proliferation following an extended cultivation (Hayflick & Moorhead, 1961; Hayflick, 1965). Discovered by Hayflick and Moorhead, senescence in normal human cells was shown to depend on telomere dysfunction originating mainly from replication‐associated telomere shortening (Harley et al., 1990; Allsopp, 1996; Bodnar et al., 1998). This type of senescence is also known as replicative senescence and is the prototypical cellular senescence state. Other forms of senescence (i.e., not linked to proliferation‐dependent telomere shortening) include a variety of prematurely developed cellular senescence phenotypes, similar but not identical to replicative senescence. Many proliferative cell types can undergo so‐called stress‐induced premature senescence (SIPS) upon exposure to subcytotoxic stresses (UV, γ‐irradiation, H2O2, hyperoxia, etc.) (Toussaint et al., 2000, 2002). Oncogene‐induced senescence (OIS) represents another complex senescence phenotype that depends on activation and/or overexpression of oncogenes (Serrano et al., 1997; Bianchi‐Smiraglia & Nikiforov, 2012). The mechanism of OIS involves DNA damage that may be a result of DNA hyper‐replication (Di Micco et al., 2006), replication fork reversal (Neelsen et al., 2013), depletion of nucleotide pools (Mannava et al., 2013), and/or increased levels of reactive oxygen species (ROS) (Lee et al., 1999). Conceptually and mechanistically, OIS is closely related to tumor‐suppressor loss‐induced senescence (Chen et al., 2005; Di Mitri & Alimonti, 2016). Cell‐to‐cell fusion‐induced senescence can also be considered a premature senescence subtype (Chuprin et al., 2013; Burton & Faragher, 2015). The distinctive phenotypic changes typical of various types of cellular senescence are cell enlargement and flattening, senescence‐associated β‐galactosidase activity (SA‐β‐gal), formation of senescence‐associated heterochromatin foci (SAHF), persistent DNA damage response (DDR), and senescence‐associated secretory phenotype (SASP). However, these and several other facultative features of cellular senescence that manifest in each particular case of cell cycle arrest greatly depend on the senescence‐inducing stimulus and the cell type (Campisi, 2013; Salama et al., 2014).

The contribution of cellular senescence to organismal aging is a question of ongoing research (van Deursen, 2014). However, strong evidence for this connection has been reported recently. Specifically, it was shown that clearance of age‐accumulated p16INK4A‐positive senescent cells in mice could extend their healthy lifespan (Baker et al., 2011, 2016). Several chemical compounds that specifically target senescent cells have been identified in the last 2 years (so‐called senolytic drugs) (Xu et al., 2015b; Zhu et al., 2015a,b). It was shown that clearance of senescent cells by such drugs may alleviate age‐related vasomotor dysfunction and frailty, enhance adipogenesis, rejuvenate haematopoietic stem cells after total‐body irradiation, and, generally, extend lifespan (Xu et al., 2015a; Zhu et al., 2015b; Roos et al., 2016). Furthermore, these studies confirm the known pathological impact of cellular senescence, exemplified by cellular dysfunction, impairment of tissue regeneration, detrimental effects on tissue microenvironment, etc. (Burton & Krizhanovsky, 2014). It is evident that along with its detrimental effects, cellular senescence has clearly defined beneficial physiological functions. For instance, it has been shown recently that cellular senescence plays a role in the differentiation of megakaryocytes (Besancenot et al., 2010), the maturation of the placenta (Chuprin et al., 2013), the restriction of fibrosis (Krizhanovsky et al., 2008; Jun & Lau, 2010; Zhu et al., 2013), tissue repair (Demaria et al., 2014), and embryonic development (Nacher et al., 2006; Munoz‐Espin et al., 2013; Storer et al., 2013). The role of cellular senescence in cancer prevention is well documented (Burton & Krizhanovsky, 2014; Munoz‐Espin & Serrano, 2014).

It is generally agreed in the field that the most important features of cellular senescence are SASP and resistance to apoptosis (Munoz‐Espin & Serrano, 2014; Burton & Faragher, 2015). SASP stimulates immune system‐dependent elimination of unwanted precancerous cells or specific embryonic cells that undergo senescence. Notably, cellular senescence may serve as an alternative to apoptosis in embryonic development as well as in cancer prevention (Childs et al., 2014). It has been shown that failure to undergo senescence triggers apoptosis in a compensatory manner to eliminate transient structures during development (Munoz‐Espin et al., 2013; Storer et al., 2013). Therefore, it may be reasonable to consider some of the cellular senescence states (e.g., SIPS), along with apoptosis, autophagy, necrosis, etc., in terms of the cell stress response rather than aging. However, it is unclear whether or not the cellular senescence that is widely implicated in normal aging, chronic diseases, tumor suppression, tumorigenesis, cell differentiation, and embryogenesis represents a single physiological cellular state.

To date, dozens of stress‐induced cellular senescence phenotypes have been reported. These senescence states may differ substantially from each other, as well as from replicative senescence, through the presence of specific senescence features. Additionally, it has been reported that some stress‐induced senescence states can be overcome, thus challenging the dogma that cellular senescence is an irreversible form of growth arrest (Romanov et al., 2001; Beausejour et al., 2003). Such caveats can lead to confusion regarding the terminology of stress‐induced cellular senescence states; it is not clear whether senescence‐like growth arrest (and variations thereof) resembles ‘true’ cellular senescence. The indispensable characteristics of this ‘true’ cellular senescence are also elusive. It can be argued that SASP (arising along with morphological changes and SA‐β‐gal) may be the most important physiologically relevant feature of cellular senescence; however, SASP has not been studied in most cases of stress‐induced senescence. Here, we attempt to catalog virtually all of the cellular senescence‐like states that can be induced by low molecular weight compounds (Table 1). We summarize the biological markers, molecular pathways involved in senescence establishment, and specific traits of cellular senescence states induced by small compounds, as well as the treatment conditions used. In total, we analyzed more than 50 chemical inducers of cellular senescence and senescence‐like states. These chemical compounds can be functionally classified into eight groups: (1) DNA replication stress inducers (aphidicolin, hydroxyurea, thymidine, bromodeoxyuridine, difluorodeoxycytidine, cyclopentenyl cytosine); (2) DNA‐damaging agents, including (2a) DNA topoisomerase inhibitors (doxorubicin, etoposide, daunorubicin, mitoxantrone, camptothecin), (2b) DNA cross‐linkers (cisplatin, mitomycin C, busulfan, cyclophosphamide, diaziquone), and (2c) drugs with complex effects (actinomycin D, bleomycin, temozolomide); (3) epigenetic modifiers that inhibit DNA methyltransferases (5‐aza‐2′‐deoxycytidine), histone deacetylases (sodium butyrate, trichostatin A, MS‐275, SAHA, LBH589, phenylbutyric acid, valproic acid), histone acetyltransferases (curcumin, C646), and histone methyltransferases (BRD4770); (4) inhibitors of telomerase activity (SYUIQ‐5, BMVC4, pyridostatin, compound 115405, perylene and indole derivatives, harmine, BIBR1532, azidothymidine); (5) inhibitors of cyclin‐dependent kinases (palbociclib, roscovitine, ribociclib); (6) activators of p53 (nutlin 3a, FL118); (7) activators of protein kinase C (TPA/PMA, PEP005, PEP008); and (8) reactive oxygen species (ROS) inducers (hydrogen peroxide, tert‐butyl hydroperoxide, phenyl‐2‐pyridyl ketoxime, phenylaminonaphthoquinones, paraquat).

Table 1.

Low molecular weight compounds that induce cellular senescence

Small compounds/Mechanism of action Cell line Cellular senescence state (as described by authors) Senescence markers documented Signaling pathways involved Cell cycle phase of the stable growth arrest Triggering conditions/Other notes (if any) References
(1) DNA replication stress inducers
 aphidicolin
Inhibitor of DNA polymerase α
HFFa
REF52
Senescence‐like arrest Growth arrest
γH2A.X foci
p53‐p21↑ c
p‐Rb↓
150–200 nm for ~10 days Marusyk et al. (2007)
MCF10
MCF7
Prolonged S‐phase senescence‐like arrest Large flattened cells with increased nuclear size (CSb‐like morphology)
SA‐β‐Gal
γH2A.X foci
increased H3K9‐trimethylation
p21↑
p‐Rb↓
S 1 μg mL−1 for 4 days
reversible state of S‐phase arrest
RPA foci
Maya‐Mendoza et al. (2014)
 hydroxyurea
Ribonucleotide reductase inhibitor
HFF Senescence‐like arrest Growth inhibition CS‐like morphology
SA‐β‐Gal
p53‐p21↑ 400–800 μm for ~3 weeks Yeo et al. (2000)
McA‐RH7777 Senescence‐like arrest Growth inhibition p21↑ G1 200–400 μm for 4 days Hong et al. (2004)
HFF
REF52
MCF10A
Senescence‐like arrest Growth inhibition CS‐like morphology
SA‐β‐Gal
γH2A.X foci
p53‐p21↑
p‐p53Ser15↑
p‐p53Ser20↑
p16‐independent
100–150 μm for ~10 days Marusyk et al. (2007)
K562 Senescence‐like arrest SA‐β‐Gal p16↑
p21↑
p27↑
50–600 μm for up to 14 days Park et al. (2000)
 thymidine
Excess of thymidine inhibits DNA replication by reducing the amount of dCTP synthesized
HeLa
TIG‐7
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
ERK1 and/or ERK2↑ 1.5 mm for 7–10 days Sumikawa et al. (2005), Kobayashi et al. (2012)
 bromodeoxyuridine
Suppresses DNA replication
HeLa S3
TIG‐7
Senescence‐like arrest p21↑ 50 μm for 4 days Eriko et al. (1999), Suzuki et al. (2001)
A549 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
p53‐p21↑
p‐Rb↓
p27↑
p57↑
S
G2
200 μm for 7 days
Chk1Ser345p↑
Chk2Thr68p↑
Masterson & O'Dea (2007)
HeLa
A549
Premature senescence Growth inhibition
CS‐like morphology
γH2AX foci
SASP
p21↑
DDR (ATM)
G1 100 μm for 48 h
elevated ROS levels
p53 activation in A549 cells
Nair et al. (2015)
 2′,2′‐difluorodeoxycytidine (gemcitabine)
Inhibits ribonucleotide reductase
Inhibits CTP synthetase
AsPC1 PANC‐1 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p21↑ Sub‐G1 100 nm for 4 days Modrak et al. (2009)
 cyclopentenyl cytosine
Inhibits CTP synthetase
MCF‐7 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53
p53, p21↑
G2
S
0.125–1 μm for 5 days Huang et al. (2011)
(2) DNA‐damaging agents
(2a) DNA topoisomerases inhibitors
 doxorubicin
DNA intercalator
Induces DSBs by poisoning DNA topoisomerase II
Induces nucleosome eviction
11 cell lines derived from different types of human solid tumors Senescence‐like phenotype Growth inhibition
CS‐like morphology
SA‐β‐gal
Can be dependent or not on p53 activation 20–50 nm for 3–6 days Chang et al. (1999a)
HCT116 Senescence‐like phenotype CS‐like morphology
SA‐β‐gal
p53‐p21
p21↑
G2 phase 50–100 nm for 1–4 days
treatment led to the appearance of a substantial fraction of polyploid nuclei in p53−/− and p21−/− lines
Chang et al. (1999b)
MCF7 Premature senescence CS‐like morphology
SA‐β‐gal
p53
p53, p21↑
1 μm for 2 h Elmore et al. (2002)
HCT116 Senescence‐like phenotype CS‐like morphology
SA‐β‐gal
p53, p21↑ G2 0.1 μM for 24 h Sliwinska et al. (2009)
Neonatal rat cardiomyocytes
H9c2
Premature senescence CS‐like morphology
SA‐β‐gal
p53↑
p‐p38↑
p‐JNK↑
p‐ERK↑
MAPK (p‐38 and JNK)
S 0.1 μm for 3 h Spallarossa et al. (2009)
WI38  Premature senescence CS‐like morphology
SA‐β‐gal
mTOR
p53, p21↑
100 ng mL−1 for 1–4 days
low p53 levels during prolonged cell cycle arrest lead to senescence, while high levels of p53—to either quiescence or cell death
Leontieva et al. (2010)
A549 Transient senescence‐like state CS‐like morphology
SA‐β‐gal
G2 50–200 nm for 72 h Litwiniec et al. (2010)
MMTV‐Wnt1 mice MCF7 Premature senescence Growth inhibition
SA‐β‐gal
SASP
p53, +/− p21 G1 (p53‐and p21‐dependent),
G2 (p21‐independent)
4 mg kg−1day−1 for 5 days,
MCF7 treated with 200 nm for 24 h
in vivo
Jackson et al. (2012)
Cardiac progenitor cells  Premature senescence CS‐like morphology
SA‐β‐gal
γH2AX
p16↑ 0.1–1 μm for 24–48 h
in vivo
Piegari et al. (2013)
DU145
LNCaP
PC3
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p21↑
p27↑
p‐Rb↓
p53‐independent
G1 10 nm for 1–5 days Park et al. (2006)
 etoposide
Poison of DNA topoisomerase II
Induces DSBs
LS174T
A2780
MCF7
Premature senescence Growth arrest
CS‐like morphology
SA‐β‐gal
G1 2 μm for 24 h te Poele et al. (2002)
WI38 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
p53
p‐p53↑
p21↑
G1 20 μm for 24 h
H2AX phosphorylation, peaked around 8 h and completely resolved at 24 h after the treatment
Probin et al. (2006)
A549 Senescence‐like phenotype Growth inhibition
CS‐like morphology
SA‐β‐gal
SAHF
p21↑ G2 0.75–3 μm for 72 h
Polyploid (higher DNA contents (>G2))
Litwiniec et al. (2013)
 daunorubicin
DNA intercalator
Poises topoisomerase II
Jurkat Senescence‐like phenotype Growth inhibition
SA‐β‐Gal
p53‐p21↑ G2 91 nm for 24 h Mansilla et al. (2003)
 mitoxantrone
Topoisomerase IIβ inhibitor
Induces DSBs
Epithelial cells in biopsies from human prostate cancer patients Premature senescence SASP p21↑
p16↑
in vivo Coppe et al. (2008)
A549
WI‐38
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
γ‐H2AX
p21↑
p‐ATM(Ser1981)↑
G1
G2
2 nm for 2–5 days Zhao et al. (2010)
 camptothecin and SN‐38
Topoisomerase I poison
Induces SSBs
LS174T
MCF‐7
A2780
HCT116
Senescence‐like arrest Growth arrest
CS‐like morphology
SA‐β‐gal
p53‐p21↑
p16↑
G1
S
G2
6–100 ng mL−1 for 24–168 h te Poele et al. (2002)
HCT116 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
p53‐p21↑ 20 nm for 24–120 h
high concentration (250 nm) of camptothecin results in apoptosis
Han et al. (2002)
H1299 Premature senescence CS‐like morphology
SA‐β‐gal
ATM/ATR G2 30–60 nm for 2–3 days
p53‐, p16‐, p38‐independent
Roberson et al. (2005)
HCT116 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
γH2AX
ATM‐Chk2‐p53‐p21
p‐ATM↑
p‐Chk2↑
p53↑
p21↑
20 nm for 72 h Zhang et al. (2014)
HeLa Senescence‐like growth arrest Growth inhibition
CS‐like morphology
SA‐β‐gal
γH2AX
p21↑ G2 10–100 nm for 1 h Velichko et al. (2015)
(2b) DNA cross‐linkers
 cisplatin
DNA‐alkylating agent
Induces DNA intrastrand cross‐links
CNE1 Senescence‐like arrest Growth inhibition
SA‐β‐Gal
S
G2
0.5 mkg mL−1 for 24 h
higher doses result in cell death
Wang et al. (1998)
Normal human lung fibroblasts Premature senescence Growth inhibition
CS‐like morphology
p53↑ G1 10 μm for 24 h Zhao et al. (2004)
Human non‐small cell lung cancer cells Senescence‐like arrest Growth inhibition
SA‐β‐gal
p16 G2 5 μm for 3 days Fang et al. (2007)
HCT116 Premature senescence Growth inhibition
SA‐β‐Gal
γH2AX foci
p53↑ 5 μm for 6 h
higher concentrations induce apoptosis
Berndtsson et al. (2007)
HepG2 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53‐p21↑ 2 μg mL−1 for 48 h
ROS↑‐dependent
Qu et al. (2014)
CCL23
CAL27
UM‐SCC1
UM‐SCC14A
Premature senescence SA‐β‐Gal
increased secretion of IL‐8
p53↑
p16↑
p‐Rb↓
6 μg mL−1 for 4 h Veena et al. (2014)
 mitomycin C
DNA‐alkylating agent
Induces DNA interstrand cross‐links
A549 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
γH2AX
p21↑ G2 0.01–0.02 μg mL−1 for 6 days McKenna et al. (2012)
 busulfan
DNA‐alkylating agent
Induces DNA intrastrand cross‐link
Murine bone marrow cells Premature senescence Growth inhibition
SA‐β‐Gal
p16↑
p19↑
30 μm for 6 h Meng et al. (2003)
WI38 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
MAPK(p38, ERK)
p‐p38↑
p‐JNK↑
p‐ERK↑
p21↑
p16↑
G2 7.5–120 μm for 24 h Probin et al., 2006, 2007
 cyclophosphamide
DNA‐alkylating agent
Induces DNA intrastrand and interstrand cross‐links
Lymphoma‐bearing C57BL/6 mice Premature senescence SA‐β‐gal  p53↑
p16↑
300 mg kg−1 day−1 for 7 days
in vivo
Schmitt et al. (2002)
TIG‐7 Premature senescence Growth inhibition
SA‐β‐gal
MAPK (p‐p38, p‐JNK, p‐ERK)↑
p21↑
p16↑
G1
G2
10 μm for 14 days Palaniyappan (2009)
 diaziquone
DNA‐alkylating agent
Induces DNA–DNA and DNA–RNA interstrand cross‐links
DU145 Premature senescence CS‐like morphology
SA‐β‐gal
0.25–10 μm for 3 days Ewald et al. (2009)
(2c) DNA‐damaging drugs with complex effects
 actinomycin D
DNA intercalator
Inhibits transcription
Can poison topoisomerases I and II, and, thus induce SSBs and DSBs
Normal human fibroblasts Premature senescence Growth inhibition p53‐p21↑ G1
G2
0.04 mg mL−1 for 12 h Robles and Adami (1998)
Human mesenchymal stem cells Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
γ‐H2AX foci
SASP
p53‐p21↑
p16↑
400 mm for 3–21 days Minieri et al. (2015)
 bleomycin
Induces DNA breaks
Normal human fibroblasts Premature senescence Growth inhibition
SA‐β‐gal
p53‐p21↑
p16↑
G1
G2
0.06 units mL−1 for 12–24 h Robles and Adami (1998)
A549 Rat primary type II cells
C57BL/6J mice
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
p21↑ 50 μg mL−1 for 120 h or 5 mg kg−1 day−1 for 7–21 days
in vivo
Aoshiba et al. (2003)
A549 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
p53‐p21↑ G2 50 mU mL−1 for 1–7 days
siRNA for caveolin‐1 reduces SA‐β‐gal
Linge et al. (2007)
BJ
293T
Premature senescence CS‐like morphology
SA‐β‐gal
SASP
100 μg mL−1 for 24 h Pazolli et al. (2012)
C57BL/6J mice Premature senescence γH2AX
p‐53BP1
SASP
p21↑
p‐ATM/ATR↑
p‐p38↑
2.5 mg kg−1 day−1 for 7–21 days
in vivo
Aoshiba et al. (2013)
 temozolomide
DNA‐alkylating agent
Alkylates/methylates DNA
Induces DNA damage
U‐87 MG Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53↑, p21↑ G2 100 μm for 3 h
the gradual appearance of hyperploid cells
Hirose et al. (2001)
Me4405
IR3
Mel‐CV
MM200
SK‐mel‐28
Mel‐FH
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53↑, p21↑ G2 25–100 μm for 72 h
the gradual appearance of hyperploid cells
Mhaidat et al. (2007)
(3) Epigenetic modifiers
 5‐aza‐2′‐deoxycytidine
Inhibitor of DNA methyltransferases
Induces DSBs
MDAH041 Premature senescence CS‐like morphology
SA‐β‐gal
p16↑ S 1 μm for 6 days Vogt et al. (1998)
HepG2
NMRI mice
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
γ‐H2AX
accumulation of H3K9me3
SASP
p53
p16↑
20–50 μm for 96 h or 0.8 mg kg−1 day−1 for 3 days
in vivo
Venturelli et al. (2013)
U2OS Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53↑
p21↑
p16 ↑
5–10 μm for 2–4 days Widodo et al. (2007)
H28 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p21↑
p27↑
ATM, ATR↑
0.1–10 μm for 2–6 days Amatori et al. (2011)
 sodium butyrate
Class I and II histone deacetylase (HDAC) inhibitor
WI38 Senescence‐like state Growth inhibition
CS‐like morphology
SA‐β‐gal
p‐ Rb ↓ G1 0.5 mm for ~20 days Ogryzko et al. (1996)
NIH3T3 Senescence‐like state CS‐like morphology p21↑ G1 5–10 mm for 48 h
activation of p21 expression may be both p53‐dependent and p53‐independent
Xiao et al. (1997)
HHUA
Hec1‐A
SKOV‐3
HeLa
SiHa
Senescence‐like state SA‐β‐gal p21↑
p‐Rb↓
G1
G2
1–4 mm for 2–5 days
activation of p21 expression may be both p53‐dependent and p53‐independent
Terao et al. (2001)
WI‐38 Senescence‐like state CS‐like morphology
SA‐β‐gal
p21↑ 4 mM for 24 h or 0.5 mM for 14 days Place et al. (2005)
E1A + Ras‐transfected rat and mouse embryonic fibroblasts Premature senescence γ‐H2AX p21↑
p16↑
G1 4 mM for 24–72 h
DDR without detectable DNA damage
Abramova et al. (2006), Pospelova et al. (2009)
BJ
293T
Premature senescence CS‐like morphology
SASP
p53‐ and RB‐independent 4 mM for 3‐6 days
SASP dependent upon ATM and NF‐κB
Pazolli et al. (2012)
 trichostatin A
Class I and II HDAC inhibitor
WI38 Senescence‐like state Growth inhibition G1 phase 10 ng mL−1 for ~30 days Ogryzko et al. (1996)
WI‐38 Senescence‐like state CS‐like morphology
SA‐β‐gal
p21↑ 2 μm for 24–72 h or 0.5 μm for 9 days Place et al. (2005)
BJ
293T
Premature senescence CS‐like morphology
SA‐β‐gal
SASP
p53‐ and RB‐independent 1 mM for 3 days
lack of DNA damage
Pazolli et al. (2012)
A549 Premature senescence Growth inhibition
CS‐like morphology
p21↑
p27↑
G1
G2
0.5–1.0 μm for 48 h Zhao et al. (2010)
 MS‐275
Class I HDAC inhibitor
Mesenchymal stem cells Premature senescence SA‐β‐gal p16↑ Predominantly G2 1 μm for 72 h Di Bernardo et al. (2009)
 SAHA (vorinostat)
Class I and II HDAC inhibitor
HCT116 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53‐ and p21‐independent G1
G2
0.4–1 μm for 5 days
induced polyploid cells
Xu et al. (2005)
 LBH589 (panobinostat)
Class I and II HDAC inhibitor
B143
MG‐63
Saos‐2
SJSA
U2OS human osteosarcoma cell lines
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
SASP
p53‐independent G1 15 nm for 21 days aysor 2–10 mg kg−1 day−1 for 17 days
in vivo (mice)
Cain et al. (2013)
 4‐phenylbutyric acid
Class I and IIa HDAC inhibitor
MCF7
HT1080
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
Akt‐p21↑ 200–500 μm for 6 days Kim et al. (2012)
 valproic acid
Class I and IIa HDAC inhibitor
D283‐Med
DAOY
PFSK
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p21↑ G1 0.6–1 mm for 3–7 days or 400 mg kg−1 day−1 for 28 days
in vivo
Li et al. (2005)
Bel‐7402
Bel‐7404
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p21↑, pRb↓ G1 0.2–0.5 mm for 2–5 days An et al. (2013)
 curcumin and C646
p300 histone acetyltransferase inhibitors
TIG3 Senescence‐like state SA‐β‐gal
SAHF
p53‐, p21‐ and p16‐independent G2 6–9 μm for 2–15 days
Global H3, H4 hypoacetylation
Lack of DNA damage
Prieur et al. (2011)
HCT116
MCF 7
U2OS
Premature senescence CS‐like morphology
SA‐β‐gal
p21↑
p53‐independent
G2 10 μm (HCT116) or 15 μm (MCF 7) or 7.5 μm (U2OS) for 24 h Mosieniak et al. (2012)
CAF myofibroblasts Premature senescence CS‐like morphology
SA‐β‐gal
p16↑
p53↑
p21↑
10 μm for 24 h Hendrayani et al. (2013)
VSMC endothelial cells derived from aorta Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐gal
SASP
p21↑
p16↑
p‐p53Ser15↑
p‐p38↑
G2 5–7.5 μm (VSMCs) and 2.5–5 μm (endothelial cells) for 3–7 days
ROS‐ and ATM‐independent
Grabowska et al. (2015)
 BRD4770
G9a histone methyltransferase inhibitor
PANC‐1 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p‐ATM↑ G2 10 μm for 24 h Yuan et al. (2012)
(4) Inhibitors of telomerase activity
 SYUIQ‐5
Stabilizes G‐quadruplexes
Induces TRF2 delocalization from telomeres
K562
SW620
Premature senescence Growth inhibition
SA‐β‐Gal
p16↑
p21↑
p27↑
0.2–0.4 μm for 16–35 days Zhou et al. (2006)
 BMVC4
Stabilizes G‐quadruplexes
H1299
MCF7
HeLa
VA13
SaoS2
U2OS
Premature senescence Growth inhibition
SA‐β‐Gal
SAHF (H3K9me3)
γ‐H2AX foci
p‐ATM↑
p‐Rb↓
S 1–10 μm for 9–12 days Huang et al. (2012)
 pyridostatin
Stabilizes G‐quadruplexes
HT1080 Premature senescence Growth inhibition
SA‐β‐Gal
0.3–40 μm for 8 days Muller et al. (2012)
 compound 115405
G‐quadruplex ligand
A549 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
0.4 μm for ~40 days Riou et al. (2002)
 perylene derivatives PM2 and PIPER
Induce G‐quadruplex formation from both telomeric DNA and hTERT promoter region
A549 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
0.4–0.8 μm for 24–78 days Taka et al. (2013)
 harmine
β‐carboline alkaloid
MCF7 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
γ‐H2AX
p53‐p21↑ 20–30 μm for 48–96 h Zhao and Wink (2013)
 indole derivatives (indole‐3‐carbinol (I3C) and indoxyl sulfate)
Phytochemicals that downregulate hTERT expression
MCF7 Premature senescence Growth inhibition
SA‐β‐Gal
G1 200 μm for 48 h Marconett et al. (2011)
HK‐2
CRF rats
Premature senescence Growth inhibition
SA‐β‐Gal
p53↑
p‐p53Ser15↑
250 μm for 48–120 h or 4 g kg−1 for 16 weeks
in vivo
Shimizu et al. (2010)
HK‐2 Premature senescence Growth inhibition
SA‐β‐Gal
NF‐κB↑ 250 μm for 48–72 h
ROS↑
Shimizu et al. (2011)
 BIBR1532
Non‐nucleosidic TERT inhibitor
NCI‐H460 Senescence‐like phenotype Growth inhibition
CS‐like morphology
SA‐β‐Gal
10 mm for 130 days Damm et al. (2001)
 azidothymidine (AZT)
Reverse transcriptase inhibitor
Inhibits telomerase activity
MCF‐7 Senescence‐like arrest Growth inhibition
SA‐β‐Gal
20 and 70 μm for ~ 50–60 population doublings (PD) Ji et al. (2005)
HTLV‐I
ATL patients
Premature senescence Growth inhibition
SA‐β‐Gal
p53↑
p21↑
p27↑
50 μm for 18 weeks
in vivo (ATL patients) the Jurkat T‐cell line, treated under the same conditions, did not enter growth arrest
Datta et al. (2006)
MASC
C57BL/6 mice
Premature senescence Growth inhibition
SA‐β‐Gal
30 μm for 48 h or 100 mg kg−1 day−1 for 2 weeks in vivo Demir and Laywell (2015)
(5) cyclin‐dependent kinase (CDK) inhibitors
 palbociclib (PD‐0332991)
CDK4 and CDK6 inhibitors
HT1080
MEL10
RPE
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
mTOR
pRb↓
0.5 μm for 3–7 days Leontieva and Blagosklonny (2013)
MEL 10
RPE
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
mTOR, MEK 1 μm for 5 days Leontieva et al. (2013)
12 sarcoma cell lines generated directly from patient samples Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
53BP1 foci
pRb↓ G1 9–27 μm for 2–4 days or 100 mg kg−1 day−1 for 3 weeks
in vivo (mice)
Perez et al. (2015)
MCF7 Premature senescence Growth inhibition
SA‐β‐Gal
G1 1 μm for 5–7 days,
0.6 or 3.0 mg kg−1 day for 10 cycles each consisting of 3 weeks
in vivo (dogs)
Hu et al. (2015)
1205Lu
WM983
WM983BR
WM451Lu
WM239A
WM3918
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
SASP
SAHF
pRb↓
mTOR↓
G1 1 μm for 8 days or 90 mg kg−1 day for 14 days
in vivo (mice)
Yoshida et al. (2016)
roscovitinE (seliciclib)
CDK2, CDK7, and CDK9 inhibitors
RTE
MDCK
WT 9‐7
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53‐p21↑ 1–10 μg mL−1 for 24 h Park et al. (2009)
ribociclib (LEE011)
CDK4 and CDK6 inhibitors
Human neuroblastoma‐derived cells Premature senescence Growth inhibition
SA‐β‐Gal
p‐Rb↓ G1 500 nm for 6 days or 200 mg kg−1 for 21 days
in vivo (mice)
Rader et al. (2013)
(6) p53 activators
 nutlin‐3a
Inhibits MDM2 binding to p53
MEF oncogenically transformed MEF murine fibrosarcoma cell lines Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53↑ 5 or 10 μm for 1–7 days no apoptosis was observed Efeyan et al. (2007)
MCF‐7 Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53 G1
G2
10 μm for 5 days Huang et al. (2011)
MyLa2000
Mac1
Mac2a
Premature senescence Growth inhibition
SA‐β‐Gal
p53↑
p21↑
G1 2.5–10 μm for 24–72 h induces apoptosis Manfe et al. (2012)
 FL118
Camptothecin analogue
Induces proteasomal degradation of MdmX
HCT116
HCT8
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p53↑
p21↑
10 nm for 3 days Ling et al. (2014)
(7) activators of protein kinase C (PKC)
 TPA/PMA (12‐O‐tetradecanoylphorbol‐13‐acetate/(phorbol‐12‐myristate‐13‐acetate)
Activates PKC
Induces DNA damage
D04
D08
MM127
MM455
Premature senescence Growth inhibition
SA‐β‐Gal
p21↑
ERK↑
p‐Rb↓
G1 0.1–1 μg mL−1 for 24 h
telomerase was selectively repressed; normal human fibroblasts were resistant to treatment
Cozzi et al. (2006)
H358
H441
H322
Premature senescence Growth inhibition
CS‐like morphology
SA‐β‐Gal
p21↑
pRb↓
G2 100 nm for 30 min reduced telomerase activity Oliva et al. (2008)
SK‐MEL‐5
MCF7
COLO‐205
Senescence‐like arrest Growth inhibition
SA‐β‐Gal
p21↑
ERK1/2↑
p‐Rb↓
G2 10‐1000 ng mL−1 for 24 h Mason et al. (2010)
 PEP005 (ingenol‐3‐angelate)
Activates PKC
D04
D08
MM127
MM455
Premature senescence Growth inhibition
SA‐β‐Gal
p21↑
ERK↑
G1 0.2–1 μg mL−1 for 24 h telomerase was selectively repressed normal human fibroblasts were resistant to treatment Cozzi et al. (2006)
PEP008 (20‐O‐acetylingenol‐3‐angelate) SK‐MEL‐5
MCF7
COLO‐205
Senescence‐like arrest Growth inhibition
SA‐β‐Gal
p21↑
ERK1↑
p‐Rb ↓
G2 10–1000 ng mL−1 for 24 h or 5–6 days Mason et al. (2010)
(8) ROS inducers
hydrogen peroxide (H2O2) F65 Senescence‐like arrest CS‐like morphology
SA‐β‐Gal
200 μm for 2 h Chen and Ames (1994)
IMR‐90 Senescence‐like arrest CS‐like morphology
SA‐β‐Gal
p53‐p21↑
p‐Rb↓
G1 300 μm for 2 h Chen et al. (1998)
IMR‐90 Senescence‐like arrest CS‐like morphology
SA‐β‐Gal
p‐p38↑
p‐Rb↓
150 μm for 2 h Frippiat et al. (2002)
2BS Premature senescence CS‐like morphology
SA‐β‐Gal
p53‐p21↑ G1 10 μm for 3 weeks accumulation of DNA damage accelerated telomere shortening Duan et al. (2005)
A549 Premature senescence CS‐like morphology
SA‐β‐Gal
p53‐p21↑
p‐Rb↓
100 μm for 2 h Yoshizaki et al. (2009)
Primary human keratinocytes Premature senescence CS‐like morphology
SA‐β‐Gal
p53‐p21↑ 50 μm for 2 h Ido et al. (2012)
HUVEC Premature senescence CS‐like morphology
SA‐β‐Gal
SASP
p53‐p21↑ 100 μm for 1 h Suzuki et al. (2013)
hMESCs Premature senescence CS‐like morphology
SA‐β‐Gal
γH2A.X and p‐53BP1 foci
p‐p38↑
p53‐p21↑
p‐Rb↓
G1 200 μm for 1 h Burova et al. (2013), Borodkina et al. (2014)
WI‐38
IMR‐90
LF1
HCA2
Premature senescence SA‐β‐Gal p21↑
p‐Rb↓
500 μm for 2 h Gorbunova et al. (2002)
IMR‐90 Premature senescence CS‐like morphology
SA‐β‐Gal
p21↑
p‐ERK↑
p‐Akt↑
150 μm for 2 h caveolin 1↑ Chretien et al. (2008)
IMR‐90 Premature senescence SA‐β‐Gal p‐p38↑
p21↑
200 μm for 2 h Zdanov et al. (2006)
HUVEC Premature senescence CS‐like morphology
SA‐β‐Gal
p53↑ 100 μm for 1 h Ota et al. (2008)
MRC‐5 Premature senescence von Zglinicki et al. (2000)
 tertbutyl hydroperoxide (tBHP) WI‐38 Premature senescence CS‐like morphology
SA‐β‐Gal
p21↑
p‐Rb↓
5 × 30 μm for 1 h day−1 Dumont et al. (2000)
HUVEC Premature senescence CS‐like morphology
SA‐β‐Gal
100 μm for 2 days Kurz et al. (2004)
WI‐38 Premature senescence CS‐like morphology
SA‐β‐Gal
SASP
5 × 30 μm for 1 h day−1 Pascal et al. (2007)
WI‐38 Premature senescence CS‐like morphology
SA‐β‐Gal
p21↑
p16↑
G1 4 × 100 μm for 1 h per every two doubling Chen et al. (2008)
Human mesangial cells Premature senescence CS‐like morphology
SA‐β‐Gal
JAK2‐STAT↑ G1 30 μm for 1 h Zhou et al. (2013)
phenyl‐2‐pyridyl ketoxime (PPKO) Primary human fibroblasts were isolated from newborn foreskins Senescence‐like arrest CS‐like morphology
SA‐β‐Gal
p53‐p21↑
p16↑
ERK1/ERK2↑
ROS‐ and NO↑‐dependent
G2 1 mm for 3 days Yang et al. (2016)
phenylaminonaphthoquinones (Q7 and Q9) T24 Senescence‐like arrest CS‐like morphology
SA‐β‐Gal
MAPK
p53‐p21↑
p27↑
G2 4 μm for 1–3 days alone or with 1 mM ascorbate Felipe et al. (2013)
paraquat TIG‐7 Premature senescence CS‐like morphology
SA‐β‐Gal
100 μm for 4 days Joguchi et al. (2004)
BALB/c mice Premature senescence SA‐β‐Gal 25 mg kg−1 for 3 days (intraperitoneal injection) Ota et al. (2008)
BJ Premature senescence CS‐like morphology
SA‐β‐Gal
p53↑
p16↑
350 μm for 16 h thioredoxin 1↑ Young et al. (2010)
a

Cell lines: 1205Lu, human lung melanoma cells; 2BS, human embryonic lung fibroblasts; A2780, human ovarian carcinoma cells; A549, human lung adenocarcinoma epithelial cells; AsPC1, human pancreas adenocarcinoma cells; B143, human osteosarcoma cells; Bel‐7402, human hepatocellular carcinoma cells; Bel‐7404, human hepatocellular carcinoma cells; BJ, normal human foreskin fibroblasts; CAF, cancer‐associated fibroblasts; CAL27, human oral adenosquamous carcinoma cells; CNE1, human nasopharyngeal carcinoma cells; D283‐Med, human medulloblastoma cells; DAOY, human cerebellar medulloblastoma cells; DU145, human prostate carcinoma cells; F65, human foreskin fibroblasts; H1299, human lung carcinoma cells; H28, human mesothelioma cells; H9c2, rat cardiomyoblast cells; HCA2, normal human foreskin fibroblasts; HCT116, human colorectal carcinoma cells; HCT8, human ileocecal colorectal adenocarcinoma cells; Hec1‐A, human uterus/endometrium adenocarcinoma cells; HeLa, human cervix adenocarcinoma cells; HepG2, human hepatocellular carcinoma cells; HFF, human foreskin normal fibroblasts; HHUA, human endometrial cells; HK‐2, human renal proximal tubule cells; hMESCs, human endometrium‐derived mesenchymal stem cells; HT1080, human connective tissue fibrosarcoma cells; HTLV‐I, human T‐cell leukemia virus type I (HTLV‐I)‐infected cells; HUVEC, human umbilical vein endothelial cells; IMR‐90, human fetal lung fibroblasts; Jurkat, human T‐cell leukemia cells; K562, human bone marrow myelogenous leukemia lymphoblasts; LF1, human embryonic lung fibroblasts; LNCaP, human prostate carcinoma cells; LS174T, human colorectal adenocarcinoma cells; Mac1, human cutaneous T‐cell lymphoma (CTCL); Mac2a, human cutaneous T‐cell lymphoma (CTCL); MASC, mouse multipotent astrocytic stem cell; McA‐RH7777, rat hepatoma cells; MCF10, human breast fibrocystic cells; MCF7, human breast adenocarcinoma cells; MDAH041, derivate from the fibroblasts of a patient with Li–Fraumeni syndrome; MDCK, canine epithelial kidney cells; MEF, mouse embryonic fibroblasts; MEL10 (SK‐MEL‐147), human melanoma cells; MG‐63, human osteosarcoma cells; MRC‐5, human lung fibroblast; MyLa2000, human cutaneous T‐cell lymphoma (CTCL); NCI‐H460, human lung carcinoma cells; NIH3T3, mouse embryo fibroblasts; PANC‐1, human pancreatic carcinoma cells; PC‐3, human prostate cancer cells; PFSK‐1, human neuroectodermal cells derived from cerebral brain tumor; REF52, rat embryonic fibroblasts; RPE, human retinal pigment epithelial cells; RTE, rat tracheal epithelial cells; Saos‐2, human osteosarcoma cells; SiHa, human cervix squamous cell carcinoma cells; SJSA‐1, human osteosarcoma cells; SKN‐SH, human neuroblastoma cells; SKOV‐3, human ovary adenocarcinoma cells; SW620, human colon cancer cells; T24, human bladder carcinoma cells; TIG‐3, human embryonic lung fibroblasts; TIG‐7, human embryonic lung fibroblasts; U2OS, human osteosarcoma cells; U‐87 MG, human glioblastoma, astrocytoma cells; UM‐SCC1, human squamous carcinoma of the oral cavity cells; UM‐SCC14A, human squamous carcinoma of the oral cavity cells; VA‐13, human lung fibroblasts; VSMC, vascular smooth muscle cells; WI38, human lung fibroblasts; WM239A, human melanoma cells; WM3918, human melanoma cells; WM451Lu, human melanoma cells; WM983, human melanoma cells; WM983BR, human melanoma cells; WT 9‐7, human cells from a patient with autosomal‐dominant polycystic kidney disease (ADPKD).

b

Abbreviations: CS, cellular senescence; DSBs, double‐stranded DNA breaks; SA‐β‐gal, senescence‐associated β‐galactosidase; SAHF, senescence‐associated heterochromatin foci; SASP, senescence‐associated secretory phenotype; SSBs, single‐stranded DNA breaks.

c

Symbols: ↑, increased activity/expression reported; ↓, decreased activity/expression reported; , involvement of the protein/pathway was verified by gene(s) knockout or knockdown, inhibitory analysis, and/or using cell lines carrying inactivating mutations.

The table highlights the fact that cancer cells can undergo cellular senescence in vitro just as well as their normal nontransformed counterparts. It is apparent that there is no senescence marker or pathway unique to normal or cancer cells. In most cases, increased SA‐β‐gal, morphological changes, and persistent DDR foci were recorded. SAHF were found in only a few cases (aphidicolin, etoposide, palbociclib, and epigenetic modifiers). SASP was also noted only in some cases; however, this is likely because SASP is not commonly analyzed as a senescence biomarker. Apparently, an implicit consensus was established that the demonstration of SA‐β‐gal, morphological changes, and persistent DDR is sufficient to document a cellular senescence‐like state. It is notable that authors designated these phenotypes as a state of premature senescence or senescence‐like cell cycle arrest, regardless of the set of biomarkers observed in each case.

Extremely prolonged drug exposure (from hours to days) was typically required to induce cellular senescence, as is evidenced by the table. In marginal situations, as in the case of aphidicolin‐induced cell cycle arrest, the full set of senescence biomarkers (SA‐β‐gal, cell enlargement, SAHF, and DDR foci) was maintained, while the drug was present in the culture medium and lost upon drug removal (Maya‐Mendoza et al., 2014). The requirement for prolonged incubation time was found for all groups of chemical compounds analyzed; however, the mechanism of senescence development appeared to differ among these groups. Whereas replication stress inducers, different DNA‐damaging agents, and telomerase inhibitors likely generate a persistent DDR following prolonged introduction of a small number of DNA lesions or telomere uncapping, long‐term incubation with epigenetic modifiers likely causes transcriptional activation of repressed loci (particularly INK4A, which encodes p16 CDK inhibitor). This hypothesis is supported by the fact that, in contrast to DNA damage‐induced cellular senescence, which depends on p21 CDK inhibitor, epigenetically induced senescence is mostly dependent on p16. This characterizes epigenetically induced senescence as ‘causeless’—epigenetic modifiers directly activate molecular pathways maintaining the cellular senescence state without generating any cell stress. In this regard, senescence induced by epigenetic modifiers can resemble developmentally programmed or organismal aging‐associated cellular senescence, while replication stress‐ and DNA damage‐induced senescence are examples of stress‐induced premature senescence states.

It follows from the table that replication stress‐ and DNA damage‐induced cellular senescence mostly depend on the p53‐p21 pathway. The same is basically true for cellular senescence induced by physical stressors such as ionizing radiation (IR) and ultraviolet (UV) (Latonen et al., 2001; Suzuki et al., 2006). It is well known that IR as well as UV can stimulate senescence in a variety of normal and cancer cell lines (Chainiaux et al., 2002; Meng et al., 2003; Jones et al., 2005; Jee et al., 2009). Mechanistically, this type of cellular senescence mostly depends on DNA damage induced by these stressors; this links IR and UV to chemical DNA‐damaging agents. Moreover, IR and UV, along with most of the DNA‐damaging agents presented in the table, induce apoptosis rather than senescence when used at higher doses. These observations further emphasize the relationship between apoptosis and senescence. Accordingly, these cell stress response pathways may operate either as alternatives or as supplement to each other. While prominent (but short term) DNA damage induces apoptosis, prolonged mild DNA damage activates cellular senescence. The p53 transcription factor emerges as a master regulator controlling these cell fate decisions (Purvis et al., 2012).

Funding

This work was supported by a Russian Science Foundation [grant number 14‐24‐00022].

Conflict of interest

None declared.

Contributor Information

Sergey V. Razin, Email: sergey.v.razin@usa.net

Omar L. Kantidze, Email: kantidze@gmail.com

References

  1. Abramova MV, Pospelova TV, Nikulenkov FP, Hollander CM, Fornace AJ Jr, Pospelov VA (2006) G1/S arrest induced by histone deacetylase inhibitor sodium butyrate in E1A + Ras‐transformed cells is mediated through down‐regulation of E2F activity and stabilization of beta‐catenin. J. Biol. Chem. 281, 21040–21051. [DOI] [PubMed] [Google Scholar]
  2. Allsopp RC (1996) Models of initiation of replicative senescence by loss of telomeric DNA. Exp. Gerontol. 31, 235–243. [DOI] [PubMed] [Google Scholar]
  3. Amatori S, Bagaloni I, Viti D, Fanelli M (2011) Premature senescence induced by DNA demethylating agent (Decitabine) as therapeutic option for malignant pleural mesothelioma. Lung Cancer 71, 113–115. [DOI] [PubMed] [Google Scholar]
  4. An HM, Xue YF, Shen YL, Du Q, Hu B (2013) Sodium valproate induces cell senescence in human hepatocarcinoma cells. Molecules 18, 14935–14947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Aoshiba K, Tsuji T, Nagai A (2003) Bleomycin induces cellular senescence in alveolar epithelial cells. Eur. Respir. J. 22, 436–443. [DOI] [PubMed] [Google Scholar]
  6. Aoshiba K, Tsuji T, Kameyama S, Itoh M, Semba S, Yamaguchi K, Nakamura H (2013) Senescence‐associated secretory phenotype in a mouse model of bleomycin‐induced lung injury. Exp. Toxicol. Pathol. 65, 1053–1062. [DOI] [PubMed] [Google Scholar]
  7. Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM (2011) Clearance of p16Ink4a‐positive senescent cells delays ageing‐associated disorders. Nature 479, 232–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Saltness RA, Jeganathan KB, Verzosa GC, Pezeshki A, Khazaie K, Miller JD, van Deursen JM (2016) Naturally occurring p16(Ink4a)‐positive cells shorten healthy lifespan. Nature 530, 184–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Beausejour CM, Krtolica A, Galimi F, Narita M, Lowe SW, Yaswen P, Campisi J (2003) Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 22, 4212–4222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Berndtsson M, Hagg M, Panaretakis T, Havelka AM, Shoshan MC, Linder S (2007) Acute apoptosis by cisplatin requires induction of reactive oxygen species but is not associated with damage to nuclear DNA. Int. J. Cancer 120, 175–180. [DOI] [PubMed] [Google Scholar]
  11. Besancenot R, Chaligne R, Tonetti C, Pasquier F, Marty C, Lecluse Y, Vainchenker W, Constantinescu SN, Giraudier S (2010) A senescence‐like cell‐cycle arrest occurs during megakaryocytic maturation: implications for physiological and pathological megakaryocytic proliferation. PLoS Biol. 8, e1000476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bianchi‐Smiraglia A, Nikiforov MA (2012) Controversial aspects of oncogene‐induced senescence. Cell Cycle 11, 4147–4151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin GB, Harley CB, Shay JW, Lichtsteiner S, Wright WE (1998) Extension of life‐span by introduction of telomerase into normal human cells. Science 279, 349–352. [DOI] [PubMed] [Google Scholar]
  14. Borodkina A, Shatrova A, Abushik P, Nikolsky N, Burova E (2014) Interaction between ROS dependent DNA damage, mitochondria and p38 MAPK underlies senescence of human adult stem cells. Aging 6, 481–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Burova E, Borodkina A, Shatrova A, Nikolsky N (2013) Sublethal oxidative stress induces the premature senescence of human mesenchymal stem cells derived from endometrium. Oxid. Med. Cell. Longev. 2013, 474931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Burton DG, Faragher RG (2015) Cellular senescence: from growth arrest to immunogenic conversion. Age 37, 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Burton DG, Krizhanovsky V (2014) Physiological and pathological consequences of cellular senescence. Cell. Mol. Life Sci. 71, 4373–4386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cain JE, McCaw A, Jayasekara WS, Rossello FJ, Marini KD, Irving AT, Kansara M, Thomas DM, Ashley DM, Watkins DN (2013) Sustained low‐dose treatment with the histone deacetylase inhibitor LBH589 induces terminal differentiation of osteosarcoma cells. Sarcoma 2013, 608964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Campisi J (2013) Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75, 685–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chainiaux F, Magalhaes JP, Eliaers F, Remacle J, Toussaint O (2002) UVB‐induced premature senescence of human diploid skin fibroblasts. Int. J. Biochem. Cell Biol. 34, 1331–1339. [DOI] [PubMed] [Google Scholar]
  21. Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, Kandel ES, Lausch E, Christov K, Roninson IB (1999a) A senescence‐like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 59, 3761–3767. [PubMed] [Google Scholar]
  22. Chang BD, Xuan Y, Broude EV, Zhu H, Schott B, Fang J, Roninson IB (1999b) Role of p53 and p21waf1/cip1 in senescence‐like terminal proliferation arrest induced in human tumor cells by chemotherapeutic drugs. Oncogene 18, 4808–4818. [DOI] [PubMed] [Google Scholar]
  23. Chen Q, Ames BN (1994) Senescence‐like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells. Proc. Natl Acad. Sci. USA 91, 4130–4134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chen QM, Bartholomew JC, Campisi J, Acosta M, Reagan JD, Ames BN (1998) Molecular analysis of H2O2‐induced senescent‐like growth arrest in normal human fibroblasts: p53 and Rb control G1 arrest but not cell replication. Biochem J. 332(Pt 1), 43–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, Cordon‐Cardo C, Pandolfi PP (2005) Crucial role of p53‐dependent cellular senescence in suppression of Pten‐deficient tumorigenesis. Nature 436, 725–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chen X, Zhang J, Fang Y, Zhao C, Zhu Y (2008) Ginsenoside Rg1 delays tert‐butyl hydroperoxide‐induced premature senescence in human WI‐38 diploid fibroblast cells. J. Gerontol. A Biol. Sci. Med. Sci. 63, 253–264. [DOI] [PubMed] [Google Scholar]
  27. Childs BG, Baker DJ, Kirkland JL, Campisi J, van Deursen JM (2014) Senescence and apoptosis: dueling or complementary cell fates? EMBO Rep. 15, 1139–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chretien A, Piront N, Delaive E, Demazy C, Ninane N, Toussaint O (2008) Increased abundance of cytoplasmic and nuclear caveolin 1 in human diploid fibroblasts in H(2)O(2)‐induced premature senescence and interplay with p38alpha(MAPK). FEBS Lett. 582, 1685–1692. [DOI] [PubMed] [Google Scholar]
  29. Chuprin A, Gal H, Biron‐Shental T, Biran A, Amiel A, Rozenblatt S, Krizhanovsky V (2013) Cell fusion induced by ERVWE1 or measles virus causes cellular senescence. Genes Dev. 27, 2356–2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J (2008) Senescence‐associated secretory phenotypes reveal cell‐nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Cozzi SJ, Parsons PG, Ogbourne SM, Pedley J, Boyle GM (2006) Induction of senescence in diterpene ester‐treated melanoma cells via protein kinase C‐dependent hyperactivation of the mitogen‐activated protein kinase pathway. Cancer Res. 66, 10083–10091. [DOI] [PubMed] [Google Scholar]
  32. Damm K, Hemmann U, Garin‐Chesa P, Hauel N, Kauffmann I, Priepke H, Niestroj C, Daiber C, Enenkel B, Guilliard B, Lauritsch I, Muller E, Pascolo E, Sauter G, Pantic M, Martens UM, Wenz C, Lingner J, Kraut N, Rettig WJ, Schnapp A (2001) A highly selective telomerase inhibitor limiting human cancer cell proliferation. EMBO J. 20, 6958–6968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Datta A, Bellon M, Sinha‐Datta U, Bazarbachi A, Lepelletier Y, Canioni D, Waldmann TA, Hermine O, Nicot C (2006) Persistent inhibition of telomerase reprograms adult T‐cell leukemia to p53‐dependent senescence. Blood 108, 1021–1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, Laberge RM, Vijg J, Van Steeg H, Dolle ME, Hoeijmakers JH, de Bruin A, Hara E, Campisi J (2014) An essential role for senescent cells in optimal wound healing through secretion of PDGF‐AA. Dev. Cell 31, 722–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Demir M, Laywell ED (2015) Neurotoxic effects of AZT on developing and adult neurogenesis. Front. Neurosci. 9, 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. van Deursen JM (2014) The role of senescent cells in ageing. Nature 509, 439–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Di Bernardo G, Squillaro T, Dell'Aversana C, Miceli M, Cipollaro M, Cascino A, Altucci L, Galderisi U (2009) Histone deacetylase inhibitors promote apoptosis and senescence in human mesenchymal stem cells. Stem Cells Dev. 18, 573–581. [DOI] [PubMed] [Google Scholar]
  38. Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, Luise C, Schurra C, Garre M, Nuciforo PG, Bensimon A, Maestro R, Pelicci PG, d'Adda di Fagagna F (2006) Oncogene‐induced senescence is a DNA damage response triggered by DNA hyper‐replication. Nature 444, 638–642. [DOI] [PubMed] [Google Scholar]
  39. Di Mitri D, Alimonti A (2016) Non‐cell‐autonomous regulation of cellular senescence in cancer. Trends Cell Biol. 26, 215–226. [DOI] [PubMed] [Google Scholar]
  40. Duan J, Duan J, Zhang Z, Tong T (2005) Irreversible cellular senescence induced by prolonged exposure to H2O2 involves DNA‐damage‐and‐repair genes and telomere shortening. Int. J. Biochem. Cell Biol. 37, 1407–1420. [DOI] [PubMed] [Google Scholar]
  41. Dumont P, Burton M, Chen QM, Gonos ES, Frippiat C, Mazarati JB, Eliaers F, Remacle J, Toussaint O (2000) Induction of replicative senescence biomarkers by sublethal oxidative stresses in normal human fibroblast. Free Radic. Biol. Med. 28, 361–373. [DOI] [PubMed] [Google Scholar]
  42. Efeyan A, Ortega‐Molina A, Velasco‐Miguel S, Herranz D, Vassilev LT, Serrano M (2007) Induction of p53‐dependent senescence by the MDM2 antagonist nutlin‐3a in mouse cells of fibroblast origin. Cancer Res. 67, 7350–7357. [DOI] [PubMed] [Google Scholar]
  43. Elmore LW, Rehder CW, Di X, McChesney PA, Jackson‐Cook CK, Gewirtz DA, Holt SE (2002) Adriamycin‐induced senescence in breast tumor cells involves functional p53 and telomere dysfunction. J. Biol. Chem. 277, 35509–35515. [DOI] [PubMed] [Google Scholar]
  44. Eriko M, Nakabayashi K, Suzuki T, Kaul SC, Ogino H, Fujii M, Mitsui Y, Ayusawa D (1999) 5‐bromodeoxyuridine induces senescence‐like phenomena in mammalian cells regardless of cell type or species. J. Biochem. 126, 1052–1059. [DOI] [PubMed] [Google Scholar]
  45. Ewald JA, Peters N, Desotelle JA, Hoffmann FM, Jarrard DF (2009) A high‐throughput method to identify novel senescence‐inducing compounds. J. Biomol. Screen. 14, 853–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fang K, Chiu CC, Li CH, Chang YT, Hwang HT (2007) Cisplatin‐induced senescence and growth inhibition in human non‐small cell lung cancer cells with ectopic transfer of p16INK4a. Oncol. Res. 16, 479–488. [DOI] [PubMed] [Google Scholar]
  47. Felipe KB, Benites J, Glorieux C, Sid B, Valenzuela M, Kviecinski MR, Pedrosa RC, Valderrama JA, Leveque P, Gallez B, Verrax J, Buc Calderon P (2013) Antiproliferative effects of phenylaminonaphthoquinones are increased by ascorbate and associated with the appearance of a senescent phenotype in human bladder cancer cells. Biochem. Biophys. Res. Commun. 433, 573–578. [DOI] [PubMed] [Google Scholar]
  48. Frippiat C, Dewelle J, Remacle J, Toussaint O (2002) Signal transduction in H2O2‐induced senescence‐like phenotype in human diploid fibroblasts. Free Radic. Biol. Med. 33, 1334–1346. [DOI] [PubMed] [Google Scholar]
  49. Gorbunova V, Seluanov A, Pereira‐Smith OM (2002) Expression of human telomerase (hTERT) does not prevent stress‐induced senescence in normal human fibroblasts but protects the cells from stress‐induced apoptosis and necrosis. J. Biol. Chem. 277, 38540–38549. [DOI] [PubMed] [Google Scholar]
  50. Grabowska W, Kucharewicz K, Wnuk M, Lewinska A, Suszek M, Przybylska D, Mosieniak G, Sikora E, Bielak‐Zmijewska A (2015) Curcumin induces senescence of primary human cells building the vasculature in a DNA damage and ATM‐independent manner. Age 37, 9744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Han Z, Wei W, Dunaway S, Darnowski JW, Calabresi P, Sedivy J, Hendrickson EA, Balan KV, Pantazis P, Wyche JH (2002) Role of p21 in apoptosis and senescence of human colon cancer cells treated with camptothecin. J. Biol. Chem. 277, 17154–17160. [DOI] [PubMed] [Google Scholar]
  52. Harley CB, Futcher AB, Greider CW (1990) Telomeres shorten during ageing of human fibroblasts. Nature 345, 458–460. [DOI] [PubMed] [Google Scholar]
  53. Hayflick L (1965) The limited in vitro lifetime of human diploid cell strains. Exp. Cell Res. 37, 614–636. [DOI] [PubMed] [Google Scholar]
  54. Hayflick L, Moorhead PS (1961) The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621. [DOI] [PubMed] [Google Scholar]
  55. Hendrayani SF, Al‐Khalaf HH, Aboussekhra A (2013) Curcumin triggers p16‐dependent senescence in active breast cancer‐associated fibroblasts and suppresses their paracrine procarcinogenic effects. Neoplasia 15, 631–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hirose Y, Berger MS, Pieper RO (2001) p53 effects both the duration of G2/M arrest and the fate of temozolomide‐treated human glioblastoma cells. Cancer Res. 61, 1957–1963. [PubMed] [Google Scholar]
  57. Hong SH, Hong B, Kim DC, Rho MS, Park JI, Rha SH, Jun HS, Jeong JS (2004) Involvement of mitogen‐activated protein kinases and p21Waf1 in hydroxyurea‐induced G1 arrest and senescence of McA‐RH7777 rat hepatoma cell line. Exp. Mol. Med. 36, 493–498. [DOI] [PubMed] [Google Scholar]
  58. Hu W, Sung T, Jessen BA, Thibault S, Finkelstein MB, Khan NK, Sacaan AI (2015) Mechanistic investigation of bone marrow suppression associated with palbociclib and its differentiation from cytotoxic chemotherapies. Clin. Cancer Res. 22, 2000–2008. [DOI] [PubMed] [Google Scholar]
  59. Huang M, Whang P, Lewicki P, Mitchell BS (2011) Cyclopentenyl cytosine induces senescence in breast cancer cells through the nucleolar stress response and activation of p53. Mol. Pharmacol. 80, 40–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Huang FC, Chang CC, Wang JM, Chang TC, Lin JJ (2012) Induction of senescence in cancer cells by the G‐quadruplex stabilizer, BMVC4, is independent of its telomerase inhibitory activity. Br. J. Pharmacol. 167, 393–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Ido Y, Duranton A, Lan F, Cacicedo JM, Chen TC, Breton L, Ruderman NB (2012) Acute activation of AMP‐activated protein kinase prevents H2O2‐induced premature senescence in primary human keratinocytes. PLoS ONE 7, e35092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Jackson JG, Pant V, Li Q, Chang LL, Quintas‐Cardama A, Garza D, Tavana O, Yang P, Manshouri T, Li Y, El‐Naggar AK, Lozano G (2012) p53‐mediated senescence impairs the apoptotic response to chemotherapy and clinical outcome in breast cancer. Cancer Cell 21, 793–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Jee HJ, Kim HJ, Kim AJ, Bae YS, Bae SS, Yun J (2009) UV light induces premature senescence in Akt1‐null mouse embryonic fibroblasts by increasing intracellular levels of ROS. Biochem. Biophys. Res. Commun. 383, 358–362. [DOI] [PubMed] [Google Scholar]
  64. Ji HJ, Rha SY, Jeung HC, Yang SH, An SW, Chung HC (2005) Cyclic induction of senescence with intermittent AZT treatment accelerates both apoptosis and telomere loss. Breast Cancer Res. Treat. 93, 227–236. [DOI] [PubMed] [Google Scholar]
  65. Joguchi A, Fujii M, Ayusawa D (2004) Increased catalase activity in mouse cell mutants resistant to paraquat. Biogerontology 5, 193–200. [DOI] [PubMed] [Google Scholar]
  66. Jones KR, Elmore LW, Jackson‐Cook C, Demasters G, Povirk LF, Holt SE, Gewirtz DA (2005) p53‐Dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int. J. Radiat. Biol. 81, 445–458. [DOI] [PubMed] [Google Scholar]
  67. Jun JI, Lau LF (2010) The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 12, 676–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kim HD, Jang CY, Choe JM, Sohn J, Kim J (2012) Phenylbutyric acid induces the cellular senescence through an Akt/p21(WAF1) signaling pathway. Biochem. Biophys. Res. Commun. 422, 213–218. [DOI] [PubMed] [Google Scholar]
  69. Kobayashi Y, Lee SS, Arai R, Miki K, Fujii M, Ayusawa D (2012) ERK1/2 mediates unbalanced growth leading to senescence induced by excess thymidine in human cells. Biochem. Biophys. Res. Commun. 425, 897–901. [DOI] [PubMed] [Google Scholar]
  70. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, Yee H, Zender L, Lowe SW (2008) Senescence of activated stellate cells limits liver fibrosis. Cell 134, 657–667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kurz DJ, Decary S, Hong Y, Trivier E, Akhmedov A, Erusalimsky JD (2004) Chronic oxidative stress compromises telomere integrity and accelerates the onset of senescence in human endothelial cells. J. Cell Sci. 117, 2417–2426. [DOI] [PubMed] [Google Scholar]
  72. Latonen L, Taya Y, Laiho M (2001) UV‐radiation induces dose‐dependent regulation of p53 response and modulates p53‐HDM2 interaction in human fibroblasts. Oncogene 20, 6784–6793. [DOI] [PubMed] [Google Scholar]
  73. Lee AC, Fenster BE, Ito H, Takeda K, Bae NS, Hirai T, Yu ZX, Ferrans VJ, Howard BH, Finkel T (1999) Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. J. Biol. Chem. 274, 7936–7940. [DOI] [PubMed] [Google Scholar]
  74. Leontieva OV, Blagosklonny MV (2013) CDK4/6‐inhibiting drug substitutes for p21 and p16 in senescence: duration of cell cycle arrest and MTOR activity determine geroconversion. Cell Cycle 12, 3063–3069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Leontieva OV, Gudkov AV, Blagosklonny MV (2010) Weak p53 permits senescence during cell cycle arrest. Cell Cycle 9, 4323–4327. [DOI] [PubMed] [Google Scholar]
  76. Leontieva OV, Demidenko ZN, Blagosklonny MV (2013) MEK drives cyclin D1 hyperelevation during geroconversion. Cell Death Differ. 20, 1241–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Li XN, Shu Q, Su JM, Perlaky L, Blaney SM, Lau CC (2005) Valproic acid induces growth arrest, apoptosis, and senescence in medulloblastomas by increasing histone hyperacetylation and regulating expression of p21Cip1, CDK4, and CMYC. Mol. Cancer Ther. 4, 1912–1922. [DOI] [PubMed] [Google Scholar]
  78. Ling X, Xu C, Fan C, Zhong K, Li F, Wang X (2014) FL118 induces p53‐dependent senescence in colorectal cancer cells by promoting degradation of MdmX. Cancer Res. 74, 7487–7497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Linge A, Weinhold K, Blasche R, Kasper M, Barth K (2007) Downregulation of caveolin‐1 affects bleomycin‐induced growth arrest and cellular senescence in A549 cells. Int. J. Biochem. Cell Biol. 39, 1964–1974. [DOI] [PubMed] [Google Scholar]
  80. Litwiniec A, Grzanka A, Helmin‐Basa A, Gackowska L, Grzanka D (2010) Features of senescence and cell death induced by doxorubicin in A549 cells: organization and level of selected cytoskeletal proteins. J. Cancer Res. Clin. Oncol. 136, 717–736. [DOI] [PubMed] [Google Scholar]
  81. Litwiniec A, Gackowska L, Helmin‐Basa A, Zuryn A, Grzanka A (2013) Low‐dose etoposide‐treatment induces endoreplication and cell death accompanied by cytoskeletal alterations in A549 cells: does the response involve senescence? The possible role of vimentin. Cancer Cell Int. 13, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Manfe V, Biskup E, Johansen P, Kamstrup MR, Krejsgaard TF, Morling N, Wulf HC, Gniadecki R (2012) MDM2 inhibitor nutlin‐3a induces apoptosis and senescence in cutaneous T‐cell lymphoma: role of p53. J. Invest. Dermatol. 132, 1487–1496. [DOI] [PubMed] [Google Scholar]
  83. Mannava S, Moparthy KC, Wheeler LJ, Natarajan V, Zucker SN, Fink EE, Im M, Flanagan S, Burhans WC, Zeitouni NC, Shewach DS, Mathews CK, Nikiforov MA (2013) Depletion of deoxyribonucleotide pools is an endogenous source of DNA damage in cells undergoing oncogene‐induced senescence. Am. J. Pathol. 182, 142–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Mansilla S, Pina B, Portugal J (2003) Daunorubicin‐induced variations in gene transcription: commitment to proliferation arrest, senescence and apoptosis. Biochem. J. 372, 703–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Marconett CN, Sundar SN, Tseng M, Tin AS, Tran KQ, Mahuron KM, Bjeldanes LF, Firestone GL (2011) Indole‐3‐carbinol downregulation of telomerase gene expression requires the inhibition of estrogen receptor‐alpha and Sp1 transcription factor interactions within the hTERT promoter and mediates the G1 cell cycle arrest of human breast cancer cells. Carcinogenesis 32, 1315–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Marusyk A, Wheeler LJ, Mathews CK, DeGregori J (2007) p53 mediates senescence‐like arrest induced by chronic replicational stress. Mol. Cell. Biol. 27, 5336–5351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Mason SA, Cozzi SJ, Pierce CJ, Pavey SJ, Parsons PG, Boyle GM (2010) The induction of senescence‐like growth arrest by protein kinase C‐activating diterpene esters in solid tumor cells. Invest. New Drugs 28, 575–586. [DOI] [PubMed] [Google Scholar]
  88. Masterson JC, O'Dea S (2007) 5‐Bromo‐2‐deoxyuridine activates DNA damage signalling responses and induces a senescence‐like phenotype in p16‐null lung cancer cells. Anticancer Drugs 18, 1053–1068. [DOI] [PubMed] [Google Scholar]
  89. Maya‐Mendoza A, Merchut‐Maya JM, Bartkova J, Bartek J, Streuli CH, Jackson DA (2014) Immortalised breast epithelia survive prolonged DNA replication stress and return to cycle from a senescent‐like state. Cell Death Dis. 5, e1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. McKenna E, Traganos F, Zhao H, Darzynkiewicz Z (2012) Persistent DNA damage caused by low levels of mitomycin C induces irreversible cell senescence. Cell Cycle 11, 3132–3140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Meng A, Wang Y, Van Zant G, Zhou D (2003) Ionizing radiation and busulfan induce premature senescence in murine bone marrow hematopoietic cells. Cancer Res. 63, 5414–5419. [PubMed] [Google Scholar]
  92. Mhaidat NM, Zhang XD, Allen J, Avery‐Kiejda KA, Scott RJ, Hersey P (2007) Temozolomide induces senescence but not apoptosis in human melanoma cells. Br. J. Cancer 97, 1225–1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Minieri V, Saviozzi S, Gambarotta G, Lo Iacono M, Accomasso L, Cibrario Rocchietti E, Gallina C, Turinetto V, Giachino C (2015) Persistent DNA damage‐induced premature senescence alters the functional features of human bone marrow mesenchymal stem cells. J. Cell Mol. Med. 19, 734–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Modrak DE, Leon E, Goldenberg DM, Gold DV (2009) Ceramide regulates gemcitabine‐induced senescence and apoptosis in human pancreatic cancer cell lines. Mol. Cancer Res. 7, 890–896. [DOI] [PubMed] [Google Scholar]
  95. Mosieniak G, Adamowicz M, Alster O, Jaskowiak H, Szczepankiewicz AA, Wilczynski GM, Ciechomska IA, Sikora E (2012) Curcumin induces permanent growth arrest of human colon cancer cells: link between senescence and autophagy. Mech. Ageing Dev. 133, 444–455. [DOI] [PubMed] [Google Scholar]
  96. Muller S, Sanders DA, Di Antonio M, Matsis S, Riou JF, Rodriguez R, Balasubramanian S (2012) Pyridostatin analogues promote telomere dysfunction and long‐term growth inhibition in human cancer cells. Org. Biomol. Chem. 10, 6537–6546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Munoz‐Espin D, Serrano M (2014) Cellular senescence: from physiology to pathology. Nat. Rev. Mol. Cell Biol. 15, 482–496. [DOI] [PubMed] [Google Scholar]
  98. Munoz‐Espin D, Canamero M, Maraver A, Gomez‐Lopez G, Contreras J, Murillo‐Cuesta S, Rodriguez‐Baeza A, Varela‐Nieto I, Ruberte J, Collado M, Serrano M (2013) Programmed cell senescence during mammalian embryonic development. Cell 155, 1104–1118. [DOI] [PubMed] [Google Scholar]
  99. Nacher V, Carretero A, Navarro M, Armengol C, Llombart C, Rodriguez A, Herrero‐Fresneda I, Ayuso E, Ruberte J (2006) The quail mesonephros: a new model for renal senescence? J. Vasc. Res. 43, 581–586. [DOI] [PubMed] [Google Scholar]
  100. Nair RR, Bagheri M, Saini DK (2015) Temporally distinct roles of ATM and ROS in genotoxic‐stress‐dependent induction and maintenance of cellular senescence. J. Cell Sci. 128, 342–353. [DOI] [PubMed] [Google Scholar]
  101. Neelsen KJ, Zanini IM, Herrador R, Lopes M (2013) Oncogenes induce genotoxic stress by mitotic processing of unusual replication intermediates. J. Cell Biol. 200, 699–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Ogryzko VV, Hirai TH, Russanova VR, Barbie DA, Howard BH (1996) Human fibroblast commitment to a senescence‐like state in response to histone deacetylase inhibitors is cell cycle dependent. Mol. Cell. Biol. 16, 5210–5218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Oliva JL, Caino MC, Senderowicz AM, Kazanietz MG (2008) S‐Phase‐specific activation of PKC alpha induces senescence in non‐small cell lung cancer cells. J. Biol. Chem. 283, 5466–5476. [DOI] [PubMed] [Google Scholar]
  104. Ota H, Eto M, Kano MR, Ogawa S, Iijima K, Akishita M, Ouchi Y (2008) Cilostazol inhibits oxidative stress‐induced premature senescence via upregulation of Sirt1 in human endothelial cells. Arterioscler. Thromb. Vasc. Biol. 28, 1634–1639. [DOI] [PubMed] [Google Scholar]
  105. Palaniyappan A (2009) Cyclophosphamide induces premature senescence in normal human fibroblasts by activating MAP kinases. Biogerontology 10, 677–682. [DOI] [PubMed] [Google Scholar]
  106. Park JI, Jeong JS, Han JY, Kim DI, Gao YH, Park SC, Rodgers GP, Kim IH (2000) Hydroxyurea induces a senescence‐like change of K562 human erythroleukemia cell. J. Cancer Res. Clin. Oncol. 126, 455–460. [PubMed] [Google Scholar]
  107. Park C, Lee I, Kang WK (2006) E2F‐1 is a critical modulator of cellular senescence in human cancer. Int. J. Mol. Med. 17, 715–720. [PubMed] [Google Scholar]
  108. Park JY, Park SH, Weiss RH (2009) Disparate effects of roscovitine on renal tubular epithelial cell apoptosis and senescence: implications for autosomal dominant polycystic kidney disease. Am. J. Nephrol. 29, 509–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Pascal T, Debacq‐Chainiaux F, Boilan E, Ninane N, Raes M, Toussaint O (2007) Heme oxygenase‐1 and interleukin‐11 are overexpressed in stress‐induced premature senescence of human WI‐38 fibroblasts induced by tert‐butylhydroperoxide and ethanol. Biogerontology 8, 409–422. [DOI] [PubMed] [Google Scholar]
  110. Pazolli E, Alspach E, Milczarek A, Prior J, Piwnica‐Worms D, Stewart SA (2012) Chromatin remodeling underlies the senescence‐associated secretory phenotype of tumor stromal fibroblasts that supports cancer progression. Cancer Res. 72, 2251–2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Perez M, Munoz‐Galvan S, Jimenez‐Garcia MP, Marin JJ, Carnero A (2015) Efficacy of CDK4 inhibition against sarcomas depends on their levels of CDK4 and p16ink4 mRNA. Oncotarget 6, 40557–40574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Piegari E, De Angelis A, Cappetta D, Russo R, Esposito G, Costantino S, Graiani G, Frati C, Prezioso L, Berrino L, Urbanek K, Quaini F, Rossi F (2013) Doxorubicin induces senescence and impairs function of human cardiac progenitor cells. Basic Res. Cardiol. 108, 334. [DOI] [PubMed] [Google Scholar]
  113. Place RF, Noonan EJ, Giardina C (2005) HDACs and the senescent phenotype of WI‐38 cells. BMC Cell Biol. 6, 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP (2002) DNA damage is able to induce senescence in tumor cells in vitro and in vivo . Cancer Res. 62, 1876–1883. [PubMed] [Google Scholar]
  115. Pospelova TV, Demidenko ZN, Bukreeva EI, Pospelov VA, Gudkov AV, Blagosklonny MV (2009) Pseudo‐DNA damage response in senescent cells. Cell Cycle 8, 4112–4118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Prieur A, Besnard E, Babled A, Lemaitre JM (2011) p53 and p16(INK4A) independent induction of senescence by chromatin‐dependent alteration of S‐phase progression. Nat. Commun. 2, 473. [DOI] [PubMed] [Google Scholar]
  117. Probin V, Wang Y, Bai A, Zhou D (2006) Busulfan selectively induces cellular senescence but not apoptosis in WI38 fibroblasts via a p53‐independent but extracellular signal‐regulated kinase‐p38 mitogen‐activated protein kinase‐dependent mechanism. J. Pharmacol. Exp. Ther. 319, 551–560. [DOI] [PubMed] [Google Scholar]
  118. Probin V, Wang Y, Zhou D (2007) Busulfan‐induced senescence is dependent on ROS production upstream of the MAPK pathway. Free Radic. Biol. Med. 42, 1858–1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Purvis JE, Karhohs KW, Mock C, Batchelor E, Loewer A, Lahav G (2012) p53 dynamics control cell fate. Science 336, 1440–1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Qu K, Lin T, Wang Z, Liu S, Chang H, Xu X, Meng F, Zhou L, Wei J, Tai M, Dong Y, Liu C (2014) Reactive oxygen species generation is essential for cisplatin‐induced accelerated senescence in hepatocellular carcinoma. Front. Med. 8, 227–235. [DOI] [PubMed] [Google Scholar]
  121. Rader J, Russell MR, Hart LS, Nakazawa MS, Belcastro LT, Martinez D, Li Y, Carpenter EL, Attiyeh EF, Diskin SJ, Kim S, Parasuraman S, Caponigro G, Schnepp RW, Wood AC, Pawel B, Cole KA, Maris JM (2013) Dual CDK4/CDK6 inhibition induces cell‐cycle arrest and senescence in neuroblastoma. Clin. Cancer Res. 19, 6173–6182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Riou JF, Guittat L, Mailliet P, Laoui A, Renou E, Petitgenet O, Megnin‐Chanet F, Helene C, Mergny JL (2002) Cell senescence and telomere shortening induced by a new series of specific G‐quadruplex DNA ligands. Proc. Natl Acad. Sci. USA 99, 2672–2677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY (2005) Escape from therapy‐induced accelerated cellular senescence in p53‐null lung cancer cells and in human lung cancers. Cancer Res. 65, 2795–2803. [DOI] [PubMed] [Google Scholar]
  124. Robles SJ, Adami GR (1998) Agents that cause DNA double strand breaks lead to p16INK4a enrichment and the premature senescence of normal fibroblasts. Oncogene 16, 1113–1123. [DOI] [PubMed] [Google Scholar]
  125. Romanov SR, Kozakiewicz BK, Holst CR, Stampfer MR, Haupt LM, Tlsty TD (2001) Normal human mammary epithelial cells spontaneously escape senescence and acquire genomic changes. Nature 409, 633–637. [DOI] [PubMed] [Google Scholar]
  126. Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, Hagler M, Jurk D, Smith LA, Casaclang‐Verzosa G, Zhu Y, Schafer MJ, Tchkonia T, Kirkland JL, Miller JD (2016) Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell Accepted Author Manuscript doi: 10.1111/acel.12458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Salama R, Sadaie M, Hoare M, Narita M (2014) Cellular senescence and its effector programs. Genes Dev. 28, 99–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, Lowe SW (2002) A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell 109, 335–346. [DOI] [PubMed] [Google Scholar]
  129. Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602. [DOI] [PubMed] [Google Scholar]
  130. Shimizu H, Bolati D, Adijiang A, Enomoto A, Nishijima F, Dateki M, Niwa T (2010) Senescence and dysfunction of proximal tubular cells are associated with activated p53 expression by indoxyl sulfate. Am. J. Physiol. Cell Physiol. 299, C1110–C1117. [DOI] [PubMed] [Google Scholar]
  131. Shimizu H, Bolati D, Adijiang A, Muteliefu G, Enomoto A, Nishijima F, Dateki M, Niwa T (2011) NF‐kappaB plays an important role in indoxyl sulfate‐induced cellular senescence, fibrotic gene expression, and inhibition of proliferation in proximal tubular cells. Am. J. Physiol. Cell Physiol. 301, C1201–C1212. [DOI] [PubMed] [Google Scholar]
  132. Sliwinska MA, Mosieniak G, Wolanin K, Babik A, Piwocka K, Magalska A, Szczepanowska J, Fronk J, Sikora E (2009) Induction of senescence with doxorubicin leads to increased genomic instability of HCT116 cells. Mech. Ageing Dev. 130, 24–32. [DOI] [PubMed] [Google Scholar]
  133. Spallarossa P, Altieri P, Aloi C, Garibaldi S, Barisione C, Ghigliotti G, Fugazza G, Barsotti A, Brunelli C (2009) Doxorubicin induces senescence or apoptosis in rat neonatal cardiomyocytes by regulating the expression levels of the telomere binding factors 1 and 2. Am. J. Physiol. Heart Circ. Physiol. 297, H2169–H2181. [DOI] [PubMed] [Google Scholar]
  134. Storer M, Mas A, Robert‐Moreno A, Pecoraro M, Ortells MC, Di Giacomo V, Yosef R, Pilpel N, Krizhanovsky V, Sharpe J, Keyes WM (2013) Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155, 1119–1130. [DOI] [PubMed] [Google Scholar]
  135. Sumikawa E, Matsumoto Y, Sakemura R, Fujii M, Ayusawa D (2005) Prolonged unbalanced growth induces cellular senescence markers linked with mechano transduction in normal and tumor cells. Biochem. Biophys. Res. Commun. 335, 558–565. [DOI] [PubMed] [Google Scholar]
  136. Suzuki T, Minagawa S, Michishita E, Ogino H, Fujii M, Mitsui Y, Ayusawa D (2001) Induction of senescence‐associated genes by 5‐bromodeoxyuridine in HeLa cells. Exp. Gerontol. 36, 465–474. [DOI] [PubMed] [Google Scholar]
  137. Suzuki M, Suzuki K, Kodama S, Watanabe M (2006) Interstitial chromatin alteration causes persistent p53 activation involved in the radiation‐induced senescence‐like growth arrest. Biochem. Biophys. Res. Commun. 340, 145–150. [DOI] [PubMed] [Google Scholar]
  138. Suzuki E, Takahashi M, Oba S, Nishimatsu H (2013) Oncogene‐ and oxidative stress‐induced cellular senescence shows distinct expression patterns of proinflammatory cytokines in vascular endothelial cells. ScientificWorldJournal 2013, 754735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Taka T, Huang L, Wongnoppavich A, Tam‐Chang SW, Lee TR, Tuntiwechapikul W (2013) Telomere shortening and cell senescence induced by perylene derivatives in A549 human lung cancer cells. Bioorg. Med. Chem. 21, 883–890. [DOI] [PubMed] [Google Scholar]
  140. Terao Y, Nishida J, Horiuchi S, Rong F, Ueoka Y, Matsuda T, Kato H, Furugen Y, Yoshida K, Kato K, Wake N (2001) Sodium butyrate induces growth arrest and senescence‐like phenotypes in gynecologic cancer cells. Int. J. Cancer 94, 257–267. [DOI] [PubMed] [Google Scholar]
  141. Toussaint O, Medrano EE, von Zglinicki T (2000) Cellular and molecular mechanisms of stress‐induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes. Exp. Gerontol. 35, 927–945. [DOI] [PubMed] [Google Scholar]
  142. Toussaint O, Royer V, Salmon M, Remacle J (2002) Stress‐induced premature senescence and tissue ageing. Biochem. Pharmacol. 64, 1007–1009. [DOI] [PubMed] [Google Scholar]
  143. Veena MS, Wilken R, Zheng JY, Gholkar A, Venkatesan N, Vira D, Ahmed S, Basak SK, Dalgard CL, Ravichandran S, Batra RK, Kasahara N, Elashoff D, Fishbein MC, Whitelegge JP, Torres JZ, Wang MB, Srivatsan ES (2014) p16 Protein and gigaxonin are associated with the ubiquitination of NFkappaB in cisplatin‐induced senescence of cancer cells. J. Biol. Chem. 289, 34921–34937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Velichko AK, Petrova NV, Razin SV, Kantidze OL (2015) Mechanism of heat stress‐induced cellular senescence elucidates the exclusive vulnerability of early S‐phase cells to mild genotoxic stress. Nucleic Acids Res. 43, 6309–6320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Venturelli S, Berger A, Weiland T, Essmann F, Waibel M, Nuebling T, Hacker S, Schenk M, Schulze‐Osthoff K, Salih HR, Fulda S, Sipos B, Johnstone RW, Lauer UM, Bitzer M (2013) Differential induction of apoptosis and senescence by the DNA methyltransferase inhibitors 5‐azacytidine and 5‐aza‐2′‐deoxycytidine in solid tumor cells. Mol. Cancer Ther. 12, 2226–2236. [DOI] [PubMed] [Google Scholar]
  146. Vogt M, Haggblom C, Yeargin J, Christiansen‐Weber T, Haas M (1998) Independent induction of senescence by p16INK4a and p21CIP1 in spontaneously immortalized human fibroblasts. Cell Growth Differ. 9, 139–146. [PubMed] [Google Scholar]
  147. Wang X, Wong SC, Pan J, Tsao SW, Fung KH, Kwong DL, Sham JS, Nicholls JM (1998) Evidence of cisplatin‐induced senescent‐like growth arrest in nasopharyngeal carcinoma cells. Cancer Res. 58, 5019–5022. [PubMed] [Google Scholar]
  148. Widodo N, Deocaris CC, Kaur K, Hasan K, Yaguchi T, Yamasaki K, Sugihara T, Ishii T, Wadhwa R, Kaul SC (2007) Stress chaperones, mortalin, and pex19p mediate 5‐aza‐2′ deoxycytidine‐induced senescence of cancer cells by DNA methylation‐independent pathway. J. Gerontol. A Biol. Sci. Med. Sci. 62, 246–255. [DOI] [PubMed] [Google Scholar]
  149. Xiao H, Hasegawa T, Miyaishi O, Ohkusu K, Isobe K (1997) Sodium butyrate induces NIH3T3 cells to senescence‐like state and enhances promoter activity of p21WAF/CIP1 in p53‐independent manner. Biochem. Biophys. Res. Commun. 237, 457–460. [DOI] [PubMed] [Google Scholar]
  150. Xu WS, Perez G, Ngo L, Gui CY, Marks PA (2005) Induction of polyploidy by histone deacetylase inhibitor: a pathway for antitumor effects. Cancer Res. 65, 7832–7839. [DOI] [PubMed] [Google Scholar]
  151. Xu M, Palmer AK, Ding H, Weivoda MM, Pirtskhalava T, White TA, Sepe A, Johnson KO, Stout MB, Giorgadze N, Jensen MD, LeBrasseur NK, Tchkonia T, Kirkland JL (2015a) Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife 4, e12997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, White TA, Johnson KO, Stout MB, Mezera V, Giorgadze N, Jensen MD, LeBrasseur NK, Kirkland JL (2015b) JAK inhibition alleviates the cellular senescence‐associated secretory phenotype and frailty in old age. Proc. Natl Acad. Sci. USA 112, E6301–E6310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Yang KE, Jang HJ, Hwang IH, Chung YH, Choi JS, Lee TH, Chung YJ, Lee MS, Lee MY, Yeo EJ, Jang IS (2016) Phenyl 2‐pyridyl ketoxime induces cellular senescence‐like alterations via nitric oxide production in human diploid fibroblasts. Aging Cell 15, 245–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Yeo EJ, Hwang YC, Kang CM, Kim IH, Kim DI, Parka JS, Choy HE, Park WY, Park SC (2000) Senescence‐like changes induced by hydroxyurea in human diploid fibroblasts. Exp. Gerontol. 35, 553–571. [DOI] [PubMed] [Google Scholar]
  155. Yoshida A, Lee EK, Diehl JA (2016) Induction of therapeutic senescence in vemurafenib‐resistant melanoma by extended Inhibition of CDK4/6. Cancer Res. 76, 2990–3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Yoshizaki K, Fujiki T, Tsunematsu T, Yamashita M, Udono M, Shirahata S, Katakura Y (2009) Pro‐senescent effect of hydrogen peroxide on cancer cells and its possible application to tumor suppression. Biosci. Biotechnol. Biochem. 73, 311–315. [DOI] [PubMed] [Google Scholar]
  157. Young JJ, Patel A, Rai P (2010) Suppression of thioredoxin‐1 induces premature senescence in normal human fibroblasts. Biochem. Biophys. Res. Commun. 392, 363–368. [DOI] [PubMed] [Google Scholar]
  158. Yuan Y, Wang Q, Paulk J, Kubicek S, Kemp MM, Adams DJ, Shamji AF, Wagner BK, Schreiber SL (2012) A small‐molecule probe of the histone methyltransferase G9a induces cellular senescence in pancreatic adenocarcinoma. ACS Chem. Biol. 7, 1152–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Zdanov S, Debacq‐Chainiaux F, Remacle J, Toussaint O (2006) Identification of p38MAPK‐dependent genes with changed transcript abundance in H2O2‐induced premature senescence of IMR‐90 hTERT human fibroblasts. FEBS Lett. 580, 6455–6463. [DOI] [PubMed] [Google Scholar]
  160. von Zglinicki T, Pilger R, Sitte N (2000) Accumulation of single‐strand breaks is the major cause of telomere shortening in human fibroblasts. Free Radic. Biol. Med. 28, 64–74. [DOI] [PubMed] [Google Scholar]
  161. Zhang JW, Zhang SS, Song JR, Sun K, Zong C, Zhao QD, Liu WT, Li R, Wu MC, Wei LX (2014) Autophagy inhibition switches low‐dose camptothecin‐induced premature senescence to apoptosis in human colorectal cancer cells. Biochem. Pharmacol. 90, 265–275. [DOI] [PubMed] [Google Scholar]
  162. Zhao L, Wink M (2013) The beta‐carboline alkaloid harmine inhibits telomerase activity of MCF‐7 cells by down‐regulating hTERT mRNA expression accompanied by an accelerated senescent phenotype. PeerJ 1, e174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Zhao W, Lin ZX, Zhang ZQ (2004) Cisplatin‐induced premature senescence with concomitant reduction of gap junctions in human fibroblasts. Cell Res. 14, 60–66. [DOI] [PubMed] [Google Scholar]
  164. Zhao H, Halicka HD, Traganos F, Jorgensen E, Darzynkiewicz Z (2010) New biomarkers probing depth of cell senescence assessed by laser scanning cytometry. Cytometry A 77, 999–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Zhou JM, Zhu XF, Lu YJ, Deng R, Huang ZS, Mei YP, Wang Y, Huang WL, Liu ZC, Gu LQ, Zeng YX (2006) Senescence and telomere shortening induced by novel potent G‐quadruplex interactive agents, quindoline derivatives, in human cancer cell lines. Oncogene 25, 503–511. [DOI] [PubMed] [Google Scholar]
  166. Zhou H, Huang B, Han Y, Jin R, Chen S (2013) Probucol inhibits JAK2‐STAT pathway activation and protects human glomerular mesangial cells from tert‐butyl hydroperoxide induced premature senescence. Can. J. Physiol. Pharmacol. 91, 671–679. [DOI] [PubMed] [Google Scholar]
  167. Zhu F, Li Y, Zhang J, Piao C, Liu T, Li HH, Du J (2013) Senescent cardiac fibroblast is critical for cardiac fibrosis after myocardial infarction. PLoS ONE 8, e74535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Zhu Y, Tchkonia T, Fuhrmann‐Stroissnigg H, Dai HM, Ling YY, Stout MB, Pirtskhalava T, Giorgadze N, Johnson KO, Giles CB, Wren JD, Niedernhofer LJ, Robbins PD, Kirkland JL (2015a) Identification of a novel senolytic agent, navitoclax, targeting the Bcl‐2 family of anti‐apoptotic factors. Aging Cell 15, 428–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O'Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann‐Stroissnigg H, Gurkar AU, Zhao J, Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD, Niedernhofer LJ, Kirkland JL (2015b) The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14, 644–658. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Aging Cell are provided here courtesy of Wiley

RESOURCES