ArticlePDF Available

Reduction of total E2F/DP activity induces senescence-like cell cycle arrest in cancer cells lacking functional pRB and p53

Rockefeller University Press
Journal of Cell Biology (JCB)
Authors:

Abstract and Figures

E2F/DP complexes were originally identified as potent transcriptional activators required for cell proliferation. However, recent studies revised this notion by showing that inactivation of total E2F/DP activity by dominant-negative forms of E2F or DP does not prevent cellular proliferation, but rather abolishes tumor suppression pathways, such as cellular senescence. These observations suggest that blockage of total E2F/DP activity may increase the risk of cancer. Here, we provide evidence that depletion of DP by RNA interference, but not overexpression of dominant-negative form of E2F, efficiently reduces endogenous E2F/DP activity in human primary cells. Reduction of total E2F/DP activity results in a dramatic decrease in expression of many E2F target genes and causes a senescence-like cell cycle arrest. Importantly, similar results were observed in human cancer cells lacking functional p53 and pRB family proteins. These findings reveal that E2F/DP activity is indeed essential for cell proliferation and its reduction immediately provokes a senescence-like cell cycle arrest.
Effects of dn-E2F on cell growth. (A) The E2F1 and DP1 expression vectors (0.6 μg) were cotransfected into 60-mm plates of SVts8 cells along with a reporter plasmid and lacZ plasmid. Where indicated, cells were also cotransfected with an increasing amount of expression plasmid encoding dn-E2F (lane 2: 0 μg; lane 3: 0.1 μg; lane 4: 0.3 μg; lane 5; 0.6 μg). Error bars indicate SD. (B) Early passage (45PDLs) TIG-3 cells infected with retrovirus encoding dn-E2F or empty vector. Cells were analyzed for expression of E2F1 and dn-E2F protein by Western blot after selection with hygromycin. The antibody against β-actin was used as a loading control. (C) EMSAs were performed using a radiolabeled probe containing the E2F consensus sequence of the adenovirus E2 gene promoter and extracts from cells infected with retrovirus encoding empty vector (lanes 1–5) or dn-E2F (lanes 6–10) in the absence or presence of antibodies as indicated. The DNA-binding activity, which is not shifted by any available E2F antibodies, was marked by an asterisk. (D and E) Cell extracts were prepared from cells at 60PDLs and subjected to RT-PCR (D) or Western blotting (E) with primers or antibodies shown at left. E2F target genes reported previously were highlighted with an asterisk. CDK4 was used here as a loading control. (F) Early passage (45PDLs) TIG-3 cells infected with retrovirus encoding dn-E2F or empty vector were selected for expression of the hygromycin selectable marker for 5 d and used in proliferation curves performed in triplicate. Error bars indicate SD.
… 
Content may be subject to copyright.
THE JOURNAL OF CELL BIOLOGY
JCB: REPORT
©
The Rockefeller University Press $8.00
The Journal of Cell Biology, Vol. 168, No. 4, February 14, 2005 553–560
http://www.jcb.org/cgi/doi/10.1083/jcb.200411093
JCB 553
Reduction of total E2F/DP activity induces
senescence-like cell cycle arrest in cancer cells
lacking functional pRB and p53
Kayoko Maehara,
1
Kimi Yamakoshi,
2
Naoko Ohtani,
2
Yoshiaki Kubo,
3
Akiko Takahashi,
2
Seiji Arase,
3
Nic Jones,
1
and Eiji Hara
1,2
1
Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, England, UK
2
Division of Protein Information, Institute for Genome Research, and
3
Department of Dermatology, School of Medicine, University of Tokushima, Tokushima 770-8503, Japan
2F/DP complexes were originally identified as po-
tent transcriptional activators required for cell prolif-
eration. However, recent studies revised this notion
by showing that inactivation of total E2F/DP activity by
dominant-negative forms of E2F or DP does not prevent
cellular proliferation, but rather abolishes tumor suppres-
sion pathways, such as cellular senescence. These obser-
vations suggest that blockage of total E2F/DP activity
may increase the risk of cancer. Here, we provide evi-
dence that depletion of DP by RNA interference, but not
E
overexpression of dominant-negative form of E2F, effi-
ciently reduces endogenous E2F/DP activity in human
primary cells. Reduction of total E2F/DP activity results in
a dramatic decrease in expression of many E2F target
genes and causes a senescence-like cell cycle arrest. Im-
portantly, similar results were observed in human cancer
cells lacking functional p53 and pRB family proteins.
These findings reveal that E2F/DP activity is indeed es-
sential for cell proliferation and its reduction immediately
provokes a senescence-like cell cycle arrest.
Introduction
As the major downstream mediator of the retinoblastoma protein
(pRB) tumor suppressor pathway, the E2F/DP transcription
factor complexes play a crucial role in cell cycle regulation
(Dyson, 1998). Recent studies suggest that E2F/DP complexes
can be broadly classified into two subgroups: a group of “acti-
vating” E2Fs (E2F1, E2F2, and E2F3) that are potent transcrip-
tional activators, and a second group of “repressive” E2Fs
(E2F4, E2F5, and E2F6) that appear to function primarily as
transcriptional repressors (Mann and Jones, 1996; Takahashi et
al., 2000; Trimarchi and Lees, 2002). This interpretation is
supported by chromatin immunoprecipitation (ChIP) analysis,
which reveals that activating E2Fs replace repressive E2Fs at
E2F-regulated promoters as cells progress from G0/G1 toward
S phase, a change that correlates with the induction of E2F-
dependent gene expression (Takahashi et al., 2000).
The combined ablation of E2F1, E2F2, and E2F3 genes
in primary mouse embryonic fibroblasts results in the reduction
of E2F target gene expression and block of cell proliferation
(Wu et al., 2001). However, inactivation of both E2F4 and
E2F5 does not inhibit cell proliferation, but instead renders
cells resistant to a p16
INK4a
-induced growth arrest (Gaubatz et
al., 2000; Ohtani et al., 2003). These results suggest that acti-
vating E2Fs and repressive E2Fs play opposing roles and that
the balance between activating E2Fs and repressive E2Fs is
likely to regulate cell cycle progression. Consistent with this
idea, inactivation of total E2F/DP activity by overexpression
of dominant-negative (dn-) forms of E2F or DP did not in-
hibit cellular proliferation, but rather abolished a variety of
growth arrest pathways, such as TGF-
–induced growth arrest,
p16
INK4a
-induced cell cycle arrest, contact inhibition, and cellular
senescence (Bargou et al., 1996; Zhang et al., 1999; Rowland
et al., 2002). These results suggest that activating E2Fs are only
required to counterbalance the effects of repressive E2Fs and
that total E2F/DP activity is not essential for cellular prolifera-
tion, but rather to promote tumor suppression mechanisms.
Thus, inactivation of total E2F/DP activity may actually increase
the risk of cancer. Because current therapeutic approaches that
target E2F/DP do not inactivate specific E2F family members,
but instead block all E2F/DP activity (Bandara et al., 1997), it
is vital to clarify the role of E2F/DP activity in normal and
cancerous human cells.
Correspondence to Eiji Hara: hara@genome.tokushima-u.ac.jp
Abbreviations used in this paper: ChIP, chromatin immunoprecipitation; dn,
dominant negative; EMSA, electrophoretic mobility shift assay; HDF, human
diploid fibroblast; pRB, retinoblastoma protein; RNAi, RNA interference; SA-
-gal,
senescence-associated
-galactosidase; SAHF, senescence-associated hetero-
chromatic foci; shRNA, small hairpin RNA.
The online version of this article includes supplemental material.
JCB • VOLUME 168 • NUMBER 4 • 2005554
Figure 1. Effects of dn-E2F on cell growth. (A) The E2F1 and DP1 expression vectors (0.6 g) were cotransfected into 60-mm plates of SVts8 cells along
with a reporter plasmid and lacZ plasmid. Where indicated, cells were also cotransfected with an increasing amount of expression plasmid encoding
dn-E2F (lane 2: 0 g; lane 3: 0.1 g; lane 4: 0.3 g; lane 5; 0.6 g). Error bars indicate SD. (B) Early passage (45PDLs) TIG-3 cells infected with retro-
virus encoding dn-E2F or empty vector. Cells were analyzed for expression of E2F1 and dn-E2F protein by Western blot after selection with hygromycin.
The antibody against -actin was used as a loading control. (C) EMSAs were performed using a radiolabeled probe containing the E2F consensus sequence
of the adenovirus E2 gene promoter and extracts from cells infected with retrovirus encoding empty vector (lanes 1–5) or dn-E2F (lanes 6–10) in the
absence or presence of antibodies as indicated. The DNA-binding activity, which is not shifted by any available E2F antibodies, was marked by an asterisk.
(D and E) Cell extracts were prepared from cells at 60PDLs and subjected to RT-PCR (D) or Western blotting (E) with primers or antibodies shown at left.
E2F target genes reported previously were highlighted with an asterisk. CDK4 was used here as a loading control. (F) Early passage (45PDLs) TIG-3 cells
infected with retrovirus encoding dn-E2F or empty vector were selected for expression of the hygromycin selectable marker for 5 d and used in proliferation
curves performed in triplicate. Error bars indicate SD.
DEPLETION OF DP PROVOKES CELLULAR SENESCENCE • MAEHARA ET AL.
555
In this paper, we address the role of E2F/DP activity in
human primary and cancer cells by comparing two approaches
to inactivating total E2F/DP activity: the use of a dn mutant
and RNA interference (RNAi) (Brummelkamp et al., 2002).
Results and discussion
To delineate the role of E2F/DP on proliferation of human pri-
mary cells, we first tested the effect of overexpressing dn-E2F
(E2F-DB), which lacks both the transcriptional activation do-
main and the pRB-family protein binding domain (Zhang et al.,
1999). As reported previously (Zhang et al., 1999; Rowland et
al., 2002), increasing amounts of dn-E2F blocked the transacti-
vating activity of cotransfected E2Fs in human fibroblasts (Fig.
1 A). The dn-E2F was then retrovirally transduced into early
passage primary human diploid fibroblasts (HDFs), TIG-3
cells. Ectopic dn-E2F expression was more than 100 times
greater than that of the endogenous E2F1 (Fig. 1 B, lane 2). As
expected, the DNA-binding activity of endogenous E2F4 was
abolished and replaced by dn-E2F (Fig. 1 C, lanes 1 and 6). In
agreement with previous reports (Zhang et al., 1999; Rowland
et al., 2002), overexpression of dn-E2F did not reduce the ex-
pression of endogenous E2F target genes in TIG-3 cells (Fig. 1,
D and E). In addition, TIG-3 cells expressing dn-E2F grew
Figure 2. Depletion of DP1 causes reduction of E2F target gene expression in HDFs. (A) 20 g of total RNA prepared from TIG-3 (50PDLs), HeLa, U2OS,
HT29, and 293T cells transfected with or without DP2 expression plasmid were subjected to Northern blot analysis using the same activity of indicated
radiolabeled probes. (B and C) Early passage TIG-3 cells (45PDLs) were infected with retrovirus encoding DP1-shRNA (lane 2) or control-shRNA (lane 1).
Cell extracts were prepared from cells at 7 d after selection with puromycin, and were subjected to RT-PCR (B) or Western blotting (C) with primers or
antibodies shown at left. E2F target genes reported previously were highlighted with an asterisk. CDK4 was used here as a loading control. (D) EMSA
was performed using extracts from cells infected with retrovirus encoding control-shRNA (lanes 1 to 5) or DP1-shRNA (lanes 6 to 10) in the absence or
presence of antibodies shown at top. The DNA-binding activity, which is resistant to any available E2F antibodies, was marked by an asterisk.
JCB • VOLUME 168 • NUMBER 4 • 2005556
faster than control TIG-3 cells, especially at late passage (Fig.
1 F; unpublished data). These and earlier results (Rowland et
al., 2002) suggest that E2F/DP activity may not be essential for
cell proliferation in mammalian cells. However, these findings
could also be explained by an incomplete inhibition of endoge-
nous E2F/DP activity or by an unforeseen side effect of over-
expression of the dn-E2F. Therefore, we sought a different ap-
proach to inactivate total E2F/DP activity in HDFs.
Although the E2F family consists of six members, the DP
family contains only two (DP1 and DP2) (Trimarchi and Lees,
2002). Because the level of DP2 mRNA is very low in TIG-3
cells (Fig. 2 A) and heterodimerization with a DP protein is es-
sential for E2F activity, we generated a retrovirus vector en-
coding a small hairpin RNA (shRNA) directed against DP1 to
deplete E2F/DP complexes in primary human cells. Early pas-
sage TIG-3 cells were infected with retrovirus encoding either
a control sequence or shRNA specific for DP1. The levels of
DP1 mRNA and protein were significantly reduced within 7 d
of infection with the virus expressing the DP1-shRNA, but not
the control virus (Fig. 2, B and C). In addition, E2F-DNA–
binding activity was almost completely abolished in extracts of
TIG-3 cells expressing DP1-shRNA (Fig. 2 D, lanes 1 and 6).
In contrast to our results with dn-E2F, the expression level of
many E2F target genes required for S phase, such as cyclin A2,
thymidine kinase, and cdc6, was dramatically reduced in DP1
knock-down cells (Fig. 2, B and C). Such reduction has also
been reported in mouse embryonic fibroblasts lacking the acti-
vating E2F genes owing to genetic ablation (Wu et al., 2001).
The levels of cdc2 mRNA and protein, an essential component
of M phase progression, were also reduced in DP1 knock-down
cells. Similar results were also observed within 2 d of viral in-
fection, suggesting that the transcriptional changes directly re-
Figure 3. Depletion of DP1 causes severe growth arrest
in HDFs. (A and B) Early passage (45PDLs) TIG-3 cells
infected with retrovirus encoding DP1-shRNA or control-
shRNA were selected for expression of the puromycin-
selectable marker for 7d and used in a proliferation assay
performed in triplicate (A) or in cell cycle profile analysis
by FACS (B). Error bars indicate SD. (C and D) Early pas-
sage TIG-3 cells (45PDLs) were infected with retrovirus
encoding flag-tagged wild-type DP1 protein containing a
mutated shRNA cleavage site (lanes 3 and 4) or empty
vector (lanes 1 and 2). After selection with hygromycin,
cells were then super-infected with retrovirus encoding
DP1-shRNA (lanes 2 and 4) or control-shRNA (lanes 1
and 3). Expression of DP1, cyclin A, and CDC2 (C) and
BrdU incorporation (D) was examined 2 d after super-
infection. CDK4 was used here as a loading control. E2F
target genes reported previously were highlighted with
an asterisk. (E) TIG-3 cells infected with retrovirus encoding
DP1-shRNA or control-shRNA were examined for SA--gal
activity and SAHF.
DEPLETION OF DP PROVOKES CELLULAR SENESCENCE • MAEHARA ET AL.
557
sult from the decrease of E2F/DP activity (Fig. S1 A, available
at http://www.jcb.org/cgi/content/full/jcb.200411093/DC1). The
levels of PCNA and MCM3 were unchanged in DP1 knock-
down cells (Fig. 2, B and C). Thus, the expression of MCM3
and PCNA might be regulated by a balance between activating
E2Fs and repressive E2Fs, as seen in
Drosophila
cells (Frolov
et al., 2003). We failed to see any increase of interferon-induc-
ible genes (STAT1 and IFITM1) or p53 levels (Fig. 2, B and
C), arguing against the possibility that the DP1 shRNA induced
an interferon response or had nonspecific untargeted effects.
Next, we measured cell proliferation in DP1 knock-down
cells. To our surprise, DP1 depletion immediately caused cell
cycle arrest (Fig. 3 A) accompanied by a significant decrease in
the S phase population and an increase in the G1 phase and G2/
M phase populations (Fig. 3 B). To confirm that these effects
were actually due to the depletion of DP1 protein, a DP1 cDNA
containing a mutated shRNA cleavage site was retrovirally
transduced into TIG-3 cells. This mutated cDNA, which is re-
sistant to DP1-RNAi, maintained the level of flag-tagged wild-
type DP1 protein, despite expression of DP1-shRNA. Strik-
ingly, it also sustained expression of E2F target genes, such as
cyclin A2 and cdc2 (Fig. 3 C, lanes 2 and 4), and entry into S
phase (Fig. 3 D, lanes 2 and 4). These results confirm that the
reduction of E2F target gene expression and cell proliferation
by DP1-RNAi were specifically dependent on the depletion of
DP1 protein. Furthermore, several features of cellular senes-
cence, such as a substantial increase in senescence-associated
-galactosidase (SA-
-gal) activity (Dimri et al., 1995) and se-
nescence-associated heterochromatic foci (SAHF) (Narita et
al., 2003) were observed in DP1 knock-down cells (Fig. 3 E),
but not in cells expressing flag-tagged DP1 that is resistant to
RNAi (unpublished data). These results suggest that the main-
tenance E2F/DP activity is required for protecting cells from
onset of premature or stress-induced senescence.
DP1 knock-down and overexpression of dn-E2F were not
equivalent with respect to either E2F target gene expression or
cell proliferation. To understand the basis for the apparent dis-
crepancy, we performed ChIP assays using an antibody against
E2F3, the most abundant activating E2F in fibroblasts, and ex-
amined the precipitated DNA for the presence of cyclin A2
promoter sequences by PCR. Importantly, we found that a sig-
nificant amount of endogenous E2F3 remained bound to the
cyclin A2 promoter in TIG-3 cells expressing dn-E2F (Fig. 4
A, lanes 3 and 4). In contrast, E2F3 binding was significantly
reduced in DP1 knock-down TIG-3 cells (Fig. 4 B, lanes 3 and
4). Similar results were observed when we used an anti-E2F4
Figure 4. Incomplete blockage of E2F/DP activity in HDFs expressing dn-E2F. (A and B) ChIP assays were performed using TIG-3 cells expressing dn-E2F
(A), or DP1-shRNA (B) and the antibodies shown. The cyclin A2 promoter was recovered by PCR using primers flanking the E2F-binding sites in the human
cyclin A2 promoter. The human -actin promoter, which does not contain E2F-binding sites, was used as a negative control for PCR. (C) TIG-3 cells
(50PDLs) expressing dn-E2F (lanes 3 and 4) or empty vector (lanes 1 and 2) were super-infected with retrovirus encoding DP1-shRNA (lanes 2 and 4) or
control-shRNA (lanes 1 and 3). Total RNAs were prepared from cells at 7 d after selection with puromycin, and were subjected to RT-PCR with primers
shown on the left of the figure. E2F target genes reported previously were highlighted with an asterisk. (D) Cells described in C were used in a proliferation
assay performed in triplicate. Error bars indicate SD.
JCB • VOLUME 168 • NUMBER 4 • 2005558
antibody for the ChIP assay (Fig. 4, A and B; lanes 5 and 6).
These findings suggest that endogenous E2F/DP activity is ef-
fectively blocked by knockdown of DP1, but not by dn-E2F.
Indeed, depletion of DP1 from TIG-3 cells expressing dn-E2F
caused a significant reduction of E2F target gene expression
(Fig. 4 C, lanes 3 and 4) and consequent growth arrest (Fig. 4
D). These results confirm that a substantial level of endogenous
E2F/DP activity is indeed present in TIG-3 cells expressing
dn-E2F. Thus, the disparate results obtained by the two ap-
proaches can be explained by the insufficient blockage of en-
dogenous E2F/DP activity by dn-E2F in HDFs.
To verify our conclusions, we compared the effects of dn-
E2F with that of DP1-RNAi in human osteosarcoma U2OS
cells, which were used in the previous dn-E2F study (Zhang et
al., 1999). Consistent with our results in TIG-3 cells, DP1
knock-down caused a significant inhibition of cell growth (Fig.
5 A), whereas overexpression of dn-E2F had little effect (un-
published data). Additionally, the interpretation of results ob-
tained by dn-E2F has recently been questioned by a study
showing that dn-E2F (E2F-DB) contains a previously uncov-
ered pRB-binding domain that could potentially inactivate en-
dogenous pRB-family proteins (Dick and Dyson, 2003). In-
deed, we observed significant interaction between dn-E2F and
pRB in U2OS cells (Fig. S1 B). Moreover, although overex-
pression of dn-DP1 in human breast epithelial cells produced
effects similar to those produced by dn-E2F, namely an in-
creased incidence of tumors (Bargou et al., 1996), different re-
sults were observed depending on the particular dn-DP1 con-
struct that was used (Wu et al., 1996). Therefore, it is important
to stress that interpretation of experiments using dn-E2F or DP
requires careful evaluation.
A recently reported knock-out study of Kohn et al.
(2004) concludes that DP1 is dispensable for growth in vari-
ous mouse embryonic tissues. Because DP2 levels are very
low in human cells tested when compared with DP1 levels
(Fig. 2 A; Fig. S2 A, available at http://www.jcb.org/cgi/
Figure 5. Depletion of DP1 causes cellular
senescence in human cancer cell lines. (A–C)
U2OS (A), HT-29 (B), and HeLa cells (C) were
infected with retrovirus encoding DP1-shRNA
or control-shRNA and selected with puromycin
for 7 d. Cell numbers were then counted tak-
ing a time course in tetraplicate. The levels of
DP1 and CDK4 (loading control) were exam-
ined by Western blotting at d 3. (D) The HeLa
cells infected with retrovirus encoding either
control-shRNA or DP1-shRNA were used for
SA--gal assay 8 d after selection with puro-
mycin. The percentages of average SA--gal–
positive cells were indicated. (E) ChIP assays
were performed in HeLa cells with or without
DP1 knocking down as described in Fig. 4, A
and B. -Actin promoter, which does not con-
tain E2F-binding sites, was used as a negative
control for PCR. (F) Formation of subcutaneous
tumors in nude mice by HeLa cells expressing
shRNA against control sequence or DP1. For
each injection, 106 cells of the indicated pop-
ulations were injected subcutaneously in a
volume of 100 l. Mice were killed when the
tumors reached a diameter of 1 cm or after
10 wk of monitoring.
DEPLETION OF DP PROVOKES CELLULAR SENESCENCE • MAEHARA ET AL.
559
content/full/jcb.200411093/DC1), it is possible that the differ-
ent responses of mouse and human cells to DP1 loss may be
due to the DP2 status in the target cells. However, it is equally
possible that other explanations exist, such as cell type speci-
ficity, acute versus stable target loss, etc.
Next, we asked if DP1 knock-down inhibits the prolifera-
tion of other human cancer cell lines. DP1 knock-down signifi-
cantly inhibited cell proliferation in HT-29 cells, which lack
functional p53 (Fig. 5 B), and HeLa cells, in which pRB-family
proteins and p53 are inactivated by viral oncoproteins (Fig. 5 C).
Interestingly, reduction of DP1 protein levels induced several
features of cellular senescence in these cancer cells, including a
large, flat morphology and expression of SA-
-gal activity
(Fig. 5 D). HeLa cells expressing either DP1-shRNA or control
shRNA were injected subcutaneously into immunocompro-
mised mice. Although the control cells efficiently formed tu-
mors in 8 wk (19/20 cases), none of the DP1 knock-down cells
developed detectable tumors during the same time period (Fig.
5 F; Fig. S2 B). These results strongly suggest that E2F/DP
activity is required for tumor development.
Several lines of evidence suggest that pRB and p53 are
critical for induction of cellular senescence (Psyrri et al., 2004).
However, we have shown here that a senescence-like cell cycle
arrest can be induced by the reduction of total E2F/DP activity
without recovering the function of pRB and p53 in HeLa cells
(Fig. 5, C and D). Because the p16
INK4a
/RB tumor suppressor
pathway is frequently deregulated in a wide range of human
cancers, it is important to identify critical downstream targets
of this pathway for cancer therapy (Drayton and Peters, 2002;
Lowe and Sherr, 2003). Although additional studies are needed
to clarify the precise roles of E2F/DP complexes, our results
show that the proliferation of cancer cells could be controlled
through targeting the E2F/DP activity.
Materials and methods
Cell culture, transfection, and retrovirus production
TIG-3, SVts8, and HEK 293T cells were grown in DME supplemented with
10% FBS and penicillin/streptomycin. Retrovirus-shRNAs were generated
as described previously (Brummelkamp et al., 2002). To generate DP1-
resistant mutant against DP1 shRNA, three-point mutations, which do not
change encoding amino acids, were introduced into the shRNA cleavage
site of the DP1 cDNA. For growth rate analysis, cells were plated on the
gridded dish at concentration of 300 cells/1 cm
2
. Cell numbers were
counted in triplicate. BrdU incorporation was measured as described pre-
viously (Ohtani et al., 2003).
Antibodies and protein analyses
Immunoblotting was performed as described previously (Ohtani et al.,
2003) with primary antibodies against CDK4 (sc-601; Santa Cruz Bio-
technology, Inc.), cyclin A (sc-751; Santa Cruz Biotechnology, Inc.), E2F1
(sc-251; Santa Cruz Biotechnology, Inc.), MCM3 (sc-9849; Santa Cruz
Biotechnology, Inc.), PCNA (sc-56; Santa Cruz Biotechnology, Inc.), and
p107 (sc-318; Santa Cruz Biotechnology, Inc.), pRB (#554136; BD Bio-
sciences), FLAG (F3165; Sigma-Aldrich),
-actin (A5316; Sigma-Aldrich),
DP1 (#W32.3, Cancer Research UK; ab-11834, Abcam), CDC2 (#17;
Cancer Research UK), p16 (NA29; Oncogene Research Products), p53
(OP43; Calbiochem), p21 (sc-397; Santa Cruz Biotechnology, Inc.), and
p27 (sc-528; Santa Cruz Biotechnology, Inc.)
Electrophoretic mobility shift assays
Electrophoretic mobility shift assays (EMSAs) were performed as de-
scribed previously (Wu et al., 1995). The specificity of the protein–DNA
interactions was conformed by competition with wild-type oligonucleotides
or addition of the antibodies specific for E2F1 (sc-251), E2F2 (sc-633x),
E2F3 (sc-879x), E2F4 (sc-866), or GFP (sc-9996).
ChIP assay
ChIP assays were performed based on a modification of previously pub-
lished methods (Kanemaki et al., 2003; Ohtani et al., 2003). In brief, 5
10
6
cells were cross-linked by addition of formaldehyde to 1% final con-
centration, and then chromatin was sonicated and immunoprecipitation
was performed with Dynabeads protein A/G (Dynal), which were incu-
bated with antibody against E2F3 (sc-878x), E2F4 (sc-1082x), or Id3 (sc-
490) beforehand. Precipitates were washed and processed for DNA puri-
fication. DNA released from precipitated complexes was amplified using
sequence-specific primers by PCR (primer sequences: see online supple-
mental information).
Semiquantitative RT-PCR analyses
Total RNA was isolated using TRIzol (Invitrogen), and 5
g was reverse
transcribed with Super-scriptase (Invitrogen). Hot-start PCR was performed,
and the linear range of amplification was determined from PCRs run with
serially diluted cDNA. The results were verified by varying the number of
PCR cycles for each cDNA and set of primers (see online supplemental in-
formation). PCR products were separated on agarose gels and visualized
by ethidium bromide staining.
SA-
-gal and SAHF analysis
TIG-3 cells infected with retrovirus encoding DP1-shRNA or control-shRNA
were examined for SA-
-gal activity and SAHF as described previously
(Dimri et al., 1995; Narita et al., 2003). Slides for SA-
-gal were imaged
using a microscope (Axiovert 35M; Carl Zeiss MicroImaging, Inc.) with an
Achrostigmat objective (10
, 0.25 NA) and a digital camera (Axiocam
MR; Carl Zeiss MicroImaging, Inc.) and software (Zeiss Axiovision). Slides
for SAHF were imaged using a microscope (BX51; Olympus) with a Plan
Apochromat objective (60
, 1.4 NA) and a digital camera (Colorview
12; Soft-Imaging System) and a software (analysis; Soft-Imaging System).
Subsequent processing of TIFF files were undertaken in Adobe Photoshop
(version 7.0.1). Images were cropped and assembled into composites for
figures after minor adjustments for contrast and color balance were ap-
plied to all parts of each image.
Online supplemental material
Fig. S1 A shows the levels of E2F target gene expression after 2 d of DP1
knock-down. Fig. S1 B shows interaction between dn-E2F and pRB. Fig.
S2 A shows the levels of DP1 or DP2 mRNA expression in various human
cancer cells. Fig. S2 B shows an example of tumor formation assay. Sup-
plemental information shows primer sequences used for RNAi and PCR
analysis. Online supplemental material available at http://www.jcb.org/
cgi/content/full/jcb.200411093/DC1.
We thank Drs. R. Agami and R. Bernards (Netherlands Cancer Institute, Amster-
dam, Netherlands), S. Chellappan (University of South Florida, Tampa, FL), S.
Gaubatz (Philipps University, Marburg, Germany), D.M. Livingston Dana-Far-
ber Cancer Institute, Boston, MA), and E. Harlow (Harvard Medical School,
Boston, MA) for providing useful materials and K. Labib, J. Campisi, D. Mann,
G. Peters, and N. Dyson for valuable discussion. We are also grateful to Dr.
M. Kanemaki for help in ChIP assay and to members in the Paterson Institute for
Cancer Research for their various technical assists. We are indebted to Dr. K.
Helin and his colleagues for exchanging results before publication.
This work was supported by grants from Cancer Research UK, Associa-
tion for International Cancer Research, Yamanouchi Foundation for Research
on Metabolic Disorders, Takeda Science Foundation, and Ministry of Educa-
tion, Science, Sports, Culture and Technology of Japan to E. Hara.
Submitted: 16 November 2004
Accepted: 16 December 2004
References
Bandara, L.R., R. Girling, and N.B. La Thangue. 1997. Apoptosis induced in
mammalian cells by small peptides that functionally antagonize the Rb-
regulated E2F transcription factor.
Nat. Biotechnol.
15:896–901.
Bargou, R., C. Wagener, K. Bommert, W. Arnold, P.T. Daniel, M.Y. Mapara, E.
Grinstein, H.D. Royer, and B. Dorken. 1996. Blocking the transcription
factor E2F/DP by dominant-negative mutants in a normal breast epithe-
lial cell line efficiently inhibits apoptosis and induces tumor growth in
SCID mice.
J. Exp. Med.
183:1205–1213.
JCB • VOLUME 168 • NUMBER 4 • 2005560
Brummelkamp, T.R., R. Bernards, and R. Agami. 2002. Stable suppression of tu-
morigenicity by virus-mediated RNA interference.
Cancer Cell.
2:243–247.
Dick, F.A., and N. Dyson. 2003. pRB contains an E2F1-specific binding domain
that allows E2F1-induced apoptosis to be regulated separately from other
E2F activities.
Mol. Cell.
12:639–649.
Dimri, G.P., X. Lee, G. Basile, M. Acosta, G. Scott, C. Roskelley, E.E. Me-
drano, M. Linskens, I. Rubelj, O. Pereira-Smith, et al. 1995. A biomarker
that identifies senescent human cells in culture and in aging skin in vivo.
Proc. Natl. Acad. Sci. USA.
92:9363–9367.
Drayton, S., and G. Peters. 2002. Immortalization and transformation revisited.
Curr. Opin. Genet. Dev.
12:98–104.
Dyson, N. 1998. The regulation of E2F by pRB-family proteins.
Genes Dev.
12:2245–2262.
Frolov, M.V., O. Stevaux, N.-S. Moon, D. Dimova, E.-J. Kwon, E.J. Morris,
E.J., and N.J. Dyson, 2003. G1 cyclin-dependent kinases are insufficient
to reverse dE2F2-mediated repression.
Genes Dev.
17:723–728.
Gaubatz, S., G.J. Lindeman, S. Ishida, L. Jakoi, J.R. Nevins, D.M. Livingston,
and R.E. Rempel. 2000. E2F4 and E2F5 play an essential role in pocket
protein-mediated G1 control.
Mol. Cell.
6:729–735.
Kanemaki, M., A. Sanchez-Diaz, A. Gambus, and K. Labib. 2003. Functional
proteomic identification of DNA replication proteins by induced proteol-
ysis in vivo.
Nature.
423:720–724.
Kohn, M.J., S.W. Leung, V. Criniti, M. Agromayor, and L. Yamasaki. 2004.
Dp1 is largely dispensable for embryonic development.
Mol. Cell. Biol.
24:7197–7205.
Lowe, S.W., and C.J. Sherr. 2003. Tumor suppression by Ink4a-Arf: progress
and puzzles.
Curr. Opin. Genet. Dev.
13:77–83.
Mann, D.J., and N. Jones. 1996. E2F-1 but not E2F-4 can overcome p16-
induced G1 cell-cycle arrest.
Curr. Biol.
6:474–483.
Narita, M., S. Nunez, E. Heard, M. Narita, A.W. Lin, S.A. Hearn, D.L. Spector,
G.J. Hannon, and S.W. Lowe. 2003. Rb-mediated heterochromatin for-
mation and silencing of E2F target genes during cellular senescence.
Cell.
113:703–716.
Ohtani, N., P. Brennan, S. Gaubatz, E. Sanij, P. Hertzog, E. Wolvetang, J. Ghys-
dael, M. Rowe, and E. Hara. 2003. Epstein-Barr virus LMP1 blocks p16
INK4a
/RB-pathway by promoting nuclear export of E2F4/5.
J. Cell Biol.
162:173–183.
Psyrri, A., R.A. DeFilippis, A.P.B. Edwards, K.E. Yates, L. Manuelidis, and D.
DiMaio. 2004. Role of the retinoblastoma pathway in senescence trig-
gered by repression of the human papillomavirus E7 protein in cervical
carcinoma cells.
Cancer Res.
64:3079–3086.
Rowland, B.D., S.G. Denissov, S. Douma, H.G. Stunnenberg, R. Bernards, and
D.S. Peeper. 2002. E2F transcriptional repressor complexes are critical
downstream targets of p19
ARF
/p53-induced proliferative arrest.
Cancer
Cell.
2:55–65.
Takahashi, Y., J.B. Rayman, and B.D. Dynlacht. 2000. Analysis of promoter
binding by the E2F and pRB families in vivo: distinct E2F proteins me-
diate activation and repression.
Genes Dev.
14:804–816.
Trimarchi, J.M., and J.A. Lees. 2002. Sibling rivalry in the E2F family.
Nat.
Rev. Mol. Cell Biol.
3:11–20.
Wu, C.L., L.R. Zukerberg, C. Ngwu, E. Harlow, and J.A. Lees. 1995. In vivo as-
sociation of E2F and DP family proteins.
Mol. Cell. Biol.
15:2536–2546.
Wu, C.L., M. Classon, N. Dyson, and E. Harlow. 1996. Expression of dominant-
negative mutant DP-1 blocks cell cycle progression in G1.
Mol. Cell.
Biol.
16:3698–3706.
Wu, L., C. Timmers, B. Maiti, H.I. Saavedra, L. Sang, G.T. Chong, F. Nuckolls,
P. Giangrande, F.A. Wright, S.J. Field, et al. 2001. The E2F1-3 transcrip-
tion factors are essential for cellular proliferation.
Nature.
414:457–462.
Zhang, H.S., A.A. Postigo, and D.C. Dean. 1999. Active transcriptional repres-
sion by the Rb-E2F complex mediates G1 arrest triggered by p16
INK4a
,
TGF
, and contact inhibition.
Cell.
97:53–61.
... To obtain insight into the mechanism responsible for the downregulation of RNaseH2A during cellular senescence, we conducted a promoter sequence analysis of human RNASEH2A. The transactivation activity of the E2F family is known to be repressed by the p16 INK4a -RB pathway in senescent cells 16,35,36 . Therefore, we hypothesised that E2F activity is crucial for RNA-SEH2A expression in HDFs. ...
... Chromatin immunoprecipitation (ChIP) analysis illustrated that E2F transcription factors, namely E2F1 and E2F3, bound to the promoter region of RNASEH2A (Fig. 2b). To confirm the importance of E2F transcription factors in RNaseH2A expression, we used small interfering RNA (siRNA) to knock down DP1, an essential activator of E2F transcription factors 35,37 . This resulted in a significant reduction in RNaseH2A mRNA and protein expression in pre-senescent HDFs (Fig. 2c, d). ...
Article
Full-text available
Cellular senescence caused by oncogenic stimuli is associated with the development of various age-related pathologies through the senescence-associated secretory phenotype (SASP). SASP is mediated by the activation of cytoplasmic nucleic acid sensors. However, the molecular mechanism underlying the accumulation of nucleotide ligands in senescent cells is unclear. In this study, we revealed that the expression of RNaseH2A, which removes ribonucleoside monophosphates (rNMPs) from the genome, is regulated by E2F transcription factors, and it decreases during cellular senescence. Residual rNMPs cause genomic DNA fragmentation and aberrant activation of cytoplasmic nucleic acid sensors, thereby provoking subsequent SASP factor gene expression in senescent cells. In addition, RNaseH2A expression was significantly decreased in aged mouse tissues and cells from individuals with Werner syndrome. Furthermore, RNaseH2A degradation using the auxin-inducible degron system induced the accumulation of nucleotide ligands and induction of certain tumourigenic SASP-like factors, promoting the metastatic properties of colorectal cancer cells. Our results indicate that RNaseH2A downregulation provokes SASP through nucleotide ligand accumulation, which likely contributes to the pathological features of senescent, progeroid, and cancer cells. The levels of catalytic subunit of the RNaseH2 enzyme, RNAseH2A, decrease during senescence promoting nucleotide accumulation and a senescence-associated secretory phenotype.
... In senescent cells, expression of the cyclin-dependent kinase (CDK) inhibitors p16INK4a and p21WAF1/CIP1 is upregulated. Inhibition of the cyclin D-CDK4/6 and cyclin E-CDK2 complexes by p16INK4a and p21WAF1/CIP1, respectively, suppresses E2F target genes and activates the cell-cycle checkpoint [142][143][144][145][146]. Overview of the cGAS-STING pathway in cancer cells and surrounding cells. ...
... In senescent cells, expression of the cyclin-dependent kinase (CDK) inhibitors p16INK4a and p21WAF1/CIP1 is upregulated. Inhibition of the cyclin D-CDK4/6 and cyclin E-CDK2 complexes by p16INK4a and p21WAF1/CIP1, respectively, suppresses E2F target genes and activates the cell-cycle checkpoint [142][143][144][145][146]. Senescent cells have unique characteristics that result in a high activation of the cGAS-STING pathway. ...
Article
Full-text available
Cancer cells exhibit the unique characteristics of high proliferation and aberrant DNA damage response, which prevents cancer therapy from effectively eliminating them. The machinery required for telomere maintenance, such as telomerase and the alternative lengthening of telomeres (ALT), enables cancer cells to proliferate indefinitely. In addition, the molecules in this system are involved in noncanonical pro-tumorigenic functions. Of these, the function of the cyclic GMP–AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which contains telomere-related molecules, is a well-known contributor to the tumor microenvironment (TME). This review summarizes the current knowledge of the role of telomerase and ALT in cancer regulation, with emphasis on their noncanonical roles beyond telomere maintenance. The components of the cGAS-STING pathway are summarized with respect to intercell communication in the TME. Elucidating the underlying functional connection between telomere-related molecules and TME regulation is important for the development of cancer therapeutics that target cancer-specific pathways in different contexts. Finally, strategies for designing new cancer therapies that target cancer cells and the TME are discussed.
... La famille des facteurs de transcription E2F joue un rôle essentiel dans le contrôle de la prolifération cellulaire en activant ou réprimant de nombreux gènes impliqués dans la progression du cycle et la réplication. Cette fonction nécessite la formation d'un hétérodimère avec une protéine DP (E2F-DP) qui leur permet d'augmenter leurs liaisons à l'ADN et leur activité de transactivation (Maehara et al., 2005;Frolov et al., 2005). Chez les animaux, trois Chez Arabidopsis thaliana, parmi les 6 gènes codant des protéines E2F (E2F a à f), trois d'entre eux, E2Fa, E2Fb et E2Fc (les E2F canoniques) sont homologues des E2F de mammifères impliqués dans la régulation du cycle cellulaire (E2F1 à E2F5), et leur protéine sont capables de former des hétérodimères avec les 2 protéines DP (DPa et DPb ; Vandepoele et al., 2002;Mariconti et al., 2002). ...
Thesis
La protéine F-box FBL17 est un régulateur clef de la progression du cycle cellulaire chez Arabidopsis thaliana. Elle est notamment impliquée dans la dégradation des protéines inhibitrices de CDK, ce qui conduit chez le mutant fbl17 à des phénotypes développementaux sévères et une réduction drastique de l'activité de division cellulaire dans les méristèmes. Si l’implication de FBL17 dans le cycle cellulaire a été bien caractérisée, son répertoire de substrats et ses autres fonctions biologiques restent encore méconnus. Durant cette thèse, l’interactome de FBL17 a été mis en évidence et son implication dans la réponse aux dommages à l’ADN (DDR) a été étudiée. Le mutant fbl17 présente une DDR constitutive associée à une augmentation du nombre de lésions de l’ADN, et l’induction de gènes de la DDR de façon SOG1-indépendante. Au niveau protéique, en condition de stress génotoxique, FBL17 est recrutée au niveau des sites de lésions de l’ADN où elle co-localise avec la protéine RBR1. Ces données supportent un rôle de FBL17 dans le maintien de l’intégrité génétique des cellules.
... In Xenopus embryonic epidermis, MCIDAS forms a tripartite complex with Dp1 and E2F4 or E2F5 to promote motile cilia formation, and E2F4 binds to sequences such as 5′-GGG CGG GAAA-3′ and 5′-AGA GCG CG-3′ to promote ciliogenesis 29 . As HeLa cells express Dp1 and E2Fs endogenously 30 , the overexpression of MCIDAS alone can result in the formation of this tripartite complex in HeLa cells. We found two sequences similar to these E2F4-binding sites, namely ZFTA (− 256 to − 249) and ZFTA (− 246 to − 239) (cyan letters in Fig. 2C). ...
Article
Full-text available
The fusion protein of uncharacterised zinc finger translocation associated (ZFTA) and effector transcription factor of tumorigenic NF-κB signalling, RELA (ZFTA-RELA), is expressed in more than two-thirds of supratentorial ependymoma (ST-EPN-RELA), but ZFTA’s expression profile and functional analysis in multiciliated ependymal (E1) cells have not been examined. Here, we showed the mRNA expression of mouse Zfta peaks on embryonic day (E) 17.5 in the wholemount of the lateral walls of the lateral ventricle. Zfta was expressed in the nuclei of FoxJ1-positive immature E1 (pre-E1) cells in E18.5 mouse embryonic brain. Interestingly, the transcription factors promoting ciliogenesis (ciliary TFs) (e.g., multicilin) and ZFTA-RELA upregulated luciferase activity using a 5′ upstream sequence of ZFTA in cultured cells. Zfta tm1/tm1 knock-in mice did not show developmental defects or abnormal fertility. In the Zfta tm1/tm1 E1 cells, morphology, gene expression, ciliary beating frequency and ependymal flow were unaffected. These results suggest that Zfta is expressed in pre-E1 cells, possibly under the control of ciliary TFs, but is not essential for ependymal development or flow. This study sheds light on the mechanism of the ZFTA-RELA expression in the pathogenesis of ST-EPN-RELA: Ciliary TFs initiate ZFTA-RELA expression in pre-E1 cells, and ZFTA-RELA enhances its own expression using positive feedback.
... As a member of E2F family, the E2F4 plays an important role in regulating cellular proliferation and development (Humbert et al., 2000;Hsu and Sage, 2016;Hsu et al., 2019). In addition, previous work demonstrated that reduction of E2F/DP activity caused cell cycle arrest, repressed DNA damage repair, and induced senescencelike phenotypes (Maehara et al., 2005;Collin et al., 2018), suggesting that E2F4 is a key regulatory TF affecting the RS program. ...
Article
Full-text available
Senescence, a stable state of growth arrest, affects many physiological and pathophysiological processes, especially aging. Previous work has indicated that transcription factors (TFs) play a role in regulating senescence. However, a systematic study of regulatory TFs during replicative senescence (RS) using multi-omics analysis is still lacking. Here, we generated time-resolved RNA-seq, reduced representation bisulfite sequencing (RRBS) and ATAC-seq datasets during RS of mouse skin fibroblasts, which demonstrated that an enhanced inflammatory response and reduced proliferative capacity were the main characteristics of RS in both the transcriptome and epigenome. Through integrative analysis and genetic manipulations, we found that transcription factors E2F4, TEAD1 and AP-1 are key regulators of RS. Overexpression of E2f4 improved cellular proliferative capacity, attenuated SA-β-Gal activity and changed RS-associated differentially methylated sites (DMSs). Moreover, knockdown of Tead1 attenuated SA-β-Gal activity and partially altered the RS-associated transcriptome. In addition, knockdown of Atf3 , one member of AP-1 superfamily TFs, reduced Cdkn2a ( p16 ) expression in pre-senescent fibroblasts. Taken together, the results of this study identified transcription factors regulating the senescence program through multi-omics analysis, providing potential therapeutic targets for anti-aging.
... In Xenopus embryonic epidermis, MCIDAS forms a tripartite complex with Dp1 and E2F4 or E2F5 to promote motile cilia formation, and E2F4 binds to sequences such as 5'-GGGCGGGAAA-3' and 5'-AGAGCGCG-3' to promote ciliogenesis 29 . As HeLa cells express Dp1 and E2Fs endogenously 30 , the overexpression of MCIDAS alone can result in the formation of this tripartite complex in HeLa cells. We found two sequences similar to these E2F4-binding sites, namely ZFTA (-256 --249) and ZFTA (-246 --239) (cyan letters in Fig. 2C). ...
Preprint
Full-text available
The fusion protein of uncharacterised zinc finger translocation associated (ZFTA) and effector transcription factor of tumorigenic NF-kB signalling, RELA (ZFTA-RELA), is expressed in more than two-thirds of supratentorial ependymoma (ST-EPN-RELA), but ZFTA’s expression profile and functional analysis in multiciliated ependymal (E1) cells have not been examined. Here, we showed the mRNA expression of mouse Zfta peaks on embryonic day (E) 17.5 in the wholemount of the lateral walls of the lateral ventricle. Zfta was expressed in the nuclei of FoxJ1-positive immature E1 (pre-E1) cells in E18.5 mouse embryonic brain. Interestingly, the transcription factors promoting ciliogenesis (ciliary TFs) (e.g., multicilin) and ZFTA-RELA upregulated luciferase activity using a 5’ upstream sequence of ZFTA in cultured cells. Zftatm1/tm1 knock-in mice did not show developmental defects or abnormal fertility. In the Zftatm1/tm1 E1 cells, morphology, gene expression, ciliary beating frequency and ependymal flow were unaffected. These results suggest that Zfta is expressed in pre-E1 cells, possibly under the control of ciliary TFs, but is not essential for ependymal development or flow. This study sheds light on the mechanism of the ZFTA-RELA expression in the pathogenesis of ST-EPN-RELA: Ciliary TFs initiate ZFTA-RELA expression in pre-E1 cells, and ZFTA-RELA enhances its own expression using positive feedback.
... Generally, multiple stress signals, such as DNA damage, inhibit two inhibitory regulators, CDKN1A (yellow module) and CDKN2A (brown module), and subsequently activate the E2F pathways of the turquoise module (Fig. 7). Total E2F reduction induces senescence-like cell cycle arrest in cancer cells [35]; particularly, E2F1 blocks cell proliferation [36]. Given that chromatin assembly is one of the hub functions of down-regulated biological functions (Fig. 3B), E2F pathways could play key roles in the down-regulation of genes related to cell cycle, transcription, splicing, and translation during HUVEC senescence. ...
Article
Full-text available
Background Endothelial cell senescence is the state of permanent cell cycle arrest and plays a critical role in the pathogenesis of age-related diseases. However, a comprehensive understanding of the gene regulatory network, including genome-wide alternative splicing machinery, involved in endothelial cell senescence is lacking. Results We thoroughly described the transcriptome landscape of replicative senescent human umbilical vein endothelial cells. Genes with high connectivity showing a monotonic expression increase or decrease with the culture period were defined as hub genes in the co-expression network. Computational network analysis of these genes led to the identification of canonical and non-canonical senescence pathways, such as E2F and SIRT2 signaling, which were down-regulated in lipid metabolism, and chromosome organization processes pathways. Additionally, we showed that endothelial cell senescence involves alternative splicing. Importantly, the first and last exon types of splicing, as observed in FLT1 and ACACA , were preferentially altered among the alternatively spliced genes during endothelial senescence. We further identified novel microexons in PRUNE2 and PSAP , each containing 9 nt, which were altered within the specific domain during endothelial senescence. Conclusions These findings unveil the comprehensive transcriptome pathway and novel signaling regulated by RNA processing, including gene expression and splicing, in replicative endothelial senescence.
Article
The TFDP1 gene codes for the heterodimeric partner DP1 of the transcription factor E2F. E2F, principal target of the tumor suppressor pRB, plays central roles in cell proliferation by activating a group of growth-related genes. E2F also mediates tumor suppression by activating tumor suppressor genes such as ARF, an upstream activator of the tumor suppressor p53, when deregulated from pRB upon oncogenic changes. Among 8 E2F family members (E2F1∼E2F8), expression of activator E2Fs (E2F1∼E2F3a) is induced at the G1/S boundary of the cell cycle after growth stimulation by E2F itself. However, mechanisms regulating DP1 expression are not known. We show here that over-expression of E2F1 and forced inactivation of pRB, by adenovirus E1a, induced TFDP1 gene expression in human normal fibroblast HFFs, suggesting that the TFDP1 gene is a target of E2F. Serum stimulation of HFFs also induced TFDP1 gene expression, but with different kinetics from that of the CDC6 gene, a typical growth-related E2F target. Both over-expression of E2F1 and serum stimulation activated the TFDP1 promoter. We searched for E2F1-responsive regions by 5' and 3' deletion of the TFDP1 promoter and by introducing point mutations in putative E2F1-responsive elements. Promoter analysis identified several GC-rich elements, mutation of which reduced E2F1-responsiveness but not serum-responsiveness. ChIP assays showed that the GC-rich elements bound deregulated E2F1 but not physiological E2F1 induced by serum stimulation. These results suggest that the TFDP1 gene is a target of deregulated E2F. In addition, knockdown of DP1 expression by shRNA enhanced ARF gene expression, which is specifically induced by deregulated E2F activity, suggesting that activation of the TFDP1 gene by deregulated E2F may function as a failsafe feedback mechanism to suppress deregulated E2F and maintain normal cell growth in the event that DP1 expression is insufficient relative to that of its partner activator E2Fs. a maximum of 6 keywords: E2F, DP1, TFDP1 gene, pRB, gene expression.
Article
Full-text available
Cellular senescence in cancer development is known to have tumor‐suppressive and tumor‐promoting roles. Recent studies have revealed numerous molecular mechanisms of senescence followed by senescence‐associated secretory phenotype induction and showed the significance of senescence on both sides. Cellular senescence in stromal cells is one of the reasons for therapeutic resistance in advanced cancer; thus, it is an inevitable phenomenon to address while seeking an effective cancer treatment strategy. This review summarizes the molecular mechanisms regarding cellular senescence, focusing on the dual roles played by senescence, and offers some direction toward successful treatments targeting harmful senescent cells.
Article
Cellular senescence is an important tumor suppression mechanism caused by various stresses. Senescent cells secrete a large number of inflammatory proteins, which is called as senescence-associated secretory phenotype (SASP). Inflammatory SASP factors secreted from senescent cells induce chronic inflammation in the surrounding tissues, thereby causing age-related diseases. Recently, it has been shown that the secretion of small extracellular vesicle (sEV) is enhanced in senescent cells, acting as one of tumorigenic SASP factors. This article reviews the basic molecular mechanisms of sEV biogenesis in senescent cells and the latest findings on the biological function of sEV secreted from senescent cells. We also discuss the possibilities for clinical application of sEV in the field of aging research.
Article
Full-text available
The mammalian transcription factor E2F plays an important role in regulating the expression of genes that are required for passage through the cell cycle. This transcriptional activity is inhibited by association with the retinoblastoma tumor suppressor protein (pRB) or its relatives p107 and p103. The first cDNA from the E2F family to be cloned was designated E2F-1, and multiple E2F family members have now been identified. They bind to DNA as heterodimers, interacting with proteins known as DP. Here we demonstrate that DP is also a family of polypeptides with at least two members (hDP-1 and hDP-2). Both hDP-1 and hDP-2 bind to all E2F family members in vivo, and each complex is capable of activating transcription. However, the various E2F/DP complexes display strong differences in the ability to bind to either pRB or p107 in vivo, and the specificity of pRB or p107 binding is mediated by the E2F subunit.
Article
Full-text available
The transcription factor E2F is regulated during the cell cycle through interactions with the product of the retinoblastoma susceptibility gene and related proteins. It is thought that E2F-mediated gene regulation at the G1/S boundary and during S phase may be one of the rate-limiting steps in cell proliferation. It was reported that in vivo overexpression of E2F-1 in fibroblasts induces S phase entry and leads to apoptosis. This observation suggests that E2F plays a role in both cell cycle regulation and apoptosis. To further understand the role of E2F in cell cycle progression, cell death, and tumor development, we have blocked endogenous E2F activity in HBL-100 cells, derived from nonmalignant human breast epithelium, using dominant-negative mutants under the control of a tetracycline-dependent expression system. We have shown here that induction of dominant-negative mutants led to strong downregulation of transiently transfected E2F-dependent chloramphenicol acetyl transferase reporter constructs and of endogenous c-myc, which has been described as a target gene of the transcription factor E2F/DP. In addition, we have shown that blocking of E2F could efficiently protect from apoptosis induced by serum starvation within a period of 10 d, whereas control cells started to die after 24 h. Surprisingly, blocking of E2F did not alter the rate of proliferation or of DNA synthesis of these cells; this finding indicates that cell-cycle progression could be driven in an E2F-independent manner. In addition, we have been able to show that blocking of endogenous E2F in HBL-100 cells led to rapid induction of tumor growth in severe combined immunodeficiency mice. No tumor growth could be observed in mice that received mock-transfected clones or tetracycline to block expression of the E2F mutant constructs in vivo. Thus, it appears that E2F has a potential tumor-suppressive function under certain circumstances. Furthermore, we provide evidence that dysregulation of apoptosis may be an important step in tumorigenesis.
Article
Full-text available
Unregulated expression of the transcription factor E2F promotes the G1-to-S phase transition in cultured mammalian cells. However, there has been no direct evidence for an E2F requirement in this process. To demonstrate that E2F is obligatory for cell cycle progression, we attempted to inactivate E2F by overexpressing dominant-negative forms of one of its heterodimeric partners, DP-1. We dissected the functional domains of DP-1 and separated the region that facilitate heterodimer DNA binding from the E2F dimerization domain. Various DP-1 mutants were introduced into cells via transfection, and the cell cycle profile of the transfected cells was analyzed by flow cytometry. Expression of wild-type DP-1 or DP-1 mutants that bind to both DNA and E2F drove cells into S phase. In contrast, DP-1 mutants that retained E2F binding but lost DNA binding arrested cells in the G1 phase of the cell cycle. The DP-1 mutants that were unable to bind DNA resulted in transcriptionally inactive E2F complexes, suggesting that the G1 arrest is caused by formation of defective E2F heterodimers. Furthermore, the G1 arrest instigated by these DP-1 mutants could be rescued by coexpression of wild-type E2F or DP protein. These experiments define functional domains of DP and demonstrate a requirement for active E2F complexes in cell cycle progression.
Article
The transcription factor E2F is regulated during the cell cycle through interactions with the product of the retinoblastoma susceptibility gene and related proteins. It is thought that E2F-mediated gene regulation at the G1/S boundary and during S phase may be one of the rate-limiting steps in cell proliferation. It was reported that in vivo overexpression of E2F-1 in fibroblasts induces S phase entry and leads to apoptosis. This observation suggests that E2F plays a role in both cell cycle regulation and apoptosis. To further understand the role of E2F in cell cycle progression, cell death, and tumor development, we have blocked endogenous E2F activity in HBL-100 cells, derived from nonmalignant human breast epithelium, using dominant-negative mutants under the control of a tetracycline-dependent expression system. We have shown here that induction of dominant-negative mutants led to strong downregulation of transiently transfected E2F-dependent chloramphenicol acetyl transferase reporter constructs and of endogenous c-myc, which has been described as a target gene of the transcription factor E2F/DP. In addition, we have shown that blocking of E2F could efficiently protect from apoptosis induced by serum starvation within a period of 10 d, whereas control cells started to die after 24 h. Surprisingly, blocking of E2F did not alter the rate of proliferation or of DNA synthesis of these cells; this finding indicates that cell-cycle progression could be driven in an E2F-independent manner. In addition, we have been able to show that blocking of endogenous E2F in HBL-100 cells led to rapid induction of tumor growth in severe combined immunodeficiency mice. No tumor growth could be observed in mice that received mock-transfected clones or tetracycline to block expression of the E2F mutant constructs in vivo. Thus, it appears that E2F has a potential tumor-suppressive function under certain circumstances. Furthermore, we provide evidence that dysregulation of apoptosis may be an important step in tumorigenesis.
Article
Repression of the endogenous human papillomavirus (HPV) type 18 E7 gene in HeLa cervical carcinoma cells by the bovine papillomavirus E2 transcription factor activates the retinoblastoma (Rb) pathway and induces cells to undergo senescence. To determine whether activation of the Rb pathway is responsible for senescence in response to HPV18 E7 repression, we tested the ability of wild-type and mutant E7 proteins to affect the activity of the Rb pathway and to modulate senescence in these cells. Enforced expression of the wild-type HPV16 E7 protein prevented Rb activation in response to E2 expression and impaired senescence. Importantly, there was an absolute correlation between the ability of mutant E7 proteins to inactivate the Rb pathway and to inhibit senescence in HeLa cells. Similar results were obtained in HT-3 cervical carcinoma cells. These results provide strong genetic evidence that activation of the Rb pathway is required for senescence in response to E7 repression. Hence, continuous neutralization of the Rb pathway by the E7 protein is required to maintain the proliferation of cervical carcinoma cells. Similarly, our results indicate that activation of the Rb pathway can prevent apoptosis induced by repression of the HPV18 E6 gene in HeLa cells.
Article
Cellular senescence is an extremely stable form of cell cycle arrest that limits the proliferation of damaged cells and may act as a natural barrier to cancer progression. In this study, we describe a distinct heterochromatic structure that accumulates in senescent human fibroblasts, which we designated senescence-associated heterochromatic foci (SAHF). SAHF formation coincides with the recruitment of heterochromatin proteins and the retinoblastoma (Rb) tumor suppressor to E2F-responsive promoters and is associated with the stable repression of E2F target genes. Notably, both SAHF formation and the silencing of E2F target genes depend on the integrity of the Rb pathway and do not occur in reversibly arrested cells. These results provide a molecular explanation for the stability of the senescent state, as well as new insights into the action of Rb as a tumor suppressor.
Article
Normal somatic cells invariably enter a state of irreversibly arrested growth and altered function after a finite number of divisions. This process, termed replicative senescence, is thought to be a tumor-suppressive mechanism and an underlying cause of aging. There is ample evidence that escape from senescence, or immortality, is important for malignant transformation. By contrast, the role of replicative senescence in organismic aging is controversial. Studies on cells cultured from donors of different ages, genetic backgrounds, or species suggest that senescence occurs in vivo and that organismic lifespan and cell replicative lifespan are under common genetic control. However, senescent cells cannot be distinguished from quiescent or terminally differentiated cells in tissues. Thus, evidence that senescent cells exist and accumulate with age in vivo is lacking. We show that several human cells express a beta-galactosidase, histochemically detectable at pH 6, upon senescence in culture. This marker was expressed by senescent, but not presenescent, fibroblasts and keratinocytes but was absent from quiescent fibroblasts and terminally differentiated keratinocytes. It was also absent from immortal cells but was induced by genetic manipulations that reversed immortality. In skin samples from human donors of different age, there was an age-dependent increase in this marker in dermal fibroblasts and epidermal keratinocytes. This marker provides in situ evidence that senescent cells may exist and accumulate with age in vivo.
Article
Background: The transition from G1 to S phase is the key regulatory step in the mammalian cell cycle. This transition is regulated positively by G1-specific cyclin-dependent kinases (cdks) and negatively by the product of the retinoblastoma tumour suppressor gene, pRb. Hypophosphorylated pRb binds to and inactivates the E2F transcription factor, which controls the expression of genes required for S-phase progression. Hyperphosphorylation of pRb in late G1 phase results in the accumulation of active E2F, a critical event in the progression to S phase. The E2F factor is not a single entity, but rather represents a family of highly related molecules, all of which bind to pRb or the pRb-related proteins p107 and p130. Results: In this study, we have used specific inhibitors of cdks to explore the requirements for cell-cycle progression from G1 to S phase. Expression of p16Ink4, which specifically inhibits cyclin D-directed cdks, blocks cells in G1 phase; this block can be overcome by expression of the viral proteins that inactivate pRb or by E2F-1. Importantly however, the G1 arrest is not overcome by overexpression of E2F-4. By using chimeric E2F proteins, containing amino-acid sequences from E2F-1 and E2F-4, we have shown that their differential abilities to overcome a p16-imposed arrest is determined by their respective amino-terminal regions. We also demonstrate that E2F-1 can promote entry into S phase without concomitant phosphorylation of pRb. In contrast to the p16-mediated G1 block, G1 arrest mediated by the cdk inhibitors p21Cip1 or p27Kip1 cannot be bypassed either by inactivation of pRb or overexpression of E2F family members. Conclusions: These data demonstrate that the role of the cyclin D-directed cdks in promoting the progression of cells from G1 into S phase is wholly to activate an E2F-1-like activity through phosphorylation, thus preventing the formation of the E2F-pRb complex. The cyclin E-cdk2 complex is also required for the G1/S transition but has a different and as yet undefined role. We also provide evidence for a functional difference between E2F-1 and E2F-4, dependant upon the region that contains the DNA-binding and dimerization domains. These results indicate that these two E2F family members are likely to regulate the expression of different subsets of E2F-responsive promoters.
Article
A variety of studies implicate the E2F transcription factor as a critical regulator of the mammalian cell cycle. The E2F pathway is aberrant in most, if not all, human tumor cells; therefore, therapeutic regimes that modulate E2F activity may provide an approach for reinstating growth control in situations where normal physiological control is lost. To elucidate the role of E2F in the cell cycle and assess its value as a therapeutic target, we have introduced peptides that functionally antagonize E2F DNA binding activity into mammalian cells. Introduction of these peptides into mammalian tumor cells caused the rapid onset of apoptosis, an outcome that correlates with the inactivation of physiological E2F.