Vous êtes sur la page 1sur 10

Review articles

E2F target genes and cell-cycle


checkpoint control
Patrizia Lavia1 and Pidder Jansen-Dürr2*

Summary
In this review, we will focus on the role played by transcription factors of the
E2F/DP family in controlling the expression of genes that carry out important
cell-cycle control functions, thereby ensuring ordered progression through the
mammalian cell division cycle. The emerging picture is that cell-cycle progression
depends on the execution of a regulatory cascade of gene expression, driven by
E2F/DP transcription factors, which are in turn regulated by the products of some
of these genes. That E2F factors are potent regulators of cell-cycle checkpoints in
mammalian cells is supported by experiments demonstrating that ectopic expres-
sion of individual E2F family members is sufficient to modulate cell proliferation
and apoptosis. It is also clear that deregulation of E2F activity will result in the loss
of particular checkpoint controls, thereby predisposing cells to malignant conver-
sion. BioEssays 21:221–230. 1999. r 1999 John Wiley & Sons, Inc.

Introduction alterations in the concentration of key regulatory proteins, the


Ordered progression through the cell cycle is continually kept level of which is controlled by phase-specific activation of
on course by regulatory pathways generally referred to as gene expression and subsequent regulated proteolysis.(1) In
checkpoint controls. Checkpoint control mechanisms monitor this concept, transcriptional regulation of cell-cycle genes can
extracellular and intracellular growth-regulating signals and be viewed as an essential level of control of cell proliferation
ensure the dependency of a cell-cycle phase upon successful and checkpoint function.
completion of the preceding one. Checkpoint systems essen- Since the discovery several years ago that the pRb
tially act as negative regulatory pathways: ‘‘sensor’’ compo- product of the retinoblastoma susceptibility gene, a major
nents interact with their monitored substrate in the cell and, tumor suppressor, acts as a repressor of transcription of
hence, contact an ‘‘effector’’ molecule, which in turn acts on a
genes that are activated by the E2F group of transcription
key target and prevents its biological activity until the events
factors, a vast amount of work has been dedicated to clarify
required for transition into the next phase are completed.
how E2F and pRb-related factors control transcription of
Typical checkpoint targets are cell-cycle phase-specific cyclin/
responsive genes. The mechanisms of transcriptional control
kinase complexes whose normal biological activity stimulates
by E2F factors have been extensively reviewed recently
progression into the next phase of the cell cycle. Thus,
(references 2–7); in the present review, instead of re-
progression through the cell cycle is regulated by periodic
examining the well-documented properties of E2F transcrip-
tion factors, we will examine how regulation of specific
E2F-target genes in a particular temporal window of the cell
1Centro di Genetica Evoluzionistica C.N.R., c/o Universita ‘‘La Sapi-
cycle affects control of further cell-cycle progression. To this
enza,’’ Rome, Italy.
2Institut f. Biomedizinische Alternsforschung der Österieichischen Akad- aim, available data on the identity and function of E2F factors
emie der Wissenschaften, Rennweg 10, A-6020 Innsbruck, Austria. and their partners will be described briefly; we will then
Funding agencies: Deutsche Forschungsgemeinschaft, CNR Progetto essentially focus on the transcriptional control of several
Strategico ‘‘Cell cycle and apoptosis,’’ Associazione Italiana per la cyclin genes, which encode G1/S regulatory proteins, and of
Ricerca sul Cancro, and Biomed 2 program of the European Union.
the RanBP1 gene, which encodes a regulator of the Ran
*Correspondence to: Pidder Jansen-Dürr, Institut f. Biomedizinische
Alternsforschung der Österieichischen Akademie der Wissenschaften, signalling network, by the E2F/DP transcription factor family.
Rennweg 10, A-6020 Innsbruck, Austria. E-mail: P.Jansen- The key hypothesis is that the ability of E2F to control
Duerr@oeaw.ac.at expression of these genes is centrally involved in the modula-
tion of cell-cycle checkpoint controls by E2F.

BioEssays 21:221–230, r 1999 John Wiley & Sons, Inc. BioEssays 21.3 221
Review articles

Levels of regulation of E2F function DP family member) have an intrinsic ability to enter the
The factor originally designated E2F is made up of DNA- nucleus and act as transcriptional activators. In contrast,
binding heterodimers containing one E2F subunit and one DP heterodimers of DP-1, which lacks a nuclear localization
subunit. These subunits are encoded by two gene families: signal, with either E2F-4 or E2F-5 normally only reach the
the E2F family includes five characterized members, E2F-1 nucleus in association with a given pocket protein, i.e., when
through E2F-5, that largely act as transcriptional activators they function as repressors.(18) This observation marks an
(references 2–7, see also below); a sixth member, isolated important distinction between the members of the E2F family
more recently, E2F-6 or EMA, is instead reported to repress (summarized in Fig. 1). These rules are bypassed, however,
transcription, because it carries a DNA-binding, but not a in cells transformed by viral oncoproteins: free E2F-5/DP-1
transcriptional activation domain, and may occupy E2F- heterodimers are localized to the nucleus in E1A-transformed
binding sequences in promoters, thereby preventing access cells;(19) similarly, free E2F-4/DP-1 heterodimers have been
to other E2F members.(8,9) The DP family includes two genes, observed in the nuclei of HPV-16 E7-transformed cells.(21) A
DP-1 and DP-2, the latter of which gives rise to two functional further level of control of E2F activity is represented by the
products generated by alternative splicing; hence, three proteolytic degradation of some E2F proteins by the ubiquitin/
functional proteins actually compose the DP family.(2–4) proteasome pathway. The association with pocket proteins,
The activity of E2F/DP heterodimers is subjected to e.g., pRb, can greatly increase the stability of the E2F
several levels of control. The expression of the genes subunits.(22)
encoding E2F-1, E2F-2, and E2F-3 is low in G0 and maximal
at the G1/S transition; in the case of the E2F-1 and E2F-2
S phase entry and apoptosis: two distinct
gene promoters, this regulation is mediated by E2F-binding functions of E2F genes
sites, which indicates an autoregulatory loop. Conversely, Many of the genes that regulate E2F function are subject to
expression of E2F-4 and E2F-5 is relatively constant during mutation in human cancers,(23) and the phenotype of E2F-1
the cell cycle; therefore, these two factors are the predomi- knockout mice suggests that, in the context of the entire
nant E2F members to be found in G0 cells, indicating that organism, the E2F-1 gene has properties of both a tumor
they play a role in repression of E2F-mediated transcription in promotor and a tumor suppressor gene.(24) This puzzling
resting cells. Thus, at least some E2F components are finding may be explained by considering that certain E2F
subjected to cell-cycle control; the DP components show no members, including E2F-1, can activate transcription of
significant fluctuations. target genes in the absence of an associated pocket protein,
E2F/DP heterodimers interact with the pRb, p107, and yet repress the same genes when complexed with pRb.
p130 members of the pocket protein family. These proteins Furthermore, E2F proteins can also directly affect cell prolif-
antagonize transcriptional activators of the E2F family with a eration in two distinct ways.
specificity that is mainly determined by the E2F component. First, ectopic expression of E2F-1 overcomes G1 arrest
E2F-1, E2F-2, and E2F-3 preferentially associate with pRb,(10) caused by the inhibition of G1 cyclin-dependent kinases and
whereas E2F-4 predominantly interacts with p107 and drives quiescent human cells into S phase.(25,26) Similarly,
p130(10–12) and E2F-5 with p130 only.(13) However, all pocket both E2F-2 and E2F-3 can induce S phase in quiescent rat
proteins can block the activity of all E2F members in vitro, and fibroblasts. The ability of E2F-3 to specifically induce S phase
under certain conditions, all members of the E2F family are seems to be directly linked to the ability of E2F-3 to activate
complexed with pRb.(10,14) Complexes of E2F/DP het- genes encoding origin recognition complex (ORC) and mini-
erodimers with pocket proteins are destabilized through the chromosome maintenance proteins, which are structural
phosphorylation of pRb family members by cyclin-dependent components required for S phase onset.(27,28) Quite differ-
kinases (reviewed in references 3, 4; see also below). ently, both E2F-4 and E2F-5 require coexpression of a DP
The biological activity of those E2F members that are partner to promote S-phase entry.(16) The amino terminus of
steadily expressed, i.e., E2F-4 and E2F-5, is critically depen- E2F-1 is sufficient to enable full-length E2F-4 to induce
dent on regulated relocalization during the cell cycle.(15–17) S-phase and to transactivate an E2F-responsive promoter;
Although E2F-1 to E2F-3 represent bona fide nuclear pro- this reflects the ability of the E2F-1 amino terminus to direct
teins, nuclear localization of E2F-4 and E2F-5 depends on proteins to the nucleus.(16)
the interaction with an appropriate protein partner, and is Second, overexpression of E2F-1 induces apoptosis, an
promoted by DP family members that contain a nuclear effect that is modulated by p53.(29) The regulated suppression
localization signal (i.e., the ␣ and ␦ isoforms of DP-3 but not of E2F-1 DNA-binding activity, mediated through the phosphor-
other DP family members; reference 18), or by association ylation of DP-1 by cyclin A-kinase, is apparently required to
with a pocket protein (Fig. 1). Association with these different prevent an apoptotic response.(30) It was concluded that
molecules has opposite effects on transcription of E2F-driven improper activation of E2F-1 may function as a specific signal
genes:(19,20) Free E2F complexes that contain E2F-1 (and any for the initiation of an apoptotic pathway that is normally

222 BioEssays 21.3


Review articles

Figure 1. Structure and functions of E2F family members. The six known members of the E2F family are shown; the known functional
domains are schematically depicted as boxes (NLS: nuclear localization signal; DBD, DNA binding domain; DD, dimerization domain;
MB, marked box; TAD/PP, transcriptional activation domain, overlapping with the binding site for pocket proteins). For each family
member, the influence of associated heterologous proteins on the subcellular localization and transcriptional capacity is indicated. The
subcellular localization of any complexes involving E2F-6 has not been reported, and the molecular mechanism by which this E2F family
member represses transcription remains to be established.

blocked.(31) Analysis of E2F-1 mutants indicates that DNA genetic effects of E2F-responsive promoters are paralleled
binding, but not necessarily transcriptional activation, is by the protein-binding features of the respective E2F binding
required for induction of apoptosis.(32) These results indicate sites in the promoter region. E2F binding sites that have been
that stimulation of cell-cycle progression and induction of identified through mutagenesis or deletion assays to confer
apoptosis are independent functions of E2F-1. negative control of transcription in G0 cells (e.g., the E2F site
in the cdc2 gene promoter) are footprinted in vivo during G0
E2F/DP target genes and early G1 phases but not upon entrance into the prolifera-
The analysis of the promoters of genes that are responsive to
tive cycle.(34) In contrast, genes whose transcription is acti-
control by E2F might help to understand how different
vated by E2F factors at the G1/S transition, exemplified by the
E2F-containing complexes exert their different biological
DHFR gene, become footprinted in vivo at the G1/S transi-
effects. Binding sites for E2F factors are characteristic of two
tion.(35) The best candidates for such a mode of action include
major classes of genes, listed in Table 1: first, a group of
E2F-1, -2, and -3, which are themselves subjected to G1/S
genes encoding essential enzymes in the nucleotide and
DNA biosynthetic pathways that are coordinately up-regu- transcriptional induction. Negative control of transcription in
lated in late G1; second, genes encoding regulators of quiescence is essentially attributable to the presence of
cell-cycle progression. Genes from both groups respond to E2F-4 and E2F-5 in nuclei, which act to bridge the transcrip-
ectopic expression of E2F-1.(33) tional repressors pRb, p107 and p130 to their target DNA
Cell-cycle regulatory genes are controlled by E2F through elements in quiescent cells.
genetically distinct mechanisms: for example, the genes There is variation in the nucleotide sequence and binding
encoding Cdc2, cyclin E, and ORC1 are up-regulated in an affinity of the E2F sites in different gene promoters (some of
E2F-dependent manner, whereas the genes encoding which are indicated in reference 36; see also Table 1). The
RanBP1, cyclin A, B-myb, and p107 are repressed in G0 and specificity of E2F complexes for particular target genes,
activated at the G1/S transition; both effects involve E2F. The identified functionally or in DNA/protein interaction assays, is

BioEssays 21.3 223


Review articles

TABLE 1. Features of E2F-Driven Promoters


Regulation by E2F/DP Regulation by E2F/DP
Protein-binding features or pocket proteins or pocket proteins
Genea of E2F sites (transient assays) (endogenous genes)

Regulatory genes
c-myc Two E2F-binding sites complexes contain Transactivation by Ad5 E1A; E2F site Expression not affected in cells lacking
pRb; in vivo protection of E2F sites cor- required for repression by pRb pocket proteinsb
relates with transcriptional activity
cyclin E Multiple E2F-binding sites; a variant site, Repression by pRb; E2F site required for Strong induction by E2F-1c; de-repression
GCGCGA complexes contain p107 and promoter activation in G1; transactiva- in pRb⫺/⫺ cellsb
p130 tion by E2F4/DP-1
B-myb In vivo protection of E2F site correlates Transcriptional repression, corepressors Induction by E2F-1c; de-repression in cells
with transcriptional repression involved? Promoter derepression by E7 lacking p107 and p130b
p107 involved in repression
p107 Two E2F-binding sites Proximal E2F site for basal promoter activ- De-repression in pRb⫺/⫺ cellsb
ity; distal E2F site for repression by pRb
and p107
pRb One E2F site, complexes contain pRb Autorepression of the Rb-1 promoter by Expression not affected in cells lacking
pRb pocket proteinsb
E2F-1 Two E2F-binding sites, each composed of Both sites required for promoter repression De-repression in cells lacking p107 and
two overlapping E2F elements in G0; both sites respond to E2F-1 and p130b
E2F-3
E2F-2 Multiple E2F-binding sites; subtle Proximal E2F sites required for repression Not determined
sequence variations in G0; proximal E2F sites required for
activation by E2F-1, -2, and -3; growth
control also requires E-box elements
cyclin A Variant E2F site AGTCGCG overlaps with Negative control of promoter activity in G0;Induction by E2F-1c; de-repression in cells
CDF-1 binding site; complexes contain E2F site required for promoter activation lacking p107 and p130b
p107; in vivo protection correlates with at G1/S; E2F site required for activation
transcriptional repression by Ad5 E1A; p107 involved in repression
p34cdc2 Nonconsensus E2F site; overlaps with Negative control of promoter activity in G0 Induction by E2F-1c; de-repression in cells
CDE/CHR elements; complexes contain cells lacking p107 and p130b
E2F-4 and p130; in vivo protection cor-
relates with transcriptional repression
RanBP1 Two E2F-binding sites; in vivo protection of Distal site required for activation at G1/S; Induction by E2F-1d
distal site in cycling cells; lack of protec- distal site required for responsiveness to
tion at proximal site in cycling cells E2F-1, E2F-4, pRb, and p107; proximal
site required for G0 repression
Structural genes
DNA pol (␣) Two E2F consensus sites Transactivation by E2F-1 Induction by E2F-1c; expression not
affected in cells lacking pocket proteinsb
HsORC1 Two overlapping E2F sites with reverse E2F sites required for G0 repression; acti- Not determined
orientation vation by E2F-1
DHFR Multiple E2F-binding sites; in vivo occu- E2F sites required for activation at G1/S; Induction by E2F-1c; premature induction
pancy of one E2F site after G1/S transi- activation by E2F-1; cooperation with in cells lacking p107 and p130b
tion; constitutive in vivo occupancy of Sp1
another E2F site
Tk Two overlapping E2F sites; constitutive in E2F sites required for activation at G1/S; Expression not affected in cells lacking
vivo protection during cell cycle cooperation with Sp1 pocket proteinsb
TS One putative E2F site E2F site part of larger element; no activa- Induction by E2F-1c; de-repression in cells
tion by E2F-1 lacking p107 and p130b
PCNA One putative E2F site in first intron Induction by E2F-1/DP-1 Induction by E2F-1c; expression not
affected in cells lacking pocket proteinsb
RRM2 Not determined Not determined Induction by E2F-1c; de-repression in cells
lacking p107 and p130b
Srp20 Two putative E2F binding sites Induction by E2F-1/DP-1 Not determined
Histone H2A One E2F-binding site E2F site required for activation in early S Not determined
activation by E2F-1; repression by pRb
and p107

aMost original references to promoter studies are indicated in the text or are quoted in references 33, 36, and 41.
bData from reference 41.
cData from reference 33.

dData from reference 71.

224 BioEssays 21.3


Review articles

listed in Table 1. Interestingly, the E2F-binding sites in the Cyclins D and E


cyclin A,(37) cdc2,(34) and cyclin E(38) gene promoters are Growth factors induce expression of the cyclin D1 gene
characterized by a variant nucleotide sequence, accompa- (reviewed in reference 23). Cyclin D1 associates with cdk4 or
nied by a lower in vitro binding affinity. The reason for the cdk6; the kinase activity associated with these complexes
presence of relatively weak E2F binding sites in the promoter phosphorylates and thereby inactivates the pRb and p107
regions of these key cell-cycle regulatory genes is unclear at members of the retinoblastoma protein family; this activity is
present; it might indicate that the expression of these genes is subjected to regulation by cdk inhibitors, like p16INK4 or
controlled by other pathways, in addition to the E2F activation p21WAF-1 (reviewed in references 5, 43). Through the
pathway. In the case of the cyclin A gene, Liu et al.(39) inactivation of pRb family members, cyclin D-associated
presented evidence that the variant E2F binding site overlaps kinase triggers transcriptional activation of E2F-driven
with the binding site for an unrelated transcription factor, genes,(44) probably including the cyclin E gene.(45,38) Once
CDF-1; according to these authors, a functional interplay of cyclin E is synthesized, it associates with cdk2; the subse-
E2F and CDF factors brings about the specific timing of quent activation of cyclin E-associated kinase is essential for
transcription of certain target genes during the cell cycle.(40) S phase entry. Cyclin E-associated kinase induces a further
The analysis of cyclin gene expression in mouse fibroblasts in activation of E2F-dependent gene expression, including, in
which specific Rb family members have been deleted indi- addition to the autoregulatory activation of the cyclin E gene,
cates that cyclin A gene activity is de-repressed in cells transcriptional activation of the cyclin A gene.(46) Similar to
lacking both p107 and p130; in contrast, expression of the cyclin E, ectopic expression of cyclin D1 also triggers transcrip-
cyclin E gene is specifically de-repressed in pRb-deficient tion of the cyclin A gene;(37) however, it is clear that the
cells.(41) These results suggest that the E2F sites in the physiologic functions exerted by D-type cyclins are quite
promoter regions of these two cyclin genes display significant distinct from those of cyclin E. Cyclin D1 is up-regulated in
functional differences, probably resulting from the coopera- response to external conditions and is dispensable in many
tion of E2F family members with additional transcription tumor cells; on the other hand, cyclin E is an essential part of
factors that bind to adjacent or overlapping sites.
the cell-cycle machinery and is required for S phase entry in
all cases studied so far. Furthermore, different molecular
E2F factors and phase-specific transcription processes are involved in activation of cyclin A transcription
of the early cyclin genes by both cyclins: whereas cyclin D1 induces phosphorylation
In the following section, we will summarize the current of p107 followed by the dissociation of E2F-p107 com-
knowledge of how E2F factors signal from the proliferation plexes,(47) cyclin E associates with the cyclin A promoter as
apparatus to the promoters of cell-cycle regulatory genes. In part of a multimeric complex containing E2F, p107, and
cells that have entered the division programme, cyclin E and cdk2(46) (see Fig. 2).
cyclin A are sequentially synthesized and associate with
cdk2. Although the abundance of cyclin-dependent kinases
(cdk) is relatively constant throughout the cycle in mammalian Cyclin A
cells, the catalytic activity of these enzymes is controlled The cyclin A protein is required for S phase entry, suggesting
through multiple regulatory mechanisms, including regulatory that regulation of cyclin A expression is one of the latest
phosphorylation and the association of specific inhibitors essential steps before the onset of DNA replication; hence,
(CKIs) (for review, see reference 42). However, activation of this gene is likely to be the target of various cell-cycle
cdk enzymes cannot proceed in the absence of an associated checkpoint controls, summarized as follows: (1) cells grown
cyclin; hence, the abundance of the various cyclins is a key to high density or treated by TGF-␤ arrest in G1, which is
determinant for the activity of cyclin-dependent kinases. accompanied by repression of cyclin A gene expression,
There is evidence that S phase entry in mammalian cells is whereas cyclins D1 and E are normally expressed under
controlled through a regulatory cascade of cyclin gene expres- these conditions.(48) Regulation of cyclin A gene transcription
sion (Fig. 2), as described in detail below. In addition to this also contributes to G1 arrest upon loss of adhesion to the
domino-like cascade, regulatory feedback loops have been extracellular matrix,(49,50) (2) when diploid human cells reach
identified that control the expression of specific cyclin genes, replicative senescence, they arrest in G1, and this effect is
and it is assumed that these feedback loops are mainly accompanied by repression of cyclin A transcription, whereas
responsible for the sharp transitions between individual cell- cyclins D1 and E are overexpressed,(51) and (3) cyclin A
cycle phases. Furthermore, the regulatory cascade of cyclin transcription is down-regulated in cells in which p53 function
gene expression, as described in Figure 2, can be interrupted is activated by, for example, DNA damage. Down-regulation
at specific points by external growth-suppressive signals, of the cyclin A promoter is not due to direct binding of p53, but
which are often mediated through the action of particular cdk occurs through the p21WAF-1/Cip1 protein, a downstream effec-
inhibitors. tor of p53, which blocks the transactivation potential of E2F

BioEssays 21.3 225


Review articles

Figure 2. The regulatory cascade of early cyclin gene expression. When mammalian fibroblasts exit from G0 to enter S-phase, a
series of cyclin-dependent kinases is consecutively activated. The addition of serum growth factors to quiescent cells triggers
transcription of the cyclin D1 gene, and cyclin D1 molecules associate with preexisting cdk4 molecules. The kinase activity that is
associated with these complexes can phosphorylate specific sites in the retinoblastoma gene product pRb, leading to inactivation of pRb
and a subsequent derepression of the cyclin E gene. Once cyclin E is made, it associates with preexisting cdk2, and the kinase activity
associated with these complexes modulates the function of at least three sets of target proteins: (1) cyclin E/cdk2 complexes associate
with E2F-p107 complexes at the cyclin A promoter and trigger its transcriptional activation, (2) cyclin E/cdk2 complexes cooperate with
cyclin D1 to phosphorylate pRb, leading to its inactivation and the subsequent activation of E2F-driven genes, including the cyclin E gene
itself; thereby, a regulatory feedback loop is established, (3) it is assumed that cyclin E/cdk2 complexes phosphorylate additional
downstream targets that are unrelated to E2F regulation but that may be involved in the initiation of DNA replication. Strong
overexpression of cyclin D1 in vitro, similar to overexpression observed in several human cancers, can trigger activation of the cyclin A
gene by a different pathway that involves the disruption of p107/E2F complexes but may be independent of cyclin E/cdk2. Once cyclin A is
made, it associates with cdk2 and the resulting kinase activity modulates the function of at least two classes of proteins: (1) cyclin A/cdk2
phosphorylates the DP1 subunit of the E2F transcription factor, which leads to a loss of DNA binding capacity and, hence, a shutoff of
E2F-driven transcription; (2) it is assumed that cyclin A/cdk2 complexes phosphorylate additional downstream targets that may be
directly involved in the initiation of DNA replication. The cascade of cyclin gene expression can be interrupted at various points through
the action of cdk inhibitors. Please note that both p16 and p27, as shown here, represent two families of related cdk inhibitors (for details,
see reference 42).

complexes,(52) possibly through direct protein/protein interac- The function of cyclin A/cdk2 complexes in cell-cycle–
tion.(53) regulated transcription is distinct from that exerted by cyclin
In these instances, down-regulation of cyclin A gene E/cdk2 or cyclin D/cdk4. Ectopic expression of cyclin A does
expression is due to a repression of cyclin A promoter activity, not induce transcription of E2F-dependent genes,(37) on the
and in most cases, the E2F-binding site in the cyclin A contrary, cyclin A/cdk2-mediated phosphorylation of E2F-1/
promoter (see Table 1) is essential for this effect. The E2F site DP-1 heterodimers results in the loss of E2F binding activ-
of the cyclin A gene promoter is also targeted by transforming ity.(55) These observations suggest that, whereas cyclins D1
oncogenes encoded by DNA tumor viruses such as adenovi- and E activate E2F-dependent transcription, cyclin A might be
rus E1A or HPV-16 E7 (for review, see reference 54). These used to turn off E2F-driven genes, probably as part of its
observations are consistent with the notion that the abroga- function as checkpoint protein.
tion of cell-cycle checkpoint controls, monitored here by the Interestingly, E2F-6, which can antagonize E2F-1-induced
deregulation of cyclin A gene expression, is involved in cell transcription, has been reported to either prolong S phase in
transformation. cells growing asynchronously(56) or inhibit the G1/S transition

226 BioEssays 21.3


Review articles

in cells that have been serum-starved and then restimu- similar to that of mammalian cells in which RanBP1 expres-
lated.(57) Although there are some discrepancies in the effects sion is deregulated (see below).
of E2F-6 in these two studies, possibly owing to the differing Growing evidence shows that the Ran network also
pRb status of the cell lines used, these observations suggest controls nucleocytoplasmic transport; this function may in fact
that this protein acts to antagonize cell-cycle progression, a underlie the role of Ran and its partners in checkpoint
function that might apply during cell-cycle checkpoint activa- controls, given that defects in nucleocytoplasmic transport
tion, which occurs, for example, after DNA damage. Although are often associated with cell-cycle abnormalities. Mutations
entirely hypothetical, two reasons suggest that E2F-6- in components of the Ran network inhibit both import and
containing complexes might have a role in preventing tran- export activities, and the Ran GTPase is itself a shuttling
scriptional activation of E2F target genes in the presence of molecule that can participate in the export of several classes
DNA-damage. First, E2F-6 lacks a cyclin A-binding domain of RNAs and cytoplasmic proteins, as well as in nuclear
and, hence, is not sensitive to cyclin A-mediated phosphoryla- import of proteins that contain a nuclear localization signal
tion. Second, E2F-6-dependent transcriptional inactivity of (see reference 62 for review). Intriguingly, the subcellular
cell-cycle genes may be prolonged in those cases where localization of several cell-cycle regulatory proteins, such as
cell-cycle arrest or delay is required to sustain the repair E2F-4,(15,16) hCdc18,(63) cdc25C, Nim1, Cdc2, and cyclin B1
processes and monitor DNA integrity before resumption of (reference 64 and references therein) is regulated stringently
progression through S and G2. during cell-cycle progression, suggesting that their intracellu-
lar redistribution might play an important role in cell-cycle
control. These lines of evidence converge to suggest that
Transcriptional regulation of the RanBP1 gene by genes that control nucleocytoplasmic transport, including
E2F: a novel aspect of checkpoint control? Ran, are required to establish orderly cell-cycle progression.
Among the genes listed in Table 1, the RanBP1 gene The genes encoding Ran and RCC1 are both induced as
represents a perhaps unexpected target of E2F regulation: immediate-early genes and are steadily expressed in cycling
the RanBP1 protein is neither enzymatically involved in DNA cells. In contrast, the RanBP1-encoding gene is transcription-
replication nor a direct regulator of cell-cycle progression. ally induced at the G1/S boundary.(65) E2F factors control both
RanBP1 acts as a major regulator of Ran, a member of the growth-dependent repression and S-phase–specific induc-
Ras superfamily of GTPases, which controls a puzzling tion of the RanBP1 gene. An in vivo footprinted E2F-binding
variety of cell-cycle events, including checkpoint controls at site in the RanBP1 promoter(66) is responsible for G1/S
the G1/S and G2/M transition (for reviews, see references. up-regulation;(67) another E2F-binding site that flanks the
58, 59; see also below). In the following two paragraphs, we transcription start site controls G0 repression (71). The
first will briefly recapitulate the known functions of the Ran finding that the timing of RanBP1 transcriptional induction is
GTPase and then summarize the available evidence concern- coupled to that of genes required for S phase by the same
ing the role of RanBP1 within the Ran regulatory network. family of E2F activators and pocket protein repressors (Fig.
The signalling activity of Ran is dependent on the switch 3), suggests that, in mammals at least, RanBP1 might act as
between the GTP-bound and GDP-bound forms of the mol- a ‘‘sensor’’ of the cell-cycle phase, connecting the Ran cycle
ecule, and the balance between both forms is modulated by to the cell cycle: As the RanBP1 protein modulates both
the antagonistic activities of RCC1 (regulator of chromosome RCC1 and RanGAP activities,(68) E2F-dependent transcrip-
condensation), which acts as the nucleotide exchange factor tional activation of RanBP1 at the G1/S transition will modify
for Ran, and of RanGAP (Ran GTPase-activating protein). the molecular ratio of RCC1 to RanBP1 and, hence, of
Although the molecular targets of Ran are not yet identified, RanGTP to RanGDP, during S phase. Regulated expression
genetic evidence suggests that the Ran GTPase functions as of RanBP1 is now emerging as a requirement for orderly
a ‘‘timer’’ molecule during cell-cycle progression. Such a role coordination of functions that are controlled by the Ran
is supported by the finding that the Ran/RCC1 signalling network. Replacing the endogenous promoter of the RanBP1
activity is absolutely required for: (1) the G2 checkpoint, as gene with cell-cycle–independent sequences resulted in pro-
indicated by the appearance of prematurely condensed duction of the RanBP1 protein throughout the cell cycle and
chromosomes independent of S-phase completion, which caused defects in all major cell-cycle transitions, failure of
represents the first recognized alteration associated with loss chromatin decondensation after metaphase and impaired exit
of function of RCC1 (reviewed in reference 60); (2) the onset from mitosis to the subsequent interphase.(69) These abnor-
of S phase (reviewed in references 59, 60; see also reference malities suggest that the molecular balance among members
61); (3) completion of the mitotic division, as indicated by of the Ran network during specific phases is crucial for
fission yeast mutants lacking RCC1 activity that are impaired orderly cell-cycle progression. Interestingly, a cell line stably
in the mitosis-to-interphase transition with hypercondensed expressing a dominant negative version of E2F-1 displays a
chromosomes (reviewed in reference. 58), a phenotype specific phenotype(70) that is similar to the phenotype ob-

BioEssays 21.3 227


Review articles

Acknowledgments
We apologize to the many colleagues whose work could not
be cited due to space constraints. Work in the laboratory of
P.J.D. was supported by a grant from the Deutsche Forsch-
ungsgemeinschaft. Work in the laboratory of P.L. was sup-
ported by grants from CNR Progetto Strategico ‘‘Cell cycle
and apoptosis’’ and Associazione Italiana per la Ricerca sul
Cancro.

References
1. Reed S. G1/S regulatory mechanisms from yeast to man. Prog Cell Cycle
Res 1996;2:15–27.
2. Johnson D, Schneider-Broussard R. Role of E2F in cell cycle control and
cancer. Front Biosci 1998;3:447–448.
3. Helin K. Regulation of cell proliferation by the E2F transcription factors.
Figure 3. E2F/DP factors are represented upstream of Curr Opin Genet Dev 1998;8:28–35.
some known target genes. Through transcriptional control of 4. Bernards R. E2F: a nodal point in cell cycle regulation. Biochim Biophys
target genes (in ovals), E2F factors control downstream Acta 1997;1333:33–39.
processes (in squares): G1 progression depends on transcrip- 5. Herwig S, Strauss M. The retinoblastoma protein: a master regulator of cell
cycle, differentiation and apoptosis. Eur J Biochem 1997;246:581–601.
tional activation of cyclin E and A genes by E2F factors;
6. La Thangue NB. E2F and the molecular mechanisms of early cell-cycle
E2F-mediated activation of genes encoding DNA replication
control. Biochem Soc Trans 1996;24:54–59.
components is required for the onset of S phase; concomitant 7. Nevins JR. Toward an understanding of the functional complexity of the
with this, transcriptional activation of the RanBP1 genes E2F and retinoblastoma families. Cell Growth Differ 1998;9:585–593.
modifies the activity of the Ran network during S phase, which 8. Morkel M, Wenkel J, Bannister AJ, Kouzarides T, Hagemeier C. An
in turn controls molecules involved in subsequent progression E2F-like repressor of transcription. Nature 1997;390:567–568.
through the G2 and M phases; the Ran network also controls 9. Trimarchi JM, Fairchild B, Verona R, Moberg K, Andon N, Lees JA. E2F-6,
nuclear transport, whose regulated activity is important for at a member of the E2F family that can behave as a transcriptional repressor.
least certain checkpoint systems. Checkpoint mechanisms Proc Natl Acad Sci USA 1998;95:2850–2855.
monitor completion of all processes (double arrowheads), or 10. Moberg K, Starz MA, Lees JA. E2F-4 switches p130 and p107 and pRb in
response to cell cycle reentry. Mol Cell Biol 1996;16:1436–1449.
trigger apoptosis when defective processes are detected.
11. Beijersbergen R, Kerkhoven R, Zhu L, Carlée L, Voorhoeve PM, Bernards
Apoptosis can also be triggered as a direct response to E2F-1.
R. E2F-4, a new member of the E2F gene family, has oncogenic activity
and associates with p107 in vivo. Genes Dev 1994;8:2680–2690.
12. Vairo G, Livingston D, Ginsberg D. Functional interaction between E2F4
and p130: evidence for distinct mechanisms underlying growth suppres-
served when the RanBP1 gene is expressed throughout the sion by different retinoblastoma protein family member. Genes Dev
1995;9:869–881.
cell cycle.(69) It is tempting to speculate that the defects 13. Hijmans EM, Voorhoeve PM, Beijersbergen RL, van’t Veer LJ, Bernards R.
observed in this cell line arise at least in part in consequence E2F-5, a new E2F family member that interacts with p130 in vivo. Mol Cell
of disruption of cell-cycle regulated RanBP1 expression. Biol 1995;15:3082–3089.
14. Ikeda M, Jakoi L, Nevins J. A unique role for the Rb protein in controlling
E2F accumulation during cell growth and differentiation. Proc Natl Acad
Sci USA 1996;93:3215–3220.
Conclusions 15. Lindeman GJ, Gaubatz S, Livingston DM, Ginsberg D. The subcellular
Faulty checkpoint controls have been observed in certain localization of E2F-4 is cell-cycle dependent. Proc Natl Acad Sci USA
1997;94:5095–5100.
hereditary cancer syndromes and at early stages of cell
16. Muller H, Moroni MC, Vigo E, Petersen B, Bartek J, Helin K. Induction of
transformation, which suggests that structural mutations, S-phase entry by E2F transcription factors depends on their nuclear
regulatory mutations, or both, in checkpoint genes might localization. Mol Cell Biol 1997;17:5508–5520.
17. Verona R, Moberg K, Estes S, Starz M, Vernon JP, Lees JA. E2F activity is
contribute to the genetic instability associated with neoplastic
regulated by cell cycle-dependent changes in subcellular localization.
growth. Genes that are regulatory targets of E2F/DP transcrip- Mol Cell Biol 1997;17:7268–7282.
tion factors are found in major cell-cycle regulatory pathways, 18. De la Luna S, Burden MJ, Lee CW, La Thangue NB. Nuclear accumula-
tion of the E2F heterodimer regulated by subunit composition and
as schematically diagrammed in Figure 3. alternative splicing of a nuclear localization signal. J Cell Sci 1996;109:
Available data suggest that there is cross-talk between 2443–2452.
E2F/DP factors and other regulatory genes, whose products 19. Allen KE, de la Luna S, Kerkhoven RM, Bernards R, La Thangue NB.
Distinct mechanisms of nuclear accumulation regulate the functional
are themselves implicated in cell-cycle control, and regulated consequence of E2F transcription factors. J Cell Sci 1997;110:2819–
transcription of particular genes by E2F factors may be 2831.
viewed as an additional level of integrated control which 20. Puri PL, Cimino L, Fulco M, Zimmerman C, La Thangue NB, Giordano A,
Graessmann A, Levrero M. Regulation of E2F4 mitogenic activity during
connects the transcriptional apparatus to control of cell terminal differentiation by its heterodimerization partners for nuclear
proliferation. translocation. Cancer Res 1998;58:1325–1331.

228 BioEssays 21.3


Review articles

21. Schulze A, Zerfass-Thome K, Mannhardt B, Zwerschke W, Jansen-Dürr P. 43. Beijersbergen RL, Bernards R. Cell cycle regulation by the retinoblastoma
Anchorage-independent transcription of the cyclin A gene induced by the family of growth inhibitory proteins. Biochim Biophys Acta - Reviews on
E7 oncoprotein of human papillomavirus type 16. J Virol 1998;72:2323– Cancer 1996;1287:103–120.
2334. 44. Schulze A, Zerfa␤ K, Spitkovsky D, Henglein B, Jansen-Dürr P. Activation
22. Hofmann F, Martelli F, Livingston DM, Wang Z. The retinoblastoma gene of the E2F transcription factor by cyclin D1 is blocked by p16INK4, the
product protects E2F-1 from degradation by the ubiquitin-proteasome product of the putative tumor suppressor gene MTS1. Oncogene 1994;9:
pathway. Genes Dev 1996;10:2949–2959. 3475–3482.
23. Sherr CJ. Cancer cell cycles. Science 1996;274:1672–1677. 45. Ohtani K, Degregori J, Nevins JR. Regulation of the cyclin E gene
24. Yamasaki L, Jacks T, Bronson R, Goillot E, Harlow E, Dyson NJ. Tumor by transcription factor E2F1. Proc Natl Acad Sci USA 1995;92:12146–
induction and tissue atrophy in mice lacking E2F-1. Cell 1996;85:537–547. 12150.
25. Johnson DG, Schwarz JK, Cress WD, Nevins JR. Expression of transcrip- 46. Zerfa␤-Thome K, Schulze A, Zwerschke W, Vogt B, Helin K, Bartek J,
tion factor E2F1 induces quiescent cells to enter S-phase. Nature Henglein B, Jansen-Dürr P. p27KIP1 blocks cyclin E-dependent trans-
1993;365:349–352.
activation of cyclin A gene expression. Mol Cell Biol 1997;17:407–415.
26. Lukas J, Petersen BO, Holm K, Bartek J, Helin K. Deregulated expression
47. Beijersbergen RL, Carlee L, Kerkhoven RM, Bernards R. Regulation of the
of E2F family members induces S-phase entry and overcomes p16(INK4A)-
retinoblastoma protein-related p107 by G(1) cyclin complexes. Genes
mediated growth suppression. Mol Cell Biol 1996;16:1047–1057.
Dev 1995;9:1340–1353.
27. Leone G, DeGregori J, Yan Z, Jajoi L, Ishida S, Williams RS, Nevins JR.
48. Feng XH, Filvaroff EH, Derynck R. Transforming growth factor-beta
E2F3 activity is regulated during the cell cycle and is required for the
(TGF-beta)-induced down-regulation of cyclin A expression requires a
induction of S phase. Genes Dev 1998;12:2120–2130.
functional TGF-beta receptor complex. Characterization of chimeric and
28. Yan Z, DeGregori J, Shohet R, Leone G, Stillman B, Nevins JR, Williams
truncated type I and type II receptors J Biol Chem 1995;270:24237–
RS. Cdc6 is regulated by E2F and is essential for DNA replication in
24245.
mammalian cells. Proc Natl Acad Sci USA 1998;95:3603–3608.
49. Guadagno T, Ohtsubo M, Roberts JM, Assoian R. A Link between Cyclin A
29. Qin XQ, Livingston DM, Kaelin WG, Adams PD. Deregulated transcription
expression and adhesion-dependent cell cycle progression. Science
factor E2F-1 expression leads to S-phase entry and p53-mediated
apoptosis. Proc Natl Acad Sci USA 1994;91:10918–10922. 1993;262:1572–1575.
30. Krek W, Xu GF, Livingston DM. Cyclin A-kinase regulation of E2F-1 DNA 50. Schulze A, Zerfass-Thome K, Berges J, Middendorp S, Jansen-Dürr P,
binding function underlies suppression of an S phase checkpoint. Cell Henglein B. Anchorage-dependent transcription of the cyclin A gene. Mol
1995;83:1149–1158. Cell Biol 1996;16:4632–4638.
31. DeGregori J, Leone G, Miron A, Jakoi L, Nevins JR. Distinct roles for E2F 51. Lucibello FC, Sewing A, Brusselbach S, Burger, C, Muller R. Deregulation
proteins in cell growth control and apoptosis. Proc Natl Acad Sci USA of Cyclin-D1 and Cyclin-E and suppression of cdk2 and cdk4 in senes-
1997;94:7245–7250. cent human fibroblasts.. J Cell Sci 1993;105:123–133.
32. Hsieh JK, Fredersdorf S, Kouzarides T, Martin K, Lu X. E2F1-induced 52. Spitkovsky D, Schulze A, Boye B, Jansen-Dürr P. Downregulation of cyclin
apoptosis requires DNA binding but not transactivation and is inhibited by A gene expression upon genotoxic stress correlates with reduced binding
the retinoblastoma protein through direct interaction. Genes Dev 1997;11: of free E2F to the promoter. Cell Growth Differ 1997;8:699–710.
1840–1852. 53. Shiyanov P, Bagchi S, Adami G, Kokontis J, Hay N, Arroyo M, Morozov A,
33. DeGregori J, Kowalik T, Nevins JR. Cellular targets for activation by the Raychaudhuri P. p21 disrupts the interaction between cdk2 and the
E2F1 transcription factor include DNA synthesis- and G(1)/S-regulatory E2F-p130 complex. Mol Cell Biol 1996;16:737–744.
genes. Mol Cell Biol 1995;15:4215–4224. 54. Jansen-Dürr P. How viral oncogenes make the cell cycle trends. Genetics
34. Tommasi S, Pfeifer GP. In vivo structure of the human cdc2 promoter: 1996;12:270–275.
release of a p130-E2F-4 complex from sequences immediately upstream 55. Krek W, Ewen ME, Shirodkar S, Arany Z, Kaelin WG, Livingston DM.
of the transcription initiation site coincides with induction of cdc2 expres- Negative regulation of the growth-promoting transcription factor E2F-1
sion. Mol Cell Biol 1995;15:6901–6913. by a stably bound cyclin A-dependent protein kinase. Cell 1994;78:161–
35. Wells J, Held P, Illenye S, Heintz NH. Protein-DNA interactions at the major 172.
and minor promoters of the divergently dhfr and rep3 genes during the 56. Cartwright P, Muller H, Wagener C, Holm K, Helin K. E2F-6: a novel
Chinese hamster ovary cell cycle. Mol Cell Biol 1996;16:634–637. member of the E2F family is an inhibitor of E2F-dependent transcription.
36. Tommasi S, Pfeifer GP. Constitutive protection of E2F recognition se- Oncogene 1998;17:611–623.
quences in the human thymidine kinase promoter during cell cycle 57. Gaubatz S, Wood JG, Livingston DM. Unusual proliferation arrest and
progression. J Biol Chem 1997;272:30483–30490.
transcriptional control properties of a newly discovered E2F family
37. Schulze A, Zerfa␤ K, Spitkovsky D, Berges J, Middendorp S, Jansen-Dürr
member, E2F-6. Proc Natl Acad Sci USA 1998;95:9190–9195.
P, Henglein B. Cell cycle regulation of cyclin A gene transcription is
58. Sazer S. The search for the primary function of the Ran GTPase continues.
mediated by a variant E2F binding site. Proc Natl Acad Sci USA
Trends Cell Biol 1996;6:81–85.
1995;92:11264–11268.
59. Rush MG, Drivas G, D’Eustachio P. The small nuclear GTPase Ran: how
38. Botz J, Zerfass-Thome K, Spitkovsky D, Delius H, Vogt B, Eilers M,
much does it run? Bioessays 1996;18:103–112.
Hatzigeorgiou A, Jansen-Dürr P. Cell cycle regulation of the murine cyclin
60. Dasso M. RCC1 in the cell cycle: the regulator of chromosome condensa-
E gene depends on an E2F binding site in the promoter. Mol Cell Biol
tion takes on new roles. Trends Biochem Sci 1993;18:96–101.
1996;16:3401–3409.
39. Liu N, Lucibello FC, Körner K, Wolfraim LA, Zwicker J, Müller R. CDF-1, a 61. Ren M, Coutavas E, D’Eustachio P, Rush MG. Effects of mutant Ran/TC4
novel E2F-unrelated factor, interacts with cell cycle- regulated repressor proteins on cell cycle progression. Mol Cell Biol 1994;14:4216–4224.
elements in multiple promoters. Nucleic Acids Res 1997;25:4915–4920. 62. Goerlich D. Transport in and out of the cell nucleus. EMBO J 1998;17:2721–
40. Lucibello FC, Liu N, Zwicker J, Gross C, Müller R. The differential binding 2727.
of E2F and CDF repressor complexes contributes to the timing of 63. Saha P, Chen J, Thome KC, Lawlis SJ, Hou ZH, Hendricks M, Parvin JD,
cell cycle-regulated transcription. Nucleic Acids Res 1997;25:4921– Dutta A. Human CDC6/Cdc18 associates with Orc1 and cyclin-cdk and is
4925. selectively eliminated from the nucleus at the onset of S phase. Mol Cell
41. Hurford RK, Cobrinik D, Lee MH, Dyson N. pRB and p107/p130 are Biol 1998;18:2758–2767.
required for the regulated expression of different sets of E2F responsive 64. Nishijima H, Nishitani H, Seki T, Nishimoto T. A dual-specificity phospha-
genes. Genes Dev 1997;11:1447–1463. tase Cdc25B is an unstable protein and triggers p34(cdc2)/cyclin B
42. Morgan D. Cyclin-dependent kinases: engines, clocks, and microproces- activation in hamster BHK21 cells arrested with hydroxyurea. J Cell Biol
sors. Annu Rev Cell Dev Biol 1997;13:261–291. 1997;138:1105–1116.

BioEssays 21.3 229


Review articles

65. Di Matteo G, Fuschi P, Zerfass K, Moretti S, Ricordy R, Cenciarelli C, RanGTPase and inhibition of GTP dissociation by Ran-GTP binding
Tripodi M, Jansendurr P, Lavia P. Transcriptional control of the Htf9-A/ protein RanBP1. EMBO J 1995;14:705–715.
RanBP1 gene during the cell cycle. Cell Growth Differ 1995;6:1213– 69. Battistoni A, Guarguaglini G, Degrassi F, Pittoggi C, Palena A, Di Matteo
1224. G, Pisano C, Cundari E, Lavia P. Deregulated expression of the RanBP1
66. Stapleton G, Somma MP, Lavia P. Cell type specific interactions of gene alters cell cycle progression in murine fibroblasts. J Cell Sci
transcription factors with a housekeeping promoter in vivo. Nucleic Acids 1997;110:2345–2357.
Res 1993;21:2465–2473. 70. Qin XQ, Barsoum J. Differential cell cycle effects induced by E2F1
67. Guarguaglini G, Battistoni A, Pittoggi C, Di Matteo G, Di Fiore B, Lavia P. mutants. Oncogene 1997;14:53–62
Expression of the murine RanBP1 and Htf9-c genes is regulated from a 71. De Fiore B, Guarguaglini G, Kerkhoven RM, Bernards R, Lavia P. Two
shared bidirectional promoter during cell cycle progression. Biochem J E2F-binding sites independently control growth-regulated and cell-cycle
1997;325:277–286. regulated transcription of the Htfga/RanBP1 gene. J Biol Chem 1999;274:
68. Bischoff RF, Krebber H, Smirnova E, Dong W, Ponstingl H. Co-activation of in press

230 BioEssays 21.3

Vous aimerez peut-être aussi