You are on page 1of 21

Chapter 15

The Cell Cycle


Vincent W. Yang

The cell cycle is a process in which a series of events In addition to the four phases of the cell cycle listed
occur in a cell that eventually leads to the reproduc- earlier, one phase that lies outside the cell cycle is called
tion of two daughter cells. By traversing through the cell the G0 (0 for zero) phase (Figure 15.1). Cells in this phase
cycle, a cell accomplishes two major tasks: duplication are in the resting phase, which is often the result of their
of its genome and division into two cells. The cell cycle leaving the G1 phase of the cell cycle. Typically, a cell
is subjected to regulation by environmental cues, such as enters the G0 phase if the environment is not conducive for
the availability of nutrients (growth factors or mitogens), the progression of the cell cycle, as in the event of depri-
and can be halted if damages to the DNA are detected to vation of essential nutrients or growth factors, or if a cell
allow sufficient time for repairing the damaged DNA. has reached a fully differentiated state such as a hepato-
Such regulation is critically important in a tissue such as cyte or neuron. In these conditions, the cell is sometimes
the gastrointestinal tract where epithelial cells undergo referred to as being in a quiescent state. Additionally, a cell
multiple rounds of cell division that are carefully orches- can enter the G0 phase and become senescent due to DNA
trated to achieve tissue homeostasis. In contrast, many dis- damage or telomere attrition. This is often an alternative to
ease processes, notably cancers, are often the consequence self-destruction of the damaged cell by apoptosis.
of dysregulated cell cycle due to mutations in key mol-
ecules that control the cell cycle. Understanding how the
15.1.1  M Phase
cell cycle is regulated is therefore essential to understand-
ing the normal and abnormal physiology of the gastroin- The M phase consists of mitosis and cytokinesis. Mitosis
testinal tract. is the process in which DNA condenses into visible chro-
mosomes, which is followed by the separation of the
chromosomes into two identical sets. Cytokinesis can
15.1  COMPONENTS OF THE CELL CYCLE be considered as the last phase of mitosis when the two
A typical eukaryotic cell cycle contains several distinct daughter cells separate, each with a nucleus and cytoplas-
phases that progress in an orderly fashion — a phase can- mic organelles. Mitosis begins with nuclear membrane
not commence without completion of the previous phase. breakdown followed by condensation of the chromo-
The four phases of the cell cycle are G1 (G for gap), S somes and separation of the centrosomes (prophase). This
(synthesis), G2, and M (mitosis) (Figure 15.1). The G1, is accompanied by the formation of mitotic spindles,
S, and G2 phases collectively make up the interphase. which are attached on one end, the centrosomes, and at
The DNA content of a cell in the G1 phase is 2N (N is the the other end, the kinetochore, a protein structure located
number of chromosomes), also known as diploid, whereas at or near the centromeres of mitotic chromosomes (pro-
the DNA content of a cell in the G2 phase is 4N (tetra- metaphase). At this point, kinetochores that are unat-
ploid). The DNA content of a cell in the S phase varies tached to the mitotic spindles generate a “wait” signal that
between 2N and 4N, depending on the stage of replication delays the onset of anaphase until all chromosomes are
of the cell. The M phase is in turn comprised of two proc- properly attached and aligned. This signal is also called
esses: mitosis, in which the cell’s chromosomes are equally mitotic checkpoint or spindle assembly checkpoint (SAC;
divided between the two daughter cells, and cytokinesis (or see Section 15.3.4). Following prometaphase, chromo-
cell division), in which the cytoplasm of the cell divides somes congregate at the equatorial plate (metaphase)
in half to form two distinct daughter cells. Typically, the before separating to the opposite poles (anaphase). This
amount of time required for a single cell cycle in actively is followed by the formation of new nuclear membranes
proliferating human cells in culture is 24 hours. Of these, around the daughter nuclei and uncoiling of the chromo-
the M phase takes approximately one hour to complete and somes (telophase), and finally into the formation of a
interphase takes up the remaining 23 hours. cleavage furrow that leads to the formation of two daughter

Physiology of the Gastrointestinal Tract, Two Volume Set. DOI: 10.1016/B978-0-12-382026-6.00015-4


© 2012 Elsevier Inc. All rights reserved. 451
452 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

FIGURE 15.1  The phases of the cell cycle.  The cell cycle begins in the G1 phase of a diploid cell (DNA content  2N; N is the number of chro-
mosomes). After DNA replication is completed in the S phase, the cell enters the G2 phase and has twice the amount of the DNA (4N) of the starting
cell. This is followed by mitosis (M) and cell division, which leads to the formation of two diploid daughter cells. Cells in either mitosis or cell divi-
sion (also called cytokinesis) are in the M phase, whereas those in the other three phases (G1, S, and G2) are in the interphase. The time in which a cell
spends in each phase varies among the cell type and is not drawn to scale.

216
FIGURE 15.2  The stages of mitosis.  The different phases of mitosis are illustrated. SAC, spindle assembly checkpoint. (Modified from with
permission.)

cells (cytokinesis). Figure 15.2 illustrates the various regulatory molecules, cyclins and cyclin-dependent kinases
stages of mitosis. (CDKs).6–18 Cyclins and CDKs form heterodimers with the
former serving as the regulatory subunits and the latter cat-
alytic subunits; CDKs are inactive in the absence of their
15.2  CONTROL OF THE CELL CYCLE cyclin partners. Specific pairs of cyclin/CDK dimers func-
The organization of the cell cycle and its control sys- tion in specific phases of the cell cycle (Figure 15.3). For
tem are highly conserved among eukaryotic organisms. example, the cyclin E-CDK2 complex becomes active late
Abundant information on how the cell cycle is regulated in the G1 phase of the cell cycle and is responsible for the
in vertebrates was derived from earlier studies in yeast.1–5 transition from G1 into S phase. When activated by a bound
These studies demonstrated that both cell cycle progres- cyclin, CDKs phosphorylate a series of downstream target
sion and cell division are driven by the sequential activa- proteins, either activating or inactivating them, which then
tion, and subsequent inactivation, of two key classes of orchestrate the coordinated entry into the next phase of
Chapter  |  15  The Cell Cycle 453

FIGURE 15.3  The cell cycle is driven by cyclin/cyclin-dependent kinase (CDK) complexes.  The various pairs of cyclin/CDK complexes and
their respective positions in the cell cycle are shown.

FIGURE 15.4  Relative expression levels of cyclins in the cell cycle.  The relative protein levels of the four different cyclins in various phases of the
cell cycle are demonstrated. The letters in parentheses indicate the classes of cyclins. The levels of cyclin D often fluctuate, depending on whether or
not growth factors or mitogens are present. (R) indicates the restriction point.

the cell cycle. The identities of the target proteins depend


15.2.1  Cyclins
on the particular combination of cyclin-CDK complexes. Cyclins were named this because they undergo a cycle of
While CDKs are constitutively expressed throughout the synthesis and degradation in each cell cycle. Depending
cell cycle, cyclins are synthesized (and destroyed) in spe- on the timing of their expression and functions in the cell
cific stages of the cell cycle (hence the name cyclin), which cycle, cyclins are divided into four classes. Three of these
is often dependent upon various signaling molecules. This classes, G1/S cyclins, S cyclins, and M cyclins, are directly
cyclic nature of cyclin expression ensures that CDKs are involved in the control of cell cycle events. The fourth
activated and inactivated in a precise manner, safeguarding class, the G1 cyclins, controls the entry into the cell cycle
the orderly progression of the cell cycle (Figure 15.4). in response to extracellular growth factors or mitogens. In
454 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

the G1 phase, growth factors are necessary to initiate and interacting with E2F, RB also recruits chromatin remod-
maintain the proper transition to the S phase. Early in the eling enzymes such as histone deacetylases (HDACs)43
G1 phase, growth factors stimulate the synthesis of G1 cyc- that often serve as transcription co-repressors.44–47 Thus,
lins, represented by the cyclin D family of cyclins, which the binding of RB to E2F does not just simply inhibit E2F
activates CDK4/6 to induce synthesis of downstream activity. The RB–E2F complex binds to promoters and
targets, one of which is cyclin E.19 The rise in cyclin E actively represses transcription by blocking activity of the
(a G1/S cyclin) levels and activity of its partner, CDK2, surrounding enhancers on the promoter.48–51
drive the cell past a restriction point (R in Figure 15.4) in The activity of RB is governed by phosphorylation
the cell cycle after which the cell is committed to proceed- catalyzed by CDKs.52–54 RB contains 16 potential phos-
ing to DNA synthesis, even if the growth factors are with- phorylation sites by CDKs and oscillates between hypo-
drawn. Subsequently, the S cyclins (represented by cyclin phosphorylated and hyperphosphorylated forms during the
A) and M cyclins (represented by cyclin B), are required cell cycle.34 The form that inhibits E2F is the hypophos-
for the initiation of DNA replication and entry into mitosis, phorylated form. CDKs, in complex with their cyclin part-
respectively (Figure 15.4). ners, phosphorylate the hypophosphorylated form of RB,
leading to the hyperphosphorylated form. At least three dif-
ferent cyclin–CDK complexes are known to phosphorylate
15.2.1.1  G1 Cyclins
RB during the cell cycle — cyclin D-CDK4/6 acts early in
The G1 cyclins are composed of the D-type cyclins that G1, cyclin E-CDK2 in late G1, and cyclin A-CDK2 in the
include cyclins D1, D2, and D3.9,20,21 Along with their S phase.22 In this way, RB becomes sequentially phospho-
partners, CDK4 and CDK6, the G1 cyclins act early in the rylated in the cell cycle. It has been shown that successive
G1 phase of the cell cycle. The levels of G1 cyclin are low phosphorylations of RB by cyclin D-CDK4/6 and cyclin
in the G0 phase and increase progressively upon addition E-CDK2 are necessary for the complete hyperphosphor-
of growth factors or mitogens to the cells.22,23 The mecha- ylation of RB.53 The hyperphosphorylation of RB prevents
nisms by which mitogens or growth factors activate cyc- its binding to E2F, thus releasing E2F from transcriptional
lin D1 are complex and occur at both transcriptional and repression.19,34 Once liberated from RB, E2F, along with its
post-transcriptional levels.19 At the transcriptional level, partner transcription factor DP,55–60 activate transcription of
induction of cyclin D1 by growth factors is dependent genes that are involved in nucleotide metabolism and DNA
on the Ras/Raf/mitogen-activated kinase kinase (MEK)/ synthesis, thus allowing entry into the S phase.35,37,61–68 It
extracellular signal-regulated kinase (ERK) signaling path- is of interest to note that one of the target genes of an active
way.24–27 Once synthesized, cyclin D1 protein has a short E2F encodes cyclin E.69–71 It was also shown that phospho-
half-life28 — its turnover is governed by ubiquitination rylation of RB by cyclin D-CDK4/6 and cyclin E-CDK2
and proteasomal degradation, which in turn are depend- triggers sequential intramolecular interactions in RB that
ent on phosphorylation of cyclin D1 by glycogen synthase progressively block RB functions as cells move through
kinase-3β (GSK-3β).29,30 Growth factors prevent cyclin D1 G1.69 The complete hyperphosphorylation of RB by cyclin
degradation by inhibiting GSK-3β-dependent phosphoryla- D-CDK4/6 and cyclin E-CDK2 coincides with the R point
tion of cyclin D1 through the Ras/phosphatidylinositol- (Figure 15.4), beyond which the cell is committed to com-
3-OH kinase (PI3K)/AKT pathway.30 It was subsequently pletion of the cell cycle.19
demonstrated that growth-factor-induced cyclin D1 gene
transcription by the MEK/ERK pathway and cyclin D1
15.2.1.2  G1/S Cyclins
protein stabilization by the PI3K/AKT pathway need to
occur in a sequential manner with the former occurring Cyclins E1 and E2 (collectively considered as cyclin E)
early and the latter occurring late in the G1 phase in order are the G1/S cyclins.9 Transcription of the cyclin E gene
to drive the progression of the cell cycle.31 is regulated by E2F,32 which, as described earlier, is acti-
One of the key targets of an activated cyclin D-CDK4/6 vated due to cyclin D-CDK4/6-stimulated phosphorylation
complex is the retinoblastoma (RB) protein.32–34 RB is one of RB. The amounts of cyclin E protein and its associated
of three “pocket protein” families of cell cycle regulator kinase (CDK2) activity are maximal in late G1 and early
proteins — the other two are p107 and p130 — and has S phases (Figure 15.4).72,73 Cyclin E-CDK2 completes RB
a major role in restraining the transition between G1 and phosphorylation in the G1 phase and the transition from
S phases of the cell cycle.35 In the absence of mitogenic cyclin D-CDK4/6 to cyclin E-CDK2 accounts for the loss
stimuli, RB interacts with and inhibits the activity of the of dependency on growth factors. Cyclin E-CDK2 phos-
transcription factor E2F.36–38 As E2F binding sites are phorylates RB on different sites from cyclin D-CDK4/6,
present in the promoters of many genes required for cell and these kinases may differentially impact the interac-
cycle progression,39–42 the inhibition of E2F by RB pre- tion between RB and E2Fs, HDACs, and other chromatin
vents entry into the cell cycle. In addition to physically remodeling proteins.34 In contrast to cyclin D-CDK4/6, the
Chapter  |  15  The Cell Cycle 455

functions of cyclin E-CDK2 are not limited to G1 control. achieved by the degradation of B-type cyclins by ubiqui-
Thus, cyclin E-CDK2 phosphorylates other substrates that tin-mediated proteolysis that is regulated by the anaphase-
are more directly involved in the control of DNA replica- promoting complex/cyclosome (APC/C).91–95
tion, centrosome duplication, replication origin licensing,
and firing.74 The timing of cyclin E-CDK2 activation and 15.2.2  Cyclin-dependent Kinases
its broader range of substrates suggest that cyclin E-CDK2
spans the interface between G1 regulation and core cell As described earlier, CDKs are the catalytic subunits of a
cycle machinery during S phase. relatively large family of serine/threonine protein kinases
In early S phase, cyclin E-CDK2 activity abruptly with a primary role in cell cycle progression. The mamma-
ceases as a consequence of cyclin E degradation (Figure lian genome contains 11 genes encoding CDKs and 9 others
15.4). This is mediated by phosphorylation by both encoding CDK-like proteins with conserved structure.8 The
GSK-3β and CDK2, which target cyclin E for ubiquitina- prototype CDK, CDK1, was first identified in yeasts and
tion by the SCFFbw7 E3 ligase, leading it to proteasomal designated as Cdc2 in Saccharomyces pombe or Cdc28 in
degradation.75–77 Like cyclin D1,30 the involvement of S. cerevisiae.96,97 The mammalian homolog of yeast Cdc2,
GSK-3β, an enzyme that is inhibited by the PI3K/AKT sig- CDK1, was subsequently identified due to its ability to com-
naling pathway, in regulating stability of cyclin E suggests plement the yeast mutants.98,99 Other mammalian CDKs
that cyclin E can be influenced by at least one mitogen- were then identified by a host of techniques including com-
dependent signaling cascade. plementation, homology probing, differential display, and
PCR amplification with degenerate primers.100–106 Again,
as described earlier, progression through the G1 phase of the
15.2.1.3  S Cyclins cell cycle requires at least three CDKs — CDK4, CDK6,
The S cyclins include both cyclins A1 and A2. Whereas and CDK2 — which, along with their regulatory cyclins,
cyclin A1 is restricted to the germ cell lineages, cyclin A2 target the RB family of proteins that include RB, p107, and
is ubiquitously expressed in all cell types.78 Low levels of p130.35,51,69 CDK2 is also important for S phase progression
cyclin A2 are first detected at the G1/S boundary. The lev- and CDK1 for G2/M transition and mitosis.
els then rise steadily as cells begin to replicate their DNA The kinase activities of CDKs are regulated at multiple
and do not decline until cyclin A is degraded in late G2 levels, including interaction with their regulatory subunits
(Figure 15.4). In S phase, cyclin A and its partner, CDK2, (cyclins), binding to negative regulators called CDK inhib-
phosphorylate substrates that commence DNA replica- itors (CKIs; see Section 15.2.4), phosphorylation-dephos-
tion from preformed replication initiation complexes.79–81 phorylation, folding, and subcellular localization.23,107
Cyclin A-CDK2 is also required to coordinate the end of S Among these, phosphorylation-dephosphorylation can
phase with activation of the mitotic cyclin-CDKs.82 have activating or inhibitory effects on CDK activity. For
example, active cyclin-CDK complexes need to be phos-
phorylated in the T-loop of the CDK subunit by CDK-
15.2.1.4  M Cyclins activating kinase (CAK), which contains a complex of
During G2, A-type cyclins (the S cyclins) are degraded by CDK7, cyclin H, and MAT1 (Figure 15.5).108–110 In con-
ubiquitin-mediated proteolysis, whereas B-type cyclins trast, cyclin-CDK complexes can be negatively regulated
(the M cyclins) are actively synthesized (Figure 15.4). As a by phosphorylation in adjacent threonine and tyrosine
consequence, CDK1 (also known as Cdc2) binds to B-type residues of the CDK subunit by the dual-specificity pro-
cyclins — an association required for the commencement tein kinases WEE1 and MYT1.111–117 Conversely, these
of mitosis. CDK1 preferentially binds to two main B-type inhibitory phosphorylations can be reversed by the ability
cyclins, cyclins B1 and B2. In contrast, the third iso- of the dual-specificity CDC25 phosphatases (CDC25A,
form, cyclin B3, may have a function in the meiotic cell CDC25B, and CDC25C) to dephosphorylate the same
cycle.83 Cyclin B-CDK1 regulates events during both the threonine and tyrosine residues and thus act as positive
G2/M transition and progression through mitosis.84 This is regulators of cyclin-CDK activity (Figure 15.5).118–120 If
accomplished by the phosphorylation of over 70 proteins both activating and inactivating phosphorylations exist in
by the cyclin B-CDK1 complexes,8 although the number the same molecule, and they result in an inactive kinase.
of substrates could be much larger.85 Phosphorylation
of target proteins leads to numerous events that include 15.2.3  In Vivo Functions of Cyclins and
separation of centrosomes,86 condensation of chromo-
somes,87,88 breakdown of the nuclear lamina,89 and disas-
CDKs as Revealed by Gene Knockout in Mice
sembly of the Golgi apparatus,90 among others. Finally, Numerous studies have investigated the in vivo functions of
inactivation of the cyclin B-CDK1 complexes is required cyclins and CDKs in knockout mice with the genes deleted
for proper exit from mitosis and this inactivation is either singly or in combination. Because it is not the intent
456 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

first class includes the inhibitors of CDK4 (INK4) proteins.


Four such INK4 proteins have been identified: p16INK4a
(also known as CDK inhibitor 2A or CDKN2A),125 p15INK4b
(CDKN2B),126 p18INK4c (CDKN2C),127,128 and p19INK4d
(CDKN2D).128,129 INK4 proteins specifically bind to and
inhibit monomeric CDK4 and CDK6 proteins.130 The sec-
ond class of CKIs includes the CDK-interacting protein/
CDK-interacting protein (Cip/Kip) family of proteins,
which are more broadly acting than the INK4 family of
proteins and do so by binding to cyclin-CDK complexes.131
There are three members of the Cip/Kip family of CKIs:
p21Cip1 (also called CDK inhibitor 1A or CDKN1A),132–137
p27Kip1 (CDKN1B),138–140 and p57Kip2 (CDKN1C).141,142
Cip and Kip inhibitors block CDK activity by forming inac-
tive trimeric complexes (cyclin E-CDK2, cyclin A-CDK2,
cyclin B-CDK1, and possibly cyclin D-CDK4 and cyclin
D-CDK6),143–148 thus exerting a much broader effect on the
progression of the cell cycle. Figure 15.5 summarizes the
multiple regulatory mechanisms by which CDK activities
are regulated.

15.3  CHECKPOINTS
FIGURE 15.5  The regulatory mechanisms of cell cycle
CDKs.  CDKs require binding to their cyclin partners for activation of The cell cycle contains several checkpoints to monitor and
kinase activity. The INK4 proteins inhibit CDK activity by directly bind- regulate its progression.149–165 Checkpoints are positioned
ing to monomeric CDK4 or CDK6. In contrast, Cip and Kip inhibitors at specific locations in the cell cycle to allow verification
inactivate CDKs by binding to cyclin-CDK complexes. Cyclin-CDK of phase processes and repair of DNA damage. A cell can-
complexes are activated by phosphorylation of the CDK subunit by
not proceed from one phase to the next without satisfying
cyclin-dependent activating kinase (CAK) that contains three subunits:
CDK7, cyclin H (cycH), and MAT1. In contrast, cyclin-CDK com- all of the checkpoint requirements. An important function
plexes are inhibited by phosphorylation in adjacent threonine and tyro- of many of the cell cycle checkpoints is to assess DNA
sine residues by the dual-specificity protein kinases, WEE1 and MYT1. damage. Upon detection of DNA damage, the checkpoint
These inhibitory phosphorylations can be reversed by the dual-specificity initiates a signal cascade to either arrest the cell cycle until
CDC25 phosphatases that dephosphorylate the CDKs at the same amino
repairs are properly made, or if repairs are not possible, to
acid residues. Cyc, cyclin. (From 8 with permission.)
target the cell for destruction via apoptosis as a means to
maintain genomic integrity. In the cell cycle, there are three
of this chapter to review all of the phenotypes of the knock- specific checkpoints for damaged or incompletely repli-
out mice, the readers are referred to the many excellent cated DNA: G1/S, G2/M, and intra-S checkpoints. These
recent review articles that summarize the findings of these checkpoints are patrolled by some of the CKIs described
studies.8–10,121–123 From these studies, it becomes clear that earlier. Another important and specific checkpoint occurs
most of the cyclins and CDKs, although previously con- in mitosis, the so-called mitotic checkpoint or SAC. This
sidered essential for cell proliferation, have turned out to checkpoint is designed to monitor proper alignment of
be dispensable. Several compensatory mechanisms were the chromosomes during mitosis. Anaphase cannot pro-
uncovered among cyclins and CDKs. The particularly unex- ceed unless this checkpoint is satisfied. The locations of
pected finding is that CDK2, thought to be a master regula- the various checkpoints in the cell cycle are illustrated in
tor of the cell cycle, is dispensable for the regulation of the Figure 15.6. The various cell cycle checkpoint pathways in
cell cycle with both CDK4 and CDK1 covering CDK2’s response to DNA damage are summarized in Figure 15.7.
functions. In fact, CDK1 alone is able to drive the mamma-
lian cell cycle,124 indicating that the regulation of the mam-
malian cell cycle is highly conserved. 15.3.1  G1/S Checkpoint
The G1/S checkpoint (also called the G1 checkpoint)
is located near the end of the G1 phase, just before the
15.2.4  CDK Inhibitors entry into S phase (Figure 15.6). In mammalian cells, the
CKIs are proteins that constrain the activities of CDKs. G1 checkpoint is the restriction or R point (Figure 15.4).
Two classes of CDK inhibitors have been described.23 The This is a point where cells typically arrest the cell cycle
Chapter  |  15  The Cell Cycle 457

if environmental conditions are unfavorable for cell divi- Here CDK4 is redistributed from cyclin D-CDK4 com-
sion, such as the presence of DNA damage or lack of plexes to INK4-CDK4 complexes, and unbound D-type
growth factors. The G1 checkpoint is controlled by both cyclin is rapidly degraded by the ubiquitination-mediated
the INK4 and Cip/Kip families of CKIs. INK4 proteins proteasomal pathway.29 Also, in early G1 phase, the cyc-
specifically bind to CDK4 and CDK6 and inhibit their lin E-CDK2 and cyclin A-CDK2 complexes are inhib-
activity.130 Enforced expression of INK4 proteins arrest ited by bound p21Cip1 and p27Kip1.148 Additionally, cyclin
cells in the G1 phase in an RB-dependent manner.166,167 D-CDK4/6 complexes bind p21Cip1 and p27Kip1.168–170
Loss of D-type cyclins therefore prevents the titration of
p21Cip1 and p27Kip1 by cyclin D-CDK4/6 complexes away
from the cyclin E-CDK2 and cyclin A-CDK2 complexes.
As a result, there is complete inhibition of cyclin E-CDK2
and cyclin A-CDK2 activities, as well as RB phosphoryla-
tion, leading to the exit from the G1 phase.171 Conversely,
growth-induced or oncogenic-induced expression of cyclin
D allows its interaction with CDK4/6 by competing with
INK4 for binding. The binding of cyclin D-CDK4/6 com-
plexes to p21Cip1 and p27Kip1 releases the inhibitors from
the cyclin E-CDK2 and cyclin A-CDK2 complexes. This
unleashes cyclin E-CDK2 and cyclin A-CDK2 activity,
allowing further RB phosphorylation, exit from G1, and
entry into S phase.171
The G1/S checkpoint is activated upon detection of
DNA damage. The mammalian DNA-damage response is
a complex network, involving a multitude of proteins that
FIGURE 15.6  The cell cycle checkpoints.  The locations of the vari- include “sensor” proteins that sense the damage and trans-
ous checkpoints (stop signs) and the primary defects the checkpoints are mit signals to “transducer” proteins, which in turn convey
designed to monitor are shown. the signals to numerous “effector” proteins implicated in

FIGURE 15.7  A simplified scheme of the cell cycle checkpoint pathways in response to DNA damage.  The black arrows indicate activating and
red lines indicate inhibitory actions.
458 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

specific cellular pathways, including DNA repair mecha- 15.3.3  Intra-S Checkpoint
nisms, cell-cycle checkpoints, cellular senescence, and
While proliferating cells respond to genotoxic stresses by
apoptosis.172–177 In response to DNA damage, signals ini-
activating checkpoint responses that impose durable cell
tiated by the sensors rapidly transduce to the ataxia tel-
cycle arrest in G1 or G2, before entry into S phase or mito-
angiectasia, mutated (ATM) and ataxia telangiectasia and
sis, respectively, cells that experience genotoxic stresses
Rad3-related (ATR) kinases, which phosphorylate a great
during DNA replication can only delay their progression
number of substrates.178–182 Among the substrates phos-
through the S phase in a transient fashion. If the damage is
phorylated by activated ATM and ATR are the checkpoint
not repaired during the delay, cells exit S phase and arrest
serine/threonine kinases, checkpoint kinase 1 (CHK1) and
upon reaching the G2 checkpoint. Nonetheless, the intra-S
checkpoint kinase 2 (CHK2). To prevent entry into S phase,
checkpoint is important for the maintenance of genomic
CHK1 and CHK2 phosphorylate the cell cycle regulatory
stability as DNA replication is a vulnerable period of the
phosphatase CDC25A, leading to its ubiquitin-mediated
cell cycle in which errors occur both endogenously or are
proteolysis.183–185 Inactivation of CDC25A leads to sus-
introduced exogenously. When the intra-S checkpoint
tained inhibitory phosphorylation of cyclin E-CDK2 com-
is activated, both replication initiation and fork progres-
plexes, thus preventing G1/S transition (Figure 15.5).183
sion are inhibited, thus reducing the rate of DNA replica-
ATM/ATR and CHK1/CHK2 kinases also target the tumor
tion.199 Studies have revealed numerous proteins that are
suppressor, p53.186–192 The phosphorylation of p53 by
involved in the control of intra-S checkpoint.199–204 Among
ATM/ATR inhibits its association with MDM2, an E3 ubiq-
these, activation of ATM and ATR is necessary to initi-
uitin ligase that normally functions to keep the p53 level
ate intra-S checkpoint upon sensing DNA double-strand
low, stabilizing the p53 protein.193 Subsequently, p53 tran-
breaks (DSBs).205–207 Activated ATM and ATR then acti-
scriptionally activates expression of the p21Cip1 gene,134
vate CHK1 and CHK2 kinases,208–211 which phosphorylate
which in turn inhibits cyclin E-CDK2 and prevents G1/S
CDC25A, leading to its ubiquitin-proteosome-dependent
transition.133 Lastly, rapid degradation of cyclin D1 in
degradation.183–185 This results in persistent inhibitory
response to DNA damage occurs independent of p53.194
phosphorylation of CDK2 in the cyclin A-CDK2 com-
The reduced cyclin D1 protein decreases the amount of
plexes, which in turn prevents firing of the replication
cyclin D-CDK4/6 complexes, resulting in the redistribution
origins.212 A second independent pathway is involved in
of p21Cip1 to cyclin E-CDK2 complexes, resulting in the
slowing down the replication rate in cells that have suffered
latter’s inactivation.23
DNA damage. This effect is mediated by ATM-dependent
phosphorylation of a cohesion protein and structural main-
15.3.2  G2/M Checkpoint tenance of chromosomes-1 (SMC-1).213–215 However, the
The G2/M checkpoint (also known as the G2 checkpoint) mechanism by which phosphorylated SMC-1 interferes
prevents cells from initiating mitosis when they experi- with DNA replication remains unknown.
ence DNA damage while in G2, when they progress into
G2 with either unrepaired DNA sustained during the pre- 15.3.4  Mitotic Checkpoint or Spindle
vious S or G1 phase, or when they possess incompletely
Assembly Checkpoint
replicated DNA from S phase.195,196 The critical target
of the G2 checkpoint is the mitosis-promoting activity of Mitosis is the process in which a cell divides itself into
the cyclin B-CDK1 complexes, whose activation after two halves, each with an identical set of chromosomes
genotoxic stresses is inhibited by ATM/ATR and CHK1/ (Figure 15.2). The central regulator of this process is the
CHK2-mediated degradation of the CDC25 family of mitotic checkpoint as SAC, which is a signaling mecha-
phosphatases, which normally activate CDK1 at the G2/M nism that arrests the progression of metaphase to anaphase
boundary.183,185,197 In addition, other regulators of CDC25 until all chromosomes are attached to the mitotic spin-
and cyclin B-CDK1, such as the polo-like kinases (PLKs), dles.94,156,163,216–219 This signal is akin to an “anaphase
are targeted by DNA damage-induced mechanisms.196 wait” signal that is generated at the kinetochores of unat-
Finally, the maintenance of the G2 checkpoint is depend- tached chromosomes and is extinguished once all kineto-
ent on the transcriptional programs regulated by p53, lead- chores are properly attached to the spindles (Figure 15.2).
ing to an induction of cell cycle inhibitors such as p21Cip1, Thus, sister chromatids are separated only when they are
growth arrest and DNA damage-inducible 45 (GADD45), in a position to be equally distributed to the two daughter
and 14-3-3σ proteins.196,198 These proteins cooperatively cells. Accordingly, the mitotic checkpoint serves to prevent
inhibit cyclin B-CDK1 activity by directly binding to cyc- chromosome missegregation.
lin B-CDK1 (p21Cip1), dissociating CDK1 from cyclin The proteins that control mitotic checkpoint were origi-
B (GADD45), and sequestering CDK1 in the cytoplasm nally identified by screens for mutations that bypassed
(14-3-3σ), resulting in G2 arrest.198 the ability of wild-type S. cerevisiae to arrest in mitosis
Chapter  |  15  The Cell Cycle 459

in the presence of spindle poisons.220,221 The genes iden- CDK6, thus maintaining RB in its active, anti-proliferative
tified include MAD (mitotic-arrest deficient), MAD1, state.125 Functional disruption of the tumor suppressors
MAD2, MAD3 (BUBR1 in humans), and budding uninhib- p16INKa and RB, or overexpression of the proto-oncogene
ited by benzimidazole 1 (BUB1).220,221 It was later found products cyclin D1 and CDK4, occur in many human can-
that these genes are conserved in all eukaryotes.163,222,223 cers, which prompts the speculation that disabling the “RB
When activated, these SAC proteins target CDC20, which pathway” is an essential part of cancer formation.22,250–255
is a cofactor of the ubiquitin ligase anaphase promoting In addition to being causative in retinoblastoma, loss of RB
complex/cyclosome (APC/C).94,224–234 Specifically, SAC has been found in many other cancers. Similarly, inactivat-
inhibits the ability of CDC20 to activate APC/C-mediated ing mutations of 16INK4a have been identified in numerous
ubiquitination of two key substrates, cyclin B and securin, tumors.256,257 The often exclusive nature of mutations that
thus preventing their degradation by the 26S proteosome. result in RB or p16INK4a loss, and/or cyclin D1 or CDK4
Proteolysis of cyclin B inactivates CDK1, allowing the overexpression, suggest that each of these cell cycle regula-
cells to exit from mitosis.235–239 On the other hand, destruc- tory genes exerts a critical function in the RB pathway of
tion of securin leads to activation of separase, which is cancer formation.254
required to cleave the cohesion complex that holds sister
chromatids together in order to execute anaphase.239–245 15.4.2  p53 and INK4a/ARF Tumor
Therefore, by keeping CDC20 in check, the SAC allows
Suppressor Genes
the chromosomes to properly align on the metaphase plate
and attach to the two spindle poles through mitotic spin- The tumor suppressor p53 is mutated in more than 50%
dles. Once the chromosomes are properly oriented, the of human cancers.258 It has been estimated that cancers
checkpoint is extinguished, relieving the mitotic arrest and derived from over 50 human cell types or tissues contain
allowing anaphase to proceed. mutations in the p53 gene.259,260 p53 is a labile protein but
accumulates in response to cellular stresses from DNA
15.4  PATHOLOGICAL CONSEQUENCES damage, hypoxia, or oncogenic activation.261,262 Upon
stabilization and activation, p53 initiates a transcriptional
OF CELL CYCLE DEREGULATION OR program that triggers either cell cycle arrest or apopto-
DYSREGULATION sis.263–269 Among the p53-responsive genes are p21Cip1,
Because regulation of the cell cycle is central to the con- BCL2-associated X protein (BAX), and mouse double-
trol of cell proliferation, it is not surprising that cancers minute 2 homolog (MDM2). While p21Cip1 regulates pro-
are often the result of deregulation or dysregulation of the gression of the cell cycle by inhibiting cyclin E-, A-, and
cell cycle (e.g., colorectal cancer). Recent genomic-scale B-CDK2 complexes, BAX causes apoptosis.270,271 The
sequencing studies have identified numerous somatic muta- transcriptional induction of MDM2 by p53 is a negative
tions in genes that possibly are involved in the formation of feedback mechanism as binding of MDM2, an E3 ubiquitin
cancer.246,247 Among these, some of the most highly ranked ligase, to p53 induces ubiquitination of p53 and subsequent
“cancer genes” are either directly or indirectly involved degradation.272–276 MDM2, in turn, is negatively regulated
in the regulation of the cell cycle. Examples include p53, by the alternative reading frame (ARF) tumor suppressor
adenomatous polyposis coli (APC), KRAS, F-box, and (p14ARF in humans and p19ARF in mice).255 ARF directly
WD40 domain protein 7 (FBXW7), and phosphatidylinosi- associates with MDM2 to block its interaction with p53,
tol 3-kinase, catalytic, α-subunit (PI3KCA).246,247 Here we therefore stabilizing p53.277–280 Thus, disruption of the
briefly review some of the common cancer genes with cell ARF-MDM2-p53 signaling pathway is a common feature
cycle regulatory functions. in cancers. This is supported by the finding that MDM2 is
overexpressed in 5–10% of human cancers, whereas ARF
15.4.1  Retinoblastoma Tumor Suppressor is silenced or deleted in many others.281–285 It is of inter-
est to note that ARF is derived from an alternative reading
Gene frame in exon 2 of the INK4a gene.286 Loss of the INK4a/
The RB tumor suppressor protein limits cell prolif- ARF locus, therefore, predisposes many tumor types due
eration by preventing entry into the S phase of the cell to the dual disruption of the RB-E2F and MDM2-p53
cycle.33,34,38,61,248,249 RB achieves its inhibitory effect pathways.
by blocking the activity of E2F. Progression into S phase
occurs when the ability of RB to suppress E2F is disrupted 15.4.3  hCDC4/FBXW7 Tumor
by the hyperphosphorylation of RB by cyclin D- and cyclin
Suppressor Gene
E-dependent CDKs in the G1 phase of the cell cycle.38 The
INK4 family of CKIs, particularly p16INKa, directly inhib- Abundant evidence indicates that ubiquitin-dependent
its activities of the cyclin D-dependent kinases, CDK4 and proteolysis regulates many aspects of the cell cycle and
460 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

that dysregulation of the ubiquitin-proteosome pathway tumors have mutations in the DNA mismatch repair
contributes to the formation of cancers.287–291 The proteo- (MMR) genes and account for approximately 15–20% of
lytic regulation of cell division is primarily controlled by sporadic CRCs.319,320 The remaining sporadic CRCs have
two ubiquitin ligase systems, APC/C and SCF. The APC/C CIN, which are frequently aneuploid, that is, they exhibit
ubiquitin ligase is a multimeric protein complex that regu- alterations in the number of chromosomes.321 It has been
lates chromosome segregation (see Section 15.3.4). SCF suggested that CIN is the driving force for the formation
ligase complexes are structurally similar to APC/C; com- of aneuploidy and tumorigenesis.156,216,322–325 Although
posed of an invariable core complex of SKP1, CUL1, and there has not been a unified mechanism responsible for
RBX1; and associated with a variable member of the F-box CIN, defects in several cellular processes have been caus-
protein family that serves as the substrate recognition com- ally linked to its formation. These include chromosome
ponent.234,292–298 Among the approximately 70 different dynamics (e.g., chromosome condensation, segregation,
F-box proteins that were identified, the human homolog cohesion, and kinetochore-spindle interaction), centrosome
of yeast CDC4 (hCDC), also known as FBXW7, has been duplication, cell cycle checkpoints (including G1, S, G2
extensively characterized and implicated in human tum- and the SACs), DNA damage repair pathway, and telomere
origenesis. For example, among the approximately 140 functions.156,158,216,326–330 Among these potential factors
mutated “cancer genes” identified in a recent genomic contributing to CIN, the mitotic checkpoint is probably
screen in colorectal cancers (CRCs), FBXW7 ranks fifth as the most important one since it is an essential part of the
the most frequently mutated gene (after APC, KRAS, p53, cell cycle that ensures equal distribution of chromosomes
and PI3K).246 upon the conclusion of cell division. Studies in mice lack-
Human FBXW7 exists in three different isoforms, ing specific components of the mitotic checkpoint support
α, β, and γ, each with a unique amino terminal end this view. Mice with genetically reduced levels of mitotic
fused to a common carboxyl-terminal.299 The interaction checkpoint proteins including MAD1, MAD2, BUB1,
between FBXW7 and its substrates depends on phospho- BUB3, BUBR1, and centromere protein E (CENP-E)
rylation of the substrate within a motif called the CDC4- all have increased levels of aneuploidy and CIN, with
phophodegredron (CPD).300 This feature enables FBXW7 the eventual formation of tumors in some animals.331–340
to simultaneously regulate a host of substrates by ubiquiti- Importantly, somatic mutations of many of the same genes
nation. Among the many substrates for FBXW7, some are have been identified in human cancers,341–349 indicating
critically involved in the regulation of the cell cycle such as the importance of the mitotic checkpoint in maintaining
cyclin E, c-Myc, c-Jun, and Notch.301–304 Mutations in the genomic integrity. Lastly, germline mutations in BUB1B
FBXW7 gene, therefore, lead to stabilization and elevated have been associated with the rare recessive condition of
levels of these substrates. It is no wonder that FBXW7 is mosaic variegated aneuploidy (MVA), which is character-
such a commonly mutated gene in human cancers.301,305 ized by growth retardation, microcephaly, and childhood
One of the best characterized substrates of FBXW7 is cancer with a mosaic pattern of chromosome gains and
cyclin E, which is essential for entry into S phase from G1 losses.350–353
phase in the cell cycle. Cyclin E level is elevated or dys-
regulated in many human cancers, resulting in dysfunc- 15.4.5  Adenomatous Polyposis Coli Tumor
tion of the cell cycle. There are many consequences of
Suppressor Gene
cyclin E deregulation including genetic instability, centro-
some amplification, and fork collapse during DNA replica- Germline mutations of the APC (which is different from
tion.81,306–314 Several studies subsequently identified cyclin APC/C) tumor suppressor gene cause familial adeno-
E as a substrate for FBXW7, which mediates the phospho- matous polyposis (FAP), an autosomal dominant disor-
rylation- and ubiquitination-dependent degradation of cyc- der characterized by the presence of numerous colonic
lin E.315–317 Thus, it appears that tumorigenesis secondary adenomas and colon cancer early in life.354–356 Somatic
to cyclin E deregulation is linked to altered function of mutations in the APC gene were subsequently found to be
FBXW7/hCDC4. present in the majority of sporadic CRCs.357 Its involve-
ment in tumor initiation leads to the hypothesis that APC
15.4.4  Chromosomal Instability and functions as a “gatekeeper” of colonic epithelial cell
Aneuploidy as Consequences of an proliferation and is responsible for the maintenance of
colonic epithelial cell renewal.357 A telling insight into the
Aberrant Mitotic Checkpoint function of APC came from the observation that it inter-
Genetic instability has long been recognized as an inte- acts with β-catenin,358,359 which is implicated in regulat-
gral part of human cancers.318 In CRC, there are two ing the Wnt signaling pathway of growth control.360–365
major forms of genetic instability: microsatellite insta- Particularly, Wnt signaling is now a well-established cen-
bility (MIN) and chromosomal instability (CIN). MIN tral mechanism for regulating proliferation of intestinal
Chapter  |  15  The Cell Cycle 461

FIGURE 15.8  Schematic presentation of the Wnt signaling pathway.  (A) Wnt is absent and (B) Wnt is present. Abbreviations: β-cat, β-catenin;
Fz, Frizzled; LRP, lipoprotein receptor-related protein; Dsh, disheveled; CK1, casein kinase 1; GSK, glycogen synthase kinase-3; TCF, T-cell factor
(Modified from 373 with permission.)

epithelial stem cells, which are the precursors to all sub- localized to the ends of microtubules embedded in kine-
sequent intestinal epithelial cell lineages.366–372 Wnt sign- tochores and forms a complex with several SAC proteins.
aling is normally absent in a quiescent, non-cycling cell. This finding suggests that APC is involved in chromosome
This is accompanied by the sequestration of β-catenin in a segregation.385–389 The role of APC in regulation of mito-
“destruction complex” in the cytoplasm that includes APC, sis is consistent with the finding that mutations in the APC
Axin, CK1, and GSK-3 (Figure 15.8A).373 This complex gene cause chromosomal instability.390–395 Thus, APC
leads to the phosphorylation of β-catenin, which is then may fulfill two requirements for a colonic epithelial cell to
degraded by ubiquitin-mediated proteasomal degrada- develop into colon cancer: the cell must acquire a selective
tion.374–381 When Wnt, a secreted glycoprotein, is present, advantage to allow initial clonal expansion, and the cell
it binds to the cell surface receptors Frizzled (Fz) and must generate genetic instability to allow multiple hits in
lipoprotein receptor-related protein (LRP). This leads to other genes that are responsible for tumor progression and
activation of the protein disheveled (Dsh) and subsequent malignant transformation.
release of β-catenin from the destruction complex, result-
ing in accumulation of free β-catenin in the cytoplasm
15.5  CONCLUSION
(Figure 15.8B). Some of this free β-catenin is shuttled into
the nucleus where it becomes associated with the tran- The cell cycle is a fundamental cellular process in which
scription factor, T-cell factor (TCF), to activate target gene a cell duplicates itself in a highly faithful manner to main-
expression (Figure 15.8B). Among the genes stimulated by tain genomic integrity. The cell cycle is divided into several
the β-catenin/TCF complexes are those encoding cyclin D1 distinct phases that are controlled by specific pairs of cyc-
and c-Myc,382–384 both of which are critical for the pro- lins and CDKs. The cell cycle also contains specific check-
gression of the cell cycle. It is important to note that muta- points with which to monitor the presence of damaged or
tional inactivation of APC or constitutional mutational incompletely replicated DNA in interphase or unaligned
activation of β-catenin, two common events in the patho- chromosomes in mitosis. Dysregulation or deregulation of
genesis of CRC, results in a similar net effect of nuclear the cell cycle often has pathological consequences and a
β-catenin accumulation to that seen during Wnt signaling. good example is cancer. Hence, many of the cell cycle reg-
These mutations then lead to unregulated cell proliferation. ulators act as tumor suppressors or oncoproteins, such as
APC has also been implicated in the regulation of Rb, p53, FBXW7, and cyclins. The relevance of cell cycle
mitosis in a β-catenin-independent manner. Here, APC is regulation also has an impact on the physiology of the
462 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

gastrointestinal tract because the epithelium is composed 19. Blagosklonny MV, Pardee AB. The restriction point of the cell cycle.
of rapidly proliferating cells. A prominent example is the Cell Cycle. 2002;1:103–110.
crucial role of the Wnt signaling pathway in regulating pro- 20. Lew DJ, Dulic V, Reed SI. Isolation of three novel human cyclins by
rescue of G1 cyclin (Cln) function in yeast. Cell. 1991;66:1197–1206.
liferation of intestinal stem cells, and that a critical com-
21. Norbury C, Nurse P. Animal cell cycles and their control. Annu Rev
ponent of the Wnt pathway, the APC protein, is mutated in
Biochem. 1992;61:441–470.
the majority of CRCs. Thus, a clear understanding of the 22. Sherr CJ. Cancer cell cycles. Science. 1996;274:1672–1677.
mechanisms regulating cell cycle progression is essential 23. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regula-
for understanding the regulation of proliferation and differ- tors of G1-phase progression. Genes Dev. 1999;13:1501–1512.
entiation of intestinal epithelial cells. 24. Filmus J, Robles AI, Shi W, Wong MJ, Colombo LL, Conti CJ.
Induction of cyclin D1 overexpression by activated ras. Oncogene.
ACKNOWLEDGMENT 1994;9:3627–3633.
25. Winston JT, Coats SR, Wang YZ, Pledger WJ. Regulation of the cell
The author wishes to thank Bing Yu for helping with some of the fig- cycle machinery by oncogenic ras. Oncogene. 1996;12:127–134.
ures. This work was in part supported by grants from the National 26. Lavoie JN, L'Allemain G, Brunet A, Muller R, Pouyssegur J.
Institutes of Health (DK052230 and CA084197). Cyclin D1 expression is regulated positively by the p42/p44MAPK
and negatively by the p38/HOGMAPK pathway. J Biol Chem.
1996;271:20608–20616.
REFERENCES
27. Aktas H, Cai H, Cooper GM. Ras links growth factor signaling to the
  1. Chang F, Nurse P. Finishing the cell cycle: control of mitosis and cell cycle machinery via regulation of cyclin D1 and the Cdk inhibi-
cytokinesis in fission yeast. Trends Genet. 1993;9:333–335. tor p27KIP1. Mol Cell Biol. 1997;17:3850–3857.
  2. Nurse P. The wellcome lecture, 1992. Cell cycle control. Philos Trans 28. Koepp DM, Harper JW, Elledge SJ. How the cyclin became a cyclin:
R Soc Lond B Biol Sci. 1993;341:449–454. regulated proteolysis in the cell cycle. Cell. 1999;97:431–434.
  3. Bartlett R, Nurse P. Yeast as a model system for understand- 29. Diehl JA, Zindy F, Sherr CJ. Inhibition of cyclin D1 phosphorylation
ing the control of DNA replication in Eukaryotes. Bioessays. on threonine-286 prevents its rapid degradation via the ubiquitin-
1990;12:457–463. proteasome pathway. Genes Dev. 1997;11:957–972.
  4. De Clercq A, Inze D. Cyclin-dependent kinase inhibitors in yeast, 30. Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase
animals, and plants: a functional comparison. Crit Rev Biochem Mol kinase-3beta regulates cyclin D1 proteolysis and subcellular localiza-
Biol. 2006;41:293–313. tion. Genes Dev. 1998;12:3499–3511.
  5. Nurse P. The josef steiner lecture: CDKs and cell-cycle control in fis- 31. Jones SM, Kazlauskas A. Growth-factor-dependent mitogen-
sion yeast: relevance to other eukaryotes and cancer. Int J Cancer. esis requires two distinct phases of signalling. Nat Cell Biol.
1997;71:707–708. 2001;3:165–172.
  6. Nigg EA. Cyclin-dependent protein kinases: key regulators of the 32. Bartek J, Bartkova J, Lukas J. The retinoblastoma protein pathway
eukaryotic cell cycle. Bioessays. 1995;17:471–480. and the restriction point. Curr Opin Cell Biol. 1996;8:805–814.
  7. Woo RA, Poon RY. Cyclin-dependent kinases and S phase control in 33. Weinberg RA. The retinoblastoma protein and cell cycle control.
mammalian cells. Cell Cycle. 2003;2:316–324. Cell. 1995;81:323–330.
  8. Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases. 34. Harbour JW, Dean DC. The Rb/E2F pathway: expanding roles and
Trends Biochem Sci. 2005;30:630–641. emerging paradigms. Genes Dev. 2000;14:2393–2409.
  9. Sherr CJ, Roberts JM. Living with or without cyclins and cyclin- 35. Cobrinik D. Pocket proteins and cell cycle control. Oncogene.
dependent kinases. Genes Dev. 2004;18:2699–2711. 2005;24:2796–2809.
10. Sanchez I, Dynlacht BD. New insights into cyclins, CDKs, and cell 36. Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR. The
cycle control. Semin Cell Dev Biol. 2005;16:311–321. E2F transcription factor is a cellular target for the RB protein. Cell.
11. Harper JV, Brooks G. The mammalian cell cycle: an overview. 1991;65:1053–1061.
Methods Mol Biol. 2005;296:113–153. 37. Nevins JR. Toward an understanding of the functional complex-
12. Murray AW. Cyclin-dependent kinases: regulators of the cell cycle ity of the E2F and retinoblastoma families. Cell Growth Differ.
and more. Chem Biol. 1994;1:191–195. 1998;9:585–593.
13. Nurse PM. Nobel Lecture. Cyclin dependent kinases and cell cycle 38. Dyson N. The regulation of E2F by pRB-family proteins. Genes Dev.
control. Biosci Rep. 2002;22:487–499. 1998;12:2245–2262.
14. Nurse P. Cyclin dependent kinases and cell cycle control (nobel lec- 39. Blake MC, Azizkhan JC. Transcription factor E2F is required for
ture). Chembiochem. 2002;3:596–603. efficient expression of the hamster dihydrofolate reductase gene in
15. Nurse P. Regulation of the eukaryotic cell cycle. Eur J Cancer. vitro and in vivo. Mol Cell Biol. 1989;9:4994–5002.
1997;33:1002–1004. 40. Thalmeier K, Synovzik H, Mertz R, Winnacker EL, Lipp M. Nuclear
16. Hartwell LH. Twenty-five years of cell cycle genetics. Genetics. factor E2F mediates basic transcription and trans-activation by E1a
1991;129:975–980. of the human MYC promoter. Genes Dev. 1989;3:527–536.
17. Doree M, Hunt T. From Cdc2 to Cdk1: when did the cell cycle kinase 41. Dalton S. Cell cycle regulation of the human cdc2 gene. EMBO J.
join its cyclin partner? J Cell Sci. 2002;115:2461–2464. 1992;11:1797–1804.
18. Minshull J, Pines J, Golsteyn R, Standart N, Mackie S, Colman A, 42. DeGregori J, Leone G, Ohtani K, Miron A, Nevins JR. E2F-1 accu-
et al. The role of cyclin synthesis, modification and destruction in the mulation bypasses a G1 arrest resulting from the inhibition of G1
control of cell division. J Cell Sci Suppl. 1989;12:77–97. cyclin-dependent kinase activity. Genes Dev. 1995;9:2873–2887.
Chapter  |  15  The Cell Cycle 463

43. Magnaghi-Jaulin L, Groisman R, Naguibneva I, Robin P, 64. Dimova DK, Dyson NJ. The E2F transcriptional network: old
Lorain S, Le Villain JP, et  al. Retinoblastoma protein represses acquaintances with new faces. Oncogene. 2005;24:2810–2826.
transcription by recruiting a histone deacetylase. Nature. 65. Bracken AP, Ciro M, Cocito A, Helin K. E2F target genes: unraveling
1998;391:601–605. the biology. Trends Biochem Sci. 2004;29:409–417.
44. Kingston RE, Narlikar GJ. ATP-dependent remodeling and 66. Ohtani K. Implication of transcription factor E2F in regulation of
acetylation as regulators of chromatin fluidity. Genes Dev. DNA replication. Front Biosci. 1999;4:D793–804.
1999;13:2339–2352. 67. Ohtani K, Iwanaga R, Nakamura M, Ikeda M, Yabuta N, Tsuruga H,
45. Kornberg RD, Lorch Y. Chromatin-modifying and -remodeling com- et  al. Cell growth-regulated expression of mammalian MCM5 and
plexes. Curr Opin Genet Dev. 1999;9:148–151. MCM6 genes mediated by the transcription factor E2F. Oncogene.
46. Collingwood TN, Urnov FD, Wolffe AP. Nuclear receptors: coactiva- 1999;18:2299–2309.
tors, corepressors and chromatin remodeling in the control of tran- 68. Helin K, Wu CL, Fattaey AR, Lees JA, Dynlacht BD, Ngwu C, et al.
scription. J Mol Endocrinol. 1999;23:255–275. Heterodimerization of the transcription factors E2F-1 and DP-1 leads
47. Wolffe AP, Hayes JJ. Chromatin disruption and modification. Nucleic to cooperative trans-activation. Genes Dev. 1993;7:1850–1861.
Acids Res. 1999;27:711–720. 69. Harbour JW, Luo RX, Dei Santi A, Postigo AA, Dean DC. Cdk
48. Weintraub SJ, Chow KN, Luo RX, Zhang SH, He S, Dean DC. phosphorylation triggers sequential intramolecular interactions that
Mechanism of active transcriptional repression by the retinoblastoma progressively block Rb functions as cells move through G1. Cell.
protein. Nature. 1995;375:812–815. 1999;98:859–869.
49. Meloni AR, Smith EJ, Nevins JR. A mechanism for Rb/p130-medi- 70. Zhang HS, Postigo AA, Dean DC. Active transcriptional repression
ated transcription repression involving recruitment of the CtBP core- by the Rb-E2F complex mediates G1 arrest triggered by p16INK4a,
pressor. Proc Natl Acad Sci U S A. 1999;96:9574–9579. TGFbeta, and contact inhibition. Cell. 1999;97:53–61.
50. Ferreira R, Magnaghi-Jaulin L, Robin P, Harel-Bellan A, Trouche 71. Geng Y, Whoriskey W, Park MY, Bronson RT, Medema RH, Li
D. The three members of the pocket proteins family share the ability T, et  al. Rescue of cyclin D1 deficiency by knockin cyclin E. Cell.
to repress E2F activity through recruitment of a histone deacetylase. 1999;97:767–777.
Proc Natl Acad Sci U S A. 1998;95:10493–10498. 72. Dulic V, Lees E, Reed SI. Association of human cyclin E with a peri-
51. Chow KN, Starostik P, Dean DC. The Rb family contains a con- odic G1-S phase protein kinase. Science. 1992;257:1958–1961.
served cyclin-dependent-kinase-regulated transcriptional repressor 73. Koff A, Giordano A, Desai D, Yamashita K, Harper JW, Elledge S,
motif. Mol Cell Biol. 1996;16:7173–7181. et al. Formation and activation of a cyclin E-cdk2 complex during the
52. Hinds PW, Mittnacht S, Dulic V, Arnold A, Reed SI, Weinberg RA. G1 phase of the human cell cycle. Science. 1992;257:1689–1694.
Regulation of retinoblastoma protein functions by ectopic expression 74. Yu Q, Sicinski P. Mammalian cell cycles without cyclin E-CDK2.
of human cyclins. Cell. 1992;70:993–1006. Cell Cycle. 2004;3:292–295.
53. Lundberg AS, Weinberg RA. Functional inactivation of the retino- 75. Clurman BE, Sheaff RJ, Thress K, Groudine M, Roberts JM.
blastoma protein requires sequential modification by at least two dis- Turnover of cyclin E by the ubiquitin-proteasome pathway is regu-
tinct cyclin-cdk complexes. Mol Cell Biol. 1998;18:753–761. lated by cdk2 binding and cyclin phosphorylation. Genes Dev.
54. Chen PL, Scully P, Shew JY, Wang JY, Lee WH. Phosphorylation of 1996;10:1979–1990.
the retinoblastoma gene product is modulated during the cell cycle 76. Won KA, Reed SI. Activation of cyclin E/CDK2 is coupled to site-
and cellular differentiation. Cell. 1989;58:1193–1198. specific autophosphorylation and ubiquitin-dependent degradation of
55. Lam EW, La Thangue NB. DP and E2F proteins: coordinating cyclin E. EMBO J. 1996;15:4182–4193.
transcription with cell cycle progression. Curr Opin Cell Biol. 77. Welcker M, Singer J, Loeb KR, Grim J, Bloecher A, Gurien-West M,
1994;6:859–866. et al. Multisite phosphorylation by Cdk2 and GSK3 controls cyclin E
56. La Thangue NB. DP and E2F proteins: components of a het- degradation. Mol Cell. 2003;12:381–392.
erodimeric transcription factor implicated in cell cycle control. Curr 78. Sweeney C, Murphy M, Kubelka M, Ravnik SE, Hawkins CF,
Opin Cell Biol. 1994;6:443–450. Wolgemuth DJ, et al. A distinct cyclin A is expressed in germ cells in
57. Adams PD, Kaelin Jr. WG. Transcriptional control by E2F. Semin the mouse. Development. 1996;122:53–64.
Cancer Biol. 1995;6:99–108. 79. Krude T, Jackman M, Pines J, Laskey RA. Cyclin/Cdk-dependent
58. La Thangue NB. E2F and the molecular mechanisms of early cell- initiation of DNA replication in a human cell-free system. Cell.
cycle control. Biochem Soc Trans. 1996;24:54–59. 1997;88:109–119.
59. Lavia P, Jansen-Durr P. E2F target genes and cell-cycle checkpoint 80. Hua XH, Newport J. Identification of a preinitiation step in DNA
control. Bioessays. 1999;21:221–230. replication that is independent of origin recognition complex and
60. Berckmans B, De Veylder L. Transcriptional control of the cell cycle. cdc6, but dependent on cdk2. J Cell Biol. 1998;140:271–281.
Curr Opin Plant Biol. 2009;12:599–605. 81. Coverley D, Laman H, Laskey RA. Distinct roles for cyclins E and A
61. Brehm A, Miska E, Reid J, Bannister A, Kouzarides T. The cell during DNA replication complex assembly and activation. Nat Cell
cycle-regulating transcription factors E2F-RB. Br J Cancer. Biol. 2002;4:523–528.
1999;80(suppl 1):38–41. 82. Mitra J, Enders GH. Cyclin A/Cdk2 complexes regulate activa-
62. Hateboer G, Wobst A, Petersen BO, Le Cam L, Vigo E, tion of Cdk1 and Cdc25 phosphatases in human cells. Oncogene.
Sardet C, et al. Cell cycle-regulated expression of mammalian CDC6 2004;23:3361–3367.
is dependent on E2F. Mol Cell Biol. 1998;18:6679–6697. 83. Nguyen TB, Manova K, Capodieci P, Lindon C, Bottega S,
63. Black AR, Azizkhan-Clifford J. Regulation of E2F: a family Wang XY, et  al. Characterization and expression of mamma-
of transcription factors involved in proliferation control. Gene. lian cyclin b3, a prepachytene meiotic cyclin. J Biol Chem.
1999;237:281–302. 2002;277:41960–41969.
464 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

  84. Nigg EA. Mitotic kinases as regulators of cell division and its 104. Ninomiya-Tsuji J, Nomoto S, Yasuda H, Reed SI, Matsumoto K.
checkpoints. Nat Rev Mol Cell Biol. 2001;2:21–32. Cloning of a human cDNA encoding a CDC2-related kinase by
  85. Ubersax JA, Woodbury EL, Quang PN, Paraz M, Blethrow JD, complementation of a budding yeast cdc28 mutation. Proc Natl
Shah K, et al. Targets of the cyclin-dependent kinase Cdk1. Nature. Acad Sci U S A. 1991;88:9006–9010.
2003;425:859–864. 105. Matsushime H, Ewen ME, Strom DK, Kato JY, Hanks SK, Roussel
  86. Crasta K, Lim HH, Giddings Jr. TH, Winey M, Surana U. MF, et al. Identification and properties of an atypical catalytic sub-
Inactivation of Cdh1 by synergistic action of Cdk1 and polo kinase unit (p34PSK-J3/cdk4) for mammalian D type G1 cyclins. Cell.
is necessary for proper assembly of the mitotic spindle. Nat Cell 1992;71:323–334.
Biol. 2008;10:665–675. 106. Xiong Y, Zhang H, Beach D. D type cyclins associate with multiple
  87. McHugh B, Heck MM. Regulation of chromosome condensation protein kinases and the DNA replication and repair factor PCNA.
and segregation. Curr Opin Genet Dev. 2003;13:185–190. Cell. 1992;71:505–514.
  88. Meyer H, Drozdowska A, Dobrynin G. A role for Cdc48/p97 and 107. Malumbres M, Barbacid M. To cycle or not to cycle: a critical deci-
Aurora B in controlling chromatin condensation during exit from sion in cancer. Nat Rev Cancer. 2001;1:222–231.
mitosis. Biochem Cell Biol. 2010;88:23–28. 108. Lolli G, Johnson LN. CAK-Cyclin-dependent Activating Kinase: a
  89. Gavet O, Pines J. Activation of cyclin B1-Cdk1 synchronizes key kinase in cell cycle control and a target for drugs?. Cell Cycle.
events in the nucleus and the cytoplasm at mitosis. J Cell Biol. 2005;4:572–577.
2010;189:247–259. 109. Kaldis P. The cdk-activating kinase (CAK): from yeast to mammals.
  90. Draviam VM, Orrechia S, Lowe M, Pardi R, Pines J. The localiza- Cell Mol Life Sci. 1999;55:284–296.
tion of human cyclins B1 and B2 determines CDK1 substrate spe- 110. Fisher RP. Secrets of a double agent: CDK7 in cell-cycle control
cificity and neither enzyme requires MEK to disassemble the Golgi and transcription. J Cell Sci. 2005;118:5171–5180.
apparatus. J Cell Biol. 2001;152:945–958. 111. Yang L, MacLellan WR, Han Z, Weiss JN, Qu Z. Multisite phos-
  91. Harper JW, Burton JL, Solomon MJ. The anaphase-promot- phorylation and network dynamics of cyclin-dependent kinase sign-
ing complex: it's not just for mitosis any more. Genes Dev. aling in the eukaryotic cell cycle. Biophys J. 2004;86:3432–3443.
2002;16:2179–2206. 112. Watanabe N, Broome M, Hunter T. Regulation of the human
  92. Noton E, Diffley JF. CDK inactivation is the only essential function WEE1Hu CDK tyrosine 15-kinase during the cell cycle. EMBO J.
of the APC/C and the mitotic exit network proteins for origin reset- 1995;14:1878–1891.
ting during mitosis. Mol Cell. 2000;5:85–95. 113. Berry LD, Gould KL. Regulation of Cdc2 activity by phosphoryla-
  93. Peters JM. The anaphase-promoting complex: proteolysis in mitosis tion at T14/Y15. Prog Cell Cycle Res. 1996;2:99–105.
and beyond. Mol Cell. 2002;9:931–943. 114. Parker LL, Piwnica-Worms H. Inactivation of the p34cdc2-cyc-
  94. Castro A, Bernis C, Vigneron S, Labbe JC, Lorca T. The anaphase- lin B complex by the human WEE1 tyrosine kinase. Science.
promoting complex: a key factor in the regulation of cell cycle. 1992;257:1955–1957.
Oncogene. 2005;24:314–325. 115. McGowan CH, Russell P. Human Wee1 kinase inhibits cell divi-
  95. Murray A. Cyclin ubiquitination: the destructive end of mitosis. sion by phosphorylating p34cdc2 exclusively on Tyr15. EMBO J.
Cell. 1995;81:149–152. 1993;12:75–85.
  96. Russell P, Nurse P. Schizosaccharomyces pombe and Saccharomyces 116. Fattaey A, Booher RN. Myt1: a Wee1-type kinase that phos-
cerevisiae: a look at yeasts divided. Cell. 1986;45:781–782. phorylates Cdc2 on residue Thr14. Prog Cell Cycle Res.
  97. Lee MG, Nurse P. Cell cycle genes of the fission yeast. Sci Prog. 1997;3:233–240.
1987;71:1–14. 117. Leise 3rd W, Mueller PR. Multiple Cdk1 inhibitory kinases regulate
  98. Draetta G, Brizuela L, Potashkin J, Beach D. Identification of the cell cycle during development. Dev Biol. 2002;249:156–173.
p34 and p13, human homologs of the cell cycle regulators of 118. Aressy B, Ducommun B. Cell cycle control by the CDC25 phos-
fission yeast encoded by cdc2 and suc1. Cell. 1987;50: phatases. Anticancer Agents Med Chem. 2008;8:818–824.
319–325. 119. Rudolph J. Cdc25 phosphatases: structure, specificity, and mecha-
  99. Lee MG, Nurse P. Complementation used to clone a human homo- nism. Biochemistry. 2007;46:3595–3604.
logue of the fission yeast cell cycle control gene cdc2. Nature. 120. Coleman TR, Dunphy WG. Cdc2 regulatory factors. Curr Opin Cell
1987;327:31–35. Biol. 1994;6:877–882.
100. Hanks SK. Homology probing: identification of cDNA clones 121. Santamaria D, Ortega S. Cyclins and CDKS in development and
encoding members of the protein-serine kinase family. Proc Natl cancer: lessons from genetically modified mice. Front Biosci.
Acad Sci U S A. 1987;84:388–392. 2006;11:1164–1188.
101. Elledge SJ, Spottswood MR. A new human p34 protein kinase, 122. Satyanarayana A, Kaldis P. Mammalian cell-cycle regulation: sev-
CDK2, identified by complementation of a cdc28 mutation in eral Cdks, numerous cyclins and diverse compensatory mecha-
Saccharomyces cerevisiae, is a homolog of Xenopus Eg1. EMBO J. nisms. Oncogene. 2009;28:2925–2939.
1991;10:2653–2659. 123. Aleem E, Kaldis P. Mouse models of cell cycle regulators: new par-
102. Paris J, Le Guellec R, Couturier A, Le Guellec K, Omilli F, adigms. Results Probl Cell Differ. 2006;42:271–328.
Camonis J, et  al. Cloning by differential screening of a Xenopus 124. Santamaria D, Barriere C, Cerqueira A, Hunt S, Tardy C, Newton
cDNA coding for a protein highly homologous to cdc2. Proc Natl K, et  al. Cdk1 is sufficient to drive the mammalian cell cycle.
Acad Sci U S A. 1991;88:1039–1043. Nature. 2007;448:811–815.
103. Tsai LH, Harlow E, Meyerson M. Isolation of the human cdk2 125. Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell-
gene that encodes the cyclin A- and adenovirus E1A-associated p33 cycle control causing specific inhibition of cyclin D/CDK4. Nature.
kinase. Nature. 1991;353:174–177. 1993;366:704–707.
Chapter  |  15  The Cell Cycle 465

126. Hannon GJ, Beach D. p15INK4B is a potential effector of TGF- 145. Nakanishi M, Robetorye RS, Adami GR, Pereira-Smith OM, Smith
beta-induced cell cycle arrest. Nature. 1994;371:257–261. JR. Identification of the active region of the DNA synthesis inhibi-
127. Guan KL, Jenkins CW, Li Y, Nichols MA, Wu X, O'Keefe CL, et al. tory gene p21Sdi1/CIP1/WAF1. EMBO J. 1995;14:555–563.
Growth suppression by p18, a p16INK4/MTS1- and p14INK4B/ 146. Warbrick E, Lane DP, Glover DM, Cox LS. A small peptide inhibi-
MTS2-related CDK6 inhibitor, correlates with wild-type pRb func- tor of DNA replication defines the site of interaction between the
tion. Genes Dev. 1994;8:2939–2952. cyclin-dependent kinase inhibitor p21WAF1 and proliferating cell
128. Hirai H, Roussel MF, Kato JY, Ashmun RA, Sherr CJ. Novel nuclear antigen. Curr Biol. 1995;5:275–282.
INK4 proteins, p19 and p18, are specific inhibitors of the cyc- 147. Lin J, Reichner C, Wu X, Levine AJ. Analysis of wild-
lin D-dependent kinases CDK4 and CDK6. Mol Cell Biol. type and mutant p21WAF-1 gene activities. Mol Cell Biol.
1995;15:2672–2681. 1996;16:1786–1793.
129. Chan FK, Zhang J, Cheng L, Shapiro DN, Winoto A. Identification 148. Russo AA, Jeffrey PD, Patten AK, Massague J, Pavletich NP.
of human and mouse p19, a novel CDK4 and CDK6 inhibitor with Crystal structure of the p27Kip1 cyclin-dependent-kinase inhibitor
homology to p16ink4. Mol Cell Biol. 1995;15:2682–2688. bound to the cyclin A-Cdk2 complex. Nature. 1996;382:325–331.
130. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin- 149. Elledge SJ. Cell cycle checkpoints: preventing an identity crisis.
dependent kinases. Genes Dev. 1995;9:1149–1163. Science. 1996;274:1664–1672.
131. Pavletich NP. Mechanisms of cyclin-dependent kinase regulation: 150. Murray A. Cell cycle checkpoints. Curr Opin Cell Biol.
structures of Cdks, their cyclin activators, and Cip and INK4 inhibi- 1994;6:872–876.
tors. J Mol Biol. 1999;287:821–828. 151. Pietenpol JA, Stewart ZA. Cell cycle checkpoint signaling: cell
132. Gu Y, Turck CW, Morgan DO. Inhibition of CDK2 activity in vivo by cycle arrest versus apoptosis. Toxicology. 2002;181–182:475–481.
an associated 20K regulatory subunit. Nature. 1993;366:707–710. 152. Xie S, Xie B, Lee MY, Dai W. Regulation of cell cycle checkpoints
133. Harper JW, Adami GR, Wei N, Keyomarsi K, Elledge SJ. The p21 by polo-like kinases. Oncogene. 2005;24:277–286.
Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin- 153. O'Connor PM. Mammalian G1 and G2 phase checkpoints. Cancer
dependent kinases. Cell. 1993;75:805–816. Surv. 1997;29:151–182.
134. el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent 154. Donjerkovic D, Scott DW. Regulation of the G1 phase of the mam-
JM, et  al. WAF1, a potential mediator of p53 tumor suppression. malian cell cycle. Cell Res. 2000;10:1–16.
Cell. 1993;75:817–825. 155. Malumbres M. Revisiting the “Cdk-centric” view of the mammalian
135. Xiong Y, Hannon GJ, Zhang H, Casso D, Kobayashi R, Beach cell cycle. Cell Cycle. 2005;4:206–210.
D. p21 is a universal inhibitor of cyclin kinases. Nature. 156. Holland AJ, Cleveland DW. Boveri revisited: chromosomal insta-
1993;366:701–704. bility, aneuploidy and tumorigenesis. Nat Rev Mol Cell Biol.
136. Dulic V, Kaufmann WK, Wilson SJ, Tlsty TD, Lees E, Harper JW, 2009;10:478–487.
et  al. p53-dependent inhibition of cyclin-dependent kinase activi- 157. Yen TJ, Kao GD. Mitotic checkpoint, aneuploidy and cancer. Adv
ties in human fibroblasts during radiation-induced G1 arrest. Cell. Exp Med Biol. 2005;570:477–499.
1994;76:1013–1023. 158. Qi W, Yu H. The spindle checkpoint and chromosomal stability.
137. Noda A, Ning Y, Venable SF, Pereira-Smith OM, Smith JR. Cloning Genome Dyn. 2006;1:116–130.
of senescent cell-derived inhibitors of DNA synthesis using an 159. Kops GJ. The kinetochore and spindle checkpoint in mammals.
expression screen. Exp Cell Res. 1994;211:90–98. Front Biosci. 2008;13:3606–3620.
138. Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, 160. Suijkerbuijk SJ, Kops GJ. Preventing aneuploidy: the contri-
Tempst P, et  al. Cloning of p27Kip1, a cyclin-dependent kinase bution of mitotic checkpoint proteins. Biochim Biophys Acta.
inhibitor and a potential mediator of extracellular antimitogenic sig- 2008;1786:24–31.
nals. Cell. 1994;78:59–66. 161. Sczaniecka MM, Hardwick KG. The spindle checkpoint: how do
139. Polyak K, Kato JY, Solomon MJ, Sherr CJ, Massague J, Roberts cells delay anaphase onset? SEB Exp Biol Ser. 2008;59:243–256.
JM, et  al. p27Kip1, a cyclin-Cdk inhibitor, links transforming 162. Decordier I, Cundari E, Kirsch-Volders M. Mitotic checkpoints
growth factor-beta and contact inhibition to cell cycle arrest. Genes and the maintenance of the chromosome karyotype. Mutat Res.
Dev. 1994;8:9–22. 2008;651:3–13.
140. Toyoshima H, Hunter T. p27, a novel inhibitor of G1 cyclin-Cdk 163. Musacchio A, Salmon ED. The spindle-assembly checkpoint in
protein kinase activity, is related to p21. Cell. 1994;78:67–74. space and time. Nat Rev Mol Cell Biol. 2007;8:379–393.
141. Lee MH, Reynisdottir I, Massague J. Cloning of p57KIP2, a cyclin- 164. May KM, Hardwick KG. The spindle checkpoint. J Cell Sci.
dependent kinase inhibitor with unique domain structure and tissue 2006;119:4139–4142.
distribution. Genes Dev. 1995;9:639–649. 165. Hartwell LH, Weinert TA. Checkpoints: controls that ensure the
142. Matsuoka S, Edwards MC, Bai C, Parker S, Zhang P, Baldini A, order of cell cycle events. Science. 1989;246:629–634.
et al. p57KIP2, a structurally distinct member of the p21CIP1 Cdk 166. Lukas J, Parry D, Aagaard L, Mann DJ, Bartkova J, Strauss M,
inhibitor family, is a candidate tumor suppressor gene. Genes Dev. et al. Retinoblastoma-protein-dependent cell-cycle inhibition by the
1995;9:650–662. tumour suppressor p16. Nature. 1995;375:503–506.
143. Chen J, Jackson PK, Kirschner MW, Dutta A. Separate domains of 167. Medema RH, Herrera RE, Lam F, Weinberg RA. Growth suppres-
p21 involved in the inhibition of Cdk kinase and PCNA. Nature. sion by p16ink4 requires functional retinoblastoma protein. Proc
1995;374:386–388. Natl Acad Sci U S A. 1995;92:6289–6293.
144. Chen J, Saha P, Kornbluth S, Dynlacht BD, Dutta A. Cyclin-binding 168. Blain SW, Montalvo E, Massague J. Differential interaction of
motifs are essential for the function of p21CIP1. Mol Cell Biol. the cyclin-dependent kinase (Cdk) inhibitor p27Kip1 with cyclin
1996;16:4673–4682. A-Cdk2 and cyclin D2-Cdk4. J Biol Chem. 1997;272:25863–25872.
466 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

169. LaBaer J, Garrett MD, Stevenson LF, Slingerland JM, Sandhu C, 193. Shieh SY, Ikeda M, Taya Y, Prives C. DNA damage-induced
Chou HS, et al. New functional activities for the p21 family of CDK phosphorylation of p53 alleviates inhibition by MDM2. Cell.
inhibitors. Genes Dev. 1997;11:847–862. 1997;91:325–334.
170. Cheng M, Olivier P, Diehl JA, Fero M, Roussel MF, Roberts JM, 194. Agami R, Bernards R. Distinct initiation and maintenance mecha-
et  al. The p21(Cip1) and p27(Kip1) CDK 'inhibitors' are essential nisms cooperate to induce G1 cell cycle arrest in response to DNA
activators of cyclin D-dependent kinases in murine fibroblasts. damage. Cell. 2000;102:55–66.
EMBO J. 1999;18:1571–1583. 195. Xu B, Kim ST, Lim DS, Kastan MB. Two molecularly distinct
171. Roussel MF. The INK4 family of cell cycle inhibitors in cancer. G(2)/M checkpoints are induced by ionizing irradiation. Mol Cell
Oncogene. 1999;18:5311–5317. Biol. 2002;22:1049–1059.
172. Schmitt E, Paquet C, Beauchemin M, Bertrand R. DNA-damage 196. Nyberg KA, Michelson RJ, Putnam CW, Weinert TA. Toward main-
response network at the crossroads of cell-cycle checkpoints, taining the genome: DNA damage and replication checkpoints.
cellular senescence and apoptosis. J Zhejiang Univ Sci B. Annu Rev Genet. 2002;36:617–656.
2007;8:377–397. 197. Mailand N, Podtelejnikov AV, Groth A, Mann M, Bartek J, Lukas J.
173. Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S. Molecular Regulation of G(2)/M events by Cdc25A through phosphorylation-
mechanisms of mammalian DNA repair and the DNA damage dependent modulation of its stability. EMBO J. 2002;21:5911–5920.
checkpoints. Annu Rev Biochem. 2004;73:39–85. 198. Taylor WR, Stark GR. Regulation of the G2/M transition by p53.
174. Motoyama N, Naka K. DNA damage tumor suppressor genes and Oncogene. 2001;20:1803–1815.
genomic instability. Curr Opin Genet Dev. 2004;14:11–16. 199. Grallert B, Boye E. The multiple facets of the intra-S checkpoint.
175. Bartek J, Lukas J. DNA damage checkpoints: from initiation to Cell Cycle. 2008;7:2315–2320.
recovery or adaptation. Curr Opin Cell Biol. 2007;19:238–245. 200. Bartek J, Lukas C, Lukas J. Checking on DNA damage in S phase.
176. Ishikawa K, Ishii H, Saito T. DNA damage-dependent cell Nat Rev Mol Cell Biol. 2004;5:792–804.
cycle checkpoints and genomic stability. DNA Cell Biol. 201. Canman CE. Replication checkpoint: preventing mitotic catastro-
2006;25:406–411. phe. Curr Biol. 2001;11:R121–124.
177. Niida H, Nakanishi M. DNA damage checkpoints in mammals. 202. Friedel AM, Pike BL, Gasser SM. ATR/Mec1: coordinating fork
Mutagenesis. 2006;21:3–9. stability and repair. Curr Opin Cell Biol. 2009;21:237–244.
178. Nojima H. Protein kinases that regulate chromosome stability and 203. Segurado M, Tercero JA. The S-phase checkpoint: targeting the rep-
their downstream targets. Genome Dyn. 2006;1:131–148. lication fork. Biol Cell. 2009;101:617–627.
179. Cimprich KA, Cortez D. ATR: an essential regulator of genome 204. Bartek J, Lukas J. Mammalian G1- and S-phase checkpoints in
integrity. Nat Rev Mol Cell Biol. 2008;9:616–627. response to DNA damage. Curr Opin Cell Biol. 2001;13:738–747.
180. Lavin MF, Kozlov S. ATM activation and DNA damage response. 205. Shiloh Y. ATM and related protein kinases: safeguarding genome
Cell Cycle. 2007;6:931–942. integrity. Nat Rev Cancer. 2003;3:155–168.
181. Paulsen RD, Cimprich KA. The ATR pathway: fine-tuning the fork. 206. Bakkenist CJ, Kastan MB. DNA damage activates ATM through
DNA Repair (Amst). 2007;6:953–966. intermolecular autophosphorylation and dimer dissociation. Nature.
182. Hurley PJ, Bunz F. ATM and ATR: components of an integrated cir- 2003;421:499–506.
cuit. Cell Cycle. 2007;6:414–417. 207. Zou L, Elledge SJ. Sensing DNA damage through ATRIP recogni-
183. Falck J, Mailand N, Syljuasen RG, Bartek J, Lukas J. The ATM- tion of RPA-ssDNA complexes. Science. 2003;300:1542–1548.
Chk2-Cdc25A checkpoint pathway guards against radioresistant 208. Helt CE, Cliby WA, Keng PC, Bambara RA, O'Reilly MA. Ataxia
DNA synthesis. Nature. 2001;410:842–847. telangiectasia mutated (ATM) and ATM and Rad3-related protein
184. Bartek J, Lukas J. Chk1 and Chk2 kinases in checkpoint control and exhibit selective target specificities in response to different forms of
cancer. Cancer Cell. 2003;3:421–429. DNA damage. J Biol Chem. 2005;280:1186–1192.
185. Donzelli M, Draetta GF. Regulating mammalian checkpoints 209. Heffernan TP, Simpson DA, Frank AR, Heinloth AN, Paules RS,
through Cdc25 inactivation. EMBO Rep. 2003;4:671–677. Cordeiro-Stone M, et  al. An ATR- and Chk1-dependent S check-
186. Farnebo M, Bykov VJ, Wiman KG. The p53 tumor suppressor: a point inhibits replicon initiation following UVC-induced DNA dam-
master regulator of diverse cellular processes and therapeutic target age. Mol Cell Biol. 2002;22:8552–8561.
in cancer. Biochem Biophys Res Commun. 2010;396:85–89. 210. Gatei M, Sloper K, Sorensen C, Syljuasen R, Falck J, Hobson K,
187. Boehme KA, Blattner C. Regulation of p53--insights into a complex et  al. Ataxia-telangiectasia-mutated (ATM) and NBS1-dependent
process. Crit Rev Biochem Mol Biol. 2009;44:367–392. phosphorylation of Chk1 on Ser-317 in response to ionizing radia-
188. Meek DW. Tumour suppression by p53: a role for the DNA damage tion. J Biol Chem. 2003;278:14806–14811.
response?. Nat Rev Cancer. 2009;9:714–723. 211. Wang XQ, Redpath JL, Fan ST, Stanbridge EJ. ATR dependent acti-
189. Vazquez A, Bond EE, Levine AJ, Bond GL. The genetics of the vation of Chk2. J Cell Physiol. 2006;208:613–619.
p53 pathway, apoptosis and cancer therapy. Nat Rev Drug Discov. 212. Broderick R, Nasheuer HP. Regulation of Cdc45 in the cell cycle
2008;7:979–987. and after DNA damage. Biochem Soc Trans. 2009;37:926–930.
190. Viadiu H. Molecular architecture of tumor suppressor p53. Curr Top 213. Kitagawa R, Bakkenist CJ, McKinnon PJ, Kastan MB.
Med Chem. 2008;8:1327–1334. Phosphorylation of SMC1 is a critical downstream event in the
191. Varley J. TP53, hChk2, and the Li-Fraumeni syndrome. Methods ATM-NBS1-BRCA1 pathway. Genes Dev. 2004;18:1423–1438.
Mol Biol. 2003;222:117–129. 214. Kim ST, Xu B, Kastan MB. Involvement of the cohesin protein,
192. Schwartz D, Rotter V. p53-dependent cell cycle control: response to Smc1, in Atm-dependent and independent responses to DNA dam-
genotoxic stress. Semin Cancer Biol. 1998;8:325–336. age. Genes Dev. 2002;16:560–570.
Chapter  |  15  The Cell Cycle 467

215. Yazdi PT, Wang Y, Zhao S, Patel N, Lee EY, Qin J. SMC1 is a 237. Irniger S. Cyclin destruction in mitosis: a crucial task of Cdc20.
downstream effector in the ATM/NBS1 branch of the human FEBS Lett. 2002;532:7–11.
S-phase checkpoint. Genes Dev. 2002;16:571–582. 238. John PC, Mews M, Moore R. Cyclin/Cdk complexes: their involve-
216. Kops GJ, Weaver BA, Cleveland DW. On the road to can- ment in cell cycle progression and mitotic division. Protoplasma.
cer: aneuploidy and the mitotic checkpoint. Nat Rev Cancer. 2001;216:119–142.
2005;5:773–785. 239. Stemmann O, Gorr IH, Boos D. Anaphase topsy-turvy: Cdk1 a
217. Rieder CL, Maiato H. Stuck in division or passing through: what securin, separase a CKI. Cell Cycle. 2006;5:11–13.
happens when cells cannot satisfy the spindle assembly checkpoint. 240. Yanagida M. Cell cycle mechanisms of sister chromatid separation;
Dev Cell. 2004;7:637–651. roles of Cut1/separin and Cut2/securin. Genes Cells. 2000;5:1–8.
218. Malmanche N, Maia A, Sunkel CE. The spindle assembly check- 241. Nagao K, Yanagida M. Regulating sister chromatid separation by
point: preventing chromosome mis-segregation during mitosis and separase phosphorylation. Dev Cell. 2002;2:2–4.
meiosis. FEBS Lett. 2006;580:2888–2895. 242. Uhlmann F. Chromosome cohesion and separation: from men and
219. Shah JV, Cleveland DW. Waiting for anaphase: Mad2 and the spin- molecules. Curr Biol. 2003;13:R104–114.
dle assembly checkpoint. Cell. 2000;103:997–1000. 243. Pellman D, Christman MF. Separase anxiety: dissolving the sister
220. Hoyt MA, Totis L, Roberts BT. S. cerevisiae genes required for bond and more. Nat Cell Biol. 2001;3:E207–209.
cell cycle arrest in response to loss of microtubule function. Cell. 244. Agarwal R, Cohen-Fix O. Mitotic regulation: the fine tuning of sep-
1991;66:507–517. arase activity. Cell Cycle. 2002;1:255–257.
221. Li R, Murray AW. Feedback control of mitosis in budding yeast. 245. Uhlmann F. Separase regulation during mitosis. Biochem Soc Symp.
Cell. 1991;66:519–531. 2003:243–251.
222. Musacchio A, Hardwick KG. The spindle checkpoint: struc- 246. Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, et al.
tural insights into dynamic signalling. Nat Rev Mol Cell Biol. The genomic landscapes of human breast and colorectal cancers.
2002;3:731–741. Science. 2007;318:1108–1113.
223. Taylor SS, Scott MI, Holland AJ. The spindle checkpoint: a quality 247. Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al.
control mechanism which ensures accurate chromosome segrega- The consensus coding sequences of human breast and colorectal
tion. Chromosome Res. 2004;12:599–616. cancers. Science. 2006;314:268–274.
224. Kim SH, Lin DP, Matsumoto S, Kitazono A, Matsumoto T. Fission 248. Beijersbergen RL, Bernards R. Cell cycle regulation by the retino-
yeast Slp1: an effector of the Mad2-dependent spindle checkpoint. blastoma family of growth inhibitory proteins. Biochim Biophys
Science. 1998;279:1045–1047. Acta. 1996;1287:103–120.
225. Hwang LH, Lau LF, Smith DL, Mistrot CA, Hardwick KG, Hwang 249. Kaelin Jr. WG. Recent insights into the functions of the retinoblas-
ES, et al. Budding yeast Cdc20: a target of the spindle checkpoint. toma susceptibility gene product. Cancer Invest. 1997;15:243–254.
Science. 1998;279:1041–1044. 250. Sellers WR, Kaelin Jr. WG. Role of the retinoblastoma pro-
226. Simpson-Lavy KJ, Oren YS, Feine O, Sajman J, Listovsky T, tein in the pathogenesis of human cancer. J Clin Oncol.
Brandeis M. Fifteen years of APC/cyclosome: a short and impres- 1997;15:3301–3312.
sive biography. Biochem Soc Trans. 2010;38:78–82. 251. Nevins JR. The Rb/E2F pathway and cancer. Hum Mol Genet.
227. Wasch R, Robbins JA, Cross FR. The emerging role of APC/CCdh1 2001;10:699–703.
in controlling differentiation, genomic stability and tumor suppres- 252. Hahn WC, Weinberg RA. Modelling the molecular circuitry of can-
sion. Oncogene. 2010;29:1–10. cer. Nat Rev Cancer. 2002;2:331–341.
228. Pesin JA, Orr-Weaver TL. Regulation of APC/C activators in mito- 253. Ortega S, Malumbres M, Barbacid M. Cyclin D-dependent
sis and meiosis. Annu Rev Cell Dev Biol. 2008;24:475–499. kinases, INK4 inhibitors and cancer. Biochim Biophys Acta.
229. Lindon C. Control of mitotic exit and cytokinesis by the APC/C. 2002;1602:73–87.
Biochem Soc Trans. 2008;36:405–410. 254. Sherr CJ, McCormick F. The RB and p53 pathways in cancer.
230. Baker DJ, Dawlaty MM, Galardy P, van Deursen JM. Mitotic Cancer Cell. 2002;2:103–112.
regulation of the anaphase-promoting complex. Cell Mol Life Sci. 255. Sherr CJ. The INK4a/ARF network in tumour suppression. Nat Rev
2007;64:589–600. Mol Cell Biol. 2001;2:731–737.
231. Peters JM. The anaphase promoting complex/cyclosome: a machine 256. Kamb A, Shattuck-Eidens D, Eeles R, Liu Q, Gruis NA, Ding
designed to destroy. Nat Rev Mol Cell Biol. 2006;7:644–656. W, et  al. Analysis of the p16 gene (CDKN2) as a candidate for
232. Acquaviva C, Pines J. The anaphase-promoting complex/cyclos- the chromosome 9p melanoma susceptibility locus. Nat Genet.
ome: APC/C. J Cell Sci. 2006;119:2401–2404. 1994;8:23–26.
233. Vanoosthuyse V, Hardwick KG. Bub1 and the multilay- 257. Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and
ered inhibition of Cdc20-APC/C in mitosis. Trends Cell Biol. its relatives. Biochim Biophys Acta. 1998;1378:F115–177.
2005;15:231–233. 258. Levine AJ. p53, the cellular gatekeeper for growth and division.
234. Vodermaier HC. APC/C and SCF: controlling each other and the Cell. 1997;88:323–331.
cell cycle. Curr Biol. 2004;14:R787–796. 259. Hollstein M, Rice K, Greenblatt MS, Soussi T, Fuchs R, Sorlie T,
235. Hershko A. Mechanisms and regulation of the degradation of cyclin et al. Database of p53 gene somatic mutations in human tumors and
B. Philos Trans R Soc Lond B Biol Sci. 1999;354:1571–1575. [dis- cell lines. Nucleic Acids Res. 1994;22:3551–3555.
cussion 1575–1576] 260. Giaccia AJ, Kastan MB. The complexity of p53 modula-
236. Wolf F, Sigl R, Geley S. ‘… The end of the beginning’: cdk1 thresh- tion: emerging patterns from divergent signals. Genes Dev.
olds and exit from mitosis. Cell Cycle. 2007;6:1408–1411. 1998;12:2973–2983.
468 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

261. Gottifredi V, Shieh SY, Prives C. Regulation of p53 after different 284. Cairns P, Mao L, Merlo A, Lee DJ, Schwab D, Eby Y, et al. Rates
forms of stress and at different cell cycle stages. Cold Spring Harb of p16 (MTS1) mutations in primary tumors with 9p loss. Science.
Symp Quant Biol. 2000;65:483–488. 1994;265:415–417.
262. Helton ES, Chen X. p53 modulation of the DNA damage response. 285. Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour
J Cell Biochem. 2007;100:883–896. AB, et  al. Frequent somatic mutations and homozygous deletions
263. Sionov RV, Haupt Y. The cellular response to p53: the decision of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet.
between life and death. Oncogene. 1999;18:6145–6157. 1994;8:27–32.
264. Bates S, Vousden KH. Mechanisms of p53-mediated apoptosis. Cell 286. Quelle DE, Zindy F, Ashmun RA, Sherr CJ. Alternative read-
Mol Life Sci. 1999;55:28–37. ing frames of the INK4a tumor suppressor gene encode two
265. Corn PG, El-Deiry WS. Microarray analysis of p53-dependent gene unrelated proteins capable of inducing cell cycle arrest. Cell.
expression in response to hypoxia and DNA damage. Cancer Biol 1995;83:993–1000.
Ther. 2007;6:1858–1866. 287. Bashir T, Pagano M. Aberrant ubiquitin-mediated proteolysis of
266. Riley T, Sontag E, Chen P, Levine A. Transcriptional con- cell cycle regulatory proteins and oncogenesis. Adv Cancer Res.
trol of human p53-regulated genes. Nat Rev Mol Cell Biol. 2003;88:101–144.
2008;9:402–412. 288. Devoy A, Soane T, Welchman R, Mayer RJ. The ubiquitin-proteas-
267. Horvath MM, Wang X, Resnick MA, Bell DA. Divergent evolu- ome system and cancer. Essays Biochem. 2005;41:187–203.
tion of human p53 binding sites: cell cycle versus apoptosis. PLoS 289. Mani A, Gelmann EP. The ubiquitin-proteasome pathway and its
Genet. 2007;3:e127. role in cancer. J Clin Oncol. 2005;23:4776–4789.
268. Jen KY, Cheung VG. Identification of novel p53 target genes in ion- 290. Reed SI. The ubiquitin-proteasome pathway in cell cycle control.
izing radiation response. Cancer Res. 2005;65:7666–7673. Results Probl Cell Differ. 2006;42:147–181.
269. Mirza A, Wu Q, Wang L, McClanahan T, Bishop WR, Gheyas 291. Yamasaki L, Pagano M. Cell cycle, proteolysis and cancer. Curr
F, et  al. Global transcriptional program of p53 target genes dur- Opin Cell Biol. 2004;16:623–628.
ing the process of apoptosis and cell cycle progression. Oncogene. 292. Cenciarelli C, Chiaur DS, Guardavaccaro D, Parks W, Vidal M,
2003;22:3645–3654. Pagano M. Identification of a family of human F-box proteins. Curr
270. Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer Biol. 1999;9:1177–1179.
DD, Schuler M, et  al. Direct activation of Bax by p53 mediates 293. Kipreos ET, Pagano M. The F-box protein family. Genome Biol.
mitochondrial membrane permeabilization and apoptosis. Science. 2000;1 [REVIEWS3002].
2004;303:1010–1014. 294. Skaar JR, Pagano M. Control of cell growth by the SCF and APC/C
271. Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in ubiquitin ligases. Curr Opin Cell Biol. 2009;21:816–824.
vivo with a conserved homolog, Bax, that accelerates programmed 295. Nakayama KI, Nakayama K. Regulation of the cell cycle by SCF-
cell death. Cell. 1993;74:609–619. type ubiquitin ligases. Semin Cell Dev Biol. 2005;16:323–333.
272. Juven-Gershon T, Oren M. Mdm2: the ups and downs. Mol Med. 296. Peters JM. SCF and APC: the Yin and Yang of cell cycle regulated
1999;5:71–83. proteolysis. Curr Opin Cell Biol. 1998;10:759–768.
273. Zhang Y, Xiong Y. Control of p53 ubiquitination and nuclear export 297. Cardozo T, Pagano M. The SCF ubiquitin ligase: insights into a
by MDM2 and ARF. Cell Growth Differ. 2001;12:175–186. molecular machine. Nat Rev Mol Cell Biol. 2004;5:739–751.
274. Michael D, Oren M. The p53-Mdm2 module and the ubiquitin sys- 298. Nishitani H, Sugimoto N, Roukos V, Nakanishi Y, Saijo M, Obuse
tem. Semin Cancer Biol. 2003;13:49–58. C, et al. Two E3 ubiquitin ligases, SCF-Skp2 and DDB1-Cul4, tar-
275. Prives C. Signaling to p53: breaking the MDM2-p53 circuit. Cell. get human Cdt1 for proteolysis. EMBO J. 2006;25:1126–1136.
1998;95:5–8. 299. Zhang W, Koepp DM. Fbw7 isoform interaction contributes to cyc-
276. Piette J, Neel H, Marechal V. Mdm2: keeping p53 under control. lin E proteolysis. Mol Cancer Res. 2006;4:935–943.
Oncogene. 1997;15:1001–1010. 300. Nash P, Tang X, Orlicky S, Chen Q, Gertler FB, Mendenhall MD,
277. Lowe SW, Sherr CJ. Tumor suppression by Ink4a-Arf: progress and et al. Multisite phosphorylation of a CDK inhibitor sets a threshold
puzzles. Curr Opin Genet Dev. 2003;13:77–83. for the onset of DNA replication. Nature. 2001;414:514–521.
278. James MC, Peters G. Alternative product of the p16/CKDN2A locus 301. Tan Y, Sangfelt O, Spruck C. The Fbxw7/hCdc4 tumor suppressor
connects the Rb and p53 tumor suppressors. Prog Cell Cycle Res. in human cancer. Cancer Lett. 2008;271:1–12.
2000;4:71–81. 302. Fujii Y, Yada M, Nishiyama M, Kamura T, Takahashi H,
279. Serrano M. The INK4a/ARF locus in murine tumorigenesis. Tsunematsu R, et  al. Fbxw7 contributes to tumor suppression by
Carcinogenesis. 2000;21:865–869. targeting multiple proteins for ubiquitin-dependent degradation.
280. Sharpless NE. INK4a/ARF: a multifunctional tumor suppressor Cancer Sci. 2006;97:729–736.
locus. Mutat Res. 2005;576:22–38. 303. Fuchs SY. Tumor suppressor activities of the Fbw7 E3 ubiquitin
281. Bueso-Ramos CE, Yang Y, deLeon E, McCown P, Stass SA, Albitar ligase receptor. Cancer Biol Ther. 2005;4:506–508.
M. The human MDM-2 oncogene is overexpressed in leukemias. 304. Minella AC, Clurman BE. Mechanisms of tumor suppression by the
Blood. 1993;82:2617–2623. SCF(Fbw7). Cell Cycle. 2005;4:1356–1359.
282. Oliner JD, Kinzler KW, Meltzer PS, George DL, Vogelstein B. 305. Akhoondi S, Sun D, von der Lehr N, Apostolidou S, Klotz K,
Amplification of a gene encoding a p53-associated protein in Maljukova A, et al. FBXW7/hCDC4 is a general tumor suppressor
human sarcomas. Nature. 1992;358:80–83. in human cancer. Cancer Res. 2007;67:9006–9012.
283. Borg A, Johannsson U, Johannsson O, Hakansson S, Westerdahl J, 306. Furstenthal L, Kaiser BK, Swanson C, Jackson PK. Cyclin E uses
Masback A, et al. Novel germline p16 mutation in familial malignant Cdc6 as a chromatin-associated receptor required for DNA replica-
melanoma in southern Sweden. Cancer Res. 1996;56:2497–2500. tion. J Cell Biol. 2001;152:1267–1278.
Chapter  |  15  The Cell Cycle 469

307. Ekholm-Reed S, Mendez J, Tedesco D, Zetterberg A, Stillman B, 328. Pihan GA, Doxsey SJ. The mitotic machinery as a source of genetic
Reed SI. Deregulation of cyclin E in human cells interferes with instability in cancer. Semin Cancer Biol. 1999;9:289–302.
prereplication complex assembly. J Cell Biol. 2004;165:789–800. 329. Tomonaga T, Nomura F. Chromosome instability and kinetochore
308. Rajagopalan H, Lengauer C. hCDC4 and genetic instability in can- dysfunction. Histol Histopathol. 2007;22:191–197.
cer. Cell Cycle. 2004;3:693–694. 330. Bharadwaj R, Yu H. The spindle checkpoint, aneuploidy, and can-
309. Rajagopalan H, Jallepalli PV, Rago C, Velculescu VE, Kinzler KW, cer. Oncogene. 2004;23:2016–2027.
Vogelstein B, et al. Inactivation of hCDC4 can cause chromosomal 331. Michel LS, Liberal V, Chatterjee A, Kirchwegger R, Pasche B,
instability. Nature. 2004;428:77–81. Gerald W, et  al. MAD2 haplo-insufficiency causes premature ana-
310. Spruck CH, Won KA, Reed SI. Deregulated cyclin E induces chro- phase and chromosome instability in mammalian cells. Nature.
mosome instability. Nature. 1999;401:297–300. 2001;409:355–359.
311. Hanashiro K, Kanai M, Geng Y, Sicinski P, Fukasawa K. Roles of 332. Kalitsis P, Earle E, Fowler KJ, Choo KH. Bub3 gene disruption in
cyclins A and E in induction of centrosome amplification in p53- mice reveals essential mitotic spindle checkpoint function during
compromised cells. Oncogene. 2008;27:5288–5302. early embryogenesis. Genes Dev. 2000;14:2277–2282.
312. Mussman JG, Horn HF, Carroll PE, Okuda M, Tarapore P, 333. Iwanaga Y, Chi YH, Miyazato A, Sheleg S, Haller K, Peloponese Jr.
Donehower LA, et  al. Synergistic induction of centrosome JM, et  al. Heterozygous deletion of mitotic arrest-deficient protein
hyperamplification by loss of p53 and cyclin E overexpression. 1 (MAD1) increases the incidence of tumors in mice. Cancer Res.
Oncogene. 2000;19:1635–1646. 2007;67:160–166.
313. Kawamura K, Izumi H, Ma Z, Ikeda R, Moriyama M, Tanaka T, 334. Babu JR, Jeganathan KB, Baker DJ, Wu X, Kang-Decker N, van
et al. Induction of centrosome amplification and chromosome insta- Deursen JM. Rae1 is an essential mitotic checkpoint regulator that
bility in human bladder cancer cells by p53 mutation and cyclin E cooperates with Bub3 to prevent chromosome missegregation.
overexpression. Cancer Res. 2004;64:4800–4809. J Cell Biol. 2003;160:341–353.
314. Simone C, Resta N, Bagella L, Giordano A, Guanti G. Cyclin E and 335. Perera D, Tilston V, Hopwood JA, Barchi M, Boot-Handford RP,
chromosome instability in colorectal cancer cell lines. Mol Pathol. Taylor SS. Bub1 maintains centromeric cohesion by activation of
2002;55:200–203. the spindle checkpoint. Dev Cell. 2007;13:566–579.
315. Koepp DM, Schaefer LK, Ye X, Keyomarsi K, Chu C, Harper JW, 336. Putkey FR, Cramer T, Morphew MK, Silk AD, Johnson RS,
et  al. Phosphorylation-dependent ubiquitination of cyclin E by the McIntosh JR, et  al. Unstable kinetochore-microtubule capture and
SCFFbw7 ubiquitin ligase. Science. 2001;294:173–177. chromosomal instability following deletion of CENP-E. Dev Cell.
316. Hao B, Oehlmann S, Sowa ME, Harper JW, Pavletich NP. 2002;3:351–365.
Structure of a Fbw7-Skp1-cyclin E complex: multisite-phospho- 337. Dai W, Wang Q, Liu T, Swamy M, Fang Y, Xie S, et al. Slippage of
rylated substrate recognition by SCF ubiquitin ligases. Mol Cell. mitotic arrest and enhanced tumor development in mice with BubR1
2007;26:131–143. haploinsufficiency. Cancer Res. 2004;64:440–445.
317. Strohmaier H, Spruck CH, Kaiser P, Won KA, Sangfelt O, Reed SI. 338. Wang Q, Liu T, Fang Y, Xie S, Huang X, Mahmood R, et  al.
Human F-box protein hCdc4 targets cyclin E for proteolysis and is BUBR1 deficiency results in abnormal megakaryopoiesis. Blood.
mutated in a breast cancer cell line. Nature. 2001;413:316–322. 2004;103:1278–1285.
318. Grady WM, Carethers JM. Genomic and epigenetic insta- 339. Baker DJ, Jeganathan KB, Cameron JD, Thompson M, Juneja
bility in colorectal cancer pathogenesis. Gastroenterology. S, Kopecka A, et  al. BubR1 insufficiency causes early onset of
2008;135:1079–1099. aging-associated phenotypes and infertility in mice. Nat Genet.
319. Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in color- 2004;36:744–749.
ectal cancers. Nature. 1997;386:623–627. 340. Jeganathan K, Malureanu L, Baker DJ, Abraham SC, van Deursen
320. Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in JM. Bub1 mediates cell death in response to chromosome missegre-
human cancers. Nature. 1998;396:643–649. gation and acts to suppress spontaneous tumorigenesis. J Cell Biol.
321. Rajagopalan H, Lengauer C. CIN-ful cancers. Cancer Chemother 2007;179:255–267.
Pharmacol. 2004;54(suppl 1):S65–68. 341. Cahill DP, Lengauer C, Yu J, Riggins GJ, Willson JK, Markowitz
322. Michor F, Iwasa Y, Vogelstein B, Lengauer C, Nowak MA. Can SD, et al. Mutations of mitotic checkpoint genes in human cancers.
chromosomal instability initiate tumorigenesis?. Semin Cancer Biol. Nature. 1998;392:300–303.
2005;15:43–49. 342. Shichiri M, Yoshinaga K, Hisatomi H, Sugihara K, Hirata Y.
323. Rajagopalan H, Lengauer C. Aneuploidy and cancer. Nature. Genetic and epigenetic inactivation of mitotic checkpoint genes
2004;432:338–341. hBUB1 and hBUBR1 and their relationship to survival. Cancer Res.
324. Rajagopalan H, Nowak MA, Vogelstein B, Lengauer C. The sig- 2002;62:13–17.
nificance of unstable chromosomes in colorectal cancer. Nat Rev 343. Gemma A, Seike M, Seike Y, Uematsu K, Hibino S, Kurimoto
Cancer. 2003;3:695–701. F, et  al. Somatic mutation of the hBUB1 mitotic checkpoint
325. Shih IM, Zhou W, Goodman SN, Lengauer C, Kinzler KW, gene in primary lung cancer. Genes Chromosomes Cancer.
Vogelstein B. Evidence that genetic instability occurs at an early 2000;29:213–218.
stage of colorectal tumorigenesis. Cancer Res. 2001;61:818–822. 344. Hempen PM, Kurpad H, Calhoun ES, Abraham S, Kern SE. A dou-
326. Thompson SL, Bakhoum SF, Compton DA. Mechanisms of chro- ble missense variation of the BUB1 gene and a defective mitotic
mosomal instability. Curr Biol. 2010;20:R285–295. spindle checkpoint in the pancreatic cancer cell line Hs766T. Hum
327. Lingle WL, Lukasiewicz K, Salisbury JL. Deregulation of the cen- Mutat. 2003;21:445.
trosome cycle and the origin of chromosomal instability in cancer. 345. Imai Y, Shiratori Y, Kato N, Inoue T, Omata M. Mutational inac-
Adv Exp Med Biol. 2005;570:393–421. tivation of mitotic checkpoint genes, hsMAD2 and hBUB1,
470 SECTION  |  I  Basic Cell Physiology, Genetics, and Growth of the GI Tract

is rare in sporadic digestive tract cancers. Jpn J Cancer Res. 365. Kikuchi A. Regulation of beta-catenin signaling in the Wnt path-
1999;90:837–840. way. Biochem Biophys Res Commun. 2000;268:243–248.
346. Ohshima K, Haraoka S, Yoshioka S, Hamasaki M, Fujiki T, 366. Haegebarth A, Clevers H. Wnt signaling, lf15-05-9780123820266,
Suzumiya J, et  al. Mutation analysis of mitotic checkpoint genes and stem cells in the intestine and skin. Am J Pathol.
(hBUB1 and hBUBR1) and microsatellite instability in adult T-cell 2009;174:715–721.
leukemia/lymphoma. Cancer Lett. 2000;158:141–150. 367. van der Flier LG, Clevers H. Stem cells, self-renewal, and dif-
347. Nomoto S, Haruki N, Takahashi T, Masuda A, Koshikawa T, ferentiation in the intestinal epithelium. Annu Rev Physiol.
Fujii Y, et  al. Search for in vivo somatic mutations in the mitotic 2009;71:241–260.
checkpoint gene, hMAD1, in human lung cancers. Oncogene. 368. Barker N, van de Wetering M, Clevers H. The intestinal stem cell.
1999;18:7180–7183. Genes Dev. 2008;22:1856–1864.
348. Tsukasaki K, Miller CW, Greenspun E, Eshaghian S, Kawabata 369. de Lau W, Barker N, Clevers H. WNT signaling in the normal intes-
H, Fujimoto T, et  al. Mutations in the mitotic check point gene, tine and colorectal cancer. Front Biosci. 2007;12:471–491.
MAD1L1, in human cancers. Oncogene. 2001;20:3301–3305. 370. Pinto D, Clevers H. Wnt control of stem cells and differentiation in
349. Wang Z, Cummins JM, Shen D, Cahill DP, Jallepalli PV, Wang the intestinal epithelium. Exp Cell Res. 2005;306:357–363.
TL, et al. Three classes of genes mutated in colorectal cancers with 371. Pinto D, Clevers H. Wnt, stem cells and cancer in the intestine. Biol
chromosomal instability. Cancer Res. 2004;64:2998–3001. Cell. 2005;97:185–196.
350. Hanks S, Coleman K, Reid S, Plaja A, Firth H, Fitzpatrick D, et al. 372. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature.
Constitutional aneuploidy and cancer predisposition caused by bial- 2005;434:843–850.
lelic mutations in BUB1B. Nat Genet. 2004;36:1159–1161. 373. Yang VW. APC as a checkpoint gene: the beginning or the end?
351. Suijkerbuijk SJ, van Osch MH, Bos FL, Hanks S, Rahman N, Kops Gastroenterology. 2002;123:935–939.
GJ. Molecular causes for BUBR1 dysfunction in the human cancer 374. Bustos VH, Ferrarese A, Venerando A, Marin O, Allende JE, Pinna
predisposition syndrome mosaic variegated aneuploidy. Cancer Res. LA. The first armadillo repeat is involved in the recognition and
2010;70:4891–4900. regulation of beta-catenin phosphorylation by protein kinase CK1.
352. Hanks S, Coleman K, Summersgill B, Messahel B, Williamson Proc Natl Acad Sci U S A. 2006;103:19725–19730.
D, Pritchard-Jones K, et  al. Comparative genomic hybridiza- 375. Marikawa Y, Elinson RP. beta-TrCP is a negative regulator of
tion and BUB1B mutation analyses in childhood cancers associ- Wnt/beta-catenin signaling pathway and dorsal axis formation in
ated with mosaic variegated aneuploidy syndrome. Cancer Lett. Xenopus embryos. Mech Dev. 1998;77:75–80.
2006;239:234–238. 376. Winston JT, Strack P, Beer-Romero P, Chu CY, Elledge SJ, Harper
353. Hanks S, Rahman N. Aneuploidy-cancer predisposition syndromes: JW. The SCFbeta-TRCP-ubiquitin ligase complex associates specif-
a new link between the mitotic spindle checkpoint and cancer. Cell ically with phosphorylated destruction motifs in IkappaBalpha and
Cycle. 2005;4:225–227. beta-catenin and stimulates IkappaBalpha ubiquitination in vitro.
354. Kinzler KW, Nilbert MC, Su LK, Vogelstein B, Bryan TM, Levy Genes Dev. 1999;13:270–283.
DB, et  al. Identification of FAP locus genes from chromosome 377. Latres E, Chiaur DS, Pagano M. The human F box protein beta-
5q21. Science. 1991;253:661–665. Trcp associates with the Cul1/Skp1 complex and regulates the sta-
355. Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A, et al. bility of beta-catenin. Oncogene. 1999;18:849–854.
Mutations of chromosome 5q21 genes in FAP and colorectal cancer 378. Sakanaka C. Phosphorylation and regulation of beta-catenin by
patients. Science. 1991;253:665–669. casein kinase I epsilon. J Biochem. 2002;132:697–703.
356. Nakamura Y, Nishisho I, Kinzler KW, Vogelstein B, Miyoshi Y, 379. Gao ZH, Seeling JM, Hill V, Yochum A, Virshup DM. Casein kinase
Miki Y, et  al. Mutations of the APC (adenomatous polyposis coli) I phosphorylates and destabilizes the beta-catenin degradation com-
gene in FAP (familial polyposis coli) patients and in sporadic color- plex. Proc Natl Acad Sci U S A. 2002;99:1182–1187.
ectal tumors. Tohoku J Exp Med. 1992;168:141–147. 380. Rubinfeld B, Albert I, Porfiri E, Fiol C, Munemitsu S, Polakis P.
357. Kinzler KW, Vogelstein B. Lessons from hereditary colorectal can- Binding of GSK3beta to the APC-beta-catenin complex and regula-
cer. Cell. 1996;87:159–170. tion of complex assembly. Science. 1996;272:1023–1026.
358. Su LK, Vogelstein B, Kinzler KW. Association of the APC tumor 381. Hart MJ, de los Santos R, Albert IN, Rubinfeld B, Polakis P.
suppressor protein with catenins. Science. 1993;262:1734–1737. Downregulation of beta-catenin by human Axin and its association
359. Rubinfeld B, Souza B, Albert I, Muller O, Chamberlain SH, with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr
Masiarz FR, et al. Association of the APC gene product with beta- Biol. 1998;8:573–581.
catenin. Science. 1993;262:1731–1734. 382. Shtutman M, Zhurinsky J, Simcha I, Albanese C, D'Amico M,
360. Gumbiner BM. Signal transduction of beta-catenin. Curr Opin Cell Pestell R, et al. The cyclin D1 gene is a target of the beta-catenin/
Biol. 1995;7:634–640. LEF-1 pathway. Proc Natl Acad Sci U S A. 1999;96:5522–5527.
361. Bienz M. beta-Catenin: a pivot between cell adhesion and Wnt sig- 383. He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa
nalling. Curr Biol. 2005;15:R64–67. LT, et al. Identification of c-MYC as a target of the APC pathway.
362. Willert K, Nusse R. Beta-catenin: a key mediator of Wnt signaling. Science. 1998;281:1509–1512.
Curr Opin Genet Dev. 1998;8:95–102. 384. Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin
363. Novak A, Dedhar S. Signaling through beta-catenin and Lef/Tcf. D1 in colon carcinoma cells. Nature. 1999;398:422–426.
Cell Mol Life Sci. 1999;56:523–537. 385. Green RA, Kaplan KB. Chromosome instability in colorectal
364. Moon RT, Bowerman B, Boutros M, Perrimon N. The prom- tumor cells is associated with defects in microtubule plus-end
ise and perils of Wnt signaling through beta-catenin. Science. attachments caused by a dominant mutation in APC. J Cell Biol.
2002;296:1644–1646. 2003;163:949–961.
Chapter  |  15  The Cell Cycle 471

386. Green RA, Wollman R, Kaplan KB. APC and EB1 function together 391. Fodde R, Kuipers J, Rosenberg C, Smits R, Kielman M, Gaspar C,
in mitosis to regulate spindle dynamics and chromosome alignment. et al. Mutations in the APC tumour suppressor gene cause chromo-
Mol Biol Cell. 2005;16:4609–4622. somal instability. Nat Cell Biol. 2001;3:433–438.
387. Kaplan KB, Burds AA, Swedlow JR, Bekir SS, Sorger PK, Nathke 392. Caldwell CM, Kaplan KB. The role of APC in mitosis and in chro-
IS. A role for the Adenomatous Polyposis Coli protein in chromo- mosome instability. Adv Exp Med Biol. 2009;656:51–64.
some segregation. Nat Cell Biol. 2001;3:429–432. 393. Tighe A, Johnson VL, Taylor SS. Truncating APC mutations have
388. Kita K, Wittmann T, Nathke IS, Waterman-Storer CM. dominant effects on proliferation, spindle checkpoint control, sur-
Adenomatous polyposis coli on microtubule plus ends in cell exten- vival and chromosome stability. J Cell Sci. 2004;117:6339–6353.
sions can promote microtubule net growth with or without EB1. 394. Abal M, Obrador-Hevia A, Janssen KP, Casadome L, Menendez M,
Mol Biol Cell. 2006;17:2331–2345. Carpentier S, et al. APC inactivation associates with abnormal mito-
389. Mogensen MM, Tucker JB, Mackie JB, Prescott AR, Nathke IS. sis completion and concomitant BUB1B/MAD2L1 up-regulation.
The adenomatous polyposis coli protein unambiguously localizes to Gastroenterology. 2007;132:2448–2458.
microtubule plus ends and is involved in establishing parallel arrays 395. Aoki K, Aoki M, Sugai M, Harada N, Miyoshi H, Tsukamoto T,
of microtubule bundles in highly polarized epithelial cells. J Cell et al. Chromosomal instability by beta-catenin/TCF transcription in
Biol. 2002;157:1041–1048. APC or beta-catenin mutant cells. Oncogene. 2007;26:3511–3520.
390. Fodde R, Smits R, Clevers H. APC, signal transduction and genetic
instability in colorectal cancer. Nat Rev Cancer. 2001;1:55–67.

You might also like