You are on page 1of 120

SECTION X OVERVIEW

T his last section of the book draws together principles to carry out specific functions and then no longer pro-
from previous chapters to explain some of the rules that liferate. How do cells decide whether to proliferate, to
govern the lifestyles of cells. Cells exhibit a remarkable stop proliferating and differentiate, or to die? This section
diversity in their patterns of growth, proliferation, and answers these and other questions.
death. For example, some human cells (neurons) are Chapter 40 begins the section with an introduction
born around the time of birth and live until the person to the language of the cell cycle. The cell cycle is driven
dies—more than 100 years in a few cases. The fate of by changing states of the cytoplasm created by shifting
other cells is to live for only a day or two (eg, cells in balances of protein phosphorylation, dephosphoryla-
the gut lining). Many differentiated cells form by elabo- tion, and degradation machinery. For the cell cycle, the
rate pathways that employ a carefully choreographed key kinases are cyclin-dependent kinases (Cdks),
series of cues from within the cell and from its neigh- which require an associated cyclin subunit for activity.
bors. Other cells, such as many in the immune system, Cdks are also regulated by phosphorylation and by
are spawned in excess, followed by selection of the few additional protein cofactors that bind and inactivate
with correctly rearranged genes or with productive con- them. Cdks are usually stable, but cyclin levels fluctuate,
nections to partner cells. The unlucky majority of their owing to targeted destruction at particular points in
siblings whose differentiation did not go so well ulti- the cell cycle. In fact, targeted proteolytic destruction
mately commit suicide. by the proteasome is a key aspect of cell-cycle control.
Very different strategies maintain populations of cells. Each cell-cycle phase is characterized by the activity of
Long-lived cells divide seldom, if at all. In contrast, the one or more E3 ubiquitin ligases. Each of these targets
cells that are involved in producing the gut lining grow particular proteins for destruction by decorating them
and divide at top speed. Most human cells differentiate with chains of ubiquitin, a protein that was introduced

Meiosis Ch 45 Mitosis Ch 44
Programmed cell death Ch 46

G2 phase and Active CDK


control of entry Normal cell arrested
into mitosis Ch 43 Introduction to the at restriction point
cell cycle Ch 40 STOP

Rb
G1 phase and the
regulation of cell
S phase and DNA proliferation Ch 41
replication Ch 42 Cancer cell passes
restriction point
GO
Rb
Danger

695
in Chapter 23. This sequential destruction of key factors has been studied since the 1800s, but technical advances
gives cell-cycle transitions their irreversible character. have considerably advanced our understanding of how
The chapters that follow explain how the cell-cycle it is accomplished at the molecular level. Division
machinery controls each step in the proliferation and requires wholesale reorganization of cellular structures,
differentiation of cells. Chapter 41 begins with newly including chromosome condensation and the assembly
born cells in the G1 phase of the cell cycle. These cells of the mitotic spindle. In many cells, the nuclear enve-
must decide whether to commit themselves to a round lope breaks down. Once the chromosomes are all
of proliferation or to withdraw from the proliferation rat attached to the microtubules of the mitotic spindle (yet
race and enter a nondividing differentiated state called another important checkpoint here), they are separated
G0. Cells that will proliferate must first pass a control equally and form two daughter nuclei. Finally, cytoki-
point known as the restriction point. This is a control nesis separates the two daughter cells.
circuit that determines whether internal and external Chapter 45 considers meiosis, a specialized form of
conditions are suitable for proliferation. Malfunctions of division that produces the gametes required for sexual
the restriction point lead to one of the most terrifying reproduction. In this division, DNA recombination is key
perturbations of the cell cycle: cancer. If DNA damage to segregation of the chromosomes. A number of arcane
is detected during G1 phase, a checkpoint halts cell terms are used to describe the specialized structures
cycle progression until either the damage is repaired or and processes involved. The chapter then explains how
the cell dies. The chapter includes a section on stem problems with meiosis can lead to genetic diseases and
cells and concludes by considering the role of one of how studies of chromosome segregation in yeast led to
the most famous cell-cycle proteins, p53, in cell-cycle an understanding of why birth defects become more
control. prevalent as human mothers age.
Cells that decide to proliferate must replicate their Chapter 46 closes the book with a discussion of what
DNA in a timely and accurate manner. Chapter 42 happens when cells commit suicide by apoptosis,
explains the mechanism of DNA replication during the necroptosis, and autophagy. This is not, strictly speak-
S phase, including the selection of sites on DNA to initi- ing, a cell-cycle event but instead represents several alter-
ate replication, the enzymes that copy the DNA, the native pathways, each with its own machinery and
regulation of replication by the cell-cycle machinery, signaling systems. Apoptosis sometimes results when it
the organization of replicating chromosomes within the all “runs off the rails” and cells receive insults from which
nucleus, and the checkpoints that help the cells to cope they cannot recover. But cell death is not always bad:
with various problems that they encounter along the apoptosis is an essential part of development of meta-
way. Chapter 43 discusses the G2 phase, during which zoan organisms, homeostasis of their organs and tissues,
cells conduct a final “cockpit check” before embarking and can be a last-ditch defense against viral infection.
on the irreversible process of division. This is also the Malfunctions of apoptotic pathways can lead to cancer.
last point in the cell cycle at which the genome is The concepts that are discussed in this section of the
scanned for damage so that it can be repaired before book build on the ideas in earlier sections. Cells are
division. A checkpoint restrains cells from entering into wonderfully complex systems whose behavior is driven
mitosis if damaged DNA is detected, and the chapter by the laws of chemistry and physics. A major challenge
briefly explains the major pathways of DNA repair. for cell biology in the future is to devise molecular expla-
Chapter 44 describes mitosis, certainly the most nations for the complex behaviors exhibited in this
dramatic and complex program in the cell cycle. Mitosis closing section of our book.

696
CHAPTER 40 

Introduction to the Cell Cycle

The cell cycle is the series of events that leads to the (Fig. 40.2). The cell cycle integrates a continuous growth
duplication and division of a cell. Research on the molec- cycle (an increase in cell mass) with a discontinuous
ular events of cell-cycle control revealed that variations division or chromosome cycle (the replication and
of similar mechanisms operate the cell cycles of all partitioning of the genome into two daughter cells). The
eukaryotes from yeasts to humans. Furthermore, the chromosome cycle is driven by a sequence of enzy-
components that regulate cell growth and division also matic cascades that produce a sequence of discrete
play key roles in the cessation of cell division that is biochemical “states” of the cytoplasm. Progress through
required for cells to differentiate. Control of the cell the cell cycle is ratchet-like and irreversible because each
cycle is of major importance to human health because new state arises not only by expression or activation of
cancer is usually caused by perturbations of cell-cycle a new cohort of activities, but also by destruction or
regulation. Based on 2010 to 2012 data, 40% of Ameri- inactivation of key activities characteristic of the pre-
cans will develop cancer during their lifetime. ceding state. Later sections of this chapter explain
Although animal cells have a wide variety of special- these mechanisms.
ized cell cycles, the cells in the stratified epithelium
that forms skin illustrate the most common types of cell
cycles (Fig. 40.1). The basal layer of the epithelium is
Phases of the Cell Cycle
composed of stem cells that divide only occasionally In describing the cell cycle, it is convenient to divide the
(see Box 41.2). They can activate the cell cycle on process into several phases. Recognition of these phases
demand and then return to a nondividing state. Stem cell began in 1882, when Flemming named the process of
populations can replenish themselves by symmetrical nuclear division mitosis (from the Greek mito, or
division, but when specific signals induce them to pro- “thread”) after he first observed the condensed chromo-
liferate, usually one daughter cell remains a stem cell and somes. Mitosis was a clear cell cycle landmark, and the
the other enters a pool of rapidly dividing cells. The rest of the cell cycle between mitoses was called inter-
dividing cells populate the upper layers of the epithe- phase (Box 40.1).
lium, stop dividing, and gradually differentiate into the Once DNA was recognized as the agent of heredity in
specialized cells that cover the surface. the 1940s, it was deduced that it must be duplicated
Like stem cells, fibroblasts of the connective tissue at some time during interphase so that daughter cells
(see Fig. 28.2) typically are in a nondividing state, but can each receive a full complement of genetic material.
they can be stimulated to enter the cell cycle following In 1953, a key experiment identified the relationship
wounding or other stimuli (see Fig. 32.11). In the most between the timing of DNA synthesis and the mitotic
extreme case, the nervous system contains a few stem cycle (Fig. 40.3). This defined the four cell-cycle phases
cells and a few dividing glial cells, but most neurons, as they are known today (see Fig. 40.2).
once differentiated, can live for more than 100 years Each cell is born at the completion of the M phase,
without dividing again. which includes mitosis, the partitioning of the chromo-
somes and other cellular components, and cytokinesis,
the division of the cytoplasm. The chromosomal DNA is
Principles of Cell-Cycle Regulation replicated during S phase (synthetic phase). The remain-
The goal of the cell cycle in most cases is to produce ing two phases are gaps between mitosis and the S
two daughter cells that are accurate copies of the parent phase. The G1 phase (first gap phase) is the interval

697
698 SECTION X  n  Cell Cycle

Death Final stage of


differentiation
in skin

Cessation
of cycling
Terminal
differentiation

Rapid
cell cycles
Expansion of
population

Infrequent
cell cycles
Renewal of
Activation stem cell
population

A B Stem cell

FIGURE 40.1  CELL CYCLES IN A STRATIFIED EPITHELIUM. A, Light micrograph of a section of skin, a stratified squamous epithelium,
stained with hematoxylin and eosin (H&E). B, Diagram showing the different types of cell cycles at the various levels of this epithelium.

A. Cell-cycle details Mitosis


B. Cycle phases in cultured cell
(not to scale)
M Cell-cycle phase Length (hours)
G1 10
Check for Check for
damaged or chromosome S 7.5
unduplicated attachment to Cytokinesis
G2 3.5
DNA mitotic spindle
M 1.0
Generation time 22
DNA
ENLARGED VIEW G0
OF CHROMOSOME

C. Time-scaled diagram
G2 (times in hours)
Growth G
1
in mass 22 0
21
Cohesion M
established
in S phase Check for G2
DNA damage
17.5
G1
Check for DNA Restriction point:
S
damage or stalled pass only if environmental
replication forks conditions favorable
S
Chromosome Centrosome
duplication duplication starts 10

FIGURE 40.2  INTRODUCTION TO THE CELL-CYCLE PHASES. A, Diagrams of cellular morphology and chromosome structure across
the cell cycle. B, Length of cell-cycle phases in cultured cells. C, Time scale of cell-cycle phases.
CHAPTER 40  n  Introduction to the Cell Cycle 699

BOX 40.1  Selected Key Terms

M phase: Cell division, comprising mitosis, when a fully


grown cell segregates the replicated chromosomes to
opposite ends of a molecular scaffold, termed the Nucleus Cell synthesizing DNA
spindle, and cytokinesis, when the cell cleaves between
the separated chromosomes to produce two daughter
cells. In general, each daughter cell receives a comple-
Add 32P, incubate briefly,
ment of genetic material and organelles identical to then wash out free 32P
that of the parent cell.
Interphase: The portion of the cell cycle when cells grow
and replicate their DNA. Interphase has three sections.
The G1 (first gap) phase is the interval between mitosis
and the onset of DNA replication. The S (synthetic)
phase is the time when DNA is replicated. The G2 Photographic
(second gap) phase is the interval between the termina- emulsion
tion of DNA replication and the onset of mitosis. In
multicellular organisms, many differentiated cells no
longer actively divide. These nondividing cells (which
Cell exposes
may physiologically be extremely active) are in the G0 photographic
phase, a branch of the G1 phase. emulsion
Checkpoints: Biochemical circuits that regulate cell-cycle
transitions in response to the physiological condition
of the cell and signals from its environment. Check- VIEW IN
MICROSCOPE
points detect damage to the DNA due to external
agents or problems that arise during DNA replication
and trigger the DNA damage response. Other check-
points detect problems that arise during attachment of
chromosomes to the spindle.

between mitosis and the start of DNA replication. The


G2 phase (second gap phase) is the interval between the 20% of cells turn the emulsion black
completion of DNA replication and mitosis. All cycling FIGURE 40.3  DISCOVERY OF CELL-CYCLE STAGES. To deter-
cells have an M phase and an S phase. The G1 and G2 mine whether cells synthesize DNA during a defined portion of the cell
phases vary in length and are very short in some early cycle or constantly throughout the entire cycle (as is the case in bac-
teria, for example), Howard and Pelc fed a radioactive component of
embryos. The following sections describe the stages of
DNA (32P) to onion root tip cells, spread the cells in a thin layer on a
the cell cycle, starting just after the birth of the cell. microscope slide, washed away the 32P that had not become incor-
porated into DNA, and overlayered the slide with photographic emul-
G1 Phase sion. After incubation in the dark, the emulsion was developed like film
G1 is typically the longest and most variable cell-cycle and examined with a light microscope. The nuclei of cells active in
replicating their DNA incorporated the radioactive 32P into DNA and
phase. When cells are “born” at cytokinesis, they are
exposed the photographic emulsion above them. Two possible out-
roughly half the size they were before mitosis, and comes were predicted. If cells synthesized DNA constantly during
during G1, they grow back toward an optimal size. interphase, then all cells would incorporate the radioactive label. Con-
During this time, many activities involved in cell-cycle versely, if each cell synthesized DNA only during a discrete portion of
progression are repressed so that the cell cannot initiate the cell cycle, then only cells engaged in active replication during the
period of exposure to 32P would expose the photographic emulsion.
a new round of proliferation. This repressive control
When the slides were examined, 20% of the interphase cell nuclei were
system is called the restriction point. If the supply of labeled, proving that cells synthesize DNA only during a discrete
nutrients is poor or if cells receive an antiproliferative portion of interphase. Mitotic cells were unlabeled. Assuming that the
stimulus such as a signal to embark on terminal differen- cells traverse the cycle at a more or less constant rate, it was possible
tiation, they delay their progress through the cell cycle to calculate the length of the synthetic phase. Overall, the time between
successive divisions—the generation time—was approximately 30
in G1 or exit the cycle to enter G0 (see “G0 and Growth
hours in the root tip cells. If approximately 20% of the cells were
Control” below). However, if appropriate positive stimuli labeled, then approximately 20% of the 30-hour generation time must
are received, cells overcome the restriction point block be spent in DNA synthesis. Thus, 0.2 × 30, or 6 hours, was spent in
and trigger a program of gene expression that commits replication. (Data from Pelc HA Sr. Synthesis of DNA in normal and
them to a new cycle of DNA replication and cell division. irradiated cells and its relation to chromosome breakage. Heredity
Suppl. 1953;6:261–273.)
Cancer cells often have defects in restriction point
700 SECTION X  n  Cell Cycle

control and continue to grow and attempt to divide even critical threshold level, the cell enters mitosis. Along the
in the absence of appropriate environmental signals. way, the chromatin and cytoskeleton are prepared for
the dramatic structural changes that will occur during
G0 and Growth Control mitosis. If damaged DNA is detected during G2, a check-
Most cells of multicellular organisms differentiate to point activates the DNA damage response and delays
carry out specialized functions and no longer divide. entry of the cell into mitosis.
Such cells are considered to be in the G0 phase. Cells
often enter G0 directly as they exit their last mitosis. G0 M Phase
cells are not dormant; indeed, they are often actively During M phase (mitosis and the subsequent cytokine-
engaged in protein synthesis and secretion, and they may sis), chromosomes and cytoplasm are partitioned into
be highly motile. Many G0 cells have a nonmotile primary two daughter cells. Mitosis is normally divided into five
cilium, which is an important sensory organelle (see Fig. discrete phases.
38.19). The G0 phase is not necessarily permanent. In Prophase is defined by the onset of chromosome
some specialized cases, G0 cells may be recruited to condensation and is actually the final part of G2 phase.
reenter the cell cycle in response to specific stimuli. TADs disassemble inside the intact nucleus and mitotic
Cell-cycle reentry involves changes in gene expression chromosomes begin to form their characteristic array
and protein stability and disassembly of the primary of loops (see Chapter 8). In the cytoplasm, a dramatic
cilium, if present. This process must be highly regulated, change in the dynamic properties of the microtubules
as the uncontrolled proliferation of cells in a multicel- decreases their half-lives from approximately 10 minutes
lular organism can lead to cancer. to approximately 30 seconds. The duplicated centro-
somes (centrioles and associated pericentriolar material
S Phase in animal cells; see Fig. 34.14) separate and form the two
Chromosomes of higher eukaryotes are so large that poles of the mitotic spindle.
replication of the DNA must be initiated at many differ- Prometaphase begins when the nuclear envelope
ent sites, termed origins of replication. In budding breaks down (in higher eukaryotes) and chromosomes
yeast, the approximately 400 origins are spaced an begin to attach randomly to microtubules emanating
average of 30,000 base pairs apart. An average human from the two poles of the forming mitotic spindle. Other
chromosome contains about 150 × 106 base pairs of microtubules originate on chromosomes and within the
DNA, approximately 10 times the size of the entire mitotic spindle. As both kinetochores on a pair of sister
budding yeast genome, so many more origins are chromatids attach to microtubules from opposite spindle
required. Each region of the chromosome that is repli- poles, the pair of chromatids slowly moves to a point
cated from a single origin is referred to as a replicon. midway between the poles. When all chromosomes are
Groups of neighboring replicons cluster in topologically properly attached, the cell is said to be in metaphase.
associating domains (TADs) (see Chapter 8). The exit from mitosis begins at anaphase with abrupt
Proliferating diploid cells replicate their DNA once, separation of the two sister chromatids from one
and only once, each cell cycle. Each origin of replication another. Most cohesion molecules linking sister chroma-
is prepared for replication by the formation of a prerep- tids are removed without cleavage during prophase in a
lication complex during G1 (a process that is referred to process initiated by mitosis-specific phosphorylation.
as licensing). As each origin “fires” during S phase, the Proteolytic cleavage of the remaining cohesin molecules
prereplication complex is dismantled and cannot be triggers the metaphase-anaphase transition. During ana-
reassembled until the next G1 phase. This ensures that phase, the separated sister chromatids move to the two
each origin fires only once per cell cycle. The cyclic spindle poles (anaphase A), which themselves move
nature of origin licensing is driven at least in part by apart (anaphase B). As the chromatids approach the
fluctuations in the activity of cyclin-dependent kinases spindle poles, the nuclear envelope reforms on the
and protein destruction machinery (discussed later). surface of the chromatin. At this point, the cell is said to
During replication, the duplicated DNA molecules, be in telophase.
called sister chromatids, become linked to each other Finally, during telophase, a contractile ring of actin
by a protein complex called cohesin (see Fig. 8.18). This and myosin assembles as a circumferential belt at the
pairing of sister chromatids is important for their orderly cortex midway between spindle poles and constricts the
segregation later in mitosis (see Fig. 44.16). equator of the cell. The separation of the two daughter
cells from one another is called cytokinesis.
G2 Phase
In most cells of metazoans, G2 is a relatively brief period
during which key enzymatic activities that will trigger
Control of Cell-Cycle Progression
the entry into mitosis gradually accumulate and are con- Control networks and checkpoints regulate progres-
verted to active forms. When these activities reach a sion of the cell cycle. Checkpoints are biochemical
CHAPTER 40  n  Introduction to the Cell Cycle 701

circuits that detect external or internal stimuli and send that trigger cell death by apoptosis. Chapters 41 and 43
appropriate signals to the cell-cycle system. The restric- discuss these proteins in detail.
tion point in G1 phase is a control network that
integrates the physiological state of the cell with its
environment, including input from other cells and
Biochemical Basis of Cell-Cycle Transitions
interactions with the surrounding extracellular matrix. Transitions between cell-cycle phases are triggered by a
Cells must receive appropriate growth stimuli from network of protein kinases and phosphatases that is
their environment to progress past this point in the linked to the discontinuous events of the chromosome
G1 phase; if not they may live on without dying or cycle by the periodic accumulation, modification, and
commit suicide by apoptosis (see Chapter 46). destruction of several key components. This section pro-
DNA damage checkpoints operate throughout vides a general introduction to the most important com-
inter­phase. If damage is detected, the DNA damage ponents of this network.
response initiates a cascade of events that blocks cell-
cycle progression and can also trigger cell death by apop- Cyclin-Dependent Kinases
tosis. Problems with DNA replication generally produce Genetic analysis of the cell cycle in the fission yeast
single-stranded DNA and activate the DNA damage Schizosaccharomyces pombe identified a gene called
response. This response stabilizes stalled replication cell division cycle–2+ (cdc2+) that is essential for cell-
forks so that they can be repaired. During mitosis, the cycle progression during both the G1 → S and G2 → M
spindle assembly checkpoint delays the onset of transitions (Box 40.2). The product of this gene, a protein
chromosome segregation until all chromosomes are kinase of 34,000 Da originally called p34cdc2, is the pro-
attached properly to the mitotic spindle. totype for a family of protein kinases that is crucial for
The DNA damage response regulates cell-cycle pro- cell-cycle progression in all eukaryotes. This mechanism
gression in a three-tier pathway (Fig. 40.4). First sensors of cell-cycle control is so well conserved that a human
detect DNA damage. These sensors activate transduc- homolog of p34cdc2 can replace the yeast protein, restor-
ers, which include both protein kinases and transcrip- ing a normal cell cycle to a cdc2 mutant yeast. Boxes
tional activators. The transducers act on effectors that 40.3 and 40.4 present a number of the key experiments
ultimately block cell-cycle progression and may also and experimental systems that led to the identification
fulfill other functions. Two key protein kinases, ataxia- of the molecules that drive the cell cycle.
telangiectasia mutated (ATM) and ataxia-telangiectasia Humans have more than 10 distinct protein kinases
and Rad9 related (ATR), lie at the head of the pathway related to p34cdc2, although only a few are involved in
and may also act as sensors of DNA damage. They acti- cell-cycle control. To be active, each enzyme must associ-
vate two transducer kinases, Chk1 and Chk2, as well as ate with a regulatory subunit called a cyclin. Thus, they
a transcription factor called p53 that induces the expres- have been termed cyclin-dependent kinases (Cdks).
sion of a cohort of genes that halt cell-cycle progression p34cdc2, now termed Cdk1, seems to function primarily
by inhibiting cyclin-dependent kinases as well as genes in the regulation of the G2 → M transition in animal cells.
Cdk2 (plus Cdk4 and Cdk6 in some cell types) is involved
in passage of the restriction point during G1. Cdk2 also
Problems at contributes to the G2 → M transition, although Cdk1 is
DNA breaks replication forks the only Cdk absolutely essential for this step (Appendix
Genotoxic
stress 40.1). Cdk7 is important for activation of other Cdks,
and also appears to participate in transcribing RNA
and repairing damaged DNA. Other Cdks participate in
diverse processes ranging from transcriptional regulation
ATM ATM ATR + cofactors Sensors
dimer monomer bind ssDNA to neuronal differentiation. Surprisingly, fibroblasts from
mice that lack Cdk2, Cdk4, or Cdk6 are viable; other
Chk2 p53 Chk1 Transducers Cdks can drive the cell cycle if necessary. The mice suffer
kinase transcription kinase developmental difficulties because those genes are
factor
needed for the differentiation of particular cell types.
Cell cycle proteins Effectors
DNA repair proteins Cyclins
Cdks require cyclin binding for catalytic activity (Fig.
Apoptosis Cell-cycle DNA repair Responses
arrest 40.13). Cyclins were discovered in rapidly dividing inver-
tebrate embryos as proteins that accumulate gradually
FIGURE 40.4  ELEMENTS OF THE DNA DAMAGE CHECK-
POINT AND RESPONSE SYSTEM. ATM, ataxia-telangiectasia during interphase and are abruptly destroyed during
mutated; ATR, ataxia-telangiectasia and Rad9 related; ssDNA, single- mitosis (see Fig. 40.11). This process of cyclic accumula-
stranded DNA. tion and destruction inspired their name.
702 SECTION X  n  Cell Cycle

BOX 40.2  Use of Genetics to Study the Cell Cycle

Studies of the distantly related budding and fission yeasts falling only when knocked down by the (n-1)th domino.
Saccharomyces cerevisiae and Schizosaccharomyces pombe According to the model, mutations in genes that are essential
(see Fig. 2.8) were important for understanding the cell for cell-cycle progression cause an entire culture of yeast to
cycle for several reasons. First, the proteins that control the accumulate at a single point in the cell cycle (the point at
cell cycle are remarkably conserved between yeasts and which the defective gene product first becomes essential).
mammals. Second, both yeast genomes are small, simplifying This is referred to as the arrest point. Fig. 40.5 shows this
the discovery of important gene products. Third, genetic by including a “mutant” domino that does not fall over when
analysis is straightforward, as both yeasts can grow as hap- struck by the upstream domino. Mutants that meet this cri-
loids, and both efficiently incorporate cloned DNA into their terion are called cell division cycle mutants or CDC
chromosomes by homologous recombination. mutants. Genetic screens for CDC mutants have identified
These two yeasts evolved very different strategies for cell many important genes involved in cell-cycle control.
division. Budding yeasts divide by assembling a single bud Because CDC genes are essential for cell-cycle progres-
on the surface of the cell every cell cycle. Fission yeasts sion, it is impossible to propagate strains of yeast carrying
divide by fission across the center of an elongated cell. A CDC mutants unless the mutants have a conditional lethal
useful feature of using yeast to study the cell cycle is that phenotype. The most commonly used conditional lethal
the stage of the cell cycle is revealed by the cellular morphol- mutations are temperature sensitive (ts). Many yeast
ogy in the light microscope. For budding yeast, unbudded temperature-sensitive mutants are viable at 23°C (the per-
cells are in G1, cells with buds smaller than the mother cell missive temperature), but cease dividing at 36°C (the
are in S phase, and cells whose buds are similar in size to restrictive temperature). Temperature-sensitive proteins
the mother cell are in G2 or M. For fission yeast, cell length often have an altered amino acid sequence, but occasionally,
provides a yardstick for estimating cell-cycle position. the lack of a gene product can cause a ts phenotype. More
The cell cycles of both yeasts differ from those of animal recently, the use of auxin-inducible degrons (see Chapters 6
cells. In budding yeast, much of the 90-minute cell cycle is and 23) has enabled experimenters to study the conse-
spent in G1. Thus, the networks controlling the G1 → S transi- quences of depleting an essential protein from yeast in a
tion are particularly amenable to study. In contrast, a fission matter of minutes.
yeast spends most of its 2-hour cell cycle in G2. S phase Fission yeasts with CDC mutants affecting the entry into
follows separation of sister chromatids and occurs prior to mitosis have distinctive morphologies. Cells mutant in Wee1
cytokinesis. Thus, the control of the G2 → M transition is (a kinase that keeps cyclin-dependent kinase–1 [Cdk1] inac-
readily studied in fission yeast. During mitosis, the nuclear tive prior to mitosis) enter mitosis prematurely and are
envelopes of both yeasts remain intact, so chromosomes shorter than normal (Fig. 40.6B). In contrast, cells lacking
segregate on a spindle inside the nucleus. Cdc25 (a phosphatase that counteracts Wee1 and activates
Genetic studies revealed that the yeast cell cycle is a Cdk1) are unable to undergo mitosis but continue their
dependent pathway whereby events in the cycle occur nor- growth cycle, therefore becoming greatly elongated (Fig.
mally only after earlier processes are completed. The cell 40.6C). This simple morphologic assay allowed straightfor-
cycle can be modeled as a line of dominoes, each domino ward classification of yeast CDC genes into those that stimu-
corresponding to the action of a gene product that is essen- late progression through mitosis and those that retard entry
tial for cell-cycle progression (Fig. 40.5) and the nth domino into mitosis.

Model of the cell cycle as a


simple dependent pathway A. Wild type B. Wee1 mutant C. Cdc25 mutant

Wild type

FIGURE 40.6  FLUORESCENCE MICROGRAPHS OF


CDC mutant FISSION YEAST CELLS ILLUSTRATING PHENOTYPES OF
CELL-CYCLE MUTATIONS. Cell walls and nuclei are stained.
A, Wild-type cells. B, A wee1 mutation that accelerates entry into
mitosis at the restrictive temperature. C, A cdc25 mutation that
delays entry into mitosis at the restrictive temperature. (Courtesy H.
Ohkura, Wellcome Trust Institute for Cell Biology, University of Edin-
FIGURE 40.5  THE CELL CYCLE MAY BE MODELED AS A burgh, United Kingdom.)
SIMPLE DEPENDENT PATHWAY. A cell division cycle (CDC)
mutation can block further progression along the pathway at a
characteristic point in the cell cycle.
CHAPTER 40  n  Introduction to the Cell Cycle 703

BOX 40.3  Studies of the Cell Cycle in Vitro

Amphibian oocytes and eggs are storehouses of most compo- by making oocytes extremely large (~500,000 times the
nents needed for cell-cycle progression. Oocytes are arrested volume of a typical somatic cell) and storing within them
in G2 until a surge of the hormone progesterone causes them vast stockpiles of the structural components needed to make
to “mature” into eggs, which are then naturally arrested in cells. As a result, only DNA and a very few proteins need be
metaphase of the second meiotic division (see Chapter 45). synthesized during early embryonic divisions. In addition to
After fertilization, the embryo of the South African clawed structural components, many factors that regulate normal
frog (Xenopus laevis) undergoes a rapid burst of cell divi- cell-cycle progression are also stockpiled in oocytes. These
sions. An initial cell cycle 90 minutes long is followed by a features make Xenopus oocytes an excellent source of mate-
rapid succession of 11 cleavages spaced only 30 minutes rial for biochemical analysis of the cell cycle.
apart to produce an embryo of 4096 cells (Fig. 40.7). Remarkably, it is possible to make cell-free extracts from
Thirty minutes per cycle is insufficient to transcribe and Xenopus eggs that progress through the cell cycle in vitro
translate all the genes needed to make the daughter cells that (Fig. 40.8). Nuclei from G1 cells or haploid sperm nuclei,
are produced at each division. The frog solves this problem when added to these extracts, efficiently replicate their DNA
and proceed through the cell cycle into mitosis, complete
with chromosome condensation, nuclear envelope break-
1 Cleavage 11 Cleavages down, chromosome alignment on a spindle, and anaphase
90 minutes 30 minutes segregation of sister chromatids without any additions to
apart
the tube. Because these events occur in a cell-free milieu,
A B C they are readily accessible to biochemical manipulation. For
Somatic cell enlarged ×10 example, antibodies and other proteins can be added to the
FIGURE 40.7  CLEAVAGES SUBDIVIDE THE EGG DURING extracts, and their effect on the cell cycle can readily be
XENOPUS EARLY DEVELOPMENT. A, Fertilized egg. B, Two-cell determined. Thus, the Xenopus extract system offers a pow-
stage. C, Multicellular embryo. Compare size of somatic cell and erful tool for testing the role of various proteins in the cell
egg. cycle in higher eukaryotes.

A. Tightly B. Eggs crushed C. Added sperm D. Reassembly E. DNA replication F. Mitosis


packed by centrifugation nucleus with of nucleus
eggs membrane
removed

Lipid

Centrifuge
hard
Extract
Xenopus
eggs

Pellet

FIGURE 40.8  USE OF XENOPUS EGG EXTRACTS TO STUDY THE CELL CYCLE. A–B, Making a Xenopus egg extract that is
competent to carry out cell-cycle oscillations in vitro. C–F, A cycling Xenopus extract undergoes alternating S and M phases. G1 and G2
phases are minimal (as they are during early development of the frog).

Humans have at least 16 different cyclin proteins that negative controls. Like other eukaryotic protein kinases
range in size from 35 to 130 kD. The highly conserved (see Fig. 25.3), Cdks have a bilobed structure with the
cyclin box domain, which docks with the Cdk partners, active site in a deep cleft between a small N-terminal and
is the defining structural feature of these proteins. Only larger C-terminal domain. Monomeric Cdks have a flex-
a handful of cyclin isoforms are involved in cell-cycle ible T loop that blocks the mouth of the catalytic pocket.
control. Of those that are, some function during G1 In addition, a short α-helix is oriented such that a glu-
phase, others during G2 phase, and still others during tamic acid required for adenosine triphosphate (ATP)
M phase. hydrolysis points away from the catalytic cleft. As a
result, ATP bound by the monomeric kinase cannot
Positive Regulation of Cyclin-Dependent Kinase transfer its α-phosphate to protein substrates (see
Structure and Function Fig. 40.13A).
Cdks monomers are intrinsically inactive, so they depend Several different mechanisms regulate Cdk activity
on activation by cyclins and are regulated by positive and (Fig. 40.14). On one hand, cyclin binding and
704 SECTION X  n  Cell Cycle

BOX 40.4  Discovery of Factors Essential for Cell-Cycle Progression

The best early evidence for the existence of positive induc- MPF might be related to the inducer of mitosis detected in
ers of cell-cycle transitions in mammals was obtained in cell the PCC experiments. In fact, extracts from mitotic tissue
fusion experiments. When cultured cells in S phase were culture cells could induce meiotic maturation when injected
fused with cells in G1, the G1 nuclei initiated DNA replication into oocytes. Similar extracts from cells in other phases
shortly thereafter. In contrast, if S phase cells were fused of the cell cycle did not cause the G2/M phase transition
with G2 cells, the G2 nuclei did not rereplicate their DNA in oocytes.
until after passing through mitosis. The most dramatic results Other cell biologists studying protein synthesis in starfish
were obtained when mitotic cells were fused with inter- and sea urchin embryos noticed a curious protein that
phase cells. This caused the interphase cells to enter into seemed to accumulate across the cell cycle but was then
mitosis abruptly (as judged by nuclear envelope breakdown destroyed during mitosis. They were well aware of the work
and chromosome condensation). The phenomenon was on MPF, and immediately suspected that their protein,
termed premature chromosome condensation (PCC). which they called cyclin, might be somehow involved in
The mitotic inducer could work in any cell-cycle phase (Fig. MPF activity (Fig. 40.11).
40.9). If mitotic cells were fused with cells in G1 phase, In a third line of investigation, geneticists working on
interphase chromosomes condensed into long, single fila- yeasts realized that the cell cycle could be dissected through
ments. If the interphase cell was in G2 phase, the duplicated the isolation of cell division cycle (CDC) mutants (see Box
chromosomes appeared as double filaments. If the inter- 40.2). The analysis of the cell cycle with these mutants
phase cell was in the S phase, the partially replicated chro- dominated cell-cycle research to such an extent that many
mosomes condensed into a complex pattern of single and human genes that are important in cell-cycle control bear
double condensed regions separated by regions of decon- the CDC name if they are related to well-characterized yeast
densed chromatin corresponding to sites where DNA was genes. The best-known genes to emerge from this analysis
actively replicating at the time of fusion. were Cdc2 (Cdk1) and Cdc25, both of which were deter-
Working independently, developmental biologists who mined genetically to encode proteins that actively promote
were interested in the control of cell division during early the G2/M transition. Other genes, such as Wee1, were found
development in frogs also discovered an activity that could to encode activities that act as antagonists that inhibit the
cause interphase cells to enter the M phase. They used a G2/M transition.
micropipette to extract a tiny bit of cytoplasm from a mature When eventually purified from Xenopus eggs (Fig. 40.12),
egg that was arrested in metaphase of meiosis II and active MPF consisted primarily of two polypeptides of 32,000
inject it into oocytes (which are in G2 phase). The oocytes and 45,000 Da. The smaller component of MPF is the
rapidly entered M phase, with concomitant chromosome Xenopus equivalent of the fission yeast Cdc2 gene product
condensation and nuclear envelope disassembly (Fig. 40.10). (now known as Cdk1). The larger component of Xenopus
This stimulation to enter M phase is called maturation, MPF is a B-type cyclin. Only 15 years later was it recognized
and the unknown factor present in the egg cytoplasm that fully functional MPF also requires Greatwall kinase
that induced oocyte maturation was termed MPF, or and its small substrates to inhibit protein phosphatase PP2A-
maturation-promoting factor (now often referred to as B55δ and give Cdk activity a chance to take off and trigger
M phase–promoting factor). It was realized early on that mitotic entry.

A. M–G1 fusion B. M–S fusion C. M–G2 fusion

5 µm

FIGURE 40.9  FUSION WITH MITOTIC CELLS CAUSES INTERPHASE CELLS TO ENTER MITOSIS PREMATURELY, NO MATTER
WHERE THEY ARE IN THE CELL CYCLE. The resulting prematurely condensed chromosomes are single threads if the interphase cell
was in G1 phase (A), double threads if the cell was in G2 phase (C) and a complex mixture of both interspersed with uncondensed regions
if the cell was in S phase (B). M, mitosis. (From Hanks SK, Gollin SM, Rao PN, et al. Cell cycle-specific changes in the ultrastructural orga-
nization of prematurely condensed chromosomes. Chromosoma. 1983;88:333–342.)
CHAPTER 40  n  Introduction to the Cell Cycle 705

BOX 40.4  Discovery of Factors Essential for Cell-Cycle Progression—cont’d

A B C D

Meiotic Nucleus
spindle Nuclear disassembly Meiotic
and chromosome spindle
condensation

Suck out Inject it Oocyte enters


cytoplasm into oocyte M phase
Egg Fully grown oocyte Egg
FIGURE 40.10  THE EXPERIMENT THAT IDENTIFIED MATURATION-PROMOTING FACTOR. A, The box shows the meiotic spindle
in a Xenopus egg arrested in metaphase II of meiosis. B, The box shows the interphase nucleus in a mature oocyte. Following injection of
MPF, the nucleus disassembles, mitotic chromosomes form (C), and the cell assembles a meiotic spindle (D). Disassembly of the oocyte
nucleus and entry into M phase is called maturation, and the factor triggering this event was named maturation-promoting factor (MPF).

A. SDS gel of
proteins in
A purified MPF
75
B 97

Molecular weight (kDa)


67
B
Intensity of bands A and B

50

43
Cyclin B
A

25

30
Cdk 1

Cleavage Induction of M phase (%) 0 0 0 0 50 85 25 0 0 0


index
0 B. H1 kinase
0 1 2
activity
Time (hours)
Fraction number 6 9 12 15
FIGURE 40.11  EXPERIMENTAL IDENTIFICATION OF A
CYCLIN. Newly synthesized proteins (labeled with 35S-methionine) FIGURE 40.12  PURIFICATION OF MATURATION-PROMOT-
in fertilized sea urchin eggs were separated by sodium dodecylsul- ING FACTOR (MPF). A, Sodium dodecylsulfate (SDS) polyacryl-
fate (SDS) polyacrylamide gel electrophoresis. It was noted that the amide gel electrophoresis of fractions from the final step of the
protein labeled A (which was named cyclin) first accumulated, was purification. The numbers at the bottom show the percentage of
greatly reduced at the metaphase/anaphase transition, and then oocytes that entered M phase when a portion of each column frac-
began to accumulate again. Protein B, which is not involved in  tion was injected (the classical MPF assay). The roughly 32-kD band
cell-cycle regulation, accumulated progressively over this time. is Cdk1 (p34cdc2). The roughly 45-kD band is cyclin B. B, Assay of
“Cleavage index” refers to the percentage of dividing cells observed the ability of the column fractions to phosphorylate histone Hl. This
in the microscope at varying times after fertilization. (From Evans T, is a standard assay for active Cdk enzymes. (Modified from Lohka
Rosenthal ET, Youngblom J, et al. Cyclin: a protein specified by M, Hayes MK, Maller JL. Purification of maturation-promoting factor,
maternal mRNA in sea urchin eggs that is destroyed at each cleav- an intracellular regulator of early mitotic events. Proc Natl Acad Sci
age division. Cell. 1983;33:389–396.) U S A. 1988;85:3009–3013.)

phosphorylation of the T loop stimulate enzyme activity. domain is altered, allowing the bound ATP to assume a
On the other, Cdks are inhibited by phosphorylation of conformation suitable for reaction with substrates.
residues adjacent to the ATP-binding site and binding of Despite these changes, the Cdk–cyclin complex has
inhibitory proteins. low catalytic activity. Full activation of most Cdks
Cyclin binding causes the T loop to retract away from requires a kinase called Cdk-activating kinase (CAK),
the mouth of the catalytic pocket (see Fig. 40.13B). In which phosphorylates a threonine in the T loop of the
addition, the secondary structure of the N-terminal Cdk (this threonine gives the loop its name). In
706 SECTION X  n  Cell Cycle

A. Cdk2 B. Cdk2–cyclin A C. Active Cdk2–cyclin A

N lobe Active site


PSTAIR ATP
helix

T loop

C lobe Cyclin A Phosphorylation


site

FIGURE 40.13  ATOMIC STRUCTURES OF CYCLIN-DEPENDENT KINASES. A, Cdk2. The PSTAIR helix, found in most Cdks, is named
after a sequence of six amino acids that binds to cyclins (one letter code). (For reference, see Protein Data Bank [PDB; www.rcsb.org] file 1DM2.)
B, Cdk2–cyclin A (kinase at basal activity level). (PDB file 1FIN.) C, Cdk2–cyclin (kinase fully active following phosphorylation of threonine160). (PDB
file 1JST.) ATP, adenosine triphosphate.

vertebrates, CAK is composed of Cdk7-cyclin H. The A. Kinase activation B. Inactive forms


phosphorylated threonine fits into a charged pocket on
the surface of the enzyme, flattening the T loop back
even farther from the mouth of the catalytic pocket (see INK4
Cyclin
Figs. 40.13C and 40.14A). This stimulates the catalytic
activity up to 300-fold, in part because the flattened Cdk
T loop forms part of the substrate-binding surface. Threo-
nine phosphorylation also stabilizes the association of Cyclin
Inactive kinase cannot
Cdk2 with cyclin A. bind
In addition to their cyclin partner, Cdk1 and Cdk2 Cyclin
bind an additional small Cdc kinase subunit (Cks) protein
to their C-terminal domain, away from the active site.
Bound Cks enables the kinase to better hold onto its INK4
substrates and increases the efficiency with which Cdks
can phosphorylate substrates at multiple sites (a hall-
mark of Cdk target phosphorylation). Interference
p27 with ATP use

Negative Regulation of Cyclin-Dependent Kinase


Structure and Function
At least two mechanisms slow or stop the cell cycle by
inactivating Cdks (see Fig. 40.14). During G2 phase, the CAK
phosphorylation
protein kinases Myt1 and Wee1 hold Cdk1 in check by
phosphorylating threonine14 and tyrosine15 in the roof of ATP
cannot
the ATP-binding site. These phosphates interfere with bind
ATP binding and hydrolysis. Threonine14 and tyrosine15 Wee1 and Myt1
phosphorylation
are accessible to the regulatory kinases only following
cyclin binding, so this phosphorylation of Cdks depends,
Cdc25
at least in part, on the availability of cyclins. phosphatase
In mammals, three Cdc25 phosphatases (see Fig. dephosphorylation
25.5) reverse these inhibitory phosphorylations. Cdc25A Interference
regulates both the G1 → S and G2 → M transitions and Active kinase with ATP use
is essential for life of the cell. Ccd25B is dispensable for FIGURE 40.14  POSITIVE AND NEGATIVE REGULATION OF
mitosis, but it is essential for the production of gametes CYCLIN-DEPENDENT KINASES. A, Pathway of activation by cyclin
in meiosis. Cdc25C is a target of the G2 DNA damage binding and phosphorylation. B, Pathways of inactivation by inhibitor
binding and phosphorylation. When INK4 binds, twisting of the Cdk
checkpoint that prevents cells from undergoing mitosis
upper lobe blocks cyclin binding or interferes with ATP hydrolysis.
with damaged DNA (see Fig. 43.11), but cells can survive When p27 binds, a loop insinuates into the upper lobe of the Cdk and
without it. blocks ATP binding. (For reference, see PDB files 1B17 [Cdk2-INK4],
A parallel mechanism for inactivating Cdks involves 1FIN and 1BI7 [Cdk2-INK4-cyclin], and 1JSU [Cdk2-p27-cyclin A].)
binding of subunits from two families of small inhibitory CAK, Cdk-activating kinase.
CHAPTER 40  n  Introduction to the Cell Cycle 707

proteins called the cyclin-dependent kinase inhibitors picture of mitotic regulation, and this is a subject of
(CKIs) and inhibitors of Cdk4 (INK4) (for their names, active study.
see Appendix 40.1). When activated in the DNA damage
response pathway, p53 turns on transcription of the CKI Role of Protein Destruction in
p21, which inhibits Cdk-cyclin A. CKI p27Kip1 inactivates
Cell-Cycle Control
complexes of Cdk2 and cyclin A by having a protein loop
invade the N-terminal domain of the Cdk, disrupting its During mitosis active Cdk1–cyclin B–Cks phosphorylates
structure and competing with ATP for binding to the key substrates leading to dramatic reorganization of the
active site (see Fig. 40.14B). cell and, ultimately, to separation of sister chromatids on
Members of the INK4 family preferentially inactivate the mitotic spindle. Once chromatids are separated, the
Cdk4 and Cdk6 in two ways (see Fig. 40.14B). First, cell must return to a state with low levels of Cdk activity
binding to monomeric Cdk distorts the orientation of so that nuclear envelope reassembly, spindle disassem-
the N- and C-terminal lobes, so cyclin D does not bind. bly, and cytokinesis can occur.
INK4 family inhibitors also inhibit preformed Cdk4/6– Exit from mitosis requires Cdk inactivation by the
cyclin D complexes by binding the Cdk and distorting ubiquitin-directed proteolytic machinery. The destruc-
the ATP-binding site so that the kinase uses ATP much tion of A- and B-type cyclins inactivates Cdk1 and Cdk2.
less efficiently. This allows PP1, PP2A-B55δ, and other phosphatases to
Cdk inhibitors are important for growth regulation reverse the action of Cdks and bring mitosis to a close.
during the G1 and G0 phases of the cell cycle (see Chapter Ubiquitylation also results in proteolysis of a protein
41). They also play a critical role in the cell-cycle arrest called securin, which regulates the onset of sister chro-
that occurs in response to DNA damage and to antipro- matid separation at anaphase.
liferative signals. Mutations in the INK4 locus are strongly Ubiquitin-mediated destruction of cyclins involves a
linked to cancer. cascade of three enzymes described in Chapter 23 (see
Fig. 23.3). First, an E1 enzyme (ubiquitin-activating
Role of Phosphatases in enzyme) activates the small protein ubiquitin by
forming a thioester bond between the ubiquitin
Counter-Balancing Cdk Activity
C-terminus and a cysteine on the enzyme. Activated ubiq-
Two important phosphatases counter-balance Cdk activ- uitin is next transferred to another thioester bond on an
ity in mitosis. Just as Cdks exhibit cyclic behavior and E2 enzyme (ubiquitin-conjugating enzyme). The E2
are activated in mitosis, these counteracting phospha- often cooperates with an E3, which is important for
tases must also be cyclic—but they are inhibited in imparting substrate specificity, to transfer ubiquitin to
mitosis. Protein phosphatase 1 (PP1), associates with the ε-amino group of a lysine on a target protein. The
numerous targets on chromosomes and the mitotic appa- resulting polyubiquitinated proteins are usually targets
ratus, and is highly active during mitotic exit. Many for destruction by the cylindrical 26S proteasome (see
target proteins have a simple loop with the sequence Fig. 23.8). This large multienzyme complex functions
RVS/TF that inserts into a groove on the enzyme. During like a cytoplasmic garbage disposal, grinding target pro-
mitosis, Cdk phosphorylation of adjacent sites often teins down to short peptides and spitting out intact
blocks this interaction with substrates. Cdk1-cyclin B ubiquitin monomers for reuse in further rounds of
also phosphorylates the PP1 catalytic subunit during protein degradation. Its role was originally thought to be
mitosis, thereby inactivating the enzyme. the removal of damaged proteins from the cytoplasm;
PP2A is more directly involved in Cdk regulation. It however, it is now recognized as a central factor in cell-
is a trimeric enzyme with catalytic, scaffolding, and cycle control.
regulatory subunits. The latter include B55α-δ and The key E3 ligase regulating cyclin proteolysis is a large
B56α-ε (see Appendix 40.1). PP2A-B55δ is largely (15-subunit) complex called the anaphase-promoting
responsible for removing phosphates added by Cdks. complex/cyclosome (APC/C) (Fig. 40.15). The APC/C
Consequently PP2A-B55δ must be inhibited to allow is inactive during the S and G2 phases of the cell cycle.
Cdks to drive the cell into mitosis. The Greatwall (Gwl) Phosphorylation by Cdk1-cyclin B-Cks1 and binding of
kinase regulates PP2A. Gwl is unusual, because 500 the protein coactivator Cdc20 activate the APC/C in
amino acids are inserted into its large lobe roughly adja- early mitosis. APC/CCdc20 then triggers the metaphase–
cent to the T loop (see Fig. 40.13). Phosphorylation anaphase transition.
by Gwl allows two small proteins, Arpp19 (cyclic An important checkpoint, the spindle assembly
adenosine monophosphate [cAMP]-regulated phospho- checkpoint, regulates APC/CCdc20 during mitosis,
protein 19) and ENSA (α-endosulfine), to bind and keeping it inactive until all kinetochores are produc-
inhibit PP2A-B55δ. Thus, Gwl confers the necessary tively attached to spindle microtubules. The checkpoint
cyclic behavior on PP2A. Understanding how Gwl is effector is the mitotic checkpoint complex whose for-
turned on and off is important for developing a full mation is triggered by unattached kinetochores. This
708 SECTION X  n  Cell Cycle

SCFSkp2, SCFβ–TrCP
APC/CCdc20 APC/CCdh1 APC
(active) (active)
Cdc20 Cdh1
E2 Cdh1 Degron
SCFSkp2

Cdk
cyclin

P M A T
P M G1 S G2 P M A T
P M G1 APC/CCdh1 surface and backbone

FIGURE 40.15  TWO FORMS OF THE ANAPHASE-PROMOTING COMPLEX/CYCLOSOME (APC/C) CONTROL THE CELL CYCLE.
At the metaphase-anaphase transition, the APC/C with associated Cdc20 triggers the onset of anaphase by signaling the degradation of securin
and cyclin B. During mitosis Cdh1 phosphorylated by Cdk1–cyclin B is unable to bind the APC/C, so APC/CCdh1 activity is low. As Cdk1–cyclin
B activity declines in anaphase, Cdh1 binds the APC/C and APC/CCdh1 drives the exit from mitosis into G1. APC/CCdh1 remains active throughout
G1 but is inactivated following synthesis of the specific inhibitor Emi1. After the onset of S phase, SCF (shown here adding a ubiquitin chain to
a docked substrate) directs the degradation of cell-cycle substrates such as p27Kip1, following their phosphorylation by protein kinases.

complex inhibits APC/CCdc20 by acting as a competitive A. Structure of SCFSkp2-E2 complex


substrate. As a result, cyclin B and securin are stable until Skp 2
the checkpoint is satisfied. A few substrates, including ~50 Å E2
cyclin A, bind directly to the APC/C without requiring
Cdc20, so they are marked for degradation even when
the checkpoint is active. Skp 1 RbX 1
Exiting from mitosis and allowing the G1 cell to
prepare chromatin for DNA replication (see Chapter 42)
requires low Cdk activity and destruction of Cdc20. This
is accomplished late in mitosis by Cdh1, a different Cullin 1
co-activator of the APC/C. Phosphorylation of Cdh1 by
Cdks blocks its binding to the APC/C early in mitosis.
Thus, APC/CCdh1 forms only after cyclin levels (and there- B. Some F-box proteins C. Their target substrates
fore Cdk activity) decline late in mitosis. As cells pass Skp 2
F E2F-1: Cell-cycle regulator
from G1 into S phase, a newly synthesized inhibitory
protein, Emi1, binds to and inactivates APC/CCdh1. This F box
p27Kip1: Cdk2 inhibitor
allows cyclins to accumulate during S and G2. Remark-
Skp 1
ably, APC/CCdh1 is also involved with regulating the
activity of synapses in nondividing neurons. β-TrCP1 Cdc25A: Cdk1 activator
After the G1 → S transition and throughout the remain- F Wee1: Cdk1 inhibitor
der of interphase members of a different family of E3
activities called SCF regulate the levels of proteins that Emi1: APC/CCdh1 inhibitor
control Cdk and other cell cycle factors. SCF is named β-Catenin: Cell-proliferation
Skp 1 regulator
after three of its four subunits: Skp1, cullin, and F-box
protein (Fig. 40.16). SCF is a molecular toolbox built on
FIGURE 40.16  STRUCTURE AND FUNCTION OF SCF.
a bow-shaped scaffold formed by the cullin subunit. The A, Structure of SCFSkp2. Left, SCF recognizes target proteins through
fourth subunit, Rbx1, binds near the C-terminus of cullin its F-box subunit (Skp2 in this case). Right, Ubiquitin is then transferred
and uses a protein motif called a RING finger to dock to from an E2 enzyme. The whole is assembled on a rigid bow-like scaf-
a ubiquitin-linked E2 enzyme. Skp1 binds to the other fold composed of the cullin subunit. Structures of F-box proteins Skp2
end of the cullin, where it provides a docking site for an and β-Trcp (B) and a list of several of their known target proteins (C).
F-box protein that recognizes and binds the substrate.
(The F-box got its name because it was first discovered
in cyclin F.) Humans have 78 F-box proteins, giving SCF SCF is fundamentally different from the APC/C,
enormous versatility. Two examples shown in Fig. 40.16 because it is constitutively active. However, it ubiquity-
are Skp2, which targets the Cdk inhibitor p27 helping to lates substrates only after they have been phosphory-
drive the G1 → S transition, and β-Trcp, which targets lated, often by Cdks. This feature links SCF activity to
Cdc25A, Wee1, and Emi1. the cell cycle.
CHAPTER 40  n  Introduction to the Cell Cycle 709

1 2 3 4 5 1 2 3 4

Increasing Cdk activity


Cdk1-cyclin B
Cdk2-cyclin A
Cdk2-cyclin E

P M A T P M A T
P M G1 S G2 P M G1
Increasing APC activity

APC/CCdh1 APC/CCdc20

Emi1
SCFSkp2, SCFβ–TrCP
PP2AB55δ, PP1

FIGURE 40.17  DIAGRAM SHOWING THE CHANGING STATES OF THE CYTOPLASM AS CELLS TRAVERSE THE CELL CYCLE.
Between G2 and G1 are shown the various stages of mitosis: P, prophase; PM, prometaphase; M, metaphase; A, anaphase; T, telophase. The
states of the cytoplasm discussed in the text are shown as green arrows across the top. Increased Cdk activity is shown as peaks upwards from
the central bar. Increasing anaphase-promoting complex/cyclosome (APC/C) activity is shown as a mirror image, with peaks going down from
the central bar. APC, anaphase-promoting complex; PP1, protein phosphatase 1; PP2A, protein phosphatase 2A.

Changing States of the Cytoplasm During Low Cdk activity is required for cytokinesis, spindle
disassembly, chromosome decondensation, nuclear
the Cell Cycle
envelope reassembly, reactivation of transcription,
The cell cycle is characterized by five discrete physiolog- reassembly of the Golgi apparatus, and assembly of
ical states of the cytoplasm (Fig. 40.17). Changing levels prereplication complexes on the chromosomes.
of Cdk activity drive transitions between these states, 4. G1–S phase transition. Growth signals from the envi-
sometimes counteracted and sometimes reinforced by ronment promote the transcription of Cyclin E. If
targeted proteolysis. the levels pass a critical threshold, a burst of Cyclin
1. Early mitosis. Cdk2-cyclin A peaks in prophase, E-Cdk2 activity allows the cell to pass the restriction
followed by Cdk1-cyclin B. When the nuclear enve- point, leading to synthesis of proteins required for
lope breaks down, the APC/CCdc20 starts degrading DNA replication and cell-cycle progression. Cdk phos-
cyclin A, but the spindle checkpoint inhibits destruc­ phorylation targets the CKI peptides for destruction
tion of other substrates. When the last chromo- by SCF allowing Cdk2 to become activated. In addi-
some has attached correctly to spindle microtubules tion, APC/CCdh1 is inactivated by newly synthesized
(metaphase), the checkpoint is satisfied, and APC/ Emi1.
CCdc20 starts to degrade cyclin B and securin, an inhibi- 5. S–G2 phase. Cdk activity remains high throughout the
tor of a key protease called separase. Their degrada- remainder of the cell cycle, and SCF continues to
tion continues throughout metaphase. degrade selected proteins tagged by Cdk phosphoryla-
2. Anaphase and mitotic exit. When securin levels fall tion. SCFβ-Trcp destruction of Cdc25A keeps Cdk1 inac-
below a critical threshold, active separase cleaves a key tive, preventing a premature entry into mitosis. The
component of the cohesin ring (see Fig. 8.18). This APC/C remains inactive, allowing mitotic cyclins to
triggers sister chromatid separation. Cyclin destruction accumulate. It is not known what ultimately triggers
continues throughout anaphase and telophase, and entry into mitosis, but an important factor may be a
falling Cdk1 activity allows the formation of APC/CCdh1, switch in the specificity of SCFβ-Trcp, which spares
which marks Cdc20 for destruction along with the Cdc25A and instead degrades the Cdk-inhibitory
remaining B-type cyclins. SCFβ-Trcp destruction of Emi1 kinase Wee1.
allows APC/CCdh1 to be active when it forms. Although this sounds complicated, the underlying
3. G1 phase. APC/CCdh1 and Cdk inhibitors of the CKI principles are actually quite straightforward. The follow-
and Ink4 families cooperate to inhibit Cdk activity. ing chapters discuss the cell-cycle transitions in greater
710 SECTION X  n  Cell Cycle

detail and show how the process is modulated in Hunt T. On the regulation of protein phosphatase 2A and its role in
response to a changing environment. controlling entry into and exit from mitosis. Adv Biol Regul. 2013;
53:173-178.
Lorca T, Castro A. The Greatwall kinase: a new pathway in the control
ACKNOWLEDGMENTS of the cell cycle. Oncogene. 2013;32:537-543.
Morgan DO. The Cell Cycle: Principles of Control. London: New
We thank Tim Hunt, David Morgan, and Jonathon Pines Science Press; 2007: 297p.
for their suggestions on revisions to this chapter. Nasmyth K. A prize for proliferation. Cell. 2001;107:689-701.
Nurse P. A long twentieth century of the cell cycle and beyond. Cell.
SELECTED READINGS 2000;100:71-78.
Primorac I, Musacchio A. Panta rhei: the APC/C at steady state. J Cell
Bartek J, Lukas J. DNA damage checkpoints: from initiation to recovery Biol. 2013;201:177-189.
or adaptation. Curr Opin Cell Biol. 2007;19:238-245. Qian J, Winkler C, Bollen M. 4D-networking by mitotic phosphatases.
Brown JS, Jackson SP. Ubiquitylation, neddylation and the DNA damage Curr Opin Cell Biol. 2013;25:697-703.
response. Open Biol. 2015;5:150018. Stukenberg PT, Burke DJ. Connecting the microtubule attachment
Craney A, Rape M. Dynamic regulation of ubiquitin-dependent cell status of each kinetochore to cell cycle arrest through the spindle
cycle control. Curr Opin Cell Biol. 2013;25:704-710. assembly checkpoint. Chromosoma. 2015;124:463-480.
Hartwell LH, Weinert TA. Checkpoints: Controls that ensure the order Wieser S, Pines J. The biochemistry of mitosis. Cold Spring Harb Per-
of cell cycle events. Science. 1989;246:629-634. spect Biol. 2015;7:a015776.

APPENDIX 40.1

Inventory of the Enzymes of the Cell-Cycle Engine


Cyclin-Dependent Kinases and Their Cyclin Partners
Kinase Cyclin (+ Other) Partner Function
Cdk1 (p34cdc2) A Mammals: triggers G2 → M transition. Yeasts: triggers G1 → S and G2 →
B1, B2 (Xenopus has 5 B-type M transitions. Cyclin A is synthesized in S and destroyed starting at
cyclins) prometaphase. Cyclins B are synthesized in S/G2 and destroyed
Cdk1–cyclin B binds Cks1 following the completion of chromosome attachment to the spindle.
(Cdc kinase subunit) Cyclins A1, B3 function preferentially in meiosis.
Cdk2 A, E Triggers G1 → S transition. Can be replaced by other Cdks in mouse.
Cdk4, Cdk6 D1–D3 Phosphorylation of the retinoblastoma susceptibility protein (pRb) in G1.
Triggers passage of the restriction point and cyclin E synthesis in some
cell types. Extracellular growth factors control synthesis of D cyclins.
Can be replaced by other Cdks in mouse.
Cdk5 CDK5R1 or CDK5R2 Neuronal differentiation, sensory pathways.
Cdk7 (CAK) H; also binds assembly factor Cdk activation by phosphorylation of the T loop. Also in TFIIH, important
MAT1 for regulation of RNA polymerase II transcription and DNA repair.
Cdk8 C Regulation of RNA polymerase II transcription.
Cdk9 T Regulation of RNA polymerase II transcription.
Cyclin Inhibitors
Inhibitor Cdk Substrates Function
CKI: p21Cip1/Waf1 most Most Cdk-cyclin complexes Induced by p53 tumor suppresser. Cell-cycle arrest after DNA damage.
Cdk-cyclin complexes Binds PCNA (proliferating cell nuclear antigen; see Chapter 42) and
inhibits DNA synthesis. Promotes cell cycle arrest in senescence and
terminal differentiation. At low levels, may help assemble active
Cdk-cyclin complexes.
CKI: p27Kip1 Most Cdk-cyclin complexes Cell cycle arrest in response to growth suppressers like TGF-β and in
contact inhibition and differentiation.
CKI: p57Kip2 Most Cdk-cyclin complexes Important in development of the palate.
INK4: p15Ink4b Cdk4, Cdk6 Cell-cycle arrest in response to transforming growth factor (TGF)-β.
Altered in many cancers.
INK4: p16Ink4a Cdk4, Cdk6 Cooperates with the retinoblastoma susceptibility protein (pRb) in
growth regulation. Cell-cycle arrest in senescence. Altered in a high
percentage of human cancers. This gene overlaps the gene for p19ARF,
an important regulator of the p53 tumor-suppresser protein.
INK4: p18Ink4c Cdk4, Cdk6 Cell-cycle arrest in response to growth suppressers.
INK4: p19Ink4d Cdk4, Cdk6 Cell-cycle arrest in response to growth suppressers.
CHAPTER 40  n  Introduction to the Cell Cycle 711

APPENDIX 40.1

Inventory of the Enzymes of the Cell-Cycle Engine­—cont’d


Other Components
Enzyme Substrates Functions
Wee1 kinase Cdk1 Y15 Nuclear kinase. Inhibits Cdk1-cyclin B in G2.
Myt1 kinase Cdk1 T14 + Y15 Cytoplasmic kinase. Inhibits Cdk1-cyclin B in G2.
Greatwall (Gwl) kinase Arpp19, ENSA (α-endosulfine) Phosphorylated Arpp19 and ENSA inhibit PP2a, allowing active Cdk1-
(MASTL in humans) cyclin B to accumulate and trigger mitotic entry
Cdc25A phosphatase Cdk1 T14, Y15 Promotes G1 → S transition and G2 → M transition. Essential for life of
the cell.
Cdc25B phosphatase Cdk1 T14, Y15 Promotes G2 → M transition. Essential in meiosis.
Cdc25C phosphatase Cdk1 T14, Y15 Promotes G2 → M transition. Dephosphorylates Cdk1 complexed to
cyclins A, B at T14 and Y15. Not essential for life.
PP2A phosphatase Many proteins phosphorylated Regulated by ENSA/Greatwall. With its targeting subunits B55α-δ and
by Cdk1-cyclin B B56α-ε it regulates many activities during mitotic exit and cytokinesis.
PP1 phosphatase Many targets Associates with many “targeting subunits,” which can be, for example,
regulatory proteins, such as RepoMan or can be structural subunits
of the kinetochore (see Chapter 8). It is inactivated by Cdk
phosphorylation during mitosis, but has a key role in mitotic exit.
APC/CCDC20 Cyclin B, securin many others E3 ubiquitin ligase active during M. Requires high Cdk activity to
function. Destruction of cyclins and other substrates essential for exit
from mitosis. Contains 15 subunits plus the specificity factor Cdc20.
APC/CCdh1 Cyclins A, B, many others E3 ubiquitin ligase active during G1. Requires low Cdk activity to
function. Keeps Cdk activity low in G1 through cyclin proteolysis.
Contains 15 subunits plus the specificity factor Cdh1.
SCF Cyclin E, many others Class of E3 ubiquitin ligases containing Skp1 + cullin + Rbx1 + an F-box
protein. Humans have 78 F-box proteins (Caenorhabditis elegans
has more than 300), acting as specificity factors for substrates
phosphorylated at specific sites, including cyclin E and Cdk inhibitors.
This page intentionally left blank
CHAPTER 41 

G1 Phase and Regulation of


Cell Proliferation
D uring the G1 phase of the cell cycle, each cell makes to the cycle from G0, and how they regulate their progress
a key decision: whether to continue through another through the G1 phase. It also considers some of the points
cycle and divide or to remain in a nondividing state at which defects in cell cycle control lead to cancer.
either temporarily or permanently. During development
of metazoans, cells exit the cell cycle as the first step
toward forming differentiated tissues. In adults, strict
G0 Phase and Growth Control
regulation of the timing and location of cell proliferation Cells stop cycling in three ways. First, they may receive
is critical to avoid cancer. external signals instructing them to withdraw from the
Cells enter G1 phase at the end of a proliferation cycle, cell cycle, enter into G0, and differentiate, as discussed
after completing mitosis. To be free to decide whether later. Second, cells may find themselves in an environment
to proliferate or differentiate, the cell must inactivate the with insufficient mitogens to drive proliferation. Under
remnants of the proliferation machinery from the pre- these conditions many cell types enter a transient nondi-
ceding cell cycle. This is initiated in late M-phase by viding state known as quiescence, while they wait for
inactivating cyclin-dependent kinases (Cdks [see Chapter conditions to improve. Third, cells that have suffered DNA
40]) via proteolytic destruction of their cyclin subunits. damage because of a loss of cell-cycle control undergo
This continues in G1 phase and is accompanied by syn- senescence, or may, in some cases, commit suicide by
thesis and stabilization of Cdk-inhibitory proteins. The apoptosis (see Chapter 46). Senescence is a permanent
absence of Cdk activity activates a regulatory network nondividing state that is physiologically distinct from G0
that represses the transcription of many genes that and from which cells normally cannot exit. One physiolog-
promote cell-cycle progression. While this repressive ical signal that can lead to senescence is a critical shorten-
network is active, the cell cannot proceed through ing of the telomere regions of the chromosomes in cells
the cell cycle. The repression can be switched off if the
cell is stimulated by specific signals from the surround- Lack of mitogens
ing medium, extracellular matrix, and other cells (see Antiproliferation signals
Chapters 27 and 30). If these signals are diffusible sub- G2 M (eg, contact inhibition,
TGF-β telomere damage)
stances, they are known as mitogens. Mitogens can
trigger another round of DNA replication and mitosis, G0
but first, the cell must pass a major decision point in G1
called the restriction point (Fig. 41.1). Restriction point:
In metazoans, many cells cease cycling, either tempo- Regulates G1 progression Proliferation signals
rarily or permanently, exiting the cell cycle into a state to S phase Nutrients
S Blocks cell-cycle progression G1
known as G0 (Fig. 41.1). This frequently accompanies
unless nutrients & mitogens
their acquisition of specialized, differentiated character- are continuously present
istics. Occasionally, it is desirable in tissues for cells in
G0 to reenter the cell cycle to replace lost cells. Special-
ized cells called stem cells fulfill this role in tissue
maintenance.
This chapter describes how cells decide whether to FIGURE 41.1  CELL CYCLE SHOWING MAJOR LANDMARKS
exit from the cell cycle into the G0 phase, how they return IN THE G1 PHASE. TGF-β, transforming growth factor–β.

713
714 SECTION X  n  Cell Cycle

TUMOR Contact
inhibition
Cdk2–cyclin E–p21
Cdk2–cyclin E–p27
(inactive)
Differentiation p27
signals Cdk2–cyclin E
p21
p16 p27
Senescence Cdk4-p15
p15
Cdk4–cyclin D–p27 Cdk4-p16
R (inactive)

Blood vessels TGF-β Cyclin D


(degraded)
Normal epithelial cells

Various signals Cdks inactive


stimulate synthesis Hypophosphorylated Rb
of Cdk inhibitors binds E2F and inactivates genes
required for cell-cycle progression

FIGURE 41.3  HOW EXTERNAL STIMULI ACT ON CDK INHIBI-


TORS TO CAUSE CELLS TO ENTER A NONDIVIDING G0 STATE.
TGF-β, transforming growth factor–β.
NORMAL

FIGURE 41.2  DISRUPTION OF NORMAL TISSUE ARCHITEC- BOX 41.1  Cdk Inhibitor Scorecard
TURE BY UNCONTROLLED PROLIFERATION OF CANCER
CELLS. Lower right, Normal thyroid tissue. Upper left, A thyroid tumor The regulation of G1 progression requires the action of
with loss of the normal gland structure. (Courtesy Clara Sambade, two families of Cdk inhibitory proteins (see Chapter 40).
IPATIMUP, Porto, Portugal.) Cyclin-dependent kinase inhibitors (CKIs), which include
p21Cip1 and p27Kip1, usually inhibit all Cdks and block cell-
cycle progression, but under special circumstances, they
that have divided more than a critical number of times. can actually activate Cdk4/6-cyclin D and promote cell
Overexpression of telomerase (see Fig. 7.14) can, when proliferation during G1. Cell-cycle blocks imposed by CKIs
combined with suitable mitogenic stimuli, prevent cells tend to be temporary. INK inhibitors, which include
from undergoing senescence in tissue culture. p15Ink4B and p16Ink4a, are specialized at inhibiting Cdk4/6-
cyclin D. These can promote a profound and often perma-
Most cells in multicellular organisms are differentiated
nent cell-cycle arrest by activating the Rb pathway of gene
(adapted to carry out specialized functions) and no
repression.
longer divide. They typically form specialized tissues,
each with a distinctive structural organization that is
important for its function. Unscheduled cell division can ongoing processes. Because of turnover, all cells must
severely disrupt the organization of such tissues (Fig. continuously synthesize housekeeping proteins. They
41.2). Accordingly, tissues strictly regulate both the loca- must also expend energy to maintain intracellular pH
tion and the frequency of cell division. In most tissues, and ionic composition and to power intracellular motil-
divisions normally occur at a low rate, producing new ity. In addition, many specialized G0 cells consume large
cells in numbers just sufficient to replace those that die. amounts of energy to synthesize and secrete protein
Under special circumstances, however, such as in products and generate action potentials. Energy metabo-
response to wounding (see Fig. 32.11), the rate of cell lism is particularly dramatic in muscle cells that are
division may increase dramatically. This highlights an responsible for all body movements. Thus, most G0 cells
important constraint on cell-cycle control in multicellular should be regarded as active cells that just happen no
organisms: To make organized tissues, cells must exit longer to be engaged in cell division.
from the cell cycle, but some cells must also retain the Transforming growth factor–β (TGF-β) is an example
ability to reenter the active cell cycle when needed to of an external signal that arrests progress through the
repair injuries or replace worn-out cells. cycle and regulates differentiation and tissue morpho-
Cells that stop cycling to differentiate are said to have genesis (Fig. 41.3). TGF-β stimulates a receptor serine/
left the cycle and entered a nonproliferating state called threonine kinase that activates SMAD transcription
G0 (Fig. 41.1). G0 may last hours or days, or even for the factors (see Fig. 27.10). SMADs suppress Cdk-4 synthesis
life of the organism, as it does for most neurons. It is and increase expression of CKI (cyclin-dependent kinase
important to note that nondividing cells are not inhibitor) and Ink4 class Cdk inhibitors (Box 41.1).
dormant: G0 cells can be biochemically very active and p15Ink4B preferentially inactivates Cdk4–cyclin D and
continue to expend large amounts of energy for many Cdk6-cyclin D complexes. It also displaces CKI class
CHAPTER 41  n  G1 Phase and Regulation of Cell Proliferation 715

inhibitors from the Cdk4–cyclin D, permitting them to Both p21Cip1 and p16Ink4a contribute to the permanent
transfer to Cdk2–cyclin E complexes in the nucleus. This cell-cycle arrest of senescent cells. p16Ink4a activates
further inhibits cell-cycle progression. inhibitory proteins of the retinoblastoma protein (Rb)
The CKI p27Kip1 helps arrest the cell cycle of normal and E2F families (see later) that bind to promoters and
cells when they become crowded by neighboring cells recruit histone methyltransferases, forming heterochro-
(contact inhibition; see ahead, Fig. 41.11) or if their matin that permanently inactivates genes required for
environment lacks mitogens. Genetic analysis in mice proliferation (see Fig. 8.7). Once cells exit the cycle,
revealed that p27Kip1 also regulates cell-cycle progression multiple redundant pathways block reentry by reinforc-
during development. Mice lacking p27Kip1 are 30% larger ing the primary inhibition of Cdk activity. In addition, a
than their normal littermates by several weeks of age. specialized histone variant H1o replaces histone H1 in
This is at least partly because cells in many organs G0 cells, resulting in more condensed chromatin. This
undergo extra rounds of cell division. represses transcription generally. However, not all gene
An analogous mechanism limits proliferation during expression is suppressed in differentiated cells, many of
the differentiation of muscle. The transcription factor which synthesize large amounts of specific proteins (eg,
MyoD drives expression of the CKI inhibitor p21Cip1, digestive enzymes secreted by the pancreas).
which helps arrest proliferation and start muscle differ-
entiation (Fig. 41.3). p21Cip1 stops cell-cycle progression
in at least two ways. First, it binds Cdk–cyclin complexes
Moving Into and Out of G0: Stem Cells
and stops them from promoting cell-cycle progression. Stem cells are professionals at moving back and forth
It also blocks DNA replication by inhibiting the DNA between G0 and proliferative cell cycles. One of their
replication factor proliferating cell nuclear antigen roles is to replace worn-out parts of tissues as differenti-
(PCNA [see Chapter 42]) that is required for DNA poly- ated cells age or die as a result of various misadventures.
merase δ activity. Box 41.2 provides a brief introduction to stem cells.

BOX 41.2  Stem Cells in Mammals

The defining feature of stem cells is their capacity to self- tissue stem cells are held in reserve unless the tissue is
renew while producing daughter cells with the capacity to damaged, when they produce daughter cells to repair the
differentiate into more specialized cells under the control of damage. Stem cells are present even in organs that have a
intrinsic and environmental cues. Stem cells play a key role limited capacity for renewal and regeneration, such as the
in the development of multicellular organisms in addition nervous system. The potential for regeneration from stem
to providing cells for the renewal and regeneration of cells has stimulated research to find ways of using embryonic
adult tissues. or tissue stem cells to repair damaged or diseased organs in
Each multicellular organism begins as a single cell (a fertil- human patients. Stem cells have also been useful for produc-
ized egg) with a genome encoding the information required tion of transgenic animals for scientific research (eg, knock-
to produce an adult. The divisions prior to implantation in out mice).
the uterine wall produce a small group of epiblast cells that
go on to form the embryo. The other cells that are produced Discovery and Defining Features of
at this stage are specialized to support the embryo. Epiblast Stem Cells
cells are termed pluripotent because their progeny can Pioneering work on blood cell development (see Fig. 28.4)
form all the specialized cells of the adult. Although the plu- established the existence of stem cells and defined many of
ripotent cells disappear as the embryo develops, epiblast the concepts that apply to all types of stem cells. The key
cells can be propagated and maintained permanently in experiment was to inject bone marrow cells from a normal
culture without losing their pluripotency if optimal condi- mouse into a mouse that had been irradiated to kill all the
tions are provided. These cell lines are called embryonic cells that produce blood cells. Transplantation of bone
stem cells (ES cells). marrow cells rescued the irradiated mice from death from
Most adult tissues set aside a few tissue stem cells that anemia, bleeding, and infections. The transplanted bone
have the capacity to renew themselves and to produce marrow contained precursor cells that formed colonies of
daughter cells that differentiate into a limited range of spe- proliferating cells that regenerated the full range of blood
cialized cells (see Figs. 28.1, 28.4, and 40.1). Adult stem cells cells. The blood-forming colonies in the spleen, each of
have diverse patterns of cell-cycle regulation. Some tissue which formed from a single stem cell, contained either one
stem cells continue the cell cycle throughout life. For or, infrequently, several types of differentiating blood cells.
example, epithelial stem cells give rise to mature cells that This experimental system first revealed the existence of
continuously replace the skin and the lining of the gastroin- several different types of progenitor cells in bone marrow
testinal tract. Hematopoietic stem cells in bone marrow with the dual capacity to proliferate and to give rise to more
give rise to both short-lived and long-lived differentiated differentiated cells (see Fig. 28.4). These committed pro-
blood cells. In other organs, such as liver and skeletal muscle, genitor cells have a limited proliferation capacity and can

Continued
716 SECTION X  n  Cell Cycle

BOX 41.2  Stem Cells in Mammals—cont’d

give rise to only to specific subsets of blood cells, such as


red blood cells, platelets, granulocytes, or lymphocytes. In Niche cell
Adherens
contrast, there are a very few multipotent hematopoietic junction
stem cells in bone marrow. They replenish the pool of com-
mitted progenitor cells, ultimately acting as a source of all Stem cell Asymmetrical Stem cell
division +
types of blood cells, while maintaining themselves through-
out the life of the individual. Antibodies for surface markers
can now be used to distinguish and purify the various types Committed
of hematopoietic progenitors as well as stem cells from mice cell
and humans. Once separated from the far more numerous
mature and differentiating cells in bone marrow, stem cells
Symmetrical Differentiated
can be used for transplantation into patients with bone division 2 stem cells progeny
marrow defects.
Most multipotent hematopoietic stem cells are in the G0 FIGURE 41.4  TWO PATTERNS OF STEM CELL DIVISION.
phase of the cell cycle. A low level of metabolic activity is Asymmetrical divisions create two daughter cells: a stem cell that
thought to contribute to their longevity, which can poten- remains associated with its niche cell to maintain the pool of stem
tially exceed the life span of the individual. When stimulated cells and one that is committed to multiply and produce differenti-
by demand for more blood cells, growth factors drive multi- ated progeny. Symmetrical divisions produce two stem cells to
potent stem cells into a cell cycle that culminates in an expand the pool of stem cells.
asymmetrical division. One daughter cell is another multipo-
tent stem cell. The second daughter cell enters the proliferat- cycle of the stem cell. Some of these factors stimulate divi-
ing pool of blood cell precursors as a committed progenitor sion; others inhibit differentiation. The niches occupied by
cell. Committed progenitor cells and their progeny prolifer- germ cells and neural stem cells from invertebrates are par-
ate vigorously and differentiate into mature blood cells. An ticularly well characterized. During asymmetrical divisions
adult human produces more than one million blood cells of these stem cells, the renewed stem cell stays behind in
every second. the niche, while the daughter that is destined to differentiate
Cytokines and other growth factors regulate proliferation into an egg, sperm, or neuron is released. In bone marrow,
and differentiation at every stage of blood cell production. osteoblasts (see Fig. 32.5) and endothelial cells (see Fig.
The later stages are best understood. For example, the cyto- 30.13) provide niches for hematopoietic stem cells.
kine erythropoietin acts through a kinase-coupled receptor
to activate a cytoplasmic transcription factor that stimulates Epidermal Stem Cells
the proliferation and differentiation of the red blood cell Skin is an example of a continuously renewing organ with a
lineage. Other cytokines guide the differentiation of granu- considerable capacity for regeneration (see Fig. 40.1). Mul-
locytes and monocytes. Hematopoietic stem cells respond to tipotential and committed stem cells contribute to both
the same families of growth factors that control other aspects renewal and regeneration. Committed stem cells reside in
of development, including Wnts (see Fig. 30.7), Notch (see the basal layer of the epidermis. Asymmetrical cell divisions
Chapter 24), fibroblast growth factor (see Fig. 24.4), and oriented at right angles to the basal lamina produce two
insulin-like growth factor (see Fig. 24.4). However, too little daughter cells. The daughter touching the basal lamina
is known about these regulatory mechanisms to grow hema- carries on as the stem cell. The apical daughter cell divides
topoietic stem cells in the laboratory. multiple times and differentiates into an ascending column
of cells, forming the superficial layers of the epidermis (see
Properties of Adult Stem Cells Fig. 35.6). Multipotent stem cells associated with hair folli-
Years of detailed analysis in the laboratory and clinic estab- cles give rise to all the cells of the hair follicle and also serve
lished hematopoietic stem cells as a model for stem cells in as a reserve for the committed epidermal stem cells in the
other tissues. General features include the capacity for self- event of injury (Fig. 41.5).
renewal and the production of daughters that proliferate and
differentiate. This dichotomy can be achieved by asymmetri- Skeletal Muscle Stem Cells
cal cell divisions guided by the same types of internal cues Small numbers of stem cells reside in a niche sandwiched
that control unequal divisions of cells in early embryos (Fig. between the basal lamina and the giant multinucleated
41.4). Symmetrical divisions yielding two daughter stem muscle cells. If the muscle is damaged, these quiescent “satel-
cells can also expand the numbers of stem cells during lite cells” multiply and produce muscle cells that regenerate
growth to maturity and during regeneration of damaged the tissue. Positive signals for proliferation and differentiation
tissues. come through receptor tyrosine kinases and the mitogen-
Stem cells depend on local environmental cues to main- activated protein (MAP) kinase pathway (see Fig. 27.6) and
tain their status as stem cells. These special environments, other pathways. Restraining signals are provided by myo-
called stem cell niches, are created by tissue cells and the statin, a member of the transforming growth factor (TGF)-β
extracellular matrix. Niche cells anchor stem cells with adhe- family (see Fig. 27.10). Inactivation of the myostatin pathway
rens junctions and provide cell surface and secreted proteins results in massive enlargement of muscles in mice and
that activate the signaling pathways that regulate the cell humans. Muscles are capable of regenerating multiple times,
CHAPTER 41  n  G1 Phase and Regulation of Cell Proliferation 717

BOX 41.2  Stem Cells in Mammals—cont’d

Epidermis

t
haf
ir s
r
Ba
sa

ye
Ha
l la
Sebaceous
gland
Dermis

Bulge

Hair
bulb
A B C
FIGURE 41.5  STEM CELLS FROM SKIN. Multipotent stem cells of the skin reside in the hair follicle bulge (green cells in A, diagram
in C). They move up and repair the epidermis during wound healing, and they move down and generate new hair growth during the hair
cycle. B, Depicts a Nude mouse grafted with the cultured cell progeny of a single “bulge” stem cell and displaying a large tuft of hair, all
derived from that single stem cell. (A and C, From Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and their niche.
Cell. 2004;116:769–778. B, From Blanpain C, Lowry WE, Geoghegan A, et al. Self-renewal, multipotency, and the existence of two cell
populations within an epithelial stem cell niche. Cell. 2004;118:635–648.)

so the stem cell population renews itself during regeneration designing the original DNA construct so that when it enters
or is augmented by stem cells that migrate through the blood the chromosome by homologous chromosome, a critical
from bone marrow or other tissues. region of a target gene is deleted or disrupted. The use of
knockout mice has revolutionized the study of developmen-
Cancer Stem Cells tal biology by allowing investigators to determine the func-
Stem cells may play a role in cancer, acting as a source for tion of specific genes in living animals.
proliferating cells that make up the bulk of the tumor. If true, Note the distinction between transgenic animals and
this concept helps explain why it is relatively easy to reduce reproductive cloning. “Cloned” animals are produced by
the size of tumors by targeting dividing cells but difficult to introducing a somatic cell nucleus into an enucleated egg.
completely eliminate residual tumor stem cells, which may Experiments first in frogs and later in mammals, such as
divide less frequently. It is thought that the spread of cancer Dolly the sheep, established that egg cytoplasm can repro-
from the primary tumor to other tissues (metastasis) gram gene expression of differentiated cell nuclei, and
requires circulating cancer cells to find locations (niches) enable the development of a cloned animal. Transfer of
where they can establish themselves as stem cells. nuclei from lymphocytes and olfactory neurons has been
used to derive healthy adult mice. This approach involves
Meristematic Stem Cells in Plants the reversal of epigenetic changes in the nucleus that drove
The growth of plants depends on carefully orchestrated pro- the differentiation of the adult cell. The molecular mecha-
liferation and differentiation of cells derived from stem cells nisms underlying reprogramming are not well understood
called meristems. Through asymmetrical divisions, these and the success rate at obtaining healthy animals through
relatively inactive cells give rise to daughters that proliferate nuclear cloning is very low. This cloning does not involve
at the tips of shoots and roots. The proliferating cells dif- the use of stem cells, but embryonic stem cells can be
ferentiate into specialized tissues such as flowers, while the derived from the blastocyst stage of the cloned embryos.
stem cells maintain a pool of slowly replicating cells in a
special niche. Therapeutic Applications of Stem Cells
Where committed stem cells can be isolated from an adult
Use of Stem Cells to Make organ, it is now possible to regenerate damaged tissues by
Transgenic Animals transplanting these stem cells from patients themselves or
Because embryonic stem cells grow in culture, they can be from donors. The best example is transplantation of bone
manipulated experimentally. They can be transfected with marrow stem cells to treat patients whose bone marrow has
DNA, and if the proper sequences are present, this DNA can been damaged by cancer, chemotherapy, or other disease.
replace a region of the endogenous chromosome by homolo- Adverse immunologic reactions are a challenge for trans-
gous recombination. If the modified embryonic stem cells plants from donors other than an identical twin. On one
are subsequently injected into developing embryos at the hand, the immune system of the recipient can reject the
blastocyst stage, they are, with low frequency, able to colo- transplanted cells. On the other hand, lymphocytes contami-
nize the cell population that will produce germ cells. When nating the donor stem cells can mount an immunologic
such chimeric embryos grow to adulthood, a proportion of attack on the recipient. Using purified hematopoietic stem
their gametes will carry a chromosome with the modification cells (ideally, the patient’s own stem cells) rather than mixed
engineered in the embryonic stem cells. Furthermore, this bone marrow cells avoids this problem. Knowing how to
chromosome will now be inherited by all progeny of that expand hematopoietic stem cells in vitro would be helpful.
embryo, giving rise to a line of transgenic animals. This This approach is already used for treating burns with epider-
method is widely used in research to knock out genes by mal stem cells. Normal skin is used as a source of committed

Continued
718 SECTION X  n  Cell Cycle

BOX 41.2  Stem Cells in Mammals—cont’d

skin stem cells, which are multiplied in culture and used to Myc. When these factors are ectopically expressed in dif-
regenerate all the layers of the skin. ferentiated cells, they can induce the expression of proteins
Stem cells might be used to regenerate other damaged required for pluripotency, as well as factors required to
tissues, including the insulin-producing cells that are lost in change the epigenetic landscape of the cell. This results in
Type I diabetes, but appropriate stem cells are not available a stable perpetuation of pluripotency. These induced plu-
for many organs, including the pancreas, brain, and heart. ripotent stem (iPS) cells, offer the promise that they can
Indeed, even with appropriate stem cells in hand, much be differentiated into any desired specific cell type for
remains to be learned about how to grow them and then medical applications. This procedure potentially eliminates
direct them to differentiate into mature tissues. the problem of the availability of autologous stem cells as
Embryonic stem cells can potentially supply all cells nec- many cell types can be reprogrammed to iPS cells. This
essary to replace any damaged tissue, but sources of human technology is rapidly developing, as researchers attempt to
embryonic stem cells are limited, and acquiring them from improve the generation of functional cell types from iPS cells
early embryos discarded by fertility clinics is unacceptable When injected into immunodeficient mice, iPS cells make
to some people. Patient-specific (autologous) embryonic tumors containing all cell types, called teratocarcinomas.
stem cells can be derived via somatic cell nuclear transfer This clearly shows that the iPS cells are pluripotent. They
(cloning) followed by expansion of epiblast cells from the can also generate all cell types in tissue culture when appro-
embryo. However, the production of such “artificial” human priate growth factors necessary for self-renewal are removed
embryos is also highly controversial. from culture media. However, this differentiation is difficult
Adult stem cells are an alternative to embryonic stem to control. We cannot yet generate specific fully functional
cells. This approach has the advantage that stem cells can cell types necessary for therapies with high purity from
be isolated from bone marrow, blood, and skin by using pluripotent stem cells, except for a few cell types such as
antibodies that recognize specific surface protein “markers”; retinal pigment epithelial (RPE) cells. iPS cell-derived RPE
however, these specialized stem cells normally do not cells were transplanted for the treatment of macular degen-
produce differentiated cells for regeneration of other tissues. eration in 2014, and appeared to be successful in blocking
An alternative method to generate pluripotent stem cells the degeneration. However, as of 2016, this was the only
is based on the transient reintroduction into a differentiated clinical trial performed with iPS cell-derived cells. Investiga-
cell of a group of specific transcription factors commonly tion on how to control iPS cell differentiation is one of the
expressed in stem cells. These include Sox2, Oct4, Klf4, and main obstacles to be overcome for regenerative medicine.

Reentry Into the Cell Cycle Immediate early Delayed


transcription factors early genes
Cells in the G0 phase may reenter the growth cycle Immediate early
Levels of expression

structural proteins
in response to specific stimulation by mitogens, often
induced by injury or normal cell turnover. Cultured fibro- Immediate early
tissue repair
blasts are favored for laboratory studies of this process, proteins
as they readily enter a quiescent state mimicking G0
0 1 2 3 4 5 6
phase when deprived of serum (ie, mitogens and growth Time (hours)
Cdk inhibitors
factors) and rapidly reenter the cell cycle when serum
is restored. This response reproduces that found in Addition
of serum or
wounded tissues. When a living tissue is wounded (see growth factor

Fig. 32.11), fibroblasts are exposed to serum that has


leaked from damaged blood vessels. In response, they FIGURE 41.6  PATTERNS OF EXPRESSION OF IMMEDIATE
divide and colonize the wound, where they lay down AND DELAYED EARLY GENES DURING THE RETURN OF
new extracellular matrix to repair the damage. GROWTH-ARRESTED FIBROBLASTS FROM G0 TO ACTIVE
PROLIFERATION AND THE CELL CYCLE.
Serum stimulates three waves of gene expression in
cultured quiescent fibroblasts (Fig. 41.6). The first
includes more than 100 “immediate early” genes. activities involved in angiogenesis (blood vessel forma-
These include transcription factors of the Jun, fos, myc, tion), inflammation, and coagulation. These proteins
and zinc finger families (see Chapter 10) that activate facilitate the movement of fibroblasts into wounds and
numerous downstream genes required for cell growth initiate the repair of tissue damage.
and division. Other immediate early genes encode tissue Expression of a second wave of “delayed early”
remodeling factors, cytokines (growth factors), extracel- genes encoding a variety of proteins that are required for
lular matrix components (fibronectin), plasma mem- cell growth and proliferation, including cyclin D, pre-
brane adhesion proteins (integrins), and cytoskeletal cedes the onset of the S phase. Genes activated after the
proteins (actin, tropomyosin, vimentin), as well as onset of the S phase are referred to as “late” genes. Both
CHAPTER 41  n  G1 Phase and Regulation of Cell Proliferation 719

delayed early and late gene transcription require synthe- amputations were stopped, the amoeba that had been
sis of the transcription factors encoded by immediate operated on divided within 38 hours. The interpretation
early genes. of this experiment was that the repeated amputations
These waves of transcription in response to mitogens prevented the experimental amoeba from ever attaining
enable the G0 cells to pass through a “gate” and reenter a size sufficient to turn on the division program. Evi-
the active cell cycle. This gate is analogous to the restric- dence suggests that some types of human cells have a
tion point, a critical aspect of G1 control that regulates similar size control while others do not.
the proliferation of all normal cells. An essential aspect of growth control during the G1
phase involves monitoring the external environment
The Restriction Point: A Critical G1 for nutrient availability and for signals to proliferate
(mitogenic signals) coming from other cells and from
Decision Point
the extracellular matrix. In a classic experiment, when
All eukaryotes have a mechanism that operates during three flasks containing populations of cultured cells were
the G1 phase to ensure that cells duplicate their genome starved by deprivation of amino acids, serum, or phos-
only when the environment is supportive and the chro- phate respectively, they all stopped cycling in G1. When
mosomes are undamaged. Healthy yeast cells do not the missing ingredients were restored, cells in all three
embark on a round of DNA replication and division until flasks resumed the cell cycle and entered the S phase at
they reach an appropriate minimum size (actually, they about the same time. This was surprising because amino
probably measure their ribosome content and ongoing acids are needed to make protein, serum provides growth
rate of protein synthesis). This is important because after factors and mitogens, and phosphate is needed for syn-
cell division, the daughter cell (bud) is smaller than the thesis of DNA and phospholipids (needed to make mem-
mother and needs more time to grow before it divides branes). This experiment was interpreted as evidence
if the population is to maintain a constant cell size. that all three types of starvation caused cells to arrest at
Whether dividing mammalian cells also monitor their a single point in the G1 phase, termed the restriction
size is not yet settled. point. The restriction point is now defined as the point
The influence of cell size on the division cycle was after which the cell cycle will proceed even if mitogenic
first demonstrated in an elegant microsurgery experi- factors are withdrawn (Fig. 41.8). This supremely impor-
ment (Fig. 41.7). Two Amoeba proteus cells were grown tant aspect of cell-cycle control prevents cells from divid-
under identical conditions in parallel cultures. Each day, ing at inappropriate times and in inappropriate places.
a portion of the cytoplasm was amputated from one Defects in restriction point control are among the most
amoeba, and the other was left untouched as a control. common causes of cancer.
Under those circumstances, the cell that suffered the Genetic analysis also revealed a point in the G1 phase
amputations did not divide for 20 days. During this time, after which budding yeast cells appear to be committed
the control amoeba divided 11 times. When the to completion of the cycle. Cells that are starved for

Amoeba A. Control

Nucleus

B. Experiment

FIGURE 41.7  A MICROSURGERY EXPERIMENT DEMONSTRATES THAT AMOEBAE WILL NOT DIVIDE IF THEY ARE PREVENTED
FROM ATTAINING A SUFFICIENT SIZE. A, Control cell continues to divide. B, Experimental cell does not divide. (For reference, see Prescott
DM. Relation between cell growth and cell division. II: The effect of cell size on cell growth rate and generation time in Amoeba proteus. Exp Cell
Res. 1956;11:86–98.)
720 SECTION X  n  Cell Cycle

Restriction point kinase (GSK)-β (see Fig. 30.7) and marked by SCF (Skp,
Cullin, F-box containing complex) for degradation (see
M G1 S
Fig. 40.16). Rb/E2F/DP represses the expression of the
genes for cyclins E and A required for Cdk2 activation.
Cell continues to cycle only if Cell committed Furthermore, high levels of the CKI class Cdk inhibitor
extracellular signals are received to cycle
p27Kip1 inhibit any Cdk2–cyclin E or Cdk2–cyclin A that
FIGURE 41.8  RESTRICTION POINT. During late G1, cells assess happens to be present (see Fig. 40.14).
external and internal stimuli and decide whether to commit to a further
round of DNA replication and division.
Signals from mitogens and the extracellular matrix
open the restriction point gate. Stimulation of receptor
tyrosine kinases (see Chapters 25 and 27) or integrins
nutrients arrest at, or just prior to, this point, termed (see Chapter 30) activates Ras and the mitogen-activated
START. The mammalian restriction point resembles protein (MAP) kinase/extracellular signal–regulated
yeast START in a number of aspects, but they are not kinase (ERK) cascade (see Fig. 27.6). The output of this
exactly equivalent, owing to differences between animal cascade stimulates transcription of D-type cyclins (Figs.
and yeast cell cycles. 41.9 and 41.10) and also inactivates GSK. This allows
cyclin D to accumulate in nuclei. Nuclear cyclin D binds
Regulation of Cell Proliferation by to and activates Cdk4 and Cdk6 (referred to hereafter as
Cdk4/6–cyclin D), producing an initial pulse of Cdk
the Restriction Point
activity that is later amplified by Cdk2–cyclin E and
The restriction point is a molecular “gate” that regu- Cdk2–cyclin A. Cdk activity in early G1 is regulated by
lates the expression of genes required for cell-cycle pro- adjusting the relative levels of the three D-type cyclins
gression. The gate is based on proteins that are related as well as the levels of Ink4 and CKI inhibitors. This
to the Rb susceptibility protein and a family of essential regulation of cyclin D levels and Cdk inhibitors provides
transcription factors known as E2F. the crucial link between extracellular mitogens and the
In brief, E2F is a master transcriptional regulator that cell cycle.
activates many of the genes whose products drive DNA Mitogens also stimulate transcription of the CKI class
replication and cell-cycle progression. Rb regulates the Cdk inhibitor p27Kip1. This protein actually activates
cell cycle by binding to E2F and converting it into a Cdk4/6–cyclin D complexes in two ways. First, the
repressor of those same cell-cycle genes. When Rb is receptor associated tyrosine kinases Jak or Src phos-
bound to E2F, the cell is said to be arrested at the restric- phorylate p27, inducing a structural change within the
tion point. Escape from this arrest involves Cdk activa- Cdk4/6–cyclin D-p27Kip1 complex that activates the
tion and subsequent Rb phosphorylation. This releases kinase. This links mitogen signaling to Cdk-4/6 activa-
E2F, which then drives cell-cycle progression. An alterna- tion. It also promotes the nuclear import of Cdk4/6–
tive way to pass the restriction point gate depends on cyclin D. This both leads to full activation of Cdks by
a potent regulator called Myc, which is discussed Cdk-activating kinase, a nuclear enzyme (see Chapter
separately later. 40), and increases the stability of cyclin D. All of this
Mammals have three Rb-related proteins (pRb, p107, depends on the continuous presence of mitogenic
and p130) and approximately eight E2F family members. signals; if these cease, then cyclin D stability rapidly
These together constitute a complex multifunctional declines again, since following dephosphorylation, p27
network. This chapter refers to the families generically and p21 again act as Cdk4/6–cyclin D inhibitors.
as Rb and E2F. Some E2F proteins form a heterodimer Cdks push the cell past the restriction point by phos-
with one of three DP (differentiation-regulated transcrip- phorylating Rb, causing it to dissociate from E2F (Fig.
tion factor-1 protein) family members. This dimer associ- 41.9B; see also Chapter 40 and Appendix 40.1). The E2F/
ates with the promoter region of E2F target cell-cycle DP heterodimer remains bound to promoter regions and
genes (Fig. 41.9A). Rb binding converts E2F/DP from a now potently activates, rather than represses, the tran-
transcriptional activator to the Rb/E2F/DP repressor. Rb scription of genes that stimulate cell proliferation. The
also recruits histone deacetylases, enzymes that remove proteins produced synthesize DNA (DNA polymerase α,
acetyl groups from a wide range of proteins, including accessory factors, and enzymes that synthesize nucleo-
the aminoterminal tails of histones (see Fig. 8.7). This tide precursors; see Chapter 42), promote cell-cycle pro-
causes compaction of chromatin structure and represses gression (cyclins E and A, Cdk1, and Cdc25), and regulate
genes required for cell-cycle progression. cell-cycle progression (pRb, p107, Emi1).
Nonproliferating cells have low Cdk activity in G1 The chain of events as mitogens break the blockade
before the restriction point. First, cyclin D messenger on cell-cycle progression imposed by Rb involves a posi-
RNA (mRNA) levels are low, so little protein is made. tive feedback loop as follows. Cdk4/6–cyclin D com-
Second, any cyclin D that is made is retained in the cyto- plexes begin to phosphorylate Rb. This releases some
plasm, where it is phosphorylated by glycogen synthase E2F and permits the initial expression of genes that
CHAPTER 41  n  G1 Phase and Regulation of Cell Proliferation 721

A. Absence of mitogens
Tyrosine External
kinase Seven-helix signals
receptor receptors
Cyclin D
Ras Cyclin D
Transcription (stable)
Restriction
point
+ Cdk 4/6
M G1 S Kinase phosphorylates p27 p27-P
tyrosine on p27
Cdk 4/6–cyclin D–p27-P
E2F/DP Rb Gene off (active kinase)

Nucleosome Histone Histone Enters


deacetylase N-terminal tails nucleus
Phosphorylates Rb
Histone deacetylation results in chromatin
compaction and repression of transcription
Passage of
restriction point
B. Mitogens present
G1 S G2 M
Tyrosine
kinase Seven-helix FIGURE 41.10  HOW GROWTH FACTORS REGULATE CDK4/6
receptor receptors
Steroid ACTIVITY: THE ROLE OF D-TYPE CYCLINS AND P21. Mitogen-
Ras receptors cAMP induced tyrosine phosphorylation of p27 determines if it assembles
Raf PKA active or inactive CDK4/6–cyclin D complexes–linking assembly of
MEK Synthesis and stability of active kinase to mitogens.
ERK cyclin D, etc

M G1 S
until the exit from mitosis. Rb is dephosphorylated at the
Histone mitosis-G0 or G1 transition. This enables it once again to
4/6 D Rb deacetylase
bind E2F and close the restriction point gate to exit from
Cdk 4/6 Cell-cycle genes the next G1.
cyclin D RNA (cyclins A, E, Cdk1)
E2F/DP pol II DNA replication genes The transcriptional regulator Myc drives an alterna-
tive pathway for G1 exit that is also stabilized by
mitogenic signals. Association of myc with one partner
AC AC AC AC AC activates the transcription of cyclins E and D2. Associa-
AC AC AC AC AC
tion with a different partner downregulates the tran-
Acetylated "open" chromatin favors transcription
scription of Cdk inhibitors of both the CKI and INK
FIGURE 41.9  REGULATION OF CELL-CYCLE PROGRESSION classes. These activities partly explain why Myc can act
BY THE E2F/DP/RB COMPLEX. A, The Rb/E2F/DP complex as an oncogene—a protein that helps transform normal
recruits histone deacetylases (see Chapter 8) and represses specific
genes that are required for cell-cycle progression. This blocks cell-
cells into cancer cells (explained further later).
cycle progression at the restriction point. B, Phosphorylation of Rb by
Cdks alleviates this block and permits passage of the restriction point.
cAMP, cyclic adenosine monophosphate; MEK, mitogen-activated
Restriction Point and Cancer
protein kinase kinase; PKA, protein kinase A. Cancer is a complex class of diseases in which genetic
changes within clones of cells lead to production of cell
populations whose uncontrolled growth can disrupt
encode cyclin E, cyclin A, and CDC25A. Active Cdk2- tissue function and ultimately kill the individual. Two in
cyclin E can initiate p27Kip1 degradation, permitting the five Americans will be affected by cancer during their
rapid accumulation of active Cdks. The restriction point lifetimes. This sounds very high, but considering the
probably is passed here, and from this point on Cdks number of cell cycles that are required to produce a
remain active until cyclin destruction in late mitosis. human composed of approximately 1014 cells, and con-
Cdk2–cyclin E participates in a second wave of Rb sidering the over 1 million cell divisions that occur per
phosphorylation on many sites, leading to the wholesale second in a healthy adult, the disease is actually remark-
liberation of E2F/DP and a surge in transcription of genes ably rare on a per cell basis. This is at least partly because
that trigger the onset of DNA replication (S phase entry) multiple genetic alterations are required to transform a
and promote progression through the cell cycle. As the normal cell into a cancer cell. Furthermore, the cell cycle
cell cycle proceeds, Rb phosphorylation is maintained is highly regulated by a web of negative feedback path-
first by Cdk2–cyclin A and then later by Cdk1–cyclin B ways that hold in check activities driving cellular
722 SECTION X  n  Cell Cycle

protein products typically inhibit products of oncogenes


Cells traversing the cell cycle or negatively regulate cell proliferation. Several genes
that are involved in restriction point control can act as
either oncogenes or tumor suppressors.
More than 100 oncogenes have been identified thus
far. Most normally function in signal transduction path-
ways that lie downstream of mitogens that stimulate
cell-cycle progression. Their inappropriate activation
Normal Cancer can mimic the effects of persistent mitogenic stimula-
tion, thereby uncoupling cells from normal environmen-
tal controls and leading to uncontrolled proliferation and
cancer. For example, Ras activates the MAP/ERK kinase
cascade and accumulation of cyclin D (Fig. 41.9). Ras
Contact inhibition:
Signaling from cadherin-based
genes are mutated in approximately 15% of human
adherens junctions stops cells Transformed cells cancers. Inappropriate activation of Ras tricks the cell
from cycling. They arrest in G1. continue to cycle into thinking that it is receiving mitogenic signals,
leading it to express cyclin D, phosphorylate Rb, and
FIGURE 41.11  LOSS OF GROWTH CONTROL IN TRANS-
FORMED CELLS. proliferate.
Fortunately, in most cells, mutations that result in
proliferation. In fact, many cancer-causing mutations uncontrolled proliferation usually lead to DNA damage
that disturb growth control pathways are actually delete- (oncogenic stress) and activate a protective pathway
rious in normal cells and cause them to undergo senes- leading to senescence. Other proteins involved in restric-
cence or commit suicide by apoptosis (see Chapter 46). tion point control can also act as oncogenes if hyperac-
Dysregulation of cell proliferation in the G1 phase tivated. These include E2F1, cyclin D (overexpressed in
causes most types of cancer. This is readily seen in the 50% of breast cancers), and Cdk4. In each case, activa-
laboratory when cells are grown on plastic tissue culture tion of the protein causes inappropriate transcription of
dishes. Most normal cells proliferate until they cover the genes promoting cell-cycle progression, bypassing the
surface completely, forming a monolayer. When the restriction point, and leading to uncontrolled cell cycles
monolayer is confluent (ie, when cells are touched by and cancerous transformation (Fig. 41.12).
other cells on all sides), signaling initiated by cadherin Rb is one of the best-characterized tumor-suppressor
proteins (see Fig. 30.7) causes cells to express p27Kip1 genes. As discussed earlier, a primary function of Rb is
and to arrest their cell-cycle progression in G1. This is to block cell-cycle progression until sustained mitogenic
called contact inhibition of growth (see Chapter 30, stimulation results in its inactivation. It is therefore not
in the section titled “Cadherin Family of Adhesion Recep- surprising that loss of Rb can lead to inappropriate cell-
tors”). Cancer cells lack this control, so they keep pro- cycle progression and cancer. Rare individuals who
liferating and piling up on top of one another as long as inherit one defective Rb gene tend to develop retinoblas-
nutrient and mitogen supplies last (Fig. 41.11). Cells that tomas as children and osteosarcomas as adults. The
lose this aspect of growth regulation are said to be cancer arises when the “good” allele is inactivated in a
transformed. proliferating cell (this is called a somatic mutation). Such
Malfunction of the restriction point is an extremely cancers are rare and occur only later in life in individuals
common contributor to transformation. Indeed, one or who inherit two good Rb genes, as two independent
more components of the p16/cyclin D/Cdk-4/Rb system somatic mutations (two “hits”) are required in the same
are mutated in most human cancers. In addition, several proliferating cell. Homozygous loss of Rb is lethal during
cancer-causing viruses, such as simian virus 40 (SV40), embryogenesis. This is partly because under some cir-
papillomaviruses, and adenovirus, make proteins that cumstances, the unleashed E2F can act as a potent
facilitate the G1 → S transition by binding Rb and inducer of apoptotic cell death (see Chapter 46).
liberating E2F. P16Ink4a is another important tumor suppressor
Most cancer cells have abnormalities in the activities involved in G1 growth control. Normally, it suppresses
of two classes of genes. Oncogenes are genes whose Cdk4/6 activity in nondividing cells (see next section;
inappropriate activation can cause oncogenic (cancer- also see Chapter 40), thereby reinforcing the ability of
ous) transformation of cells. The protein products of Rb to maintain the growth arrest of G1 cells (Fig. 41.12).
most oncogenes regulate cellular growth and prolifera- Mutations in the p16Ink4a gene are very common in
tion. They are typically components of signal trans­ cancer, but this is partly because this gene is fascinat-
duction pathways that are controlled by feedback ingly complex (Fig. 41.16). Mutations in other INK4 Cdk
mechanisms. Tumor suppressors are genes whose inhibitors and the CKI protein p27Kip1 are also found in
inactivation can lead to cancerous transformation. Their cancer, although less frequently.
CHAPTER 41  n  G1 Phase and Regulation of Cell Proliferation 723

KEY Transcription
=

Increasing Cdk activity


prevented Normal cell arrested Nutrients
Transcription of cyclins A, E, = +1 at restriction point Mitogens
Cdk 1 DNA replication genes
STOP Cdk2–cyclin E
No external signals
Rb
Cdk4/6–cyclin D

Normal cell passes M G1 S


Cdk 4/6–cyclin D restriction point

Increasing ubiquitin ligase activity


External signals (active) Restriction point
Rb GO

+1

APC/CCdh1 Emi1
Rb mutant cell passes SCFSkp2
restriction point SCFβ-TrCP

No external signals GO
Degraded proteins: Cyclins Cdc25A p27Kip1
Rb Danger

+1
FIGURE 41.13  PROTEOLYTIC ACTIVITIES IN G1 PHASE.

No external signals Cell with active oncogene Phosphorylated p27Kip1 is recognized by a specific E3
passes restriction point
Active oncogene Cdk 4/6–cyclin D ubiquitin ligase called SCFSkp2 (see Figs. 40.15 and 40.16).
mimics external (active)
signals Rb GO The resulting destruction of p27Kip1 permits a burst of
Danger
Cdk2-cyclin E activation in a positive feedback loop that
+1 rapidly amplifies Cdk activity at the initiation of the S
phase. Later in the S phase, phosphorylation of the DP
Cdk 4/6–p16Ink4a Differentiated cell expressing
subunit of E2F causes its dissociation from DNA, recogni-
(inactive) p16 does not cycle tion by SCF, and destruction. This is essential to com-
External signals plete S phase. SCF also targets cyclins D1 and E for
may be present
STOP destruction, the former when mitogens are limiting and
Rb the latter following autophosphorylation during progres-
sion through the S phase.
Progression throughout G1 requires the activity of
*p16Ink4a
a second E3 ubiquitin ligase known as the anaphase-
Differentiated cell mutant for promoting complex or cyclosome (APC/C). A specific
Cdk 4/6 p16 passes restriction point
cofactor, Cdc20, activates the APC/C to trigger the
metaphase-to-anaphase transition (see Chapter 40). As
External signals Cdk 4/6–cyclin D the cell leaves mitosis, Cdh1 replaces Cdc20 and targets
may be present (active)
Rb GO many cell-cycle regulatory proteins for degradation,
Danger
including Cdc20, A- and B-type cyclins, and factors
+1 involved in DNA replication. Destruction of these target
proteins during mitotic exit and G1 (Fig. 41.13) likely
contributes to the requirement for transcription and de
FIGURE 41.12  HOW ACTIVATED ONCOGENES OR MUTA-
TIONS IN THE RB OR P16 TUMOR SUPPRESSOR PROTEINS novo synthesis of these proteins at the moment cells
CAN LEAD TO ABNORMAL PASSAGE OF THE RESTRICTION decide whether to enter or not a new cycle of genome
POINT AND CANCER. replication.

Proteolysis and G1 Cell-Cycle Progression Integrity of Cellular DNA Monitored by


a G1/S Checkpoint
Just as controlled destruction of proteins is key to the
transition of cells from mitosis to the G1 phase (see The S phase is a point of no return in the history of any
Chapter 40), proteolysis also fulfills several key roles dividing cell. Given the semiconservative mechanism of
during progression from G1 into the S phase (Fig. 41.13). DNA replication, whereby existing DNA strands serve as
For example, when Cdk2–cyclin E is activated following templates for newly synthesized strands, any DNA defect
cyclin D synthesis, it phosphorylates its inhibitor p27Kip1. that passes unnoticed through the S phase becomes
724 SECTION X  n  Cell Cycle

Undamaged DNA dsDNA breaks ssDNA

(ATM)2 ATR not (ATM)2 ATM ATR active


inactive signaling active localized to
damage site

E2F drives p53 present E2F Mdm2 p53 Chk1


expression of in low amounts active inactive active kinase
cell cycle genes in cytoplasm in nucleus
Cdc25A

p21 Degraded

Normal cell-cycle progression Apoptosis Stable cell- Rapid cell-


cycle arrest cycle arrest
FIGURE 41.14  THE G1/S CHECKPOINT AND DNA DAMAGE RESPONSE.

perpetuated as a mutation that is transmitted to all future DNA double-strand breaks vigorously activate ATM,
progeny of the cell. Furthermore, any single-stranded which directly and indirectly stabilizes and activates a
nick in DNA may become a full-fledged chromosome critical tumor suppressor, p53. This transcription factor
break if present during replication. To avoid these prob- has been called the “guardian of the genome,” because
lems, cells have a quality control mechanism to block in response to DNA damage it also applies a rapid brake
entry into the S phase if damaged DNA is detected. to cell cycle progression. In some cases this arrest leads
This quality control mechanism involves a check- to senescence, a permanent cessation of the cell cycle
point that operates throughout the G1 and S phases (putting the car up on blocks).
(Fig. 41.14). Checkpoints are biochemical circuits super- p53 is very powerful medicine for the cell cycle and
imposed on the normal cell cycle. When activated, the must be carefully regulated by a ubiquitin ligase (E3)
G1/S checkpoint triggers a DNA damage response called Mdm2 (mouse double-minute 2; the human ortho-
that blocks cell-cycle progression. The block may be log is Hdm2) that keeps p53 levels low when the cell
temporary but, in some cases, checkpoint activation cycle is running normally (Fig. 41.15A). Both p53 and
leads to senescence or cell death by apoptosis. Sensor Mdm2 protein shuttle in and out of the nucleus (see
proteins detect DNA damage and activate this check- Chapter 9). When the two proteins associate in the cyto-
point. In the subsequent DNA damage response, the plasm, Mdm2 promotes rapid degradation of p53
sensor proteins activate protein kinases and a key tran- by the ubiquitin/proteasome system (see Chapter 23).
scriptional regulator that block cell-cycle progression Because p53 directly stimulates expression of Mdm2, a
(see Fig. 40.4). feedback loop keeps levels of p53 low. Loss of the Mdm2
The DNA damage response has fast and slow compo- gene in mice is lethal unless the p53 gene is also lost.
nents: The former is analogous to applying the brakes in Both p53 and Mdm2 are phosphorylated following
a car; the latter is analogous to removing the wheels and DNA damage (Fig. 41.15B). These phosphorylations
putting it up on blocks. Both components start with prevent Mdm2 from binding, so p53 is stabilized, and its
the protein kinases, ATM and ATR (see Fig. 40.4). ATM concentration in the nucleus increases dramatically. The
and ATR are related to the lipid kinase phosphatidylino- phosphorylations also make p53 a more potent transcrip-
sitol 3-kinase (see Fig. 26.7), but their only known tional activator. The result is a burst of transcription of
substrates are proteins (see Chapter 40). People lacking p53-regulated genes.
ATM have the disease ataxia-telangiectasia, which is p53 is rapidly activated in response to DNA damage
characterized by immunodeficiency, photosensitivity, resulting from hyperproliferation of cells following loss
cerebellar degeneration, and an elevated incidence of of restriction point control (oncogenic stress). It also
leukemias and lymphomas. Loss of ATR is fatal. responds to DNA damage induced by the environment.
DNA damage that disrupts ongoing DNA replication If the damage is rapidly repaired, cells continue to cycle,
and produces single-stranded DNA activates ATR, which but if the damage is too severe, p53 induces senescence
phosphorylates and activates a downstream kinase called or apoptotic cell death (see next paragraph).
Chk1. Chk1 targets the essential phosphatase CDC25A In the case of oncogenic stress following loss of
(Fig. 41.14), marking it for destruction. Because CDC25A restriction point control (eg, by Ras mutations) E2F stim-
is required to remove inhibitory phosphate groups from ulates the expression of the tumor suppressor protein
inactive Cdks, its destruction applies a rapid brake to p19Arf (alternate reading frame). This binds and seques-
cell-cycle progression. ters Mdm2 in the nucleolus (Fig. 41.15C), allowing p53
CHAPTER 41  n  G1 Phase and Regulation of Cell Proliferation 725

A. Healthy cell B. Irradiation C. Oncogene activation


Mdm2 ubiquitin ligase ATM phosphorylates and activates First the irradiation pathway is
directs destruction of p53, which blocks Mdm2 binding activated, then phosphorylation
p53 in cytoplasm of E2F promotes transcription of
p19Arf, which sequesters Mdm2
in nucleoli, releasing active p53

DNA
damage E2F p19Arf
Nucleolus Mdm2
ATM p19Arf
p53 Mdm2 Mdm2
p53-Mdm2 activated cannot p53
bind activated

p53-Mdm2 p53
Ub p53
Ub Active nuclear p53 drives expression Active nuclear p53 drives expression
Ub of proteins that arrest the cell cycle of proteins that arrest the cell cycle
p53-Mdm2 and promote cell death (apoptosis) and promote cell death (apoptosis)

5 3 + Mdm2 p53
p

activated Active p53 also drives expression of the ubiquitin


p53 levels low ligase Mdm2, providing negative feedback on p53
throughout cell Mdm2

FIGURE 41.15  P53 REGULATION AND THE DNA DAMAGE CHECKPOINT IN G1. A, Healthy cell. B, After irradiation, Mdm2 (mouse
double-minute 2) can no longer bind p53, which accumulates in active form in the nucleus. C, After oncogene activation, Mdm2 is sequestered
in the nucleolus, and active p53 accumulates in the nucleus. Activated p53 can induce either cell-cycle arrest or cell death.

to accumulate in the nucleoplasm. There, it activates A. One gene, two promoters


transcription of the CKI p21, stopping the cell cycle. If 1β 1α 2 3
this arrest is prolonged, p16INK4A is induced, activating Ink4a
Arf
Rb and leading to permanent cell cycle arrest (senes-
cence). p53 activation can also induce apoptosis by acti-
vating transcription of genes for proapoptotic proteins, B. Two key proteins Ink4a/Arf
including Bax, BH3-domain proteins, Puma, CD95 (Fas/ p16Ink4a p19Arf
Apo1), and Apaf-1 (Fig. 41.15; also discussed in Chapter
46). The decision whether to induce senescence or
Cdk4-cyclin D1 Rb Mdm2 p53
death is very complex and may be regulated by different
posttranslational modifications of p53. One way or the FIGURE 41.16  DUAL CONTROL OF G1 PROGRESSION BY
other, p53 serves its function as guardian, as the outcome THE P16INK4A/P19ARF GENE. This gene encodes two completely dif-
ferent proteins that are key to avoiding cancer. A, The intron–exon
is that aberrantly proliferating cells are either perma- structure of the p16Ink4A/p19Arf gene. B, p16Ink4A and p19Arf negatively
nently silenced or removed, and the body is protected. regulate the restriction point via Rb and the DNA damage checkpoint
p53 is mutated or deleted in about half of all human via p53, respectively.
cancers. Families that carry a mutated p53 allele have
Li-Fraumeni syndrome with an elevated risk of cancers.
Mice lacking p53 are viable but a defective G1 DNA p16Ink4a (Fig. 41.16). Despite having different promoters
damage checkpoint results in cancers while young. This (that respond to different stimuli), the two genes not
illustrates a common theme that in many cases check- only overlap but also share a common exon. Neverthe-
point components are not essential for life as long as less, the two proteins have no common amino acid
nothing untoward occurs. Checkpoints exist primarily sequences because the shared exons are read in different
to deal with problems that arise during cell-cycle pro- frames in the mature mRNAs (mRNAs) for the two pro-
gression. However, the elevated cancer rates in teins. Thus, the p16Ink4a/p14Arf locus encodes two vital
Li-Fraumeni syndrome patients indicate that although protective factors with different jobs. It is not surprising
p53 is not essential for the passage of every cell cycle, it that mutations in this key locus are found in between
is essential for long-term genetic stability and for main- 25% and 70% of human cancers.
taining a proper balance among cell proliferation, dif-
ferentiation, and death during the lifetime of a mammal.
The p19Arf protein (in humans, the protein is smaller
G1 Regulation: A Matter of Life and Death
and so is called p14Arf) is extremely unusual, as it is To commit to a new cycle of proliferation, cells must
encoded in a common gene with the Cdk inhibitor pass through the restriction point gate. The key to this
726 SECTION X  n  Cell Cycle

gate is the phosphorylation of Rb by Cdks, so signals Childs BG, Baker DJ, Kirkland JL, Campisi J, van Deursen JM. Senes-
such as mitogens that activate Cdks set up a feedback cence and apoptosis: dueling or complementary cell fates? EMBO
Rep. 2014;15:1139-1153.
loop that promotes passage of the gate. Of course, in the Clevers H. The intestinal crypt, a prototype stem cell compartment.
real world, accidents happen, and the G1/S checkpoint Cell. 2013;154:274-284.
and DNA damage response provide a way to block Dick FA, Rubin SM. Molecular mechanisms underlying RB protein func-
cell-cycle progression even in the presence of growth tion. Nat Rev Mol Cell Biol. 2013;14:297-306.
factors and mitogens. The complex G1 regulatory net- Fuchs E, Tumbar T, Gausch G. Socializing with the neighbors: Stem
cells and their niche. Cell. 2004;116:769-778.
works have a potential impact on all of us. If they are Goldstein M, Kastan MB. The DNA damage response: implications for
disrupted by mutations or damage, the result is cancer. tumor responses to radiation and chemotherapy. Annu Rev Med.
In fact, very few cancers have intact restriction point 2015;66:129-143.
control networks. Jackson SP, Bartek J. The DNA-damage response in human biology and
disease. Nature. 2009;461:1071-1078.
Johnson A, Skotheim JM. Start and the restriction point. Curr Opin Cell
ACKNOWLEDGMENTS Biol. 2013;25:717-723.
Rando TA. Stem cells, ageing and the quest for immortality. Nature.
We thank Jiri Bartek, Ludger Hengst, Keisuke Kaji, and 2006;441:1080-1086.
Marcos Malumbres for their suggestions on revisions to Sage J. The retinoblastoma tumor suppressor and stem cell biology.
this chapter. Genes Dev. 2012;26:1409-1420.
Scadden DT. The stem-cell niche as an entity of action. Nature.
2006;441:1075-1079.
SELECTED READINGS Sherr CJ. The INK4a/ARF network in tumour suppression. Nat Rev Mol
Cell Biol. 2001;2:731-737.
Blanpain C, Fuchs E. Epidermal stem cells of the skin. Annu Rev Cell Shi X, Garry DJ. Muscle stem cells in development, regeneration and
Dev Biol. 2006;22:339-373. disease. Genes Dev. 2006;20:1692-1708.
Bryder D, Rossi DJ, Weissman IL. Hematopoietic stem cells: The para- Silverman JS, Skaar JR, Pagano M. SCF ubiquitin ligases in the mainte-
digmatic tissue-specific stem cell. Am J Pathol. 2006;169:338-346. nance of genome stability. Trends Biochem Sci. 2012;37:66-73.
Cardozo T, Pagano M. The SCF ubiquitin ligase: Insights into a molecu- Sperka T, Wang J, Rudolph KL. DNA damage checkpoints in stem cells,
lar machine. Nat Rev Mol Cell Biol. 2004;5:739-751. ageing and cancer. Nat Rev Mol Cell Biol. 2012;13:579-590.
Chandler H, Peters G. Stressing the cell cycle in senescence and aging. Takahashi K, Yamanaka S. A decade of transcription factor-mediated
Curr Opin Cell Biol. 2013;25:765-771. reprogramming to pluripotency. Nat Rev Mol Cell Biol. 2016;17:
Chen HZ, Tsai SY, Leone G. Emerging roles of E2Fs in cancer: an exit 183-193.
from cell cycle control. Nat Rev Cancer. 2009;9:785-797. Veit B. Stem cell signalling networks in plants. Plant Mol Biol. 2006;
Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. 60:793-810.
Nat Rev Mol Cell Biol. 2013;14:329-340.
CHAPTER 42 

S Phase and DNA Replication

A ccurate replication of DNA, which is crucial for cel- protein complex associated with the fork that is actively
lular propagation and survival, occurs during the S phase replicating the DNA is known as the replisome. Accumu-
(DNA synthesis phase) of the cell cycle. This chapter lating evidence suggests that the replisome is stationary at
begins with a brief primer on the events of replication the fork as it “reels in” replicating DNA rather than moving
and then discusses its regulation. Next, the chapter along the DNA like a train on a track.
covers the proteins at origins of replication that ensure
that each region of DNA is replicated once and only once Replicating DNA
per cell cycle. It closes by discussing how the structure
of the nucleus influences replication.
Mechanism of
chain elongation
DNA Replication: A Primer
O O
One of the most exciting byproducts of the Watson- P
O O–
Crick model for the structure of DNA was a predicted H2C O Base 1
mechanism for DNA replication. Because DNA strand Growing DNA chain
pairing is determined by complementary base pairing, H H H H

it was logical to propose the existence of “DNA poly- H


O O
merases,” enzymes that would move along a single strand P
O O–
of DNA, recognize each base in turn, and insert the H2C O Base 2 Free 3'-hydroxyl end
proper complementary base at the end of the growing
chain. Thus, one might have surmised that only a H H H H

single enzyme was required for DNA synthesis. In fact, H


O O
DNA replication in cells involves a complex macromo- P
O O–
lecular machine. H2C O Base 3
In the chemical reaction of DNA replication, the 3′
hydroxyl at the end of the growing DNA strand makes a H H H H

nucleophilic attack on the α-phosphate of the incoming OH H O O–


P
nucleoside triphosphate to form a phosphodiester bond. Nucleophilic O O O–
This incorporates the nucleotide into the growing chain attack P
O O O–
and releases pyrophosphate (Fig. 42.1). This reaction P
requires the presence of a template strand of DNA that O O–
specifies, via base pairing, which of the four nucleoside H2C O Base 4 Entering dNTP
triphosphates is added to the growing molecule. H H H H
The exact site on the chromosomal DNA where replica- OH H
tion begins is termed the origin of bidirectional repli-
FIGURE 42.1  MECHANISM OF DNA POLYMERIZATION. A 3′
cation. As the term bidirectional implies, two sets of
OH group at the end of a growing DNA chain makes a nucleophilic
DNA replication machinery head off in opposite directions attack on the α-phosphate of a triphosphate precursor in the active
from the origin. At the replication fork, one parental site of polymerase (enzyme not shown here). dNTP, deoxynucleoside
DNA molecule splits into two daughters (Fig. 42.2). The triphosphate.

727
728 SECTION X  n  Cell Cycle

Lagging strand
polymerase complex

Lagging strand Polymerase/


5' primase
3'
RNA RPA CMG
Nascent DNA primer Replication Helicase
(Okazaki fragment) fork 3'
Origin of
5'
bidirectional Replisome
replication
5' Nascent DNA
Leading strand
3' Leading strand polymerase complex

FIGURE 42.2  KEY COMPONENTS AND EVENTS AT THE REPLICATION FORK. All enzymes are closely associated in the replisome
complex, but are shown separate here for clarity.

The bidirectional nature of DNA replication causes a A. E. coli chromosome


fundamental problem, as the chemical reaction of DNA
Origin
synthesis invariably proceeds in a 5′ to 3′ direction. oriC
Replication of the so-called leading strand, in which
DNA polymerase ε moves in a 3′ to 5′ direction along
the template (laying down nascent DNA in a 5′ to 3′
direction) poses no problems; the polymerase simply
chases behind the replication fork (Fig. 42.2). However,
the other template strand faces in the opposite direction, 500,000 bp
apparently requiring a DNA polymerase to synthesize
DNA in the wrong direction as the replication fork B. Portion of eukaryotic chromosome
progresses away from the origin (ie, adding nucleotides Active Active
Dormant Dormant
origin origin
in a 3′ to 5′ direction). No DNA polymerase with this origins origins
polarity has been found. Instead, this lagging strand
replicates in a series of short segments. Every time
the DNA strands have been peeled apart (unwound)
by 200 nucleotides or so, probably corresponding to 130,000 bp
the unwinding of a single nucleosome, a polymerase/ FIGURE 42.3  A, The Escherichia coli chromosome is a simple
primase complex (Figs. 42.2 and 42.12) initiates DNA replicon with a single origin of replication. In cells, this chromosome
synthesis on the lagging strand, and DNA polymerase has a complex, highly supercoiled structure. B, Eukaryotic chromo-
δ runs away from the fork, back toward the replication somes have multiple origins of replication, most of which remain
dormant unless needed.
origin, again synthesizing nascent DNA in a 5′ to 3′
direction. Thus, lagging strand synthesis proceeds in
bursts in a direction opposite to the overall direction their large genomes during the S phase, which can be as
of fork movement. Synthesis of each lagging strand short as a few minutes in some early embryos. These
fragment stops when DNA polymerase runs into the 5′ numerous origins are distributed along the chromosome:
end of the previous fragment, displacing the RNA primer up to 600 to 700 in budding yeast and more than 100,000
with which it was initiated (see later). Thus, the lagging in human cells. The origins are distributed so that all the
strand is copied in a highly discontinuous fashion into DNA is replicated in the available time, and many more
short fragments known as Okazaki fragments (named origins are prepared than are actually needed.
after their discoverer [Fig. 42.2]). Fig. 42.12 describes How is the “firing” of all these origins orchestrated so
the enzymes and events at the replication fork in that each is used no more than once per S phase? Cells
greater detail. manage this problem by a mechanism termed licensing,
which ensures that each segment of DNA is replicated
just once per cell cycle. Replication of the origin removes
Origins of Replication the license, which cannot normally be renewed until the
Bacteria such as Escherichia coli replicate their circular cell has completely traversed the cycle and has passed
chromosomes using two replication forks starting from through mitosis.
a single origin of replication (Fig. 42.3A), but eukary- The portion of chromosomal DNA replicated by the
otes must use multiple origins of replication to duplicate two bidirectional forks initiated at a single origin is
CHAPTER 42  n  S Phase and DNA Replication 729

termed a replicon. The classic replicon is the E. coli A


chromosome (which is 4 × 106 base pairs [bp] in size)
with a single genetically defined replicator site called
oriC (Fig. 42.3). An initiator protein (product of the E.
Plasmid
coli DnaA gene [Fig. 42.13]) binds to this origin and
either directly or indirectly promotes melting of the DNA
duplex, giving the replication machinery access to two Grow cells
Selectable
single strands of DNA. Other factors unwind the DNA, marker gene Introduce into under selective
leading to the full assembly of the replisome, which yeast cells conditions
powers a wave of DNA replication proceeding outward
in both directions along the DNA (a replication “bubble”) B
at approximately 750 to 1250 bases per second. ARS
An average human chromosome contains approxi-
mately 150 × 106 bp of DNA. Because the replication
Plasmid
machinery in mammals moves only approximately 20 to
40 bases per second (partly reflecting the fact that the
DNA is packaged into chromatin and partly reflecting the Grow cells
Selectable
slower speed of the eukaryotic replisome), it would take marker gene Introduce into under selective
up to 2000 hours to replicate this length of DNA from a yeast cells conditions
single origin. In most human cells the S phase takes FIGURE 42.4  PLASMID ASSAY FOR IDENTIFICATION OF AN
approximately 10 hours. This means that at least 25 AUTONOMOUSLY REPLICATING SEQUENCE (ARS) ELEMENT
(ORIGIN OF DNA REPLICATION) IN BUDDING YEAST. The
to 125 origins of replication are required to replicate
plasmid has a selectable marker gene (eg, a gene required for the
an average chromosome in the allotted time. In fact, synthesis of an essential amino acid) plus (in panel B) an ARS element.
origins of replication are much more closely spaced This plasmid is transferred into growing yeast cells that are defective
than this. It is estimated that 30,000 to 50,000 origins of in the marker gene carried by the plasmid; these cells are then plated
replication “fire” during each cell cycle to replicate out on agar medium that lacks the essential amino acid. A, A plasmid
lacking an ARS fails to replicate and is lost from the cells. These cells
the entire human genome, but many more dormant
cannot grow into colonies on plates that lack the essential amino acid.
origins are licensed for use if necessary. These dormant B, If the plasmid contains an ARS element, it replicates along with the
origins are essential for resolving replication stress chromosomal DNA and is maintained in the population. These cells
(see later). grow into colonies in the absence of the essential amino acid.
To explain the events at origins of replication, the
budding yeast Saccharomyces cerevisiae serves as a
good example. Its DNA replication is better understood the S phase. Because the number of origins exceeds the
than that of any other eukaryote and although its origins number required to replicate the genome within the
of replication are specialized, the proteins that act on allotted time, some origins need not “fire” every cell
them are conserved across metazoa. cycle. The probability that any given origin will be used
in a given cell cycle ranges from less than 0.2 to greater
Replication Origins in Saccharomyces cerevisiae than 0.9. It is important to note that replication by a
Approximately 400 origins of replication participate in passing fork coming from an adjacent origin inactivates
replicating the budding yeast genome. A major break- dormant origins. This prevents re-replication of genomic
through in understanding DNA replication in S. cerevi- regions during the cell cycle.
siae was the identification of short (100 to 150 bp) The ARS element does two things to establish an
segments of DNA that act as replication origins in vivo origin of replication. First, its conserved sequences act
when cloned into a yeast plasmid (circular DNA mole- as binding sites for a protein complex that marks it as
cule). These autonomously replicating sequences a potential origin. Second, it has nearby sequences that
(or ARS elements) allow yeast plasmids to replicate in are readily induced to unwind by separating the base-
parallel with the cellular chromosomes (Fig. 42.4). ARS paired strands.
elements are often, although not always, bona fide rep- Budding yeast ARS elements share a common DNA
lication origins in their native chromosomal context. sequence motif called the ARS core consensus
Replication always initiates within ARS elements, but not sequence: 5′-(A/T)TTTAT(A/G)TTT(A/T)-3′ (Fig. 42.5).
all ARS elements act as origins of DNA replication in Single base mutations at several locations within this
every cell cycle. sequence completely inactivate ARS activity. Other, less
Yeast replication origins are spaced approximately well-conserved DNA sequences also contribute to the
every 30,000 bp, with a maximum separation of approxi- activity of the ARS as a replication origin. One of these,
mately 130,000 bp. Even this longest interval should termed B1, together with the ARS core, forms the bind­
replicate easily within the 30 minutes available during ing site for a complex of six proteins termed the origin
730 SECTION X  n  Cell Cycle

ORC EdU PCNA (t + 30 min) Merge


complex
ABF-1 OBR
(origin of
bidirectional 90°
replication) ATP ATP

B3 B2 DNA B1 ARS
unwinding core
A A A
element T T T T A TG T T TT 5 µm

FIGURE 42.5  ORGANIZATION OF THE AUTONOMOUSLY FIGURE 42.6  SUPERRESOLUTION VIEW OF ACTIVE REPLI-
REPLICATING SEQUENCE (ARS)-1 ELEMENT. The ORC (origin CONS IN A HELA (HENRIETTA LACKS) CELL. EdU (red) was used
recognition complex) binds to the ARS core sequence plus element to label sites of active replication for 15 minutes followed by washing
B1. B2 is a sequence that can readily be induced to unwind. The OBR out for 15 minutes. Then antibody recognizing proliferating cell nuclear
(origin of bidirectional replication) is the site where DNA synthesis antigen (PCNA) was used to stain all active replicons in the cell.
actually begins. B3 is a binding site for an auxiliary factor called ABF-1 Thousands of replicons can be seen, and it is clear that in the 30
that is both a transcriptional activator and an activator of the ARS minutes between the labelling of the red and green channels, many
element. ATP, adenosine triphosphate. (For reference, see Protein Data new origins of replication have been activated. Scale bar = 5 µm. OMX
Bank [PDB; www.rcsb.org] file 4X6C.) superresolution. (Microscopy by Vadim Chagin and Cristina Cardoso,
Technical University of Darmstadt, Germany.)

recognition complex (ORC [see later section]). The


DNA unwinding element is another short sequence (B2) Replication Origins in Mammalian Cells
located a bit further along the DNA. DNA synthesis Less is known about the structure and function of mam-
begins at an origin of bidirectional replication midway malian origins of DNA replication than about ARS ele-
between the ORC binding site and the DNA unwinding ments in budding yeast. Attempts to develop a mammalian
element. equivalent to the yeast ARS assay had few successes.
ORC was identified by its ability to bind the 11-bp ARS Over the years approximately 30 metazoan origins of
core sequence (Fig. 42.5). This binding has two notewor- replication were identified by painstaking methods, but
thy features. First, the subunits of the ORC complex are more recently, high throughput methods, including the
AAA adenosine triphosphatases (ATPases; see Box 36.1) sequencing of short nascent strands of replicating DNA
and adenosine triphosphate (ATP) hydrolysis is required have identified thousands of mammalian replication
for ORC to bind ARS DNA. Second, in yeast, the ORC origins. Superresolution microscopy can now resolve the
complex remains bound to the origins of replication thousands of replicons active at any one time in a human
across the entire cell cycle. Thus, something other than cell (Fig. 42.6).
the presence of ORC must regulate the periodic activa- Overall, the chromatin landscape influences mam-
tion of origins in the S phase (Fig. 42.14). In some malian origins. Important factors include DNA sequence,
metazoan cells, ORC behavior is more complex—for DNA modifications, chromatin structure, and nuclear
example, the largest subunit, Orc1, is degraded during organization. Although high throughput sequencing
part of the cell cycle. revealed no sequences as specific as the yeast ARS ele-
ARS elements often contain binding sites for other ments, many origins are associated with short regions of
sequence-specific DNA binding proteins, including tran- G-rich DNA. These G-rich regions can form specialized
scription factors. For example, a transcription factor structures that have fewer nucleosomes and are there-
called ARS-binding factor 1 (ABF-1) binds to the B3 fore more accessible to the DNA replication machinery.
sequence within the ARS1 element (Fig. 42.5). Deletion Indeed, replication origins tend to be located near gene
of the ABF-1 binding site only slightly reduces the ability promoters, where the density of nucleosomes is low.
of ARS1 to act as a replication origin in vivo and other Such actively transcribed regions of the genome are
transcription factors can substitute for ABF-1. packaged into euchromatin with modified histones and
In addition to their role in DNA replication, several tend to replicate earlier during S phase than regions of
ORC subunits also regulate heterochromatin formation the genome that are not transcribed.
and transcription (see Chapters 8 and 10). This crosstalk A typical mammalian replicon encompasses about
between the machinery used for transcription and DNA 130 kb and contains approximately four or five licensed
replication may explain why regions of chromosomes origins-only, one of which is typically used. The first
with actively transcribed genes typically replicate early replicon to be mapped lies just downstream of the
in the S phase (see the next section below). In some hamster gene for dihydrofolate reductase, an enzyme
metazoan cells, the Orc6 subunit also functions during that is essential for biosynthesis of thymidine. Investiga-
cytokinesis, apparently via interactions with cytoskel- tors selected cells with this chromosomal region ampli-
etal filament proteins independent of its role at replica- fied as hundreds or even thousands of copies (Fig. 42.7)
tion origins. and looked for the first regions of the amplified DNA to
CHAPTER 42  n  S Phase and DNA Replication 731

Mapping a cellular replication origin


Add high CYTOPLASM
concentration Cdt1 Geminin
of methotrexate Cells die
Geminin degraded Cdk phosphorylation
Cdks inactive triggers other CMG
Cultured cells components to assemble
Add gradually Prereplication
Mcm2-7
increasing concentration Cells live in high complex
double
of methotrexate over concentration of hexamer
ORC Cdt1
many generations methotrexate 3’
Cdc6 5’

Cdc45 3’ 5’

GINS
Loop domain containing CMG helicases
Restriction
dihydrofolate reductase gene NUCLEUS point
unwind DNA
Normal Chromosome with G1 S
chromosome amplified domain

Base of FIGURE 42.8  COMPONENTS OF THE PREREPLICATION


Potential OBRs
domain (origins of bidirectional COMPLEX BEFORE AND AFTER THE INITIATION OF DNA
replication) REPLICATION. CMG, Cdc45, Mcm2–7, and GINS (Go-Ichi-Ni-San).
DHFR gene
(For reference, see PDB files 3JA8 [for CMG helicase with Cdk45
bound], 3JC5 [for Mcm2–7], and 5F9R [for ORC].)

Dormant Dormant
origins Active origins
Dormant solutions to this problem that have been reached by
Active origins
origin origin yeasts and vertebrates.
Recall that yeast ORC is stably bound to replication
DHFR gene β γ Next gene origins throughout the cell cycle. However, ORC is not
–30 –20 –10 0 10 20 30 40 50 60 70 the trigger for DNA replication. Rather, it acts as a
Number of bases (kb)
“landing pad” for assembly of a prereplication complex
Initiation zone of other proteins that initiates DNA replication.
Formation of the prereplication complex “licenses”
FIGURE 42.7  MAP OF DNA REPLICATION LANDSCAPE NEAR
THE DIHYDROFOLATE REDUCTASE (DHFR) GENE. Normal cells each origin for a single initiation event as follows. During
are killed by exposure to methotrexate, but it is possible to select late anaphase or very early G1 phase, several proteins,
resistant cell lines by growing them in progressively increasing concen- including Cdc6 and Cdt1, bind to the ORC complex at
trations of the drug, selecting at each stage for cells that survive. Use origins of replication (Table 42.1). Before the onset of
of this procedure on hamster cells resulted in a cell line that contains
the S phase ORC-Cdc6-Cdt1 loads a double hexamer of
approximately 1000 copies of a 230,000-bp chromosomal domain
containing the DHFR gene. This region of DNA is replicated using Mcm proteins (minichromosome maintenance) on the
origins found within a 55,000-bp region adjacent to the DHFR gene. origin DNA (Fig. 42.8). Mcm proteins were identified in
Most initiation occurs at two specific origins, called β and γ, but other a screen for genes of budding yeast that are required for
dormant origins are scattered throughout the entire 55,000-bp region. the stability of small artificial chromosomes. Six of these
Mcm genes encode a structurally related group of pro-
replicate. They found that DNA replication can initiate teins, termed Mcm2–7, that is required for DNA replica-
with low efficiency at a number of sites distributed tion. ORC-Cdc6–Cdt1 uses ATP hydrolysis to crack open
across a broad region of approximately 55,000 bp. Two two hexameric Mcm2-7 rings so they can wrap around
of these sites, termed Ori-β and Ori-γ (Fig. 42.7) account DNA in a head-to-head orientation.
for approximately 20% of all initiation in the region. Replication starts when each Mcm2–7 hexamer asso-
The emerging view is that the replication machinery ciates with the GINS (Go-Ichi-Ni-San; 5-1-2-3 in Japanese)
is highly conserved between budding yeasts and verte- complex and the Cdc45 protein to form the replicative
brates but that the location of replication origins is more CMG helicase (Cdc45-Mcm-GINS). This helicase forms
flexible in vertebrates. This might reflect both the differ- the core of the replisome and uses ATP hydrolysis to
ing chromatin landscapes across metazoan genomes (see separate DNA strands (Fig. 42.8). As the two CMG heli-
Chapter 8) and the diverse range of cell cycles required cases move away from each other, the origin is converted
to make a complex metazoan. from a licensed to an unlicensed state. Because origin
licensing by Mcm2–7 loading can only occur prior to
Origin Licensing and Assembly of entry into S phase, origins only fire once per cell cycle.
In mammals, licensing occurs in the early G1 phase
the Prereplication Complex
before passage of the restriction point (see Chapter 41).
To preserve the integrity of the genome, each origin of At the exit from mitosis, destruction of cyclins and syn-
replication must “fire” no more than once per cell cycle. thesis of inhibitory proteins inactivates Cdks. This creates
We now have a reasonable understanding of the various a window of time between anaphase and the restriction
732 SECTION X  n  Cell Cycle

TABLE 42.1  Biochemical Activities Required for Replication of DNA in Eukaryotes


Activity Name of Protein
Origin recognition ORC (origin recognition complex; five of six subunits are AAA ATPases)
Prereplication complex Cdc6 (recruits Mcm2–7)
Cdt1 (recruits Mcm2–7)
Origin activation Cdc7-Dbf4
Cdk (in human, Cdk2-cyclin A) phosphorylation of Sld2/RecQ4 (yeast/human names)
and Sld3/treslin recruits Dbp11/TopBP1 and initiates CMG assembly to trigger the
actual start of replication
DNA unwinding (helicase) CMG helicase is made up of:
• Mcm2–7 (assemble double hexamer before other CMG components)
• Cdc45 and the GINS complex (4 proteins)
Stabilization of single-stranded DNA RPA (binds single-stranded DNA)
Replicative polymerases DNA polymerase α/primase (no editing function) starts synthesis of the leading
strand and each Okazaki fragment
DNA polymerase δ replicates the lagging strand
DNA polymerase ε replicates the leading strand (both δ and ε have 3′–5′ exonuclease
editing capability)
Processivity factor PCNA (ring-shaped clamp that slides along the DNA. Keeps polymerases δ and ε
attached to the template strand so that they make longer chains; coordination of
cell-cycle control and replication; role in repair)
PCNA loader RF-C (Binds primer: template junction. AAA ATPase. Loading factor for PCNA,
important for polymerase switch)
Closing Factors
Removal of RNA primer Fen1 5′–3′ exonuclease
Dna2 helicase and endonuclease
RNase H
Ligation of discontinuous DNA fragments DNA ligase I
Releasing superhelical tension DNA topoisomerase I
Disentangling daughter strands DNA topoisomerase II

ATPase, adenosine triphosphatase; CMG, Cdc45, Mcm2–7, and GINS; GINS, Go-Ichi-Ni-San; PCNA, proliferating cell nuclear antigen; RPA, replication
protein A.

point for licensing replication origins (Fig. 42.9). Mam- Different cell types use various combinations of several
malian cells pass the restriction point when the levels of different pathways to downregulate the activity of the
Cdk2–cyclin E, and subsequently, Cdk2–cyclin A rise licensing system once cells enter S phase. In metazoans,
(see Fig. 40.17). This prevents the relicensing of replica- the most important pathways reduce Cdt1 activity by
tion origins until after the next mitosis. In yeasts, the degradation in S and G2 phases. After the cell passes
single Cdk that is complexed with B-type cyclins inhibits the restriction point, Cdks phosphorylate Cdt1. This
origin licensing. Experimental inactivation of CdkCdc2 promotes its ubiquitylation by the SCFSkp2 and degrada-
during the G2 phase in the fission yeast Schizosaccharo- tion. In addition, the key replisome component prolif-
myces pombe demonstrated the importance of Cdk erating cell nuclear antigen (PCNA) recruits another
activation: Cells lacking CdkCdc2 activity loaded Mcm2–7 ubiquitin ligase that causes Cdt1 degradation during S
onto already replicated DNA and then carried out further phase. In some cell types, Cdks phosphorylate Cdc6 and
rounds of “illegal” DNA replication without division. Cdk Orc1, leading to their ubiquitylation and degradation.
regulation of origin licensing during G1 phase actually Vertebrates use a protein called geminin as an alter-
provides one explanation for oncogenic stress (see native regulator of origin licensing by Cdt1 (Fig. 42.8).
Chapter 41), in which inappropriate activation of onco- Geminin binds to Cdt1 and prevents it from loading Mcm
genes leads to cell-cycle arrest followed by death or proteins onto DNA. The anaphase-promoting complex/
senescence. Inappropriate oncogene activation can lead cyclosome (APC/C; see Fig. 40.15) triggers degradation
to premature stabilization of cyclins D and cyclin E, of geminin, keeping its concentration very low from
resulting in premature activation of Cdks. This, in turn, anaphase through the late G1 phase, allowing prereplica-
can lead to an insufficient number of replication origins tion complexes to assemble. Accumulation of geminin
being licensed, thereby inhibiting the cell’s ability to starting in the S phase inhibits the assembly of new
deal with replication stress (see later) and its associated prereplication complexes until after the next mitosis.
DNA damage. Yeasts control origin licensing without geminin.
CHAPTER 42  n  S Phase and DNA Replication 733

HUMAN CERVICAL EPITHELIUM STAINED WITH ANTI-MCM5


Cdk2-cyclin E
A. Normal B. Low-grade C. High-grade
Origin licensing Cdk2-cyclin A lesion lesion
Cdk activity
Increased

Cdk4/6-cyclin D

G1 S
Restriction point
APC activity
Increased

Geminin
APC/CCdh1 Emi1
SCFSkp2
SCFβ-TrCP

APC/CCdh1 SCFβ-TrCP SCFSkp2 Other SCF


Geminin Emi1 p27Kip1 Cyclin D
Cdc6 Cdc25A Orc1 DP1
Cyclins Cdt1

FIGURE 42.9  PROTEIN DEGRADATION REGULATES DNA


REPLICATION. Degradation of geminin, Cdc6, Cdc25A, and cyclins
during the G1 phase keeps Cdk activity low and allows prereplication
complex formation. Degradation of p27Kip1 and inactivation of the
APC/CCdh1 by Emi1 allows the activation of Cdks to levels sufficient for
the initiation of the S phase. Once cells enter the S phase, the G1/S FIGURE 42.10  SECTIONS OF HUMAN CERVIX STAINED
regulatory machinery (cyclins D and E and the E2F cofactor DP1) is WITH ANTIBODIES TO MCM5. A, Normal G0 cells in this stratified
degraded. Degradation of Cdt1 and accumulation of geminin block epithelium lack Mcm5 and other replication proteins. B–C, Cancer
reassembly of prereplication complexes. APC, anaphase-promoting cells express Mcm5 at higher levels as they become more malignant.
complex; SCF, Skp2 (S-phase kinase-associated protein), cullin, and Bound antibodies were detected with peroxidase coupled to a sec-
F-box proteins. ondary antibody (brown reaction product) and lightly counterstained
with hematoxylin (blue). (Modified from Williams GH, Romanowski P,
Morris L, et al. Improved cervical smear assessment using antibodies
A third way to regulate origin licensing is to sequester against proteins that regulate DNA replication. Proc Natl Acad Sci
molecules required to assemble the prereplication U S A. 1998;95:14932–14937.)
complex in the cytoplasm following the onset of S phase.
This was first suggested by studies of DNA replication the transcription of genes involved in DNA replication
in Xenopus egg extracts (see Fig. 40.8) in which intact (see Fig. 41.9). In addition to cyclin E itself, E2F drives
nuclei replicate their DNA only once but re-replicate if expression of cyclin A, Cdc25A, enzymes required for
the nuclear membrane is disrupted. In different cell synthesis of DNA precursors (dihydrofolate reductase,
types, nuclear exclusion of ORC, Cdc6, Cdt1, or Mcm2–7 thymidine kinase, and thymidylate synthase), origin-
all contribute to limiting licensing during S and G2 phases. binding proteins (Cdc6, Orc1, Cdt1 and its inhibitor
Components of the prereplication complex are absent geminin), and two components of the replication
from nondividing differentiated cells. In fact, detection machinery (DNA polymerase α and PCNA; see Fig. 42.9).
of these proteins with antibodies in cells from cervical In the S phase, the SCFSkp2 ubiquitin ligase complex
smears can be used as a sensitive method for the early targets the Cdk inhibitor p27Kip1 for destruction by pro-
detection of cancer cells (Fig. 42.10). teasomes (see Fig. 40.16). SCF gets its name from three
of its components: Skp2, cullin, and F-box proteins (see
Fig. 40.16). Skp2, which is short for “S-phase kinase-
Signals That Start Replication associated protein,” was identified in a complex with
A classic experiment (Fig. 42.11) demonstrated that (a) Cdk2–cyclin A. SCF recognizes and ubiquitylates many of
a cytoplasmic inducer triggers the transition into the S its substrates only after they are phosphorylated by Cdk2-
phase and (b) this inducer triggers DNA replication in a cyclin A. Cdk2–cyclin A also targets E2F/DP and cyclin E
G1 nucleus but not in a G2 nucleus. The inducer is a for degradation when cells enter the S phase (Fig. 42.9).
combination of protein kinases, including Cdk–cyclin For DNA replication to start, the paired strands of the
pairs, as well as a specialized kinase, Cdc7-Dbf4. In double helix must be separated so DNA polymerase can
mammals, Cdk2–cyclin E activity peaks at the G1/S transi- recognize the bases and begin synthesizing the daughter
tion (Fig. 42.9) and phosphorylates pRb, thereby opening strand. The multi-subunit CMG DNA helicase uses ATP
the restriction point “gate.” This allows the E2F/DP hydrolysis to peel apart the paired strands of the DNA
dimer to function as a transcription factor and stimulate double helix. Prior to S phase each origin of replication
734 SECTION X  n  Cell Cycle

called a primase, which, like other RNA polymerases,


A can initiate synthesis de novo without the need for a
+
3′OH group. In eukaryotes, all DNA chains are started by
Cell in S phase Cell in G1 phase Heterokaryon a complex of DNA polymerase α and a primase subunit,
(cell with two nuclei) collectively known as Pol α/Primase. Primase synthe-
sizes an RNA chain of approximately seven nucleotides
B to which DNA polymerase α adds another 20 to 25
100
Fusion of nucleotides of so-called initiator DNA (iDNA) (Fig.
% G1 cells that enter S phase

S and G1 cells
42.12D–E). These initiating reactions are potentially
G1 cell alone risky, because DNA polymerase α lacks proofreading
ability. To avoid errors created by mismatching of an
50 Advancement incoming base, the RNA primer and most or all the iDNA
due to inducer laid down by Pol α/Primase are subsequently replaced.
Once Pol α/Primase has done its job, two further
essential factors act to complete replisome assembly. A
pentameric protein complex called replication factor
C (RFC) binds the 3′ end of the iDNA. RFC uses energy
0
0 4 8 12 16 from ATP hydrolysis to load the trimeric protein PCNA
Hours after fusion onto the DNA (Fig. 42.12E–G). The PCNA trimer is
FIGURE 42.11  CELL FUSION EXPERIMENT REVEALS THE
doughnut-shaped, and when the DNA is inserted into its
EXISTENCE OF A POSITIVE INDUCER OF THE S PHASE. central hole, it is topologically locked onto the DNA. RFC
A, Synchronized cells in different stages of the cycle were fused to binding and PCNA loading displace Pol α/Primase from
yield two nuclei in a single cytoplasm. B, If the fusion involved G1 and the DNA, and PCNA then recruits DNA polymerases δ
S cells, the G1 nucleus was induced to enter the S phase sooner than and ε to the DNA. With PCNA acting as a sliding platform,
expected. If the fusion involved S and G2 cells, the G2 nucleus failed
to rereplicate its DNA (not shown). (Modified from Rao PN, Johnson
DNA is reeled through the replisome and polymerase
RT: Mammalian cell fusion: studies on the regulation of DNA synthesis ε synthesizes DNA continuously on the leading strand
and mitosis. Nature. 1970;225:159–164.) (Fig. 42.12G).
On the lagging strand, polymerase δ synthesizes DNA
is licensed by assembly of a double hexamer of Mcm2–7 in bursts of approximately 250 bp, each initiated by Pol
complexes. Cdc7 kinase with its associated regulatory α/Primase and roughly correlating with the passage of
subunit Dbf4 phosphorylates the Mcm complex. Next, one nucleosome by the replication fork. The replisome
Cdks phosphorylate other proteins that recruit GINS contains a chromatin remodeling activity that removes
and Cdc45 to the phosphorylated Mcm2–7 hexamer, nucleosomes from the DNA ahead of the fork and
thereby producing the CMG helicase (Table 41.1 and replaces them after the fork passes.
Fig. 42.12A). Subsequent activation of the helicase initi- Both polymerases δ and ε have associated exonuclease
ates replication. activities. This enables them to proofread the newly
synthesized DNA and correct most mistakes that they
have made. The combination of selecting the correct
Mechanism of DNA Synthesis nucleotides and efficient correction of mistakes may
Replication initiates when the activated CMG helicase explain the amazing fidelity of DNA replication, with
starts to separate the DNA strands, which move outward typically only one error per 109 bp polymerized.
in both directions as the replisome assembles at the Locally, the final steps of DNA replication are removal
origin of bidirectional replication. The newly unwound of the RNA primer (and probably iDNA) and ligation of
DNA binds the single-strand DNA-binding protein, RPA adjacent stretches of newly synthesized DNA. Removal
(replication protein A), ensuring that the separated of the primer can be accomplished in two ways (Fig.
strands do not base-pair with one another again (Fig. 42.12H). On one hand, an RNA exonuclease called
42.12B). Table 42.1 describes several other proteins that RNase H can chew in from the 5′ end of the primer.
also bind at this time. Box 42.1 provides an introduction However, this enzyme cannot remove the last ribonucle-
to DNA replication in E. coli. otide that is joined to iDNA. That requires the nucleases
The separated DNA strands are ready for replication, Fen1 (Flap endonuclease) or Dna2. Fen1 requires a
but DNA synthesis always involves addition of an incom- helicase to peel the RNA (and possibly the iDNA) away
ing nucleoside triphosphate to a free 3′ OH group at the from the template, creating a sort of flap. Fen1 then
terminus of a preexisting nascent polynucleotide (Fig. cleaves at the junction where the flap is anchored to the
42.1). In the absence of a nascent DNA chain, how does DNA template, removing the oligomer of unwanted
DNA polymerase get started? This problem is solved on nucleotides in one step. Alternatively, the Dna2 nuclease
the lagging strand by a DNA-dependent RNA polymerase can do the whole job itself because it also has helicase
CHAPTER 42  n  S Phase and DNA Replication 735

A. Activation of origin F. RFC binds iDNA, evicting polymerase α / primase,


loading PCNA
Cdc7
Dbf4
Cyclin A
Cdk2
RFC PCNA
ORC ATP

Mcm2-7
double hexamer
Cdc6 and Cdt1 leave Polymerase α and primase leave

G. PCNA recruits polymerase ε to leading strand


B. Binding of Cdc45, GINS and RPA
Polymerase ε
RPA Cdc45
GINS
5’

3’ Active
CMG helicase

C. Cdc45 recruits polymerase α / primase H. Processive DNA


synthesis starts

RFC PCNA
Nascent
DNA

Primase
Polymerase α

I. Primer and
D. Synthesis of RNA primer i-DNA removal

RNA RNase H
primer

Fen1

J. DNA synthesis completed by polymerase ε


E. Synthesis of i-DNA followed by ligation of strands;
p97/Cdc48 removes
Mcm2-7 and replisome
RNA
primer disassembles
5’ i-DNA p97/Cdc48

3’

FIGURE 42.12  MAIN EVENTS OF DNA REPLICATION ON THE LEADING STRAND. For a more detailed description, including events on
the lagging strand, see the text. (For reference, see PDB files 1A76 [for Fen1], 1SXJ [for RFC/PCNA], 2ZXX [for Cdt1], and 3CF2 [for p97/Cdc48].)

activity. Most of these maturation/processing enzymes reticulum-associated degradation [ERAD] pathway; see
are recruited by interacting with the PCNA trimer. The Chapter 23) recognizes this ubiquitin and then actively
individual interactions are transient while PCNA remains separates the Mcm2–7 hexamer from Cdc45 and GINS,
as a stable loading platform. causing the replisome to fall apart.
Following removal of initiator RNA, the Pol δ/PCNA
complex extends the upstream nascent chain until it Higher-Order Organization of DNA
runs into the downstream 5′ end created by Fen1. DNA
Replication in the Nucleus
ligase I then joins the two stretches of DNA together
(Fig. 42.12I). The term S phase gives the impression that all DNA
When the DNA is ligated, the replisome must be disas- replicates more or less synchronously, but this is far from
sembled. This is an active process triggered by ubiquity- true. At any given time during the S phase, only 10% to
lation of the Mcm7 subunit of the CMG helicase. The 15% of the replicons actively synthesize DNA. Some
AAA-ATPase p97/Cdc48 (which also extracts proteins replicate early, others late. This pattern of replication
from the endoplasmic reticulum in the endoplasmic is not random; some segments of DNA consistently
736 SECTION X  n  Cell Cycle

BOX 42.1  DNA Replication in Escherichia coli

The DNA replication system of Escherichia coli has been that requires the histone-like proteins. Next, DnaC binds to
reconstituted entirely from purified components. Analysis of DnaB and escorts it to the unwound DNA. DnaB is the key
this system reveals many similarities with eukaryotic replica- helicase that will drive DNA replication by unwinding the
tion, indicating that this process is highly conserved. E. coli double helix, but it binds DNA poorly on its own in the
DNA replication can be subdivided into three phases: initia- absence of its DnaC escort. Once DnaB has docked onto
tion, elongation, and termination. Thus far, at least 28 the DNA, DnaC is released, and the helicase can then start
polypeptides are known to be involved. to unwind the DNA, provided that ATP, SSB, and DNA gyrase
are present. SSB is a single-stranded DNA binding protein
Initiation that stabilizes the unwound DNA, and DNA gyrase is a
E. coli chromosomal DNA replication initiates within a topoisomerase (see Chapter 8) that removes the twist that
245-bp region, termed oriC. This region contains four 9-bp is generated when the two strands of the double helix are
binding sites for the E. coli initiator protein, DnaA. Nearby separated.
are three repeats of a 13-bp A/T-rich sequence. oriC also
contains specific binding sites for two small histone-like Elongation
proteins called HU and IHF. Replication is initiated with the As in eukaryotes, E. coli DNA replication involves a leading
cooperative binding of 10 to 20 DnaA monomers to their strand, with the daughter DNA synthesized as a single con-
specific binding sites (Fig. 42.13). To be active, these mono- tinuous molecule, as well as a lagging strand, with the DNA
mers must each have bound ATP. Binding of DnaA permits synthesized as discontinuous Okazaki fragments. All daugh-
unwinding of the DNA at the 13-bp repeats, in a reaction ter strands are started by an RNA primase that deposits
primers of 11 ± 1 nucleotides. The enzyme that actually
A R1 R2 R3 R4 oriC DNA synthesizes the DNA is the polymerase III holoenzyme,
which has at least 10 subunits. This contains polymerase and
13-mers DnaA sites
proofreading subunits and is held to the DNA by a doughnut-
DnaA + ATP like “sliding clamp” (β). The β is loaded onto the DNA by a
HU or IHF pentameric complex in a process that requires ATP. The
B parallel with PCNA and RFC in eukaryotes is striking. Activi-
ties specific for the lagging strand include RNase H, which
DnaC
removes the RNA primers; DNA polymerase I, which fills in
+ ATP DnaB•DnaC the gaps left behind by primer removal; and DNA ligase,
DnaB complex which links the Okazaki fragments together. DNA replication
DnaC in E. coli is significantly faster than it is in eukaryotes, with
C the fork moving at a rate of approximately 1000 bp per
second. This higher speed is presumed to be at least partially
SSB attributable to the absence of nucleosomes on the bacterial
ATP chromosome, but the eukaryotic enzymes may also be
ADP
DnaA
intrinsically slower.

D Termination
A specialized termination zone is found on the circular E.
coli chromosome opposite oriC. This zone contains binding
FIGURE 42.13  FACTORS INVOLVED IN THE INITIATION OF sites called ter sites, to which the ter binding protein binds.
DNA REPLICATION IN ESCHERICHIA COLI. A, DNA sequences
This protein appears to block the movement of DNA heli-
at OriC. B, Unwinding of the origin. C, Binding of helicase. D, The
template, now ready for binding of DNA polymerase. ADP, adenos-
cases, such as DnaB, thereby stalling the DNA replication
ine diphosphate; ATP, adenosine triphosphate; SSB, single-stranded fork. Following termination of replication, a specialized
DNA binding protein. (Modified from Baker TA, Wickner SH. Genet- topoisomerase, the product of the parC and parE genes,
ics and enzymology of DNA replication in Escherichia coli. Annu Rev is required to separate the daughter chromosomes from
Genet. 1992;26:447–477.) one another.

replicate early in the S phase, whereas others consis- of these foci. There are roughly 60,000 origins in a mam-
tently replicate late in the S phase. malian cell, so each replication focus represents five or
Up to 1000 sites of replication, called replication six replication origins that are activated coordinately.
foci are active at any one time during the S phase in a The replication foci correspond to structural domains of
mammalian cell nucleus (Figs. 42.6 and 42.14B–C and chromosomes that are now called TADs (topologically
E–F). Given that each of these replication foci is active associating domains) (see Fig. 8.13). The function of
for only about 30 minutes out of the 8- to 10-hour S each TAD, measured by replication timing, chromatin
phase, a cell will replicate DNA at approximately 10,000 composition and transcriptional output, can change
CHAPTER 42  n  S Phase and DNA Replication 737

FIGURE 42.14  VISUALIZATION OF DNA REPLICATION


A. BrdUTP
WITHIN THE NUCLEUS. A, BrdUTP is introduced into DNA in place
O– O– of dTTP to label newly replicated DNA. The incorporated BrdU (bro-
P
O O modeoxyuridine) molecules are detected with fluorescent antibodies.
O– Incorporated B, In a related approach, green-dUTP (deoxyuridine triphosphate) and
P O into cellular DNA
O O Br red-dUTP, when added together, show the many sites of DNA replica-
O– NH by replication
P tion in a cell nucleus. Because both UTP (uridine triphosphate) analogs
O O CH are incorporated simultaneously into the DNA, the sites of replication
2 O N O
appear yellow. C, Green-dUTP is followed by red-dUTP added 3 hours
H H
H H later. The later sites of DNA replication show very little overlap with the
Fed to cells
OH H earlier sites. D, Mitotic chromosome from a cell that was labeled early
in the S phase with IdU (green), and then 4 hours later with CldU (red).
B C D The late-replicating and early replicating regions of the chromosome
are segregated into discrete bands. E, CldU (green) added early in the
S phase and IdU (red) added 4 hours later show relatively little overlap.
F, CldU (green) added early in the S phase and IdU (red) added 6
hours later show no overlap. The large red blocks of labeling seen with
the IdU are characteristic of the pattern of replicating heterochromatin
seen late in the S phase. Bodipy-TR-dUTP, a red fluorescent form of
5 µm 5 µm dUTP; BrdUTP, bromodeoxyuridine triphosphate; CldU, chlorine-
Fluorescein-dUTP Fluorescein-dUTP IdU dUTP; Fluorescein-dUTP, a green fluorescent form of dUTP; IdU,
Bodipy-TR-dUTP Bodipy-TR-dUTP CIdU iodine-dUTP. All are used in place of dTTP (deoxythymidine triphos-
at same time 3 hrs later 4 hrs later phate) in DNA synthesis. (B–C, Courtesy P.R. Cook, University of
Oxford, United Kingdom. From Manders EMM, Kimura H, Cook PR.
Direct imaging of DNA in living cells reveals the dynamics of chromo-
E F
some formation. J Cell Biol. 1999;144:813–821, copyright The
Rockefeller University Press. D, Courtesy A.I. Lamond, University of
Dundee, United Kingdom. From Ferreira J, Paolella G, Ramos C, et al.
Spatial organization of large-scale chromatin domains in the nucleus:
a magnified view of single chromosome territories. J Cell Biol.
1997;139:1597–1610, copyright The Rockefeller University Press.
E–F, From Ma H, Samarabandu J, Devdhar RS, et al. Spatial and
temporal dynamics of DNA replication sites in mammalian cells. J Cell
Biol. 1998;143:1415–1425. Copyright 1998 The Rockefeller University
Press.)

5 µm 5 µm
CIdU added for 2 min CIdU added for 2 min
IdU added 4 hrs IdU added 6 hrs
later for 5 min later for 5 min
fluorescent dye allow the newly replicated DNA to be
observed directly in living cells.
coordinately during differentiation. Thus, TADs with The time at which each replicon fires during the S
similar function tend to end up close to each other phase can be seen clearly by synchronizing cells at the
forming larger compartments—for example, euchroma- beginning of the S phase, releasing them from cell-cycle
tin and heterochromatin. arrest, and then exposing them to BrdU at various times
Evidence for the replication of clusters of replicons thereafter. This experiment reveals very distinctive pat-
within the nucleus was first obtained by fiber autoradi- terns of DNA synthesis occurring at different times
ography. Cells were fed radioactive precursors for DNA during the S phase (Fig. 42.14B–C and E–F). Early on,
synthesis and then examined by electron microscopy. euchromatin replicates throughout the nucleus. Later,
(For an explanation of this technique, see Fig. 40.3.) replicating regions are concentrated around nucleoli
Clusters of replicating DNA regions were observed. and other areas of constitutive and facultative hetero-
Several methods are available to observe the spatial chromatin (see Chapter 8). Toward the end of the
distribution of DNA replication during the S phase. One S phase, replication is largely concentrated in blocks
can employ a pulse of nucleotide base analogs that are of heterochromatin. These observations show that
incorporated into DNA and later detected by fluores- DNA replication occurs throughout the nucleus, wher-
cence microscopy. For example, thymidine analogs such ever DNA is located. DNA does not move to a small
as BrdU (bromodeoxyuridine triphosphate), IdU (iodo- number of discrete sites to be replicated (as was once
deoxyuridine), and CldU (chlorodeoxyuridine) can be thought).
detected in fixed cells by reaction with fluorescent The timing of replication of particular replication
antibodies (BrdU is called by its correct name BrdUTP origins was studied in detail in budding yeast using a
in Fig. 42.14). Alternatively analogs labeled with a modification of the BrdU labeling method. First, a
738 SECTION X  n  Cell Cycle

procedure was developed whereby all cells in a popula- density) during the S phase. This experiment demon-
tion entered S phase synchronously. Next, the shift in strated that each ARS element replicates at a character-
the density of the DNA following BrdU incorporation istic time during the S phase. More recently, genome-wide
was used to distinguish DNA that had replicated from maps of replication timing have been constructed by
DNA that had not (Fig. 42.15). Incorporation of BrdU isolating newly replicated DNA at various times during S
into DNA makes the newly synthesized daughter DNA phase and identifying the chromosome regions involved
strand heavier, allowing its separation from the parental by high-throughput DNA sequencing.
DNA by centrifugation on a cesium chloride density The most striking aspect of these patterns of DNA
gradient (see Chapter 6). It was then relatively simple to synthesis is their reproducibility from one cell cycle to
take DNA probes from different regions of the chromo- the next. For example, regions of DNA labeled early in
some and determine when each replicated (changed its the S phase overlap little or not at all with DNA labeled

A 1 2 3 4 5 = Restriction enzyme
* cutting site
Origin

1 2 3 4 5
Initiation
H:L
3 H:H
1 2 4 5
Replication H:L
G force
H:H

3
1 2 4 5 Isolated DNA
digested with
Heavy: Heavy:Heavy + Heavy:Light + Heavy: restriction enzyme
Heavy Heavy:Light Heavy:Heavy Heavy
Heavy:
Light

B C D
100 100 100 Centromeres
replicate early
Amount of DNA

% Replicated
% In H:L peak

a
1,5 3
1,5 b
50
50 50
Telomeres
replicate late
3
2,4
0 0 0
HL HH 0 2 4 6 8 0 14 28 42 56
Top Density Bottom Time of replication (min) Time in S phase (min)

FIGURE 42.15  MEASUREMENT OF THE TIME OF REPLICATION OF PARTICULAR CHROMOSOMAL REGIONS IN SACCHARO-
MYCES CEREVISIAE. A–C, This protocol is based on a classic density shift experiment of Messelson and Stahl that proved that DNA replication
is semiconservative. S. cerevisiae cells are grown for several generations in a medium containing 13C and 15N so that their DNA is fully substituted
with heavy isotopes. At the beginning of the experiment, the cells are synchronized so that they enter the S phase in a single wave. At the same
time, the heavy (H) isotope medium is removed and replaced with “light medium” (L) containing 12C and 14N. At various times after the initiation
of the S phase, aliquots of cells are removed, and the DNA is isolated. The DNA is then cleaved with restriction enzymes so that the chromosomes
are cut into many fragments. DNA from each time point is then subjected to CsCl density gradient centrifugation. When any local region of DNA
is replicated, its density alters from heavy/heavy to heavy/light. After very short incubations with light isotopes, only DNA near the origin of replication
will be heavy/light; all other DNA will be heavy/heavy. These two populations of molecules are separated from one another by density gradient
centrifugation. To examine the timing of replication of a specific gene, a cloned segment of DNA corresponding to the region of interest is used
to probe (by DNA hybridization) the heavy/heavy and heavy/light peaks from each gradient. The time of replication of each locus is the time at
which the restriction fragment being detected by DNA hybridization moves from the heavy/heavy peak to the heavy/light peak. The numbers in
panels B and C refer to the numbered regions of the chromosomes shown in A. D, Data from a replication timing experiment show that in
budding yeast, centromeres replicate early in the S phase and telomeres replicate late. To generate curve a, fractions from a gradient like that
shown in panel B were hybridized to a cloned centromere region. To generate curve b, fractions from the same gradient were hybridized to a
cloned telomere region probe. Note that in mammalian cells, centromeres replicate late and telomeres replicate earlier. (Based on the work of the
laboratory of B.J. Brewer and W.L. Fangman. For reference, see Meselson M, Stahl FW. The replication of DNA in Escherichia coli. Proc Natl
Acad Sci U S A. 1958;44:671–682.)
CHAPTER 42  n  S Phase and DNA Replication 739

3 hours later (Fig. 42.14C and E). However, DNA labeled following completion of DNA synthesis at the early
at corresponding points of the S phase in two successive replicons and disassembly of those replisomes.
cell cycles superimposes almost entirely. This strongly
suggests that particular TADs initiate DNA synthesis Replication Stress and
during preferred “windows” during S phase.
At least three mechanisms may explain the sequence
the Intra-S Checkpoint
of replication patterns for different chromosomal regions: The textbook-smooth DNA double-helix is actually lit-
1. Local chromatin structures, established as a result of tered with problems and obstacles, including DNA
gene expression influence the time of replication, damage, ribonucleotides inserted by mistake into the
particularly in metazoans. Thus, transcriptionally DNA, and awkward secondary structures caused, for
active loci (where transcription factors are already example, by base-pairing within single DNA strands as
bound) have a head start over other regions of the opposed to between strands. In addition, if cell-cycle
chromosomes, permitting them to initiate DNA repli- regulation is perturbed, cells can simply run out of either
cation first. In general, euchromatin (which has low the deoxynucleotide triphosphate (dNTP) precursors
DNA methylation and high histone acetylation) repli- that they need to synthesize DNA or a number of key
cates first, followed by facultative heterochromatin proteins that are also present in very low amounts. The
(including the inactive X) and, finally, the constitutive replication fork may stall as result of these problems and
pericentric heterochromatin (which has heavy DNA often needs help to complete replication.
methylation and low histone acetylation). This mecha- An intra-S checkpoint responds to stalled replication
nism can explain why a particular locus may replicate forks, probably as a result of detecting excessive single-
at different times in two cell types. For example, one stranded DNA (Fig. 42.16). Replication forks stall if they
origin in mammalian genomes is located just upstream encounter a damaged DNA base, bases that the repli-
of the DNA encoding the β-globin gene (encoding a some cannot “read” or a DNA secondary structure that
protein subunit of hemoglobin). This region of more it cannot unfold. DNA damage can result from ultraviolet
than 200-kb of DNA replicates early in erythroid cells light, mutagens or chemicals such as aldehydes produced
that express the β-globin gene, but later in other cells as a by-product of ethanol metabolism.
where the gene is inactive. Replication blockage leads to a condition known
2. The higher-order packing of chromatin in the nucleus as replication stress in which single-stranded DNA
may influence when origins replicate. It is likely that accumulates. Replication stress with stalled forks can
replication origins fire in a sequence that is imposed also result from inappropriate activation of genes that
on them at the same time that the TAD organization promote cell cycle progression (oncogenic stress; see
of interphase chromosomes is reestablished during Chapter 41). Possible reasons include insufficient licens-
mitotic exit and early G1 phase. ing of origins during the G1 phase, depletion of nucleo-
3. Limiting amounts of certain essential proteins may side triphosphate pools, or insufficient pools of essential
contribute to the sequential replication of different replication factors. Surprisingly, cells make a number
chromatin domains. These proteins accumulate of essential components—including the dNTPs—in
on replisomes assembled on early-firing replicons amounts just sufficient for replication, so there is little
and only become available to later-firing replicons margin for error.

dsDNA Unreplicated RPA


breaks DNA DNA damage
*
RPA

ATR actived by Stalled fork Fork approaches


excess ssDNA from activated
(ATM)2 2x ATM stabilizes stalled dormant origin
active replication forks
Chk1/Chk2

Cdc25A
Block initiation of new replication foci
Degraded
Block cell cycle progression

FIGURE 42.16  REPLICATION STRESS AND THE INTRA-S CHECKPOINT. If DNA breaks are detected, the ATM (ataxia-telangiectasia
mutated) kinase activates downstream kinases Chk1 and Chk2, leading to phosphorylation of Cdc25A and its subsequent ubiquitin tagging and
degradation. This blocks the initiation of new replication forks as well as cell-cycle progression more generally. If DNA persists in unreplicated
form, or if replication forks stall, the ATR (ataxia-telangiectasia and Rad3–related) kinase activates a similar downstream response. ATR stabilizes
stalled replication forks to give nearby dormant origins time to fire and replicate the DNA downstream of the block.
740 SECTION X  n  Cell Cycle

Transcription Processing of mRNA Stability of the mRNA


increases increases (on free ribosomes)
increases
Primary Mature mRNA
transcript exported to Degrading
cytoplasm enzymes

Histone
gene 3' ends of pre-mRNA
remain behind

NUCLEUS

FIGURE 42.17  THREE MECHANISMS THAT ELEVATE HISTONE EXPRESSION DURING THE S PHASE.

In conditions of replication stress, the ATM (ataxia- are made during a relatively brief period. Histone synthe-
telangiectasia mutated) and ATR (ataxia-telangiectasia sis apparently keeps pace, in part, because there are
and Rad3–related) kinases activate the DNA damage approximately 40 sets of histone genes.
response (see Box 43.1). The activities of these kinases Synthesis of histones during the S phase is tightly
and their downstream effectors result in the degradation coupled to ongoing DNA replication and a controlled
of Cdc25A, the phosphatase that triggers entry of the cell supply of histones is essential for normal replication. If
into mitosis. The resulting inactivation of Cdks during replication is blocked either by addition of drugs or by
the S phase prevents replication initiation within other temperature-sensitive mutants, histone synthesis declines
unreplicated domains. abruptly shortly thereafter, possibly because nucleosome
A key aspect of this response unique to S phase is that deposition appears to be linked to lagging strand synthe-
ATR-activated by the presence of excessive levels of sis. Indeed, the chaperone that assembles histones into
single-stranded DNA associated with RPA protects the nucleosomes is associated with the replisome. This link
stalled forks from disassembly, known as replication between histone synthesis and DNA replication appears
fork collapse. This response is critical, because replica- to involve at least three processes (Fig. 42.17).
tion forks with unwound and nicked DNA molecules can First, transcription of the histone genes rises three-
turn into breaks if the fork disassembles before replica- fold to fivefold as cells enter the S phase. Each histone
tion is complete and all the DNA is ligated. Error-prone gene has a cell-cycle-responsive element in its promoter
mechanisms tend to repair DNA damage that arises to which a transcription factor binds specifically during
during replication stress. The resulting mutations are the S phase.
thought to contribute to oncogenic transformation. Second, the processing of histone messenger RNAs
Of course, stalled replication forks must not only be (mRNAs) increases six- to 10-fold as cells enter the S
protected, but they must also somehow be rescued. At phase. Histone mRNAs are not polyadenylated, and the
moderate levels of replication stress, ATR activation primary transcripts are considerably longer than the
blocks the activation of new replication foci, but allows mature forms. Processing of the 3′ end of histone pre-
dormant origins to fire near stalled forks in replication mRNAs involves the U7 small nuclear ribonucleoprotein
domains that are already active. This results in the arrival (snRNP) (see Chapter 11), a portion of which recognizes
of a converging fork on the downstream side of the histone mRNA and base-pairs with it during processing.
damaged DNA, leaving only a very small gap that can be Cell-cycle-dependent regulation of processing appears to
repaired by a specialized DNA polymerase in a process involve changes in the accessibility of the necessary
known as translesion synthesis. Thus, in the real world portion of U7 small nuclear RNA (snRNA). This region
where replication forks routinely encounter problems is inaccessible in G0 cells but becomes accessible
that cause them to stall, a pool of dormant licensed when cells that reenter the cycle and begin the S phase.
replication origins is essential for integrity of the genome. The mechanism for this change in RNA conformation
is not known.
Third, changes in the stability of the mRNA also regu-
Synthesis of the Histone Proteins late histone synthesis. Normally, the level of histone
Chromatin contains approximately equal masses of DNA mRNA on free polysomes drops rapidly by approximately
and core histones. Human cells require about 62 × 106 35-fold as cells enter the G2 phase. If DNA synthesis is
copies of each core histone, assuming a genome size of interrupted during the S phase, a region at the 3′ end of
6.2 × 109 bp and 200 bp per nucleosome. Because the mature message targets the mRNA for degradation.
approximately 90% of histone transcription occurs If this region is removed from the 3′ terminus of the
during the S phase, enormous amounts of these proteins histone mRNA, the normal link between ongoing
CHAPTER 42  n  S Phase and DNA Replication 741

replication and mRNA stability is lost. Furthermore, this genome has been replicated completely and that no
sequence, transposed onto the 3′ terminus of a globin harmful DNA damage is present. These checks, together
mRNA, renders that mRNA sensitive to degradation if with other ongoing preparations for mitosis, are the
DNA synthesis is blocked. Degradation of histone mRNA principal events of the G2 phase (see Chapter 43).
requires ongoing protein synthesis, and it has been
speculated that histones themselves participate in the
ACKNOWLEDGMENTS
control.
As discussed in Chapter 8, specialized variant forms We thank Hiro Araki, Julian Blow, Cristina Cardoso,
of histones are synthesized and inserted into the chro- David Gilbert, and Julian Sale for suggestions on revi-
matin outside of S phase. These histones are encoded sions to this chapter.
by mRNAs with introns and normal poly(A) tails and
are therefore not processed by the specific S phase–
associated pathway (see Chapter 11). Their insertion SELECTED READINGS
into chromatin is typically correlated with RNA tran- Boos D, Frigola J, Diffley JF. Activation of the replicative DNA helicase:
scription rather than DNA replication and involves spe- breaking up is hard to do. Curr Opin Cell Biol. 2012;24:423-430.
cific chromatin-remodeling factors. Costa A, Hood IV, Berger JM. Mechanisms for initiating cellular DNA
replication. Annu Rev Biochem. 2013;82:25-54.
Deegan TD, Diffley JF. MCM: one ring to rule them all. Curr Opin
Other Events of the S Phase Struct Biol. 2016;37:145-151.
Hills SA, Diffley JFX. DNA replication and oncogene-induced replicative
Although the bulk of attention on the S phase focuses stress. Curr Biol. 2014;24:R435-R444.
on the duplication of the chromosomes, centrosomes Labib K. How do Cdc7 and cyclin-dependent kinases trigger the initia-
also duplicate at this time. Duplication of the centro- tion of chromosome replication in eukaryotic cells? Genes Dev.
2010;24:1208-1219.
somes is essential for stability of the genome, because McIntosh D, Blow JJ. Dormant origins, the licensing checkpoint, and
they set up the poles of the mitotic spindle that is the response to replicative stresses. Cold Spring Harb Perspect Biol.
responsible for accurate partitioning of the replicated 2012;4:a012955.
chromosomes. Interestingly, Orc1 functions indepen- Méchali M, Yoshida K, Coulombe P, Pasero P. Genetic and epigenetic
dent of DNA replication with cyclin A to limit centriole determinants of DNA replication origins, position and activation.
Curr Opin Genet Dev. 2013;23:124-131.
duplication to once per cell cycle. (See Fig. 34.17 for a O’Donnell M, Langston L, Stillman B. Principles and concepts of DNA
discussion of centrosome duplication.) replication in bacteria, archaea, and eukarya. Cold Spring Harb
With the completion of DNA replication and duplica- Perspect Biol. 2013;5:a010108.
tion of the centrosomes, the cell is ready to divide. As O’Donnell M, Li H. The eukaryotic replisome goes under the micro-
the levels of Cdk activity rise toward the threshold that scope. Curr Biol. 2016;26:R247-R256.
Rhind N, Gilbert DM. DNA replication timing. Cold Spring Harb Per-
is sufficient to trigger mitotic entry and other factors spect Biol. 2013;5:a010132.
necessary for mitosis accumulate, the cell continues to Zeman MK, Cimprich KA. Causes and consequences of replication
screen the integrity of the DNA to ensure that the stress. Nat Cell Biol. 2014;16:2-9.
This page intentionally left blank
CHAPTER 43 

G2 Phase, Responses to DNA


Damage, and Control of Entry
Into Mitosis

T he G2 phase is the gap between the completion of multifaceted regulation of Cdk1 (see Fig. 40.14) includes
DNA replication and the onset of mitosis. This chapter binding of cyclin cofactors, inhibition and activation
begins with the biochemical basis for the G2/mitosis (M) by phosphorylation, binding of inhibitory molecules,
transition and discusses how the G2/M checkpoint delays changes in subcellular localization and changes in the
this transition if DNA damage is detected. Finally, the activities of competing protein phosphatases (Fig. 43.1).
chapter introduces the major pathways that cells use to These various factors are finely balanced, until a positive
repair damaged DNA. feedback loop enables the cells to make an abrupt and
decisive entry into mitosis.
Mammals have at least three B-type cyclins: B1, B2,
Enzymology of the G2/Mitosis Transition and B3. Cyclin B1 is essential for triggering the G2/M
The transition from the G2 phase into mitosis is the most transition. Cyclin B1, newly synthesized during the latter
profound morphologic and physiological change that part of the cell cycle, binds Cdk1 and shuttles it in and
occurs during the life of a proliferating cell. Entry into out of the nucleus. Importin β carries the Cdk1–cyclin
mitosis is controlled by a network of stimulatory and B1 complex into the nucleus, and then Crm1 rapidly
inhibitory protein kinases and phosphatases, presided exports it back to the cytoplasm (see Chapter 9). Cdk1–
over by Cdk1–cyclin B1. (Chapter 40 introduced the cyclin B2 associates with the Golgi apparatus during
components involved in the G2/M transition.) interphase and might function in Golgi disassembly
Cdk1, the driving force for entry into mitosis, is during mitosis (see Fig. 44.4). Cyclin B3 may function
present at a constant level throughout the cell cycle. The only during meiosis in mammals.
Wee1/Myt1 phosphorylates Cdk1
on T14 & Y15 (inhibitory)

Cyclin B1 Y15 Cyclin B1


Cyclin B1
Wee1 Myt1 T 14
T161 T161
Cdk1 Cdk1 Cdk1 Cdk1 Cdk1
Inactive Inactive Inactive Active Inactive
kinase kinase CAK kinase Cdc25A,B,C kinase kinase
CAK phosphorylates Cdc25 Cyclin B1
Cyclin B1 Cdk1 on T161 (stimulatory) dephosphorylates degraded
T14 and Y15

S G2 M G1

FIGURE 43.1  REGULATION OF CDK1 BY CYCLIN BINDING AND PROTEIN PHOSPHORYLATION FROM THE LATE S PHASE
THROUGH MID-MITOSIS. (For reference, see Protein Data Bank [PDB; www.rcsb.org] file 1X8B and Squire CJ, Dickson JM, Ivanovic I, et al.
Structure of human Wee1A kinase: kinase domain complexed with inhibitor PD0407824. Structure. 2005;13:541–550.)

743
744 SECTION X  n  Cell Cycle

As cells approach the G2/M transition, phosphoryla- into mitosis (Fig. 43.2). Together, Wee1 and Myt1 ensure
tion simultaneously activates and inhibits Cdk1 bound to that Cdk1 remains inactive as it shuttles into and out of
cyclin A or B. Cdk-activating kinase (CAK-actually Cdk7– the nucleus (Fig. 43.3).
cyclin H), located in the nucleus, phosphorylates Cdk1 This combination of stimulatory and inhibitory phos-
on T161 (Fig. 43.1). This triggers a refolding of the active phorylations holds Cdk1–cyclin complexes poised for a
site cleft that allows the enzyme to bind substrates (see burst of activation.
Fig. 40.13). This phosphorylation activates Cdk1-cyclin That burst occurs when one of three Cdc25 protein
kinases in the nucleus (Fig. 43.1). phosphatases removes the inhibitory phosphates from
At the same time, Wee1 and Myt1 kinases phosphory- T14 and Y15 (Figs. 43.1 and 43.8). Cdc25s are “dual-
late T14 and Y15 to turn the enzyme off. Wee1 in the specificity” protein phosphatases (see Fig. 25.5) that
nucleus phosphorylates Cdk1 on Y15 adjacent to the remove phosphates from serine (S), threonine (T), and
adenosine triphosphate (ATP)-binding site. This stops tyrosine (Y) residues. Of the three Cdc25 phosphatases,
the kinase from using ATP to phosphorylate substrates. only Cdc25A is indispensable for life. It functions at both
While Cdk-cyclin complexes are in the cytoplasm, they the G1/S and G2/M transitions, whereas Cdc25B and
are phosphorylated on T14 and Y15 by Myt1 kinase associ- Cdc25C have roles only in the G2/M transition.
ated with the Golgi apparatus and endoplasmic reticu- Because Cdc25 phosphatases trigger mitotic entry,
lum. The role of Wee1 as a mitotic inhibitor is clearly their regulation is both important and elaborate.
demonstrated in Schizosaccharomyces pombe: overex- Cdc25A and Cdc25C are held inactive during interphase
pression of Wee1 delays or prevents the entry of cells by mechanisms involving stimulatory and inhibitory

A. Wee1  B. Wee1+ C. Wee1+ (3x) D. Wee1+ (5x)

FIGURE 43.2  EFFECT OF CHANGING CELLULAR LEVELS OF WEE1 PROTEIN ON CELL-CYCLE PROGRESSION IN FISSION
YEAST. A, Cells that lack functional Wee1 protein enter mitosis too soon in the cell cycle and are smaller than wild-type cells (B). C–D, Cells
that express excess Wee1 protein are too effective at inactivating Cdk1 and are severely delayed in their ability to enter mitosis (hence their larger
size). (From Russell P, Nurse P. Negative regulation of mitosis by Wee1+, a gene encoding a protein kinase homolog. Cell. 1987;49:559–567.)

AT
ADP
14-3-3 Cyclin B1 Myt1
docking (Active) Y15 Cyclin B1 Myt1
site Cyclin B1 docking site (Inactive)
T14 blocked
T161
Cdc25C Cdk1 NES
(Inactive) (Inactive)
CYTOPLASM
NUCLEUS 14-3-3 AT
ADP
14-3-3 Cyclin B1
Y15
Cyclin B1 T161
Wee1 T14 Wee1
14-3-3 (Active) T161 Cdk1 NES
Cdc25C Cdk1 NES (Inactive) (Active)
(Inactive) (Inactive)
Cdc25A Cyclin B1 nuclear
(Inactive/unstable) export signal inactivated

S phase G2 phase Mitosis

FIGURE 43.3  CDK REGULATION IN INTERPHASE AND MITOSIS. Inhibitory phosphorylation and shuttling of components between the
nucleus and cytoplasm regulate Cdk1 activity in interphase and mitosis. Cdc25, which would remove the inhibitory phosphates, is held inactive
until its activation by Polo kinase and Cdk1-cyclin B stimulates mitotic entry (not shown here). ADP, adenosine diphosphate; ATP, adenosine
triphosphate; NES, nuclear export sequence. (Based on an original figure by Helen Piwnica-Worms. For reference, see PDB file 1YWT.)
CHAPTER 43  n  G2 Phase, Responses to DNA Damage, and Control of Entry Into Mitosis 745

Interphase Mitosis
Low Cdc25C activity, cytoplasmic Enhanced Cdc25C activity, nuclear,
S216 phosphorylation blocked
14-3-3 Phosphorylated
S216 binds
14-3-3 protein Y15
Cyclin B1 Cdc25C Cyclin B1
Cdc25C T14
T161 T161
Cdk1 Cdk1
14-3-3 (Inactive) (Active)
Cdc25A
Cdc25A Cyclin B1/Cdk1 Cdc25A accumulates due to increased stability
docking site Enhanced interaction with Cdk1-cyclin B1
Low Cdc25A protein levels due to proteolysis complexes due to loss of 14-3-3 binding
Reduced interactions with Cdk1-cyclin B1 Enhanced activity due to
due to 14-3-3 binding at the Cdc25 mitotic-specific phosphorylation
C-terminus
FIGURE 43.4  REGULATION OF CDC25A AND CDC25C ACTIVITY IN INTERPHASE AND MITOSIS. (Based on an original figure by Helen
Piwnica-Worms.)

phosphorylation, binding to a 14-3-3 adapter protein, For cells to enter and complete mitosis, protein phos-
alterations in their subcellular localization, and ubiquitin- phatase 2A (PP2A, a protein serine/threonine phospha-
mediated proteolysis (Fig. 43.4). tase; see Fig. 25.6) must be inactivated. PP2A removes
Phosphorylation on a serine residue of Cdc25 creates the phosphates that Cdk1 puts on its target proteins. If
a binding site for a 14-3-3 adapter protein that interacts PP2A is not inhibited, cells enter mitosis, but they then
with the phosphoserine and several flanking amino acids slip back into interphase. PP2A inhibition involves three
(see Fig. 25.10). Association with 14-3-3 interferes with steps. Newly active Cdk1 phosphorylates a protein
Cdc25A binding to Cdk1–cyclin B1. Phosphorylation at kinase called Greatwall (a name from Drosophila genet-
other sites also targets Cdc25A for ubiquitination by ics). Greatwall phosphorylates a small target protein that
SCFβTrCP and destruction by proteasomes. Cdc25C has a then binds to the substrate-binding pocket of PP2A,
nuclear export sequence (see Fig. 9.17), so Cdc25C acting as a competitive inhibitor. Cdk1 activation at the
bound to 14-3-3 is primarily cytoplasmic (Fig. 43.3). onset of anaphase eventually leads to dephosphorylation
Following the theme of phosphorylation having both of the small protein and activation of PP2A, thereby
positive and negative effects, full activation of Cdc25s allowing cells to exit mitosis.
requires phosphorylation of other residues in the amino-
terminal region. A protein kinase called Polo (see next Changes in Subcellular Localization at
paragraph) initiates this phosphorylation of Cdc25,
the G2/M Transition
followed by more extensive phosphorylation by
Cdk1–cyclin B1, the substrate of Cdc25. This action of Late in G2, phosphorylation inactivates the nuclear
Cdk1–cyclin B1 on Cdc25 creates a powerful positive export signal of cyclin B1 (see Chapter 9). As a result,
feedback amplification loop that provides a burst of Cdk Cdk1–cyclin B1 accumulates in the nucleus within 5
activity and triggers entry into mitosis (Fig. 43.7). minutes (Figs. 43.3 and 43.5). Cdc25C also stops shut-
A molecular trigger is required to start the ampli- tling at the G2/M transition, probably as a result of Polo
fication cycle in which Cdk1–cyclin B1 and Cdc25 kinase phosphorylation. The Cdk1–cyclin B that accumu-
activate each other. Members of the Polo family of lates in the nucleus is thought to be already active, so
protein kinases are candidates for this role, by activat- the reason for this rapid nuclear localization is not
ing Cdc25. These kinases possess amino acid motifs entirely clear. The concentration of Cdk–cyclin B com-
called “polo boxes” that bind to protein partners after plexes together with Cdc25A and Cdc25C in the con-
they have been “primed” by phosphorylation by another fined volume of the nucleus may contribute to the final
kinase. Interestingly, the most common “priming burst of Cdk1–cyclin B1 activation.
kinases” for polo are Cdk–cyclin pairs. This creates
another positive amplification loop that allows for Cdk1 Activity and the Initiation
additional levels of control and rapid activation of Cdk–
of Prophase
cyclin complexes. In addition to activating Cdks by phos-
phorylating Cdc25, polo family kinases are involved in a Cdk2–cyclin A plays a critical role during the S phase
variety of mitotic events, including formation of a bipolar (see Chapter 42), but also helps trigger the G2/M transi-
spindle, cytokinesis, and passage through certain cell- tion. Cdk–cyclin A activity peaks at G2/M, before the
cycle checkpoints. peak of Cdk1–cyclin B1 activity, and inactivation of
746 SECTION X  n  Cell Cycle

cyclin A in Drosophila or mammalian cultured cells These events occur while most Cdk1–cyclin B1 is in
arrests the cell cycle in G2. Furthermore, G2 cells enter the cytoplasm. It appears most likely, therefore, that
mitosis prematurely if injected with active Cdk2–cyclin Cdk1–cyclin A triggers at least the nuclear events of
A complexes just completing S phase. Finally, microin- prophase (Fig. 43.6). Commitment to mitosis appears
jection of a selective inhibitor of Cdk–cyclin A causes to be irreversible only after Cdk1–cyclin B1 enters
prophase cells to return rapidly to interphase; chromo- the nucleus.
somes decondense, rounded prophase cells flatten, and
the interphase microtubule network returns. Recap of the Main Events of
Cdk activity regulates several events during the G2-to-
the G2/M Transition
prophase transition. In the cytoplasm, the half-life of
microtubules drops dramatically from approximately 10 Synthesis of cyclin B1 in the latter portion of the S and
minutes to approximately 30 seconds late in G2 (see G2 phases leads to assembly of Cdk1–cyclin B heterodi-
Table 44.1). This, coupled with an enhanced ability of mers that shuttle into and out of the nucleus, spending
centrosomes to initiate microtubule polymerization, most of their time in the cytoplasm associated with
completely transforms the organization of the microtu- microtubules. In late G2 phase, Cdk1–cyclin A activity
bule cytoskeleton. Centrosomes take on the appearance initiates mitotic prophase, beginning with changes in
of spindle poles and migrate apart over the surface of microtubule dynamics and chromosome condensation.
the nucleus. While this is happening, chromatin begins Several events trigger entry into the active phase of
to condense in the nucleus. A protein complex called mitosis. Cdc25A accumulates and no longer binds 14-3-3
condensin is required for this condensation to begin proteins, allowing it to interact more effectively with
during prophase (see Fig. 8.18). Cdks activate condensin Cdk1–cyclin B. The phosphoserine-binding site for 14-3-3
by phosphorylation of two of its subunits in late G2. proteins on Cdc25C is dephosphorylated, allowing
Cdc25C to accumulate in the nucleus. In addition, phos-
phorylation of cyclin B1 blocks its export from the
G2 phase Prophase Metaphase nucleus and promotes its import, thus causing Cdk1–
cyclin B1 to accumulate rapidly in the nucleus. The
inhibitory kinase Wee1 is also phosphorylated, causing
its activity to drop. Cdc25A and Cdc25C activate Cdk1–
cyclin B1 by removing inhibitory phosphates on T14 and
Y15. This starts in the cytoplasm and then may be stimu-
lated as the proteins concentrate in the nucleus. There,
the action of Cdk1–cyclin B1 on the nuclear lamina trig-
FIGURE 43.5  CYCLIN B1 LOCALIZATION DURING MITOSIS. gers nuclear envelope breakdown and drives the cell
Cyclin B1 moves rapidly from the cytoplasm into the nucleus at the
onset of prophase and subsequently associates with the spindle
into mitosis. Fig. 43.7 highlights the positive feedback
during mitosis. (Courtesy Christina Karlsson and Jonathon Pines, loop between Cdc25 and Cdk1–cyclin B that drives the
Wellcome/CRC Institute, Cambridge, UK.) cell into mitosis.

G2/M transition events:


Microtubule stability drops
Cyclin B1 Cyclin B1 Cyclin B1
Centrosomes migrate apart
Chromosomes condense
Cdk1 Cdk1 Kinetochore assembly starts
CYTOPLASM
Shuttling
NUCLEUS Cyclin B1
Cyclin B1 Cyclin B1
degraded
Cdk1
Cdk1 Cdk1
Cdk1
Cyclin A

Cyclin A Cyclin A Cyclin A


degraded
Cdk2 Cdk2 Cdk1

G1 S G2 Prophase M

FIGURE 43.6  CDK1 REGULATION. Locations and patterns of activation of Cdk1 complexed to cyclin A versus cyclin B across the cell cycle.
CHAPTER 43  n  G2 Phase, Responses to DNA Damage, and Control of Entry Into Mitosis 747

Cell-cycle arrest in Rad9+ and rad9 cells


Cdc25 phosphatase with DNA damage
dephosphorylation
cdc13tsRad9+ cdc13tsRad9-
Wee1 and Myt1
phosphorylation

T161 Cells on
Inactive Active Cdk1 CAK
kinase protein kinase plates
Myt1,
Activated by Polo kinase Cdc25 Wee1
and amplified by protein protein
positive feedback phosphatase kinases
from Cdk1–cyclin B
Bud

S G2 M
e
DNA otyp
damage pe phen
FIGURE 43.7  SUMMARY OF THE CDK1 FEEDBACK REGULA-
W ild-ty Nucleus
TION MECHANISM AT THE G2/M TRANSITION.
G2 arrest

No delay
Why did such an elaborate system evolve to regulate Phe
n
Cells otype o
k f
the G2/M transition? The answer appears to lie in with eep divi rad9 mu
fragm ding tan
the exquisite sensitivity provided by the interlocking ente and d t
d nu i
network of stimulatory and inhibitory activities. On the clei e
one hand, this network ensures a rapid, almost explo-
sive, final transition into mitosis. On the other, it pro- FIGURE 43.8  GENETIC IDENTIFICATION OF THE G2/M
vides a number of ways to delay the G2/M transition if CHECKPOINT. Cells defective in the G2/M checkpoint (Rad9 mutants
the cell detects damage to chromosomes. Attempting of budding yeast) cannot delay their entry into mitosis in the presence
of damaged DNA and therefore divide themselves to death. Budding
mitosis with chromosomal damage can lead to cell death yeast Rad9 is an ortholog of the ATM (ataxia-telangiectasia mutated)
or contribute to cancer. adapter protein 53BP1 (see text). Confusingly, budding yeast Rad9 is
not related to fission yeast Rad9, which gives the 9-1-1 complex its
name (see text). (Courtesy Ted Weinert, University of Arizona, Tucson.)
G2/M Checkpoint
Separation of sister chromatids during mitosis is a poten-
tial danger point for a cell. After DNA is replicated each
chromosome consists of paired sister chromatids held detected and repaired in the daughter cells after division
together by cohesin. Therefore, if the DNA is damaged, (see later).
the cell can use information present in the undamaged Studies of radiation-induced G2 delay in budding
chromatid to guide the repair process. However, once yeast identified a major cell-cycle checkpoint that is sen-
sisters separate, this corrective mechanism can no longer sitive to the status of the cellular DNA. Cells defective in
operate. In addition, if a cell enters mitosis before com- this checkpoint are more sensitive than wild-type cells
pleting replication of its chromosomes, attempts to sepa- to radiation injury because they continue to divide,
rate sister chromatids damage the chromosomes. To despite the presence of broken or otherwise damaged
minimize these hazards, a checkpoint operates in the G2 chromosomes (Fig. 43.8). The cells die, presumably from
phase to block mitotic entry if DNA is damaged or DNA chromosomal defects or loss. In metazoans, the G2/M
replication is incomplete. checkpoint delays entry into mitosis until the damage is
Just as DNA damage can arrest the cell cycle in G1 either fixed, triggers cell suicide by apoptosis, or causes
phase, damaged or unreplicated DNA also halts the cell cells to enter a nonproliferating (senescent) state. The
cycle temporarily in the G2 phase. Interestingly, the G1 checkpoint works by modulating the activities of the
checkpoint—which can be activated by a single DNA components that control the G2/M transition.
break in human cells—is more sensitive than the G2/M
checkpoint, which requires 10 to 20 breaks to block
cell-cycle progression. The G2/M checkpoint may be less
DNA Damage Response
sensitive then the G1 checkpoint, because G2 cells are Considering that it is the blueprint for life, DNA
already primed to enter mitosis. Consequently, human is remarkably accident-prone, and organisms have an
cells can enter mitosis with limited amounts of damaged elaborate network of mechanisms to repair those acci-
or unreplicated DNA. These problem regions can be dents. This complex network is known as the DDR
748 SECTION X  n  Cell Cycle

(DNA damage response). This section discusses a few biochemical signal as a result of the detected damage,
key components of the DDR, and Box 43.1 (see later) and EFFECTORS (both protein kinases and transcriptional
explains several mechanisms that repair damaged DNA. activators) that coordinate repair pathways to block cell-
The minimal machinery of a DNA damage checkpoint cycle progression and repair the damage.
(see Fig. 40.4) involves SENSORS that detect DNA damage, When DNA is damaged, several proteins concentrate
TRANSDUCERS (usually protein kinases) that produce a at the damage site within seconds to minutes, forming a
focus that activates the G2/M checkpoint and repairs the
damage. The use of high-energy lasers to “draw” patterns
of DNA damage onto cell nuclei revolutionized our
γH2AX understanding of the DDR by allowing a minute-by-
BRCA1
minute mapping of the events during focus formation
and leading to DNA repair (Fig. 43.9).
If a single strand of DNA is broken, the SENSOR enzyme
poly(adenosine diphosphate [ADP]-ribose) polymerase,
binds to the broken end and immediately starts polymer-
izing a chain of ADP-ribose (which it makes from nico-
tine adenine dinucleotide [NAD]). Several proteins bind
to poly(ADP-ribose) chains and recruit the critical TRANS-
DUCER ATM kinase (ataxia-telangiectasia mutated) to the
break together with its partner NBS1, a subunit of the
MRN complex (Fig. 43.10A). ATM is usually present in
the nucleus as an inactive dimer. Interactions with MRN
G2 nucleus separate the dimer into ATM monomers that are acti-
G1 nucleus vated by MRN docked to the broken end of a DNA
molecule.
FIGURE 43.9  INDUCTION OF DNA DAMAGE FOCI USING A The MRN complex is a SENSOR that detects double-
HIGH-ENERGY LASER. Laser-induced DNA damage results in acti- strand DNA breaks. In this case, the NBS1 subunit of
vation of the DDR with production of γH2AX (red) at damage sites MRN recruits ATM kinase to the break site. MRN is also
followed by binding of other factors, including BRCA1 (green). DNA is
blue. The left cell is not yellow because BRCA1 is not present in G1-
a nuclease with a key role in repairing those breaks. Its
phase cells. Micrograph taken 30 minutes after induction of damage. job is to chew back (resect) one DNA strand in a 5′ →
(Micrograph courtesy Martin Mistrik and Jiri Bartek.) 3′ direction at the site of a DNA break, leaving a stretch

A B DNA damage

DNA breaks
DNA strand
with damage
removed
ATM
dimer (inactive)
MRN complex RPA
binds to DNA break binds ssDNA

AT
ATM ADP ATRIP
autophosphorylation
ATRIP binds
ATM monomer to RPA
Active ATM binds ATR
(active) MRN complex

ATRIP

γ-H2AX ATR active


ATR
phosphorylated

FIGURE 43.10  MECHANISMS FOR LOCALIZING ATM AND ATR TO SITES OF DNA DAMAGE. ATR, ataxia-telangiectasia and Rad3
related; ssDNA, single-stranded DNA.
CHAPTER 43  n  G2 Phase, Responses to DNA Damage, and Control of Entry Into Mitosis 749

of single-stranded DNA that becomes coated with RPA, ovarian cancer coexists with breast cancer. BRCA1 is a
the protein that also coats single-stranded DNA at repli- very large, complex protein with numerous roles in DNA
cation forks (Fig. 42.12). RPA then attracts a protein repair that are still being determined.
called ATRIP plus its partner, the second key TRANSDUCER, ATM (Table 43.1) is dispensable for life, though
ATR kinase (ataxia-telangiectasia and Rad3 related). people lacking it have ataxia-telangiectasia—a disorder
ATM and ATR stay bound to the chromatin, which is associated with degeneration of cerebellar neurons, dila-
remodeled by reactions involving protein phosphoryla- tion of blood vessels, a very high predisposition for
tion, acetylation/deacetylation, methylation, and ubiqui- cancer, and a number of other symptoms. The NBN gene
tylation. The response is very complex. For example, is mutated in humans with Nijmegen breakage syn-
several hundred phosphorylation events have been drome, a rare inherited disorder featuring chromosomal
detected during the DDR. These chromatin changes are instability and a predisposition to cancer. ATR is essen-
likely part of a response to suppress transcription so that tial for life, presumably because it has a key role in ensur-
RNA polymerase does not collide with the damage and ing that replication forks are stabilized until all the
cause more problems. The histone modifications associ- single-stranded DNA created during S phase is replicated.
ated with the DDR generally tend to create “open” chro-
matin that is more accessible to the DNA repair ATM/ATR
machinery. Repair foci are often found just adjacent to
heterochromatin, and indeed, DNA repair appears to be
more efficient in euchromatin than heterochromatin. H2AX Chk1 Chk2 MDM2
ATR tends to stay on the single-stranded DNA close activated activated sequestered
to the break, but ATM spreads along the chromatin
γ-H2AX p53 Other
through a cycle of reactions involving its most famous Chromatin effects:
stabilized kinases
substrate, the specialized histone isoform H2AX (Figs. Cohesin binding Cdc25A
Focus formation
43.9 and 43.11). H2AX phosphorylated by ATM is known Recruitment of
inhibited p21 14-3-3σ
repair machinery degraded transcribed transcribed
as γ-H2AX. γ-H2AX forms very rapidly in a focus of modi-
fied chromatin immediately surrounding the damage. Wee1 Cdc25C
This amplifies and spreads the response to damage, activated inhibited
sequestered
because γ-H2AX binds a SENSOR protein, MDC1 that Cdk1–cyclin A
inhibited
recruits more ATM and repair factors. The response Cdk1–cyclin B1
spreads along the chromatin as ATM creates more γ- inhibited
H2AX, so that after a few minutes the γ-H2AX focus S G2 M G1
covers about a megabase of DNA.
MDC1 has two BRCT domains that bind the phos- FIGURE 43.11  THE G2/M CHECKPOINT BLOCKS THE G2/M
phorylated site on γ-H2AX. BRCT domains were discov- TRANSITION FOLLOWING ACTIVATION OF ATM AND/OR ATR
BY DNA DAMAGE. Dotted lines show activities that are switched off
ered in BRCA1, one of the first genes whose mutations by the checkpoint. The dashed line between ATM/ATR and the kinase
were linked to familial breast cancer. BRCA1 is mutated that inhibits Cdc25C indicates that this pathway is not yet known.
in 90% of families where inherited predisposition to (Based on an original figure by Helen Piwnica-Worms.)

TABLE 43.1  Key DNA Repair Gene Defects Associated With Human Disease
Human Disease Pathway Defective Genes*
Ataxia-telangiectasia (AT) Checkpoint ATM
Seckel syndrome Checkpoint ATR
Xeroderma pigmentosum NER XP-A, XP-B, XP-C, XP-D, DDB-2, XP-F, XP-G, POLH
Cockayne syndrome NER XP-B, XP-D, CSA, CSB
Trichothiodystrophy NER XP-D
Hereditary nonpolyposis colon cancer MMR MSH2, PMS2, MLH1
AT-like disorder DSB repair MRE11
Nijmegen breakage syndrome DSB repair NBN
Breast cancer predisposition HR BRCA1, BRCA2
LIG4 syndrome NHEJ LIGIV
Severe combined immune deficiency NHEJ ARTEMIS
ATM, ataxia-telangiectasia mutated; ATR, ataxia-telangiectasia and Rad3 related; DSB, double-strand break; HR, homologous recombination;
MMR, mismatch repair; NER, nucleotide excision repair; NHEJ, nonhomologous end joining.
*This list is an outline. For an updated list of the human DNA repair genes known to date, the reader is referred to http://sciencepark.mdanderson.org/labs/
wood/.
750 SECTION X  n  Cell Cycle

Table 43.1 shows only a few of the diseases associated From the DNA Damage Response to
with mutations in proteins involved in repairing DNA
breaks. (See Box 43.1 for a discussion of the major DNA
the G2/M Checkpoint
repair pathways.) It is likely that new components of this ATM and ATR at damage foci cooperate with adapters to
pathway remain to be identified. phosphorylate and activate the two key EFFECTOR kinases

BOX 43.1  DNA Repair in Vertebrates*

Every human cell experiences approximately 105 DNA repair mechanisms act in concert with the apoptotic machin-
damage events each day. These are mostly repaired accu- ery to ensure that the cell will die if the DNA damage cannot
rately, so only about 100 mutations are passed on in each be repaired (see Chapter 46). DNA damage checkpoints are
new human generation. In cancer, the spontaneous muta- a critical component of the cellular response to DNA damage
tion frequency can be at least 200-fold higher. Cell division (see Fig. 40.4), as they impose a delay in the cell cycle during
must not occur with inaccurately replicated or damaged which cells have a chance to repair their genomes. Promis-
genomes, as this may cause cell death or heritable mutation. ing new strategies for cancer treatment include use of agents
A number of systems have evolved to repair particular forms that actually cause DNA damage, with the goal of over-
of DNA damage sustained by the cell (Fig. 43.12). These whelming the already defective defenses in cancer cells, and
causing them to activate cell death pathways.

Base Excision Repair


Bases in DNA can become oxidized, reduced, alkylated, or
deaminated owing to endogenous activities or environmen-
A. Type of DNA B. Repair
lesion pathway tal stress. Damaged bases are cut away from the DNA sugar–
Base damage: phosphate backbone by a damage-recognizing glycosylase,
U deamination of leaving an abasic site (Fig. 43.13A). Abasic sites, which also
cytosine to uracil
can be generated directly by DNA damage, are then removed
Base by cleavage of the sugar–phosphate backbone mediated
excision by certain glycosylases and endonucleases. The missing
repair
sequence is then reconstructed from its complementary
G Base damage: strand by DNA polymerase β, with DNA ligase III-Xrcc1
methylation of
guanosine completing the repair by sealing the gaps in the backbone.

Nucleotide Excision Repair


Incorporation Mismatch
C of mismatched repair Bulky DNA adducts caused by chemical agents or environ-
A
base mental stress (particularly UV radiation from sunlight) are
excised by a complex, though well-understood, reaction
T Formation of UV Nucleotide (Fig. 43.13B). Defects in nucleotide excision repair genes
photoproduct: excision
6-4 thymidine cause the human genetic disease xeroderma pigmento-
dimer repair
T sum (XP), which is characterized by hypersensitivity to
sunlight and predisposition to skin cancer. Eight proteins
encoded by genes mutated in xeroderma pigmentosum
(Table 43.1) take part in nucleotide excision repair, provid-
3' OH Double-strand Double-strand ing one of the best examples in which human genetics has
break break repair
helped to unravel a complicated biological process. Recog-
3' OH • Homologous
recombinational nition of the DNA lesion involves the heterotrimeric rep-
G C repair lication protein RPA, XPA, and XPC, and the nine-subunit
• Nonhomologous
end joining transcription factor TFIIH, which contains XPB and XPD.
ATP-dependent unwinding of the DNA by XPB and XPD
forms a preincision complex. XPG, which replaces XPC
in the complex, makes an incision six to nine bases 3′ of
the damaged base, and XPF-Ercc1 cuts 20 to 25 bases 5′ of
DNA double the damage site. This releases a short single-stranded DNA
helix expanded fragment containing the damaged DNA. After excision, DNA
polymerases δ or ε fill in the gap by copying the undamaged
strand. Prokaryotes have a similar system of adduct recogni-
FIGURE 43.12  INTRODUCTION TO DNA REPAIR. Examples tion, removal, and repair involving the UvrA, UvrB, and UvrC
of DNA damage and the repair pathways that respond to different proteins; however, the enzymes that are involved are not
types of lesion. conserved between kingdoms.
CHAPTER 43  n  G2 Phase, Responses to DNA Damage, and Control of Entry Into Mitosis 751

BOX 43.1  DNA Repair in Vertebrates*—cont’d

A. Base excision repair B. Nucleotide excision repair C. Mismatch repair


DNA damage DNA damage Mismatch

RPA, XPA, XPC


Glycosylase Recognition TFIIH (contains XPB Recognition MSH2/MSH6 dimer
removes
damaged base and XPD)
Nick
Abasic site
ATP-dependent
unwinding by RPA, XPA, XPC, TFIIH Sliding clamp
DNA polymerase β XPB, XPD searches for nick Msh2/Msh6 dimer plus
DNA ligase III-Xrcc1 (Okazaki frag.) Mlh1/Pms2 dimer
form sliding clamp
Preincision
complex forms
May travel thousands
of bases
Exonuclease
XPF-Ercc1 nicks XPG nicks 6–9 bp degrades back Exo1
20–25 bp to 5' to 3' to mismatch

DNA polymerase δ, ε
DNA polymerase δ, ε

FIGURE 43.13  PATHWAYS FOR THE REPAIR OF BASE DAMAGE, BULKY ADDUCTS SUCH AS THYMIDINE DIMERS FORMED
BY ULTRAVIOLET LIGHT, OR MISMATCHED BASES. For detailed descriptions, see the text. The inset in panel A shows human
3-methyladenine DNA glycosylase complexed to DNA. This enzyme scans the DNA for bases that are not strongly H-bonded, uses its
“finger” to swing them up into the pocket for scanning, and, if they are damaged, catalyzes excision of that base. (Inset illustration by Graham
Johnson [www.fivth.com] for the Howard Hughes Medical Institute, copyright 2004, all rights reserved. For reference, see PDB file 1BNK
and Lau AY, Scharer OD, Samson L, et al. Crystal structure of a human alkylbase-DNA repair enzyme complexed to DNA: mechanisms for
nucleotide flipping and base excision. Cell. 1998;95:249–258.)

Mismatch Repair Double-Strand Break Repair


Errors in DNA replication missed by the proofreading activity DNA double-strand breaks can be caused by ionizing radia-
of the DNA polymerase are recognized by a dimer consisting tion or radiomimetic drugs or arise spontaneously after
of the MSH2 and MSH6 proteins. When a mismatch is replication. They are particularly hazardous forms of damage,
detected, this heterodimer undergoes an ATP-dependent as they carry the risk of losing chromosomal material or,
transition to a sliding clamp and recruits a second heterodi- if misrepaired, causing chromosomal translocations. Two
mer, consisting of MLH1 and PMS2 (Fig. 43.13C). To distin- major pathways repair double-strand breaks. Homologous
guish between the original (“correct”) sequence and the recombinational repair uses undamaged DNA as a template
newly synthesized DNA strand, this sliding clamp complex for the accurate repair of double-strand breaks, this sequence
can then translocate along the DNA until a break is reached, usually being derived from the sister chromatid after replica-
such as that found between Okazaki fragments. The broken tion. Nonhomologous end joining (NHEJ) repairs double-
strand is therefore identified as the newly synthesized DNA strand breaks with no requirement for homology. NHEJ is
strand. The mismatch repair complex then recruits the exo- the predominant activity that repairs double-strand breaks in
nuclease EXO1 and degrades the newly synthesized DNA the G1 and early S phase, whereas homologous recombina-
strand all the way back to the misincorporated base. The tion becomes more important in the late S and G2 phase.
resultant long, single-stranded region is stabilized by binding Homologous recombinational repair is normally extremely
of RPA and eventually filled in by the replicative DNA poly- accurate, but NHEJ often introduces errors, as it can join
merases δ and/or ε. A second type of “mismatch” involves both related and unrelated DNA ends together. Both path-
mistaken incorporation of ribonucleotides into DNA by ways require the activity of the MRN protein complex
either DNA polymerases δ and ε. Specialized ribonuclease H (Mre11/RAD50/Nbs1), which localizes to DNA double-strand
(RNAse H) enzymes detect and cleave DNA/RNA hybrids and breaks and is also found at telomeres. The exonuclease activ-
remove the ribonucleotides. ity of this complex resects (chews back) broken DNA ends

Continued
752 SECTION X  n  Cell Cycle

BOX 43.1  DNA Repair in Vertebrates*—cont’d

to provide single-stranded DNA substrates for the repair A number of proteins, including BRCA1 and BRCA2, control
systems. These mechanisms repair also repair double-strand the activity of mammalian Rad51 in homologous recombina-
breaks during genome editing (see Fig. 6.16). tional repair. Inactivation of BRCA1 and BRCA2 predisposes
The key protein required for homologous recombina- to cancer. The helicase Rad54 facilitates strand invasion,
tional repair in cells is Rad51, the eukaryotic homologue when the single-stranded region forces its way into the com-
of Escherichia coli RecA (Fig. 43.14A). Rad51 forms an plementary DNA duplex on the undamaged sister chromatid.
extended nucleoprotein filament on single-stranded DNA, Following invasion of the recombining DNA strands, poly-
replacing RPA, which acts like a placeholder when single- merase activity extends the DNA beyond the site of the
stranded DNA is produced. Rad51 catalyses the search for double-strand break, leading to the formation of a Holliday
homologous sequences, strand pairing, and strand exchange. junction (Fig. 43.14B). Resolution of the Holliday junction

A. Homologous recombinational repair C. Nonhomologous end joining


DNA break

Double-strand break
Binds ends
Recruits DNA- Ku70/
dependent Ku80 ring
protein kinase
5' to 3' resection MRN complex
Rad50/Mre11/Nbs1 MRN complex
5' 3'
Aligns broken ends Other repair factors

3' 5'
5' 3'

3' 5' DNA ligase IV/


Rad51 Ends sealed Xrcc4
Initial strand invasion
New DNA synthesis

B. Holliday junction structure


3' 5'
5'
Second end capture, = Resolvase cutting sites 3'
synthesis, ligation

Holliday
junction

Holliday junction resolution 3'


5' 3' 5'

5' 5' 3'


3' +
5' 3'
Repair complete
3' 5'

FIGURE 43.14  PATHWAYS FOR THE REPAIR OF DNA DOUBLE-STRAND BREAKS. A double-strand break is recognized by the
MRN complex, which recruits ATM (ataxia-telangiectasia mutated). The break is then bound by repair factors involved in either the homolo-
gous recombination or nonhomologous end-joining pathway of DNA repair. A, Homologous recombination pathway of DNA repair. The MRN
complex in conjunction with other nucleases chews back (resects) the DNA at a break, leaving a single-stranded overhang that is stabilized
by RPA (not shown). This recruits ATR (ataxia-telangiectasia and Rad3 related) to the damage site. It is believed that the MRN complex also
plays a role in keeping the broken ends close to one another. Next, RAD51 forms a nucleoprotein filament on the single-stranded DNA,
displacing the RPA. The Rad51 nucleoprotein filament then initiates homology searching and repairs the DNA break by inserting the extended
single-stranded DNA into homologous sequences (usually on the sister chromatid [blue]) and allowing homologous recombination and DNA
repair/resynthesis to occur. Capture of the second single-stranded DNA end allows the formation of a joint molecule with a double Holliday
junction. Resolution of this Holliday junction structure results in accurate, templated repair of the double-strand break. B, A Holliday junction
formed by four complementary oligonucleotides complexed to the enzyme Cre (not shown). The Holliday junction is a dynamic structure
(arrows) that can migrate along the DNA. C, Nonhomologous end-joining pathway of DNA repair. This pathway is initiated by break recogni-
tion by the Ku70/Ku80 heterodimer, which recruits DNA-PK and tethers the broken ends. The breaks are then processed in a reaction
involving the MRN complex and other repair factors. DNA-PK’s precise role is not yet entirely clear. Next, DNA ligase IV/XRCC4 is recruited
to the processed double-strand break, which is ligated back together. (B, For reference, see PDB files 3CRX and 2CRX and Gopaul DN,
Guo F, Van Duyne GD: Structure of the Holliday junction intermediate in Cre-loxP site-specific recombination. EMBO J 1998;17:
4175–4187.)
CHAPTER 43  n  G2 Phase, Responses to DNA Damage, and Control of Entry Into Mitosis 753

BOX 43.1  DNA Repair in Vertebrates*—cont’d

and filling-in of the repaired DNA sequences results in com- catalytic subunit of the DNA-dependent protein kinase,
plete repair of the lesion. One of two known human Holliday stimulating other repair factors and aligning the broken
junction resolvases is the Gen1 nuclease, a relative of the ends of the DNA (Fig. 43.14C). A complex of the protein
Fen1 flap endonuclease that functions during DNA replica- XRCC4 with DNA ligase IV seals the ends of the broken
tion (see Fig. 42.12). (The other more complex resolvase DNA. NHEJ is also necessary for V(D)J recombination
is the product of four genes.) Cells must have one or the and therefore for the development of the immune system
other of these enzymes or they die during mitosis because (see Fig. 28.10).
of an inability to separate DNA strands that have undergone Given the importance of accurate transmission of the
repair in the previous cell cycle. Note that Fig. 43.14 shows genetic material, deficiencies in DNA repair and checkpoint
only a subset of the proteins involved in homologous recom- genes are associated with a number of diseases (Table 43.1).
binational repair. It is likely that new factors remain to Note that several DNA repair activities are essential for
be identified. life, so their inactivation has not been described in any
NHEJ is initiated at a DNA double-strand break by binding human diseases.
of the Ku70 and Ku80 heterodimer as a ring that binds the

*By Ciaran Morrison, National University of Ireland, Galway.

Chk1 and Chk2. Phosphorylation of the ATM adapter, p53 stimulates transcription of the Cdk inhibitor p21.
53BP1 (p53-binding protein 1), recruits Chk2 for activa- Although p21 is best known for promoting cell-cycle
tion by ATM. The trimeric 9-1-1 complex is required arrest in G1 cells as part of the G1 DNA damage check-
for Chk1 activation by ATR. This complex, which gets point, it can also act in G2. Expression of p21 is an effec-
its name from its subunits Rad9, Hus1, and Rad1, resem- tive way of blocking the initiation of prophase, because
bles proliferating cell nuclear antigen (PCNA), the it inhibits Cdk1–cyclin A approximately 100-fold better
doughnut-shaped processivity factor that is indispens- than it inhibits Cdk1–cyclin B1.
able during DNA replication (see Fig. 42.12). PCNA is Active p53 also drives the expression of 14-3-3σ, an
loaded onto DNA by replication factor C (RFC, a penta- adapter protein that interferes with shuttling of Cdk1–
meric AAA-ATPase) and anchors DNA polymerases and cyclin B1 between the nucleus and cytoplasm (Fig.
other factors to DNA. A similar ATPase composed of one 43.11). Binding of 14-3-3σ maintains the Wee1 inhibitory
special subunit, Rad17, plus the four small subunits of kinase in a more active state, ensuring that the Cdk1–
RFC loads the 9-1-1 complex onto DNA at or near sites cyclin B1 complex remains inactive. Disruption of the
of damage. Mutants in those four RFC subunits are defec- gene for 14-3-3σ is fatal for cells with DNA damage.
tive in G2/M checkpoint control in yeasts, Drosophila Instead of activating their G2/M checkpoint, they enter
and Caenorhabditis elegans. RPA stimulates loading of an aberrant state with characteristics of both mitosis and
the 9-1-1 complex at damage sites, making it specific for apoptosis, and then die.
regions of single-stranded DNA. Typically, G2/M checkpoint activation has one of three
Phosphorylation releases Chk1 and Chk2 from chro- outcomes. If DNA damage is so extensive that it cannot
matin, so they can diffuse throughout the nucleus and be repaired, the cell either enters a non-proliferating state
cell to implement the DDR. They also trigger the G2/M known as senescence or commits suicide by apoptosis
checkpoint response by altering the cell cycle machinery (see Chapter 46). Less-serious damage can be repaired
and inducing the transcription of key EFFECTORS. In some by one of the systems described in Box 43.1.
cases their actions trigger cell death by apoptosis.
Activation of Chk1 is important to establish the G2/M
checkpoint response because Chk1 phosphorylates the
Transition to Mitosis
Cdc25A and Cdc25C protein phosphatases thereby The complex web of stimulatory and inhibitory activities
blocking cell-cycle progression (Fig. 43.11). Phosphory- in the G2 phase poises Cdk1–cyclin B in a state ready for
lation produces binding sites for a 14-3-3 protein that the explosive burst of activation that triggers the G2/M
blocks Cdc25A from activating Cdk1–cyclin B. Chk1 transition. These complex regulatory pathways are the
phosphorylation also targets Cdc25A for ubiquitin- basis of the G2/M checkpoint control that prevents cells
mediated proteolysis ensuring that levels of Cdc25A from segregating their chromosomes if genomic DNA
remain low. cannot meet stringent quality control standards. Eventu-
The transcription factor p53 (see Fig. 41.15), another ally, however, if all goes well, Cdk1–cyclin A and Cdk1–
EFFECTOR of the G2/M checkpoint, is phosphorylated and cyclin B1 are activated, and the cell embarks on mitosis,
activated following DNA damage (Fig. 43.11). Activated probably the most dramatic event of its life.
754 SECTION X  n  Cell Cycle

ACKNOWLEDGMENTS Morgan DO. The Cell Cycle: Principles of Control. London: New
Science Press; 2007.
We thank Anton Gartner, Ciaran Morrison, and Ashok Parrilla-Castellar ER, Arlander SJ, Karnitz L. Dial 9-1-1 for DNA damage:
Venkitaraman for their suggestions on revisions to this the Rad9-Hus1-Rad1 (9-1-1) clamp complex. DNA Repair (Amst).
chapter. 2004;3:1009-1014.
Paull TT. Mechanisms of ATM activation. Annu Rev Biochem. 2015;84:
711-738.
SELECTED READINGS Pearl LH, Schierz AC, Ward SE, et al. Therapeutic opportunities within
the DNA damage response. Nat Rev Cancer. 2015;15:166-180.
Ciccia A, Elledge SJ. The DNA damage response: making it safe to play Polo SE, Jackson SP. Dynamics of DNA damage response proteins at
with knives. Mol Cell. 2010;40:179-204. DNA breaks: a focus on protein modifications. Genes Dev. 2011;25:
Hartwell LH, Weinert TA. Checkpoints: Controls that ensure the order 409-433.
of cell cycle events. Science. 1989;246:629-634. Smits VA, Medema RH. Checking out the G(2)/M transition. Biochim
Jackson SP, Bartek J. The DNA-damage response in human biology and Biophys Acta. 2001;1519:1-12.
disease. Nature. 2009;461:1071-1078. Wieser S, Pines J. The biochemistry of mitosis. Cold Spring Harb Per-
Mehta A, Haber JE. Sources of DNA double-strand breaks and models spect Biol. 2015;7:a015776.
of recombinational DNA repair. Cold Spring Harb Perspect Biol. Wood RD, Mitchell M, Sgouros J, et al. Human DNA repair genes.
2014;6:a016428. Science. 2001;291:1284-1289.
Melo J, Toczyski D. A unified view of the DNA-damage checkpoint.
Curr Opin Cell Biol. 2002;14:237-245.
CHAPTER 44 

Mitosis and Cytokinesis

M itosis is the division of a somatic cell (a vegetative


Prophase Early prometaphase
cell in yeast) into two daughter cells. The daughters are CDK activity high
usually identical copies of the parent cell, but the process
can be asymmetrical. For example, division of some stem
cells gives rise to one stem cell and another daughter cell
that goes on to mature into a differentiated cell. See Box
41.2 for examples.
Traditionally, mitotic events are subdivided into six Late prometaphase Metaphase
phases: prophase, prometaphase, metaphase, ana-
phase, telophase, and cytokinesis (Fig. 44.1). The
dramatic reorganization of both the nucleus and cyto-
plasm during the mitotic phases is brought about by
activation of a number of protein kinases, including
Cdk1–cyclin B–cks (abbreviated here as “Cdk1 kinase”;
Anaphase Telophase
see Chapter 40). After activation by Cdc25 phosphatase,
APC/C activity high
Cdk1 kinase accumulates in the nucleus, where it joins
Cdk1–cyclin A–cks, which was activated somewhat
earlier (see Fig. 43.6). These two Cdk1 kinase complexes
operate as both master controllers and workhorses that
directly phosphorylate many proteins whose functional
and structural status is altered during mitosis. Their
Early cytokinesis Late cytokinesis
progressive inactivation following the correct attach-
ment of the chromosomes to spindle microtubules drives
the orderly exit of cells from mitosis.
Mitosis is an ancient process, and a number of varia-
tions emerged during eukaryotic evolution. Many single-
celled eukaryotes, including yeast and slime molds,
undergo a closed mitosis, in which spindle formation FIGURE 44.1  OVERVIEW OF THE PHASES OF MITOSIS.
and chromosome segregation occur within an intact APC/C, anaphase-promoting complex/cyclosome.
nuclear envelope to which the spindle poles are
anchored. This chapter focuses on open mitosis, as
Prophase
used by most plants and animals, in which the nuclear
envelope disassembles before the chromosomes segre- Prophase, the transition from G2 into mitosis, begins
gate. Fig. 44.2 summarizes some of the important events with the first visible condensation of the chromosomes
during the various mitotic phases. and disassembly of the nucleolus (Fig. 44.3). In the

755
756 SECTION X  n  Cell Cycle

A. Interphase B. Prophase C. Prometaphase D. Metaphase


Nucleus
Centrosome
Chromosomes
NE

Microtubules

Centrosomes separate Begins with nuclear Chromosomes align


Chromosomes condense envelope (NE) break-down on spindle equator
Chromosomes attach
to spindle

E. Anaphase A F. Anaphase B G. Telophase H. Cytokinesis

NE

CS

Midbody
CF CF CS remnant
CS
Pole
Sister chromatids separate Organized central spindle (CS) Cleavage furrow (CF) constricts Chromosomes decondense
and move to poles assembles Nuclear envelope (NE) Interphase microtubule
Poles (arrows) separate reassembles network reforms
Cleavage Furrow (CF)
assembles Daughter cells separate

FIGURE 44.2  KEY EVENTS OF MITOSIS. A–C, Prophase–prometaphase: Cdk1 kinase triggers condensation of replicated sister chromatids,
disassembly of the nuclear envelope and Golgi, and a dramatic reorganization of the cytoskeleton. As the barrier between the chromosomes and
cytoplasm is abolished, microtubules contact the condensed chromosomes and attach at the kinetochores (see Fig. 8.21). Interaction of kineto-
chores with dynamic microtubules culminates with the chromosomes aligned at the midplane of the bipolar spindle. D–F, Metaphase–anaphase:
Once all chromosomes achieve a bipolar attachment to the spindle, an inhibitory signal is silenced, leading to activation of a proteolytic network
that destroys proteins responsible for holding sister chromatids together and also inactivates Cdk1 by destroying its cyclin B cofactor (see Fig.
40.15). Sister chromatids separate and move toward opposite spindle poles, which themselves move apart. G–H, Telophase–cytokinesis: Targeting
of nuclear envelope components back to the surface of the chromatids leads to the reformation of two daughter nuclei. In most cells, the two
daughter nuclei and the surrounding cytoplasm are partitioned by cytokinesis. (Micrographs courtesy William C. Earnshaw.)

cytoplasm, the interphase network of long microtubules a century ago, the biochemical mechanism remains a
centered on a single centrosome (see Fig. 34.18) is mystery. Protein kinases trigger mitotic chromosome
converted into two radial arrays of short microtubules condensation and onset of condensation is correlated
called asters. Most types of intermediate filaments disas- with phosphorylation of histones H1 by Cdk1 kinase and
semble, the Golgi apparatus and endoplasmic reticulum H3 by Aurora-B protein kinase. However, chromosomes
fragment, and both endocytosis and exocytosis are still condense when both of these phosphorylation
curtailed. events are blocked. It is possible that a combination of
histone modifications promotes mitotic chromatin con-
Nuclear Changes in Prophase densation (see Fig. 8.3).
Chromosome condensation, the landmark event at the Two pentameric protein complexes, condensin I
onset of prophase, often begins in isolated patches of and condensin II are major regulators of mitotic chromo-
chromatin at the nuclear periphery. Later, chromosome some architecture. These complexes share the SMC2 and
condense into two threads termed sister chromatids SMC4 (structural maintenance of chromosomes) ABC
that are closely paired along their entire lengths. Although adenosine triphosphatases (ATPases), but have two dif-
chromosome condensation was first observed more than ferent sets of three auxiliary proteins (see Fig. 8.18).
CHAPTER 44  n  Mitosis and Cytokinesis 757

A. Prophase Chromosome B
condensation
Phosphorylated begins
condensin enters nucleus
Histone H3 Duplicated
phosphorylation centrioles begin
begins to separate
Cell surface Microtubule half-
markers life decreases
internalized and asters form
Intracellular membrane DNA
Cell begins to Microtubules
networks remodeled round up Centrosomes

FIGURE 44.3  INTRODUCTION TO PROPHASE. A, Summary of the major events of prophase. B, Distribution of DNA (blue), microtubules
(red), and γ-tubulin (centrosomes [green]) in a prophase human cell. (B, Images were recorded by Dr. Melpomeni Platani on the University of
Dundee’s School of Life Sciences Imaging Facility OMX 3DSIM Microscope and stored and processed in OMERO.)

Condensins are required to disassemble the topologically


TABLE 44.1  Comparison of Microtubule Dynamics
associating domains (TAD) of interphase chromatin (see
in Interphase and Mitotic Newt Lung Cells
Fig. 8.13) and promote the formation of the linear array
of loops that characterizes mitotic chromosomes (see Parameter Interphase Mitosis
Fig. 8.14). The decisive experiment was to target SMC2 Elongation rate 7 µm/min 14 µm/min
for rapid degradation, with the result that no well- Elongation time before 71 s 60 s
organized mitotic chromosomes formed. The chromatin catastrophe
compacted but was less organized. Slower depletion of Shortening rate 17 µm/min 17 µm/min
condensins by RNA interference (RNAi) or conditional Probability of rescue 0.046/s 0
gene disruption gave less-dramatic results. from catastrophe*
Condensin complexes can encircle DNA and also Length 100 µm 14 µm
promote its supercoiling in vitro, but how these activi- *Most cellular microtubules grow constantly by addition of subunits to
ties help them to orchestrate the changes in chromatin their free ends but they occasionally stop growing and begin shrinking
rapidly (a “catastrophe”). Unless shrinking is reversed (a “rescue”), the
architecture is not known. Condensin II is nuclear during microtubule completely disappears.
interphase, and has an important role in prophase chro- Data from Gliksman NR, Skibbens RV, Salmon ED: How the transition
mosome formation following its activation by phos- frequencies of microtubule dynamic instability regulate microtubule
dynamics in interphase and mitosis. Mol Biol Cell. 1993;4:1035–1050.
phorylation. Condensin I acts both in prophase and
prometaphase in further compacting the chromosomes.
DNA topoisomerase II and other scaffold proteins also properties of the microtubules (Table 44.1; also see
function during mitotic chromosome formation, but Chapter 34). Interphase microtubules have a high prob-
condensin appears to play the major role. ability of recovering from catastrophes, so they grow
quite long. Mitotic microtubules grow more rapidly
Cytoplasmic Changes in Prophase but exist only transiently, because rescues are rare fol-
Most of the cytoskeleton reorganizes during prophase. lowing a catastrophe. Thus, they usually shorten all
The microtubule array changes from an extensive the way back to the centrosome, with little chance of
network permeating the cytoplasm into two dense, rescue. These differences in dynamic instability can be
radial arrays of short, dynamic microtubules around the reproduced in vitro in mitotic and interphase cellular
duplicated centrosomes (see Fig. 34.17). Each of these extracts. They appear to arise, at least in part, from
asters eventually becomes one pole of the mitotic counterbalancing interactions between microtubule-
spindle. During prophase, the two asters usually migrate associated proteins that promote microtubule stability,
apart across the surface of the nuclear envelope, signal- and kinesin-13 (see Fig. 36.13), which promotes micro-
ing the start of spindle assembly (Fig. 44.3). tubule disassembly.
Mitotic microtubules behave like interphase microtu- Other cytoskeletal elements that disassemble during
bules in many ways (see Chapter 34). They are mostly prophase include many, but not all, classes of interme-
nucleated at their minus ends, they grow by addition of diate filaments (including the nuclear lamins) (see
tubulin subunits at their plus ends, and they undergo Fig. 35.1A) and specialized actin filament structures,
random catastrophes during which they rapidly shorten. such as stress fibers. However, the junctional complexes
To a large extent, the prophase changes in microtubule between adjoining cells are maintained in epithelial
organization can be explained by two simple biochemi- cells. As a result of the cytoskeletal reorganization, most
cal changes: (a) increased microtubule-nucleating activ- cells round up during prophase. This is particularly
ity of centrosomes and (b) altered dynamic instability evident for animal cells that are cultured on a flat
758 SECTION X  n  Cell Cycle

substrate, but cells in tissues also change their shape bly of new ribosomes. Phosphorylation of several nucleo-
dramatically during mitosis. lar proteins leads to disassembly of the nucleolus.
RNA transcription of the chromosomes stops during The Golgi apparatus and endoplasmic reticulum frag-
mitosis except for highly specialized transcription at ment and disperse during prophase (Fig. 44.4). Several
centromeres. Phosphorylation of components of the kinases, including Cdk1 drives Golgi apparatus disas-
transcriptional machinery by Cdk1 kinase appears to be sembly, the first step being fragmentation into smaller
responsible for this shutoff. Cdk1 kinase phosphoryla- ministacks following phosphorylation of Golgi stacking
tion of ribosomal elongation factor 2a (EF2a) also stops proteins and tethers. Later steps are still being investi-
most (but not all) ongoing protein synthesis and assem- gated. Many lines of evidence argue that Cdk1 phos-
phorylation of key components prevents the fusion of
transport vesicles back into Golgi stacks (see Chapter
21), the net result being that the Golgi buds away into
Interphase Mitosis
small vesicles that disperse throughout the mitotic cell
cytoplasm. Other evidence suggests that an imbalance of
vesicle flow between the Golgi and the endoplasmic
Golgi Ministacks reticulum results in the Golgi being absorbed into the
Golgi stacking GM130 endoplasmic reticulum during mitosis. Whatever the
proteins Cdk1–
cyclin B–p9 mechanism of its disassembly, Golgi reassembly begins
p115
again during late anaphase/early telophase, following
inactivation of Cdk1 kinase.

Prometaphase
In cells that undergo an open mitosis, prometaphase
begins abruptly with disassembly of the nuclear envelope
FIGURE 44.4  GOLGI APPARATUS DYNAMICS IN INTER- (Fig. 44.5). Microtubules growing outward from the
PHASE AND MITOSIS. Disassembly in mitosis is driven by phos- spindle poles penetrate holes in the nuclear envelope,
phorylation of components blocking fusion of Golgi membranes. make contact with the chromosomes, and attach to them

A. Early prometaphase C. Late prometaphase E. Kinetochore attachments


to the spindle
2 4
1 Tension

3 5
(+) ends

1. Nuclear envelope disassembles 4. Chromosome slides rapidly poleward Amphitelic (correct)


along microtubule
2. Microtubules grow and shrink in aster 5. Microtubule from opposite pole is
3. Kinetochore captures microtubule captured by sister kinetochore
6. Chomosome attached to both poles
congresses to middle of spindle

B D
Syntelic (errors)

DNA
Microtubules
Centrosomes Merotelic (errors)

FIGURE 44.5  INTRODUCTION TO PROMETAPHASE. A, Summary of the key events of early prometaphase. B, Distribution of DNA (blue),
microtubules (red), and γ-tubulin (centrosomes [green]) in early prometaphase human cells. C, Summary of the key events of late prometaphase.
D, Distribution of DNA, actin, microtubules, and centrosomes in late prometaphase PtK1 cells. E, Terms used to describe the orientation of
kinetochore attachments to the mitotic spindle. (B and D, Images were recorded by Dr. Melpomeni Platani on the University of Dundee’s School
of Life Sciences Imaging Facility OMX 3DSIM Microscope and stored and processed in OMERO.)
CHAPTER 44  n  Mitosis and Cytokinesis 759

at specialized structures called kinetochores (see Fig. as an extensive tubular (or flattened cisternal network—
8.19). Interactions of the two opposing kinetochores of another source of discussion) throughout mitosis.
paired sister chromatids with microtubules from oppo- Further experiments are required to answer this ques-
site poles of the spindle ultimately result in alignment of tion, and both mechanisms could contribute. Lamin B
the chromosomes in a group midway between the poles. remains associated with the dispersed nuclear envelope,
An important cell-cycle checkpoint (see Chapter 40) whereas lamins A and C and many proteins of the nuclear
known as the spindle assembly checkpoint (SAC) pore complexes disperse as soluble subunits.
delays the onset of chromosome segregation until all During prophase, kinetochores transform from non-
kinetochores are attached to microtubules. descript balls of condensed chromatin into structures on
the surface of the chromosomes. By early prometaphase,
Nuclear Envelope Disassembly in Prometaphase the characteristic trilaminar disk structure (see Fig. 8.19)
Nuclear envelope disassembly involves the removal of can be seen. Each sister chromatid has a kinetochore.
two membrane bilayers coupled with disassembly of the Sister kinetochores are located on opposite faces of
nuclear pores and the fibrous nuclear lamina mesh- the mitotic chromosome.
work that underlies the inner bilayer (Fig. 44.6). Phos-
phorylation causes the nucleoporin Nup98 to dissociate Organization of the Mitotic Spindle
from nuclear pores. This removes the permeability The mature metaphase spindle is a bilaterally sym-
barrier between nucleus and cytoplasm. Phosphoryla- metrical structure with centrally located chromosomes
tion of other proteins causes the pore to disassemble to flanked by arrays of microtubules radiating from the
soluble subcomplexes. Phosphorylation of the nuclear poles (Fig. 44.7).
lamins at two sites flanking the coiled-coil causes the Three predominant classes of microtubules are
lamina network to disassemble into subunits. Interaction present in the metaphase spindle (Fig. 44.12). Kineto-
between microtubules and dynein associated with the chore microtubules have their plus ends embedded in
nuclear envelope can rip holes in the envelope, although the kinetochore and their minus ends at or near the
this is not required for nuclear envelope disassembly. spindle pole. They characteristically form bundles, called
Nuclear envelope membranes are dispersed in the kinetochore fibers, which contain anywhere from 1
cytoplasm from prometaphase until telophase (Fig. microtubule in the budding yeast to more than 200
44.6), but the mechanism is not settled. Some experi- microtubules in some higher plants. Each human kineto-
ments suggest that the nuclear membranes break up into chore binds approximately 20 microtubules. Up to
small vesicles that disperse in the cytoplasm. Other approximately 80% of the approximately 2200 spindle
experiments suggest that the nuclear envelope is microtubules in humans may be present in kinetochore
absorbed into the endoplasmic reticulum, which remains fibers, but not all microtubules in those fibers stretch all

A. Interphase B. Mitosis C D. Lamins A and C E. Lamin B


Nuclear Endoplasmic
membrane: reticulum
Outer
Inner
HO PO4– O PO4– O
Lamina
Interphase O O
disassembly C C
OCH3 O–
Nuclear
lamina Cdk1– G2 / M G2 / M
cyclin B–p9
Condensing
chromosomes
PO4 PO4
O O
Eggs? Somatic C C
cells? Earliest OCH3 O–
prometaphase OH O PO4– OH O PO4–

Lamin B

+ or

Chromosomes Vesicles derived Lamin B


from the nuclear dispersed through the Soluble Subunits on
Metaphase Lamina Lamina
envelope endoplasmic reticulum subunits membrane vesicles

FIGURE 44.6  DISASSEMBLY OF THE NUCLEAR ENVELOPE DURING MITOSIS. A–B, Two contrasting models to explain the fate of the
nuclear envelope during the transition from interphase to mitosis in a higher eukaryote. C, Micrographs showing solubilization of lamin A fused
to green fluorescent protein (GFP) (green) during mitosis. DNA is blue. Scale bar is 10 µm. D–E, Reversible disassembly of lamins A, C, and B
is driven by posttranslational modifications of the lamin polypeptides. (C, Courtesy William C. Earnshaw.)
760 SECTION X  n  Cell Cycle

A. Metaphase–forces balanced, spindle length stable B. Anaphase–forces elongating the spindle dominate

Cytoplasmic dynein moves


toward minus (–) end
Microtubule disassembly at kinetochores
Microtubule (+)
plus flux moves sister chromatids toward poles
(+) (+)
(+) (+)
(+) Flux
(+) (+)

(+)
Ordered central
(+) spindle assembles
(–) (–) (–) (+) (–) (+)
(–) (–) (–) (–)
(–) (–)
(–) (–) (–) (–) (–)
(+) Kinesin-13
(+)
(+) (–)
(+) (–)
(+)

(–) (+)
(–) (+) NuMA
Bipolar plus-end–directed kinesin-5 dominates
Inward force from minus-end–directed kinesin-14 and microtubule to elongate spindle, pushing poles apart
disassembly at poles by kinesin-13 is balanced by outward force
from kinesin-5 plus dynein stretching poles apart
Microtubule assembly at kinetochores and disassembly at poles causes tubulin subunit flux along kinetochore microtubules

FIGURE 44.7  ROLE OF MOTOR PROTEINS IN SPINDLE DYNAMICS. Mitotic spindle structure depends on microtubule assembly/
disassembly plus balanced forces that slide microtubules relative to one another and to pull the poles together or push them apart. A, In
metaphase, the structure is at steady state. Forces that tend to elongate the spindle, including cytoplasmic dynein (which moves toward
microtubule minus ends, pulling the poles out toward the cell cortex) and bipolar kinesin-5 (which moves toward microtubule plus ends, pushing
the poles apart), are counterbalanced by cohesion between sister chromatids and kinesin-14, which moves toward microtubule minus ends (and
pulls the poles together) and microtubule disassembly at the spindle poles. Dynein and its associated protein, NuMA (nuclear mitotic apparatus),
also help to organize a focused spindle pole. B, In anaphase, sister chromatids separate, the balance of kinesin activity shifts, microtubule disas-
sembly at the poles declines, and the spindle undergoes a dramatic elongation. During anaphase, bipolar kinesin-5, chromokinesin KIF4A, and
protein regulated in cytokinesis 1 (PRC1) also have important roles in organizing the central spindle, which is essential for subsequent assembly
and function of the cleavage furrow.

the way from the kinetochores to the spindle poles. microtubules. As a result, the highly dynamic spindle
Interpolar microtubules are distributed throughout changes shape as a delicate balance of forces shifts
the body of the spindle and do not attach to kineto- between the various motors. For example, inactivating
chores. Many interpolar microtubules penetrate between one or more kinesins with drugs or switching a
and through the chromosomes and extend for some temperature-sensitive mutant to the nonpermissive
distance beyond them. Thus, the central spindle contains temperature can cause the spindle to collapse rapidly
a large number of interdigitated antiparallel microtu- on itself.
bules. Tracking these spindle microtubules by electron
microscopy revealed a tendency for the interdigitated Spindle Assembly
microtubules of opposite polarity to pack next to one In metazoans, spindle assembly starts in prophase with
another. During late anaphase, these antiparallel micro- the separation of the asters. In most cells, each aster is
tubules bundle to form a structure called the central organized around a centrosome, consisting of a centriole
spindle that plays important roles in signaling during pair and associated pericentriolar material. γ-Tubulin
cytokinesis. Astral microtubules project out from the ring complexes in the pericentriolar material efficiently
poles and have a role in orienting and positioning the nucleate microtubules (see Fig. 34.16), so each centro-
spindle in the cell through interactions with the cell some acts as a microtubule organizing center
cortex in somatic cells. All microtubules within each (MTOC). By the end of prophase, the spindle consists of
aster have the same polarity, with their minus ends two asters linked by a few interpolar microtubules.
proximal to the pole. Each unit of a spindle pole, with Cytoplasmic dynein at the cell cortex exerts an outward
its associated kinetochore and interpolar and astral force separating the centrosomes, whereas kinesin-14
microtubules, is referred to as a half-spindle. motors (which move toward microtubule minus ends)
Spindle structure is largely determined by a combina- on the interpolar microtubules exert a counterbalancing
tion of microtubule dynamics plus the action of at least force holding the asters together.
seven different types of kinesins and cytoplasmic dynein This balance of forces changes when the nuclear
(see Chapter 36). These motors often work in opposition envelope breaks down. Bipolar kinesin-5 motors phos-
to one another. Furthermore, forces exerted by motors phorylated by Cdk1 kinase concentrate in the central
can influence the dynamic assembly/disassembly of spindle, where they crosslink adjacent antiparallel
CHAPTER 44  n  Mitosis and Cytokinesis 761

interpolar microtubules. Kinesin-5 moves toward the enable chromosomes to control the activity of key
plus ends of microtubules, so when attached to two spindle assembly factors. The nuclear import receptors
adjacent antiparallel microtubules, its action will cause importin α and β bind these factors, as though they were
them to slide and push the spindle poles apart (Fig. going to transport them into the nucleus. This blocks
44.7). However, the two half-spindles do not separate their spindle assembly activity. During mitosis, chromo-
because they are physically linked via the chromo- somes bind high levels of RCC1, the guanine exchange
somes, with sister kinetochores attached to opposite factor for Ran (Ran-GEF in Fig. 9.18). This RCC1 creates
spindle poles. a gradient of the active guanosine triphosphatase
Also at this time, the asters mature into focused (GTPase) Ran-GTP (guanosine triphosphate) around the
spindle poles. The pericentriolar material efficiently chromosomes. During interphase, Ran-GTP is confined
nucleates the assembly of new microtubules with their to the nucleus, where it releases importins from their
minus ends at the pole. Microtubule assembly at two cargo. In mitosis the chromosome-associated cytoplas-
other sites also contributes to spindle morphology. At mic Ran-GTP gradient locally liberates the spindle assem-
kinetochores, a complex derived from interphase nuclear bly factors including motor proteins and NuMA from
pores recruits γ-tubulin. This locally nucleates microtu- sequestration by importins. They then stabilize nearby
bules that grow by inserting subunits at the kinetochores, microtubules and organize them into a bipolar spindle.
pushing the minus ends with γ-tubulin outwards toward If centrosomes are removed or destroyed experimen-
the spindle poles. These preformed kinetochore fibers tally in cells about to enter mitosis, somatic cells can also
are then incorporated into the spindle. In the central use motor proteins to organize microtubules into bipolar
spindle, microtubules are nucleated on the walls of other spindles that lack asters but are otherwise remarkably
microtubules by γ-tubulin recruited by the multi-subunit normal. Most treated cells manage to complete mitosis
augmin complex. The action of augmin creates branched successfully but normal mammalian cells then either
microtubules throughout the central spindle, contribut- arrest in the next cell cycle prior to replicating their DNA
ing to an even fir tree-like distribution of microtubules. or commit suicide. Both outcomes depend on the pres-
The daughter microtubules may be released from their ence of the important tumor suppressor protein p53 (see
nucleation site, free at both ends, and move toward the Fig. 43.11). Thus, centrosomes are not required to form
spindle poles as part of the flux of tubulin from the spindles, but they contribute to cell-cycle progression in
center of the spindle toward the centrosomes. many cells. This dependence on centrosomes is not
The microtubule array focuses at the poles partly universal; Drosophila, for example, can live without
because centrosomes tether microtubules, and partly centrosomes.
due to the concerted action of various motors and micro-
tubule crosslinking proteins such as dynein and nuclear Chromosome Attachment to the Spindle
mitotic apparatus (NuMA) protein. NuMA is released Dynamic microtubules of prometaphase asters scan the
from the nucleus when the nuclear envelope breaks cytoplasm effectively “searching” for binding sites that
down, and it accumulates near the poles at the minus will capture and stabilize their distal plus ends. Captured
ends of microtubules. microtubules are approximately fivefold less likely to
Large cells that lack centrosomes, such as eggs, form depolymerize catastrophically than free microtubules.
spindles by an alternative pathway that also functions in When catastrophes do occur, the microtubules depoly-
the background in cells with centrosomes (Fig. 44.8). merize back to the pole, recycling tubulin subunits for
This mechanism hijacks the nuclear trafficking system to incorporation into other, growing microtubules.

Unstable microtubule
g radient
TP Microtubule
G
stabilized by
n-
Ra

Ran-GTP
gradient

Stabilized Motors sort Other motors and


microtubules microtubules, (–) end-binding proteins
accumulate on which bind to organize spindle poles
chromosomes kinetochores (note absence of asters)

FIGURE 44.8  ASSEMBLY OF A BIPOLAR SPINDLE IN THE ABSENCE OF CENTROSOMES. A gradient of Ran-guanosine triphosphate
(GTP) stabilizes microtubules around chromosomes, which contain high concentrations of bound Ran GEF RCC1. This releases spindle assembly
factors from importin α and β. Microtubules that accumulate around the chromosomes are sorted, organized, and focused to make poles by
motors and (−) end-binding proteins such as NuMA. These spindle poles lack prominent astral microtubules.
762 SECTION X  n  Cell Cycle

Breakdown of the nuclear envelope makes the con- chromosome toward the middle of the spindle. These
densed chromosomes accessible to the microtubules. movements are accompanied by coordinated shrinkage
Chance encounters allow kinetochores to capture of the microtubules at the leading kinetochore and
microtubule plus ends. Capture probably involves the growth of microtubules at the trailing kinetochore.
nine-component KMN network, which includes the rod- More recent studies revealed that chromosomes
shaped Ndc80 complex (see Fig. 8.21) that binds along attached to only one spindle pole can move away from
the sides of microtubules near their plus ends. Another that pole if the unattached kinetochore associates with
member of the complex, the scaffolding protein Knl1 the kinetochore fiber of a chromosome already aligned
(its name in vertebrates—the “K” of KMN; see later), at the spindle equator. In this case, the kinetochore of
anchors Ncd80 in the kinetochore. the mono-oriented chromosome glides toward the
Historically, it was thought that forces generated by equator, where it is more likely to capture microtubules
bipolar attachment of the kinetochores of sister chro- emanating from the opposite pole. This motion of one
matids center chromosomes midway between the two chromosome along the kinetochore fiber of another
spindle poles. This hypothesis was based on the observa- chromosome requires the kinesin-7 motor centromere
tion that when a kinetochore first attaches to a microtu- protein E (CENP-E) (see Fig. 36.13) associated with the
bule, the chromosome moves along the side of that kinetochore of the moving chromosome. Recognition of
microtubule toward the spindle pole (Fig. 44.9). Subse- a tubulin posttranslational modification leads CENP-E to
quent capture of a microtubule emanating from the move the chromosome toward the spindle equator,
opposite spindle pole by the sister kinetochore would rather than out into the aster.
provide a counterforce pulling the chromosome in the The attachment of microtubules to kinetochores can
opposite direction. Chromokinesin family motor proteins be reconstituted in vitro from mixtures of chromosomes,
distributed along the chromosome arms were also isolated centrosomes, and tubulin subunits. The plus
thought to contribute to the gradual movement of the ends of microtubules grow out from centrosomes and

A E
Spindle
pole
Spindle pole
4 Microtubule
Kinetochore
Distance traveled toward

3
spindle pole (µm)

B 1

–1 Start of
movement
–2
0 20 40 60 80
Time (seconds)

C D
Kinetochore Microtubule
10 µm

FIGURE 44.9  INITIAL CHROMOSOMAL MOVEMENTS DURING PROMETAPHASE. A–B, Capture of a microtubule by the kinetochore
results first in movement along the side of the microtubule toward the pole from which that microtubule originated. These images come from a
study in which living cells, observed by differential interference microscopy, were subjected to rapid chemical fixation just after a chromosome
had attached to the spindle (arrow). C, Attachment of the chromosome to the spindle was confirmed by indirect immunofluorescence staining
for tubulin and thin-section electron microscopy (D). E, The graph shows the movements of the chromosome before and after attachment. (From
Rieder CL, Alexander SP, Rupp G. Kinetochores are transported poleward along a single astral microtubule during chromosome attachment to
the spindle in newt lung cells. J Cell Biol. 1990;110:81–95, copyright The Rockefeller University Press.)
CHAPTER 44  n  Mitosis and Cytokinesis 763

attach to the chromosomes. Surprisingly, chromosome- Chromosome Attachment Errors: The Spindle Assembly
bound microtubules can either lengthen or shorten at Checkpoint” below) delays mitotic progression to allow
the attached end without detaching from the chromo- the correction process to occur.
some. Similar experiments with kinetochores isolated When syntelic attachments occur, one or both kineto-
from budding yeast cells showed that kinetochores can chores must detach for the chromosome to achieve a
remain attached to a shortening microtubule plus end bipolar orientation. Chromosome attachment to oppo-
even against an applied force of 9 pN (piconewtons). site spindle poles is more stable than attachment to a
Physiological levels of tension actually stabilize the single pole, because the tension generated by bipolar
attachments of kinetochores to microtubules in vitro, as attachment (where forces pull a chromosome simultane-
in vivo. This tethering of kinetochores to disassembling ously toward opposite spindle poles) preferentially sta-
microtubules is essential for chromosome movements bilizes microtubule connections to both kinetochores.
during mitosis. Merotelic attachments are more dangerous, as the kineto-
chore is under tension and the attachments are therefore
Correcting Errors in Chromosome Attachment to stable. Merotelic attachments are the most common
the Spindle cause of chromosome segregation errors in cultured
The goal of mitosis is to partition the replicated chromo- mammalian cells.
somes accurately between two daughter cells. Therefore, Syntelic and merotelic chromosome attachments are
all chromosomes must attach correctly to both spindle corrected through the action of Aurora B protein kinase,
poles (known as amphitelic attachment; Fig. 44.5) which forms the chromosomal passenger complex
before being segregated. Three other sorts of attachment (CPC), along with inner centromere protein (INCENP),
are seen: (a) chromosomes with one kinetochore lacking survivin, and borealin (Fig. 44.10). The other subunits
attached microtubules (known as monotelic attach- target Aurora B to its various sites of action during mitosis
ment; this is a normal intermediate), (b) chromosomes and regulate the kinase activity. The complex concen-
with both sister kinetochores attached to the same trates at inner centromeres (the heterochromatin beneath
spindle pole (known as syntelic attachment), and (c) and between the two sister kinetochores) during pro-
chromosomes with a single kinetochore attached simul- metaphase and metaphase. At anaphase onset, the CPC
taneously to both spindle poles (known as merotelic moves to the overlapping interpolar microtubules of the
attachment). Correcting monotelic and syntelic errors central spindle and to the cell cortex, where the cleav-
takes time, and the SAC (see “Finding Time to Fix age furrow will form, ultimately winding up in the

A Borealin
B C

DNA
Survivin
Microtubules

D E. CPC Aurora-B F
INCENP

Borealin

Survivin

Histone H3 phosphorylation
Kinetochore targets
Central spindle targets
Cleavage furrow targets

FIGURE 44.10  CHROMOSOMAL PASSENGER COMPLEX (CPC) REGULATES MITOTIC EVENTS. The CPC (green) is present at
centromeres in prometaphase and metaphase (B), but transfers to the spindle midzone at anaphase (C) and midbody at anaphase (D).
E, Diagram of Aurora B protein kinase complexed with INCENP (inner centromere protein), survivin, and borealin with some key targets of the
CPC. F, If CPC function is inhibited (in this case by RNA interference [RNAi] depletion of borealin), chromosome attachment errors are common
and many chromosomes fail to segregate properly in anaphase. Distribution of DNA (blue), microtubules (red), and survivin–green fluorescent
protein (GFP) (green) in human mitotic cells. Inset in A, Distribution of kinetochores (red), and borealin (green) in a prometaphase cell.
(A–D, Micrographs by Sally Wheatley and William C. Earnshaw. F and Inset in A, Micrographs by Ana Carvalho, Reto Gassmann, and William
C. Earnshaw. Insets in A and F, From Gassmann R, Carvalho A, Henzing AJ, et al. Borealin: a novel chromosomal passenger required for stability
of the bipolar mitotic spindle. J Cell Biol. 2004;166:179–191, copyright The Rockefeller University Press. A–C, From Wheatley SP, McNeish IA.
Survivin: a protein with dual roles in mitosis and apoptosis. Int Rev Cytol. 2005;247:35–88.)
764 SECTION X  n  Cell Cycle

intercellular bridge during cytokinesis. The CPC regu- lose a chromosome only once in 100,000 cell divisions.
lates mitotic events from prophase through cytokinesis. The frequency of chromosome loss may be 20-fold to
Along the way it contributes to the correction of 400-fold higher for human cells grown in culture. To
chromosome attachment errors and to the operation of achieve even this level of accuracy, most cells delay
the checkpoint that delays the cell cycle in response to entry into anaphase until all chromosomes have achieved
those errors. amphitelic attachment to the spindle. This delay is
Aurora B corrects chromosome attachment errors by caused by the spindle assembly checkpoint (SAC), which
phosphorylating Ndc80 in the microtubule-binding KMN senses the completion of microtubule binding to kineto-
complex (see Fig. 8.21). Aurora B phosphorylation chores at metaphase (Fig. 44.11). The spindle checkpoint
strongly inhibits Ndc80 binding to microtubules, causing differs from DNA damage checkpoints in that its default
the kinetochore to release attached microtubules. When setting is “on” as cells enter mitosis. It is silenced only
a chromosome is correctly attached to both spindle when every chromosome is properly attached to the
poles, tension stretches the kinetochore away from the spindle.
CPC buried in the chromatin beneath. This can stabilize The SAC involves the products of the mitotic arrest–
chromosome–microtubule interactions by preventing defective (MAD) genes, the budding-uninhibited-by-
the kinase from phosphorylating Ndc80. benzimidazole (BUB) genes, the monopolar spindle
(Mps1) kinase and the CPC. The MAD and BUB genes
Finding Time to Fix Chromosome Attachment Errors: were identified in yeast genetic screens for cells that
The Spindle Assembly Checkpoint continued to divide (and die) when the spindle was
Segregation of replicated chromosomes into daughter disassembled by drugs. These genes are conserved
cells is extremely accurate. For example, budding yeasts from yeast to humans. SAC proteins accumulate at

A D

C
23 minutes
Time

B
Seat
belt
N

Time Mad1 • Mad2


Destroy free kinetochore 23 minutes
with blast of laser light
APC/C
substrate

C
APC/C APC/C
it

Go
Wa

MCC Cdc20APC/C
Kinetochore•
Mad1 • Mad2 Mad2
APC/C
Microtubule Cdc20APC/C

Checkpoint proteins
not on kinetochore BubR1

Cdc20MCC U Ub Ub
Go! Wait! Ub b

Mad2 MCC
Proteins not to scale relative to kinetochore Substrate

FIGURE 44.11  SPINDLE CHECKPOINT. Signaling by unattached kinetochores stops the cell from entering anaphase until all chromosomes
have made a proper bipolar spindle attachment. A, As long as there is a chromosome that is not properly attached to the spindle (beige cells),
the cell does not enter anaphase. The cell enters anaphase approximately 20 minutes after chromosome attachment is complete (green cells).
B, In a cell with a persistently maloriented chromosome, anaphase entry is delayed (beige cells). If the unattached kinetochore is destroyed with
a high-powered laser, the cell enters anaphase about 20 minutes later. This proves that the unattached kinetochore sends an inhibitory signal.
C, Overview of the spindle assembly checkpoint (see text for details). D, Structure of the Mad1/Mad2 complex.
CHAPTER 44  n  Mitosis and Cytokinesis 765

kinetochores early in mitosis, when the checkpoint is Silencing of the checkpoint involves several pathways.
“on” (ie, during prophase or prometaphase), and are Protein phosphatase 2A (PP2A) and protein phosphatase
gradually displaced as microtubules bind and the kineto- 1 (PP1) are recruited to the kinetochore in feedback
chores come under tension. loops involving the CPC and other checkpoint compo-
The target of the SAC is the APC/CCdc20, the anaphase- nents. They dephosphorylate Knl1 so that it releases
promoting complex/cyclosome (APC/C) ubiquitin- Mad1. When microtubules bind, cytoplasmic dynein
protein ligase (an E3 enzyme; see Figs. 23.3 and 40.16) motors actively strip checkpoint components from the
with its substrate recognition factor Cdc20 bound, APC/ kinetochore, dragging them away toward the centro-
CCdc20 ubiquitylates target proteins to mark them for somes. In yeast, access of Mps1 to its target sites on
destruction by proteasomes. Key APC/CCdc20 substrates Knl1 is physically blocked when microtubules bind.
are proteins that must be degraded for the cell to move Exactly how this interaction is regulated in metazoans is
from metaphase to anaphase, including cyclin B and still actively studied. In addition, the SAC appears to
securin, an inhibitor of the enzyme that triggers separa- crosstalk with the DNA damage response (see Chapter
tion of sister chromatids at anaphase (Fig. 44.16). 43), since DNA damage response components activate
During mitosis, Cdk1 activates the APC/C by phos- SAC components and vice versa. However, the network
phorylating an auto-inhibitory loop, allowing Cdc20 to of interactions is very complex and details are still being
bind. The SAC is an additional regulatory circuit that worked out.
inactivates APC/CCdc20 until all kinetochores attach to Experimental inactivation of the spindle checkpoint
spindle microtubules. Kinetochores without microtubules causes a catastrophic, premature entry into anaphase,
attract proteins that assemble the mitotic checkpoint regardless of the status of chromosome alignment. This
complex (MCC), the inhibitor that inactivates APC/CCdc20. leads to an unequal distribution of sister chromatids and
Checkpoint activation starts when Aurora B in the genetic imbalance between daughter cells known as
CPC activates Mps1 kinase, allowing it to phosphorylate aneuploidy. Yeasts can live without the checkpoint
Knl1 in the kinetochore at several sites. Mps1 phos- genes, but their loss is lethal for mice, which die early
phorylation of Knl1 creates a binding site that results in during embryogenesis. Mice heterozygous for various
Mad1 recruitment to the kinetochore. Mad1 then recruits checkpoint components show increased aneuploidy.
Mad2 to form a stable complex (Fig. 44.11). A loop on Humans with mutations in BubR1 have mosaic variegated
Mad2 wraps around Mad1 like a safety belt making aneuploidy syndrome (extra copies or loss of various
the complex particularly stable. This form of Mad 2 is chromosomes in a variety of tissues), which is associated
known as “closed” Mad2. Mad1/Mad2 can transiently with microcephaly (decreased brain size) and an
bind soluble Mad2 molecules (known as “open” Mad2), increased cancer risk.
load them onto Cdc20 in the closed safety belt conforma-
tion (this loading probably occurs at kinetochores), then
release them to form the soluble MCC of Mad2/Cdc20/
Metaphase
BubR1/Bub3. The MCC associates with the APC/CCdc20, When all the chromosomes have attained amphitelic
interfering with binding of cyclin B and other key sub- orientations and moved to positions roughly midway
strates. As each chromosome becomes attached to both between the two spindle poles, the cell is said to be in
poles of the spindle it stops producing MCC. When the metaphase (Fig. 44.12). The compact grouping of chro-
last chromosome has achieved a proper attachment, the mosomes at the middle of the spindle is referred to as
last source of MCC is extinguished, and entry into ana- the metaphase plate. In many cells, even though chro-
phase can proceed. mosomes remain, on average, balanced at the middle of

A. Metaphase B
Kinetochore
microtubules

Astral
microtubules

Interpolar
microtubules DNA
Cyclin B and Microtubules
securin degraded Chromosomes oscillate Centrosomes

FIGURE 44.12  INTRODUCTION TO METAPHASE. A, Summary of the major events of metaphase. B, Distribution of DNA (blue), microtu-
bules (red), and gamma tubulin (centrosomes [green]) in a metaphase human cell. (B, Images were recorded by Dr. Melpomeni Platani on the
University of Dundee’s School of Life Sciences Imaging Facility OMX 3DSIM Microscope and stored and processed in OMERO.)
766 SECTION X  n  Cell Cycle

A B C D

Spindle pole
E F G

P AP

AP P

2 µm
2 min
Spindle pole

FIGURE 44.13  KINETOCHORE OSCILLATIONS BETWEEN P (POLEWARD) AND AP (AWAY FROM THE POLE) MOVEMENT DURING
LATE PROMETAPHASE AND ANAPHASE IN PTK1 (RAT KANGAROO) CELLS. A–D, Images showing the movements of several pairs of sister
kinetochores, labeled with green fluorescent protein (GFP)-Cdc20 (green), combined with phase-contrast images of the cell (red). E and G, Higher-
magnification views of sister kinetochores (marked with dashed lines) in prometaphase and anaphase, respectively. F, Kymograph (collage of images
of a vertical strip showing the same two kinetochores at various time points during the movie) showing the movements of these two kinetochores.
Movements toward (P) and away from (AP) spindle poles are indicated. P movement involves microtubule shrinkage at the leading kinetochore and
microtubule growth at the trailing kinetochore (which is undergoing AP movement away from its associated kinetochore). Spindle poles are near
the top and bottom of panels E to G. (Micrographs courtesy E.D. Salmon, University of North Carolina, Chapel Hill.)

the spindle, they jostle one another and undergo numer-


A B C
ous small excursions toward one pole or the other
throughout metaphase (Fig. 44.13). 0 sec
Metaphase can also be defined biochemically as the
time of destruction of cyclin B and securin (Fig. 44.16),
Fluorescent tubulin
because this begins as soon as the last chromosome 10 sec speckles move
achieves amphitelic orientation. Degradation of cyclin A toward poles
begins earlier, at the entry into prometaphase, and is
largely complete before metaphase. Loss of securin initi- 20 sec
P P
ates a process leading to the separation of sister chroma-
tids and the onset of anaphase. P P P P
30 sec
Microtubule Flux Within the Metaphase Spindle
FIGURE 44.14  MICROTUBULE FLUX IN METAPHASE. A, Cells
Although the average length of the kinetochore micro- entering mitosis were injected with tubulin subunits modified chemi-
tubules is roughly constant during metaphase, the cally by attachment of a caged fluorescent dye. This dye becomes
microtubules change continuously in three ways. First, fluorescent after being irradiated with UV light. When cells entered
there is constant net addition of new tubulin subunits metaphase, the spindle was illuminated with a narrow stripe of UV
light, activating a narrow band of fluorescent tubulin subunits. With
(approximately 10 subunits per second) to the plus end
time, these subunits approach the spindle poles (P). Because the
of the microtubules, where they are attached to the length of kinetochore microtubules is constant during this time, the
kinetochore. Second, a comparable number of tubulin labeled tubulin molecules must migrate along the microtubules toward
subunits is continuously lost from the minus end of the pole (arrows). This can occur if new subunits are added to the
the kinetochore tubules at the spindle poles. There­ microtubule at the kinetochore and old subunits are removed at the
pole. B, Microtubule flux at metaphase in a Drosophila embryo visual-
fore, tubulin subunits slowly migrate through kineto-
ized by fluorescence speckle microscopy. Embryos were injected with
chore microtubules from the kinetochore to the pole very low levels of fluorescent tubulin, which appears as speckles dis-
(Fig. 44.14). This subunit flux or treadmilling in kineto- tributed along the microtubules. If a very sensitive camera is used,
chore microtubules is caused by microtubule depolymer- these speckles can be seen to move toward the poles, reflecting the
ization at the poles driven by kinesin-13 family members. flux in the underlying microtubules. Scale bar is 5 µm. C, Movement
of labeled tubulin speckles toward the spindle poles. (A, Courtesy
In addition, tubulin moves toward the poles as by micro-
Arshad Desai and the MBL Cell Division Group, Marine Biology Labo-
tubules are transported towards the pole (many nucle- ratory, Woods Hole, MA; from Mitchison TJ, Salmon ED. Mitosis: a
ated by the augmin complex) within the kinetochore history of division. Nat Cell Biol. 2001;3:E17–E21. B, Courtesy Paul
fiber. All microtubules attached to each kinetochore Maddox and Arshad Desai, University of California, San Diego.)
CHAPTER 44  n  Mitosis and Cytokinesis 767

change coordinately in length during chromosomal omitted here for simplicity) and Scc3. Additional proteins
oscillations. are required to stabilize the loading of this complex onto
DNA. Cells with mutations in cohesin components sepa-
rate sister chromatids prematurely in mitosis, resulting
Anaphase in chaotic chromosome missegregation. This system is
The separation of sister chromatids at anaphase is one of very ancient, as bacteria depend on an SMC-related
the most dramatic events of the entire cell cycle (Fig. protein for orderly chromosome segregation.
44.15). Sister chromatids move to opposite spindle poles A variety of evidence suggests that cohesin forms a
(anaphase A), and the poles move apart (anaphase B). ring with a diameter of 35 nm, large enough to encircle
Anaphase is also the time when the mitotic spindle two sister chromatids like a lasso. In yeast, the complex
activates the cell cortex in preparation for cytokinesis. functions only if it binds chromosomes during DNA
Two forms of the APC/C (see Fig. 40.15) trigger the replication. Cohesin accumulates at preferred sites on
transition from metaphase to anaphase by degrading key the chromosomes, often near centromeres in budding
proteins. APC/CCdc20 targets cyclin B for degradation, yeast or in regions of heterochromatin in fission yeast.
causing Cdk activity to fall (see Fig. 40.17). This decline In vertebrates, most cohesin dissociates from the chro-
in Cdk activity allows for activation of APC/CCdh1, because mosome arms by late metaphase, owing to the action of
Cdh1 phosphorylated by Cdk1 kinase cannot bind to the the protein kinases Plk1 and Aurora B. Importantly, a
APC/C. APC/CCdh1 targets polypeptides whose destruc- critical fraction remains associated with heterochromatin
tion by the proteasome is required for the cell to exit flanking centromeres where it is protected from cleav-
from mitosis and return to interphase. APC/CCdh1 remains age by shugoshin until the onset of anaphase (see follow-
active during G1 phase, where it is essential for the ing paragraphs and Chapter 45).
licensing of DNA replication origins (see Fig. 42.28). Sequential cleavage of two key proteins triggers sister
chromatid separation at anaphase. This proteolysis makes
Biochemical Mechanism of anaphase onset an irreversible transition. The first target,
Sister Chromatid Separation securin, inhibits the separase protease. After the last
Separation of sister chromatids is regulated by the chro- chromosome forms an amphitelic attachment to the
mosomes themselves, not by the mitotic spindle. Under spindle, the spindle checkpoint is silenced. This allows
certain circumstances, sister chromatids can separate in APC/CCdc20 to tag securin with ubiquitin, leading to its
the absence of microtubules, ruling out a requirement destruction by proteasomes throughout metaphase. When
for forces from the spindle in the process. securin levels fall below a critical threshold, separase is
Three factors regulate sister chromatid separation: a unleashed to cleave the Scc1 subunit of cohesin. Cleavage
protein complex known as cohesin, a protease known of Scc1 breaks the cohesin ring, allowing the sister
as separase, and an inhibitor of separase known chromatids to separate triggering the onset of anaphase
as securin (Fig. 44.16). This system is conserved from (Fig. 44.16B).
yeast to human. Chapter 8 discusses the functions of Efficient Scc1 cleavage requires that the protein be
cohesin in interphase. phosphorylated near its cleavage site. This allows a
Cohesin is a complex of four proteins that resembles mode of regulation where shugoshin (Japanese for
the condensin complex (see Fig. 8.18). Like condensin, “guardian spirit”) recruits PP2A to centromeres. PP2A
cohesin has two large subunits from the SMC ATPase keeps Scc1 dephosphorylated. This inhibits its cleavage
family. These proteins, SMC1 and SMC3, are complexed and protects cohesin until shugoshin is released follow-
with proteins called Scc1 (which has other names ing amphitelic attachment of the chromosome. This

A. Anaphase B
Sister chromatids separate
Cohesin Anaphase A: Chromatids approach
degrades poles (APC/CCdc20 active)
Interdigitated interpolar
microtubules bundled by PRC1
and stem body material to
form central spindle
DNA
Anaphase B: Spindle poles Microtubules
migrate apart (APC/C Cdh1active) Centrosomes

FIGURE 44.15  INTRODUCTION TO ANAPHASE. A, Summary of the major events of anaphase. B, Distribution of DNA (blue), microtubules
(red), and gamma tubulin (centrosomes [green]) in a mid-anaphase human cell. APC/C, anaphase-promoting complex/cyclosome; PRC1, protein
regulated in cytokinesis 1. (B, Images were recorded by Dr. Melpomeni Platani on the University of Dundee’s School of Life Sciences Imaging
Facility OMX 3DSIM Microscope and stored and processed in OMERO.)
768 SECTION X  n  Cell Cycle

Hinge
A. S phase

Replication
fork
Cohesin
complex

Smc3

Smc1
Scc1

SA1

B C D E
Separase
Chromatin
loops Securin
APC/C
Ubiquitin Sister
Cohesin chromatids
Proteasome separate
Sister
chromatids

Active
Mitotic separase
chromosome cleaves Scc1
Metaphase/
anaphase
S phase Early mitosis transition Anaphase

FIGURE 44.16  REGULATION OF SISTER CHROMATID PAIRING BY THE COHESIN COMPLEX. A–B, The cohesin complex forms a
35-nm diameter ring that links sister chromatids during DNA replication. At anaphase onset, degradation of its securin inhibitor liberates active
separase enzyme. C–E, Separase then cleaves cohesin subunit Scc1, and the two sister chromatids are freed to separate from one another and
move toward opposite spindle poles.

mechanism is absolutely essential during meiosis, as Microtubule disassembly on its own can move chro-
without it, it would not be able to segregate homologous mosomes (see Fig. 37.8). Energy for this movement
chromosomes from each other (see Fig. 45.12). comes from hydrolysis of GTP bound to assembled
Securin can act as an oncogene in cultured cells tubulin, which is stored in the conformation of the
and is overexpressed in some human pituitary tumors. lattice of tubulin subunits. Microtubule protofilaments
Overexpression of securin may dis­rupt the timing of are straight when growing, but after GTP hydrolysis
chromosome segregation, leading to chromosome loss protofilaments are curved, so they peel back from the
and, ultimately, contributing to cancer progression. ends of shrinking microtubules (see Fig. 34.6). Several
kinesin “motors” influence the dynamic instability of the
Mitotic Spindle Dynamics and Chromosome spindle microtubules. Members of the kinesin-13 class,
Movement During Anaphase which encircle microtubules near kinetochores and at
Anaphase is dominated by the orderly movement of spindle poles, use adenosine triphosphate (ATP) hydro-
sister chromatids to opposite spindle poles brought lysis to remove tubulin dimers and promote microtubule
about by the combined action of motor proteins and disassembly rather than movement.
changes in microtubule length. There are two compo- Kinetochores are remarkable in their ability to hold
nents to anaphase chromosome movements (Fig. 44.15). onto disassembling microtubules. In straight (growing)
Anaphase A, the movement of the sister chromatids to microtubules, the Ndc80 complex is mostly responsible
the spindle poles, requires a shortening of the kineto- for microtubule binding. It binds to the interface between
chore fibers. During anaphase B, the spindle elongates, α and β tubulin subunits. This interface bends in curved
pushing the spindle poles apart. The poles separate (shrinking) microtubules, so Ndc80 cannot bind. This
partially because of interactions between the antiparallel could allow it to redistribute onto straight sections of
interpolar microtubules of the central spindle and par- the lattice and thereby move away from the curved
tially because of intrinsic motility of the asters. Most cells protofilaments at the disassembling end. In metazoans
use both components of anaphase, but one component the Ska complex in the outer kinetochore binds α and β
may predominate in relation to the other. tubulin subunits away from the interface, so it can bind
CHAPTER 44  n  Mitosis and Cytokinesis 769

to curved (disassembling) protofilaments. At yeast kineto- kinetochore microtubules toward the pole. In these
chores the Dam1 ring (green in Fig. 8.21) couples the cells, subunit flux accounts for only 20% to 30% of
kinetochore to disassembling microtubules. chromosome movement during anaphase A, and this flux
Anaphase A chromosome movement involves a com- is dispensable for chromosome movement. In Drosophila
bination of microtubule shortening and translocation of embryos, in which subunit flux accounts for approxi-
the microtubule lattice that result from flux of tubulin mately 90% of anaphase A chromosome movement, the
subunits (Fig. 44.14). The contributions of the two chromosomes catch up with a marked region of the
mechanisms vary among different cell types. When living kinetochore fiber slowly, if at all.
vertebrate cells are injected with fluorescently labeled Anaphase B appears to be triggered at least in part by
tubulin subunits, the spindle becomes fluorescent (Fig. the inactivation of the minus-end–directed kinesin-14
44.17). If a laser is used to bleach a narrow zone in the motors, so that all the net motor force favors spindle
fluorescent tubulin across the spindle between the elongation. Four factors contribute to overall lengthen-
chromosomes and the pole early in anaphase, the chro- ing of the spindle: release of sister chromatid cohesion,
mosomes approach the bleached zone much faster than sliding apart of the interdigitated half-spindles, microtu-
the bleached zone approaches the spindle pole. This bule growth, and intrinsic motility of the poles them-
shows that the chromosomes “eat” their way along the selves (Fig. 44.7). During the latter stages of anaphase B,
the spindle poles, with their attached kinetochore micro-
tubules, appear to move away from the interpolar
microtubules as the spindle lengthens. This movement
A of the poles involves interaction of the astral microtu-
Photobleached
zone bules with cytoplasmic dynein molecules anchored at
the cell cortex.
Time Anaphase B spindle elongation is accompanied by
reorganization of the interpolar microtubules into a
Microtubule
disassembly
highly organized central spindle between the separat-
ing chromatids (Fig. 44.15). Within the central spindle,
an amorphous dense material called stem body matrix
stabilizes bundles of antiparallel microtubules and holds
B 42 70 212 422 together the two interdigitated half-spindles. Proteins
concentrated in the central spindle help regulate cytoki-
nesis. One key factor, PRC1 (protein regulated in cytoki-
nesis 1), is inactive when phosphorylated by Cdk kinase
and functions only during anaphase when Cdk activity
declines and phosphatases remove the phosphate groups
5 µm
placed on target proteins by Cdks and other mitotic
kinases. PRC1 directs the binding of several kinesins to
C 40 92 210 436
the central spindle. The kinesin KIF4A targets Aurora B
kinase to a particular domain of the central spindle,
where phosphorylation of key substrates then regulates
spindle elongation and cytokinesis.
How can protein kinases such as Aurora B continue
to function during anaphase while protein phosphatases
are removing phosphate groups placed there by Cdks
and, indeed, Aurora B during early mitosis? One answer
FIGURE 44.17  CHROMOSOMES MOVE ON SHRINKING is that the phosphatase activity is highly localized, con-
MICROTUBULES DURING ANAPHASE. A, Mitotic cells were trolled by specific targeting subunits. Cdk phosphoryla-
injected with a fluorescently labeled tubulin that was incorporated into tion can inhibit targeting subunits such as the exotically
the spindle. Just after anaphase onset, a laser was used to photo-
named Repo-Man (recruits PP1 onto mitotic chromatin
bleach a stripe (white) across the spindle near the upper pole. The live
cell was monitored over time by fluorescence (B) and phase-contrast at anaphase) from binding protein phosphatase 1 or
(C) microscopy. In this mammalian cell, the chromosomes approach localizing to targets, such as chromatin in early mitosis.
the bleached stripe much faster than the stripe approaches the spindle When Cdk activity drops, Repo-Man (and other similar
pole. In other organisms with higher rates of microtubule flux in their targeting subunits) is dephosphorylated, and now targets
spindles, the bleached zone would also move appreciably toward the
PP1 to chromatin, where it removes phosphates placed
pole. The numbers are time in seconds. (B–C, From Gorbsky GJ,
Sammak PJ, Borisy GG. Microtubule dynamics and chromosome there by Aurora B in the CPC. As long as phosphatases
motion visualized in living anaphase cells. J Cell Biol. 1988;106:1185– are not specifically targeted to the cleavage furrow,
1192, copyright The Rockefeller University Press.) Aurora B can continue to control events there during
770 SECTION X  n  Cell Cycle

mitotic exit by phosphorylating key target proteins scaffold protein ELYS to chromatin. ELYS can recognize
required for cytokinesis. DNA regions rich in A : T base pairs, so it is likely to
bind directly to the DNA. ELYS then recruits other
components of the nuclear pore scaffold and nuclear
Telophase pore trans-membrane proteins. The pore subsequently
During telophase, the nuclear envelope reforms on the matures as various peripheral components and elements
surface of the separated sister chromatids, which typi- of the permeability barrier are added.
cally cluster in a dense mass near the spindle poles (Fig. The mechanism of nuclear membrane reassembly is
44.18). Some further anaphase B movement may still debated. In cells where nuclear membranes fragments
occur, but the most dramatic change in cellular structure into vesicles during mitosis, a Ran-GTP–dependent
at this time is the constriction of the cleavage furrow and pathway directs at least two discrete populations of
subsequent cytokinesis. vesicles to chromatin where they fuse to reform the
nuclear envelope. In cells where the nuclear membrane
Reassembly of the Nuclear Envelope is absorbed into the endoplasmic reticulum during
Nuclear envelope reassembly begins during anaphase mitosis, reassembly involves lateral movements of mem-
and is completed during telophase (Fig. 44.19). As in brane components within the membrane network and
spindle assembly, Ran-GTP promotes early steps of their stabilization at preferred binding sites at the periph-
nuclear envelope assembly at the surface of the chromo- ery of the chromosomes.
somes by releasing key components sequestered by Lamin subunits disassembled in prophase are recycled
importin β. These include several nuclear pore com­ to reassemble at the end of mitosis. Lamina reassembly
ponents, and one of the earliest events in nuclear enve- is triggered by removal of mitosis-specific phosphate
lope reassembly involves binding of the nuclear pore groups and methyl-esterification of several COOH side

A. Telophase B
Cleavage plane
specified
Nuclear envelope
reassembles around Organized
chromosomes central spindle
assembles

Poles DNA
continue Microtubules
to separate Centrosomes

FIGURE 44.18  INTRODUCTION TO TELOPHASE. A, Summary of the major events of telophase. B, Distribution of DNA (blue), microtubules
(red), and γ-tubulin (centrosomes [green]) in a telophase human cell. (B, Images were recorded by Dr. Melpomeni Platani on the University of
Dundee’s School of Life Sciences Imaging Facility OMX 3DSIM Microscope and stored and processed in OMERO.)

A B C

0 min 8–10 min 333 nm ≥ 25 min

FIGURE 44.19  SCANNING ELECTRON MICROSCOPY OF THE STAGES OF ASSEMBLY OF MEMBRANE VESICLES ON THE
SURFACE OF CHROMOSOMES IN A XENOPUS EGG CYTOSOLIC EXTRACT. A cell lysate containing membrane vesicles was added to
isolated chromatin from Xenopus sperm, fixed, and then imaged by scanning electron microscopy. Each panel shows the time of incubation prior
to fixation. (Micrographs courtesy of K.L. Wilson, Johns Hopkins Medical School, Baltimore, MD. A and C, From Wiese C, Goldberg MW, Allen
TD, et al. Nuclear envelope assembly in Xenopus extracts visualized by scanning EM reveals a transport-dependent “envelope smoothing” event.
J Cell Sci. 1997;110:1489–1502.)
CHAPTER 44  n  Mitosis and Cytokinesis 771

chains on lamin B (Fig. 44.6). Together with ELYS, B-type


lamins are among the earliest components of the nuclear
Cytokinesis
envelope to target to the surface of the chromosomes Cytokinesis divides a mitotic cell into two daughter cells
during mid-anaphase. Either at this time or shortly there- (Fig. 44.20). Cytokinesis depends on signals to specify
after, other proteins associated with the inner nuclear the cleavage plane (Fig. 44.21), assembly and constric-
membrane, including BAF, LAP2, and lamin B receptor tion of the contractile apparatus, specific alterations of
(see Fig. 9.10), join the forming envelope. Later during the cell membrane, and the final separation (abscission)
telophase when nuclear import is reestablished, lamin of the two daughter cells.
A enters the reforming nucleus and slowly assembles In animals, protozoa, and most fungi, a contractile
into the peripheral lamina over several hours in the ring of actin filaments and myosin-II guides the separa-
G1 phase. If lamin transport through nuclear pores is tion of daughter cells at the end of mitosis (Fig. 44.2).
prevented, chromosomes remain highly condensed fol- Myosin-II pulls on the ring of actin filaments, applying
lowing cytokinesis, and the cells fail to reenter the next tension to the plasma membrane, much like contraction
S phase. of smooth muscle (see Figs. 39.23 and 39.24). Because

A. Early cytokinesis B. Late cytokinesis C


Chromatin decondenses
New membrane Actomycin
inserted Nuclear substructures
contractile reform
Actomycin ring forms
Interphase microtubule
Midbody array reassembles
begins
to form Midbody

ESCRT III action leads to


separation (abscission) of the two cells

FIGURE 44.20  INTRODUCTION TO CYTOKINESIS. A–B, Summary of the major events of cytokinesis. C, Distribution of DNA (blue),
microtubules (red), and γ-tubulin (centrosomes [green]) in a human cell undergoing cytokinesis. ESCRT, endosomal sorting complexes required
for transport. (C, Images were recorded by Dr. Melpomeni Platani on the University of Dundee’s School of Life Sciences Imaging Facility OMX
3DSIM Microscope and stored and processed in OMERO.)

A. Evidence that the cleavage furrow is positioned B. An organized central spindle is required for
midway between asters in eggs cleavage furrow formation and/or function
TOP VIEW
Glass rod pushed
down into egg

90°

Sand dollar egg Microtubules


Chromosomes

Ectopic furrow Actin


ring

Profilin
Metaphase 1 Cytokinesis 1 Metaphase 2 Cytokinesis 2 Wild type mutant

FIGURE 44.21  IN EGGS, A CLEAVAGE FURROW FORMS MIDWAY BETWEEN SPINDLE ASTERS. IN ANIMAL CELLS, THE CENTRAL
SPINDLE IS IMPORTANT. A, A classic experiment in which a sand-dollar egg is caused to adopt a toroid shape. At cytokinesis 2, the egg
cleaves into four cells, and a furrow forms between the back sides of the two spindles. (For a description of this and other classic experiments
in cytokinesis, see the book by Rappaport in the “Selected Readings” list.) B, Left, A wild-type Drosophila spermatocyte undergoing cytokinesis,
with the contractile ring stained in yellow. Right, In a profilin mutant, no central spindle forms, and the cell fails to form a contractile ring.
(Micrographs courtesy Professor Maurizio Gatti, University of Rome, Italy. B, From Giansanti MG, Bonaccorsi S, Williams B, et al. Cooperative
interactions between the central spindle and the contractile ring during Drosophila cytokinesis. Genes Dev. 1998;12:396–410.)
772 SECTION X  n  Cell Cycle

the contractile ring is confined to a narrow band of central spindle appeared to modulate the behavior of the
cortex around the equator, it forms a cleavage furrow, furrow signaled by the poles. We now know that the
constrict­ing the plasma membrane locally like a purse central spindle does emit a positive signal directing a
string (Fig. 44.20). Signals from the mitotic spindle and cleavage furrow to form above it, while the poles con-
cell cycle machinery control the position of this ring (ie, tribute by focusing that furrow at a point on the cortex
the relative sizes of the two daughter cells) and the midway between them.
timing of its constriction. The molecular nature of the cleavage stimulus is now
Protozoa, animals, fungi, and plants use an evolution- beginning to be understood in animals. The following is
arily conserved set of components to implement differ- a simplified scenario:
ent strategies to separate daughter cells. For example, 1. During anaphase, overlapping microtubules between
both fission yeast and metazoan cells use signals from the separating chromatids establish an ordered array
polo kinase and a Rho-GTPase to direct the assembly of known as the central spindle. A key protein compo-
a contractile ring of actin, myosin-II, and other conserved nent of this array is a protein heterodimer known as
components, even though the yeast has a closed mitosis centralspindlin. Centralspindlin is normally seques-
and the metazoans have an open mitosis. In animal tered in the cytoplasm, but phosphorylation by the
cells, contractile ring constriction provides the force that CPC enables it to target to the central spindle. Dro-
remodels the cortex to generate the two daughter cells. sophila mutants that fail to form a central spindle
In contrast, in yeasts, which have a cell wall, contractile cannot initiate cytokinesis (Fig. 44.21B). In contrast,
ring constriction is thought to guide the orderly centrip- C. elegans embryos that lack a central spindle can
etal growth of the cell wall septum, which contributes initiate but not complete the process.
force to overcome turgor pressure and invaginate the 2. One of the components of centralspindlin recruits a
plasma membrane. Plants lack myosin-II, so they divide GEF (guanine exchange factor; see Figs. 4.6 and 4.7)
by targeted fusion of membrane vesicles to build a new for the small GTPase RhoA. This Rho-GEF, Ect2, also
cell wall rather than constricting a cleavage furrow (Box has a motif for targeting to the inside surface of the
44.1 and Fig. 44.26). These differences reflect the fact equatorial plasma membrane.
that widely divergent eukaryotes use variations of similar 3. Membrane associated Ect2 locally activates RhoA,
themes for cytokinesis. Cytokinesis in prokaryotes is which then stimulates localized actin filament assem-
genuinely different, since completely different proteins bly and activation of myosin-II to begin assembly of
are involved (Box 44.2 and Fig. 44.27). the contractile ring.
Although cytokinesis has been studied for more Signals from the poles of the mitotic spindle contribute,
than 100 years, it has posed a number of challenges due particularly in large invertebrate embryos, by confining
to its complexity at the molecular level. For example the zone of active RhoA to a narrow equatorial band
genetic analysis of fission yeast revealed more than between the separating sister chromatids.
150 genes that contribute to cytokinesis. RNAi-based
protein knockdown and molecular replacement analy­ Assembly and Regulation of the Contractile Ring
sis indicates that similar proteins participate in cytoki- Exposure of the cell cortex to the cleavage stimulus
nesis of Caenorhabditis elegans, Drosophila, and culminates in the assembly of a contractile ring consist-
vertebrate tissue culture cells. Cytokinesis research typi- ing of a very thin (0.1 to 0.2 µm) array of actin filaments
cally employs living cells, although progress is being attached to the plasma membrane at many sites around
made toward reconstituting some aspects of the process the equator (Fig. 44.22). Polymerization of the actin fila-
in cell-free systems. ments depends on formins (see Fig. 33.14). Small, bipolar
filaments of myosin-II are interdigitated with actin fila-
Signals Regulating the Position of ments. The plasma membrane adjacent to this actin-
the Cleavage Furrow myosin ring undergoes alterations in its lipid composition
Elegant experimental data from classic studies on fertil- that may help recruit proteins important for the function
ized echinoderm eggs suggest that a cleavage stimulus, of the contractile ring.
emitted by the mitotic spindle, specifies the position of Membrane furrowing requires actin and the motor
the cleavage furrow midway between the poles and activity of myosin-II (see Fig. 36.7). In animals, the small
perpendicular to the long axis of the spindle, thereby GTPase RhoA regulates actin polymerization by formins
ensuring that the cleavage process separates the daugh- as well as constriction of the ring. Many other proteins
ter nuclei (Fig. 44.21). In fertilized eggs, the poles, with are required for cytokinesis to go to completion. In their
their large astral arrays of microtubules (see Fig. 6.4B), absence, furrowing begins, but the cleavage furrows
were regarded as the source of the cleavage stimulus, as ultimately regress, producing binucleated cells. These
furrows can be induced to form midway between two supporting proteins include anillin, actin filament cross-
poles, even when no chromosomes are present. In addi- linking proteins, the CPC (Fig. 44.10) and the central-
tion, a signal emitted by the bundled microtubules of the spindlin complex, among many others. Anillin helps
CHAPTER 44  n  Mitosis and Cytokinesis 773

A. Early anaphase B. Late anaphase C

INCENP
Myosin II

Actin pointed ends

INCENP
Myosin II

Actin barbed ends


E. Myosin confocal

Central optical section


H. Contractile mechanism I. Equatorial section

G Actin
Myosin II

J. Grazing saggital section

FIGURE 44.22  ORGANIZATION OF THE CONTRACTILE RING. A, Organization of actin at the cell cortex prior to cytokinesis. B, Distribution
of actin and myosin at the start of ring contraction. C, INCENP (inner centromere protein) (red) concentrates at the site where the cleavage furrow
will form just before myosin (green). D, INCENP and myosin concentrate in the contractile ring during contraction. E, Confocal micrograph shows
the distribution of myosin in an optical cross-section contracting contractile ring. F–G, Dividing invertebrate egg with DNA (blue) and actin (red)
in the contractile ring. H, Organization of actin and myosin filaments during cytokinesis. I–J, Electron micrographs showing actin filaments in
the contractile ring. Note the thick filaments that are thought to be myosin-II filaments (red arrowheads) and the thinner actin filaments (yellow
arrows). (C–D, Courtesy William C. Earnshaw. E, I, and J, Courtesy P. Maupin, Johns Hopkins Medical School, Baltimore, MD. F–G, Courtesy
Professor Issei Mabuchi, University of Tokyo, Japan. For reference, see Maupin P, Pollard TD. Arrangement of actin filaments and myosin-like fila-
ments in the contractile ring and actin-like filaments in the mitotic spindle of dividing HeLa cells. J Ultrastruct Res. 1986;94:92–103; Maupin P,
Phillips CL, Adelstein RS, et al: Differential localization of myosin-II isozymes in human cultured cells and blood cells. J Cell Sci. 1994;107:3077–3090;
and Eckley DM, Ainsztein AM, MacKay AM, et al. Chromosomal proteins and cytokinesis. J Cell Biol. 1997;136:1169–1183.)

keep active myosin-II focused into an organized contrac- required for transport) complex, which has a key role in
tile ring throughout cytokinesis. the final separation of daughter cells (see later).
The CPC and the centralspindlin complex are both In fission yeast, with closed mitosis, the nucleus
required for animal cells to assemble the central spindle. determines the position of cleavage. Fission yeast assem­
Consequently, if either of the two complexes is elimi- ble a contractile ring along a well-defined pathway by
nated in C. elegans, a contractile ring fails to form. Thus, recruiting proteins from cytoplasmic pools (Fig. 44.23).
they appear to contribute to the cleavage stimulus, and During interphase, assemblies of proteins called nodes
indeed, both require microtubules to localize to the site form on the inside of the plasma membrane around the
of cleavage furrow formation as originally shown for the middle of cell. Prior to mitosis, an anillin-like protein
cleavage stimulus. In addition, the CPC regulates the leaves the nucleus and joins these nodes. During pro-
timely completion of cytokinesis by blocking premature phase, myosin-II, a formin and other contractile ring
activation of the ESCRT (endosomal sorting complexes proteins join the nodes. When the formin polymerizes
774 SECTION X  n  Cell Cycle

Interphase
-60 Mid1p (anillin-like actin patches
protein) exits nucleus
Anillin-like
Cell wall

Formin Myosin-II
-10 Nodes containing
anillin, myosin-II and formin Profilin
assemble around equator

0 SPBs separate Nodes condense into


a contractile ring of
+5 Anaphase A actin filaments and myosin-II
Time (min)

+10 Anaphase B
elongates mitotic Contractile ring
spindle matures by addition of
actin binding proteins

+30 End Anaphase B

Signal from cell cycle via SIN


pathway triggers constriction of
+40 Constriction begins
contractile ring and deposition of
cell wall material to form a septum

Plasma membrane fusion


completes cytokinesis
+70 Constriction ends

FIGURE 44.23  CYTOKINESIS IN FISSION YEAST SCHIZOSACCHAROMYCES POMBE. During interphase, microtubules (red) position
the nucleus in the middle of the cell. Actin filaments concentrate in small patches (yellow) in the cortex at the two growing ends of the cell (see
Fig. 33.1). The mitotic spindle is inside the nucleus, as the nuclear membrane does not break down during mitosis. As the cell enters mitosis,
an anillin-like protein moves from the nucleus to the equatorial cortex, where it sets up nodes of proteins, including myosin-II and a formin. The
formin grows actin filaments (yellow), and myosin-II pulls the nodes together into a continuous contractile ring. At the end of anaphase a signaling
system consisting of a GTPase and three protein kinases (the septation initiation network [SIN]) triggers constriction of the contractile ring and
associated synthesis of new cell wall to form a septum. The septum is a three-layered structure, with the primary septum flanked by two secondary
septae. Digestion of the primary septum separates the daughter cells. (For reference, see Wu J-Q, Kuhn JR, Kovar DR, et al. Spatial and temporal
pathway for assembly and constriction of the contractile ring in fission yeast cytokinesis. Dev Cell. 2004;5:723–734.)

actin filaments, myosin-II pulls the nodes together into a


ring around the equator of the cell (Fig. 44.23). Constriction of the Cleavage Furrow
Contractile ring assembly in animal cells shares many Contractile rings of echinoderm eggs produce enough
properties with fission yeast, but is less completely force to invaginate the plasma membrane and form the
understood. The decline in the activity of cell cycle cleavage furrow, although many details are still being
kinases at the onset of anaphase is part of the trigger, studied. Constriction of the ring probably involves a
since they inhibit centralspindlin components through sliding filament mechanism similar to muscle (see Figs.
metaphase. INCENP and anillin move from the inter- 39.9 and 39.23), but little is known about how the
phase nucleus to the cortex around the cell equator in contractile ring is attached to the plasma membrane.
early anaphase (Fig. 44.22). Formins and profilin polym- During the early stages of furrowing, contractile rings
erize some new actin filaments, but preexisting actin fila- maintain a constant volume, but then disassemble as
ments are recruited into the contractile ring from they constrict further.
adjacent areas of the cortex. Myosin-II is dispersed The role of myosin-II as the motor for cytokinesis was
throughout the cytoplasm until anaphase, when it con- established by microinjection of inhibitory antibodies
centrates in the cortex, especially around the equator into echinoderm embryos and confirmed by genetic
where the furrow forms. The myosin-II is derived from inactivation in the slime mold Dictyostelium. Slime mold
various interphase structures including stress fibers (see amoebas lacking the myosin-II heavy chain round up
Fig. 33.1) that break down during prophase. during mitosis and complete nuclear division but cannot
CHAPTER 44  n  Mitosis and Cytokinesis 775

form a normal cleavage furrow. Mutant cells accumulate


A
many nuclei, because the mitotic cycle continues. Mutant
cells can divide on a substratum using pseudopods to
pull themselves apart into smaller cells.
Constriction of the contractile ring is regulated so that
it does not begin until after the onset of anaphase B,
when sister chromatids are well separated. In fission Midbody ring
yeast, a signaling pathway called the septation initiation B. Abscission (centralspindlin, anillin, cep55)
ESCRT I and ESCRT II
network (SIN) initiates constriction. Much less is known ESCRT III
Midbody
in other cells. Vps4
Microtubules

Abscission
As the contractile ring pulls the cell membrane inward, ESCRT I

the single cell that entered mitosis is gradually trans-


formed into two daughters joined by a thin intercellular
bridge (Fig. 44.20). This process requires a significant
net increase in the surface area of the cell. New plasma
FIGURE 44.24  ABSCISSION IN ANIMAL CELLS. Proteins
membrane is inserted adjacent to the leading edge of associated with overlapping bundles of microtubules form the midbody.
the furrow. The source of the new membrane appears The midbody ring links the midbody to the membrane, and nucleates
to be recycling endosomes (see Chapter 22), so addition the formation of ESCRT (endosomal sorting complexes required for
of membrane to the cleavage furrow is a specialized form transport) III filaments that constrict the membrane to divide the
daughter cells.
of exocytosis. Fusion of vesicles providing the new
membrane depends on specific syntaxins, t-SNAREs
(soluble N-ethylmaleimide-sensitive factor attachment
protein receptors) (see Fig. 21.15) that promote vesicle
fusion along the secretory pathway.
The plasma membrane in the cleavage furrow has a
discrete composition. In budding yeast, this compart-
ment is delineated by rings made from polymers of
septins, a family of GTP-binding proteins recruited by
anillin. Septins are essential for cytokinesis in Saccharo-
myces cerevisiae but not fission yeast.
In most animal cells, constriction of the cleavage
furrow ultimately reduces the cytoplasm to a thin inter-
cellular bridge between the two daughter cells. The
FIGURE 44.25  INCOMPLETE CYTOKINESIS IN A DRO-
intercellular bridge contains a highly ordered, antiparal-
SOPHILA EGG CHAMBER LEAVES CELLS JOINED BY RING
lel array of microtubules derived from the spindle with CANALS. Colocalization of actin (red) and the ring canal protein
a dense knob, the midbody, at its center (Fig. 44.20). HtsRC (green) in the ring canals makes them appear yellow. In the
Isolated midbodies contain more than 160 proteins, with Drosophila egg chamber, ring canals connect nurse cells to each other
approximately one-third involved in various aspects of and to the oocyte. Late in oocyte development, nurse cell contraction
forces their cytoplasmic contents through the ring canals and into the
membrane trafficking.
oocyte. This helps the oocyte gain the stockpile of components that
The midbody is encircled by a dense ring of proteins is needed for early development of the fly embryo. (Courtesy Andrew
that includes centralspindlin, anillin, and a centrosomal Hudson and Lynn Cooley, Yale University, New Haven, CT.)
protein known as Cep55. Interactions between anillin
and membrane-associated septin filaments tether the
membrane to the ring. Cep55 recruits the ESCRT (endo- Ultimately, disassembly of the ESCRT filaments leads to
somal sorting complexes required for transport) III separation of the two daughter cells—abscission.
complex, proteins with important roles in vesicle In some tissues, intercellular bridges remain open as
budding events such as formation of multivesicular ring canals. After several rounds of nuclear division
bodies (see Chapters 22 and 23). In the intercellular with incomplete cytokinesis, the network of cells main-
bridge, ESCRT III forms a helical filament that spirals tains cytoplasmic continuity as each former contractile
around the inner surface of the membrane, becoming ring matures into a larger ring canal. During Drosophila
more and more constricted as it grows away from the oogenesis, four rounds of nuclear division with persis-
midbody (Fig. 44.24). ESCRT III also recruits factors that tent ring canals creates 15 nurse cells, all in continuity
disassemble the bundled microtubules in the intercellu- with the oocytes (Fig. 44.25). The cytoplasmic continu-
lar bridge, allowing the membrane to constrict further. ity through ring canals allows nurse cells to transfer
776 SECTION X  n  Cell Cycle

BOX 44.1  Cytokinesis in Plants

Chromosome segregation is similar in plants and animals, but materials (see Fig. 32.13), move along phragmoplast micro-
cytokinesis is very different because plants lack myosin-II tubules to the equator, where they fuse due to the action of
and do not form a conventional contractile ring (Fig. 44.26). cytokinesis-specific soluble N-ethylmaleimide-sensitive factor
Myosin-II appeared during evolution in the common ances- (NSF) attachment protein receptor (SNARE) proteins (see
tor of amoebas, fungi and animals, after branching from Fig. 21.15), forming a membrane network that becomes the
plants (see Fig. 2.4B). Plants also lack dynein, so microtubule new plasma membrane and laying down the material that
dynamics in mitosis are regulated by some of the more than will become the new cell wall. Dynamin-related proteins also
20 different plus-end– and minus-end–directed kinesins that participate in shaping the newly forming plasma membrane.
are expressed in mitotic cells. In a further difference from Thus, the membrane fusion machinery used for cytokinesis
animals, plants also lack centrosomes, and during interphase, by eukaryotes likely came from the last eukaryotic common
microtubules radiate out from the surface of the cell nucleus ancestor. Actin filaments polymerized by formins and
in all directions. In mitosis, the spindle does not focus to myosin-VIII help position the phragmoplast in the cell. As
sharp poles at metaphase; instead, it assumes a barrel shape the zone of newly deposited membrane expands radially, the
with broad, flat poles. Early in mitosis, a band of microtu- ring of microtubules surrounding it similarly expands. Even-
bules and actin filaments forms around the equator of the tually, the new membrane reaches the lateral cell periphery,
cell adjacent to the nucleus. This so-called preprophase band and fusion with the plasma membrane separates the two
disassembles as cells enter prometaphase. Because the entire daughter cells. The cortical division site, not the spindle,
cell cortex is covered by a meshwork of actin filaments, determines the site of cleavage. This was shown by centri-
disassembly of the preprophase band actually leaves an actin- fuging mitotic cells to displace the spindle from the central
poor zone in a ring where cytokinesis will ultimately occur. location where it initially formed. Late in mitosis, the phrag-
This is called the cortical division site, and it is marked by moplast formed at the midzone of the displaced spindle, but
the tethering of specific kinesin motors. In late anaphase, this phragmoplast then migrated to the plane of the prepro-
two nonoverlapping, antiparallel arrays of microtubules phase band, where cytokinesis occurred. Since plant cells
form over the central spindle. This structure, the phragmo- have cell walls and do not move, the orientation of cleavage
plast, gradually expands laterally until it makes a mirror- planes critically determines the morphology of the organism.
symmetric double disk of short microtubules oriented The hormone auxin can influence cleavage, giving rise to
parallel to the spindle axis with their plus ends abutting the asymmetric division of daughter cells, but the underlying
plane of cell cleavage. Golgi vesicles, containing cell wall mechanism is not yet known.

Chromosomes Cytokinesis in higher plants Early phragmoplast Late phragmoplast

Cortical
actin

Preprophase Cortical
band actin- Golgi
(microtubules) depleted vesicles
zone

Prophase Metaphase Early cytokinesis Late cytokinesis Daughter cells

FIGURE 44.26  CYTOKINESIS IN HIGHER PLANTS. See the text for details.

their cytoplasm into the developing egg, thus greatly Yeast cells use a signaling pathway to terminate
increasing its stockpile of proteins and messenger RNAs mitosis, promote contraction of the contractile ring, and
available for use in early development. In mammals, initiate septation. These pathways, called the mitotic exit
incomplete cytokinesis in the testis results in ring canals network (MEN) in budding yeast and the SIN in fission
connecting several hundred developing sperm cells. yeast, involve a small GTPase and protein kinases. Cdk
kinase activity suppresses the pathway until anaphase,
when Cdk activity drops sharply. The MEN GTPase is
Exit From Mitosis associated with one spindle pole body (the yeast version
To exit from mitosis, cells must inactivate the Cdk1 of the centrosome), while its key regulator, a GTP
kinase. This reverses the biochemical and structural exchange factor, is located in the bud. Elongation of the
changes that are characteristic of mitosis and prepares mitotic spindle during anaphase B moves the GTPase
the cell for proliferation in the next cell cycle. into the bud, where it is activated.
CHAPTER 44  n  Mitosis and Cytokinesis 777

BOX 44.2  Cytokinesis in Bacteria

The strategy for cytokinesis in bacteria is similar to that in to the cell cortex, where it inhibits Z-ring formation. MinE
animal cells (Fig. 44.27), but the molecules are completely is an antagonist of MinC/MinD action. This system works in
different. Cleavage of most bacterial cells depends on a truly remarkable way. MinE forms a ring at the cell equator
a ring of the FtsZ protein (filamentous temperature-sensitive; that migrates along the inner surface of the cell membrane
mutants in fts genes cannot divide and make long filaments until it reaches the end of the cell, at which point it disas-
on cells). This is called the Z ring. FtsZ is the prokaryotic sembles. The ring then reforms in the center of the cell and
homolog of eukaryotic tubulins, but it assembles into fila- sweeps toward the other end of the cell. As it moves, MinE
ments rather than tubules. As for tubulins (see Fig. 34.4), inactivates the MinC/MinD inhibitory complex on the cell
FtsZ polymerization requires bound GTP and hydrolysis of cortex. The inhibitory complex rapidly reestablishes itself on
this GTP destabilizes the polymers. Although purified FtsZ the cell cortex behind the moving MinE ring. It takes
forms rings that use energy from GTP hydrolysis to deform approximately 2 minutes for each sweep of the MinE ring
lipid vesicles, the main function of the Z ring seems to be to along half of the cell, and this cycle is repeated continuously
coordinate the assembly of a complex of proteins (divisome) until the FtsZ ring assembles at the cell center. Bacillus
including an actin homolog FtsA and number of transmem- subtilis uses an alternative mechanism to position the Z ring
brane proteins. The transmembrane proteins synthesize cell for cytokinesis.
wall materials to form the cleavage furrow. Chloroplasts use a homolog of FtsZ for their division, and
The Z ring is positioned at the cell equator of Escherichia FtsZ has been detected in mitochondria of certain primitive
coli by the action of three gene products: MinC, MinD, and eukaryotes. Mitochondria of higher eukaryotes appear to use
MinE (minicell mutants divide at inappropriate locations and another GTPase, dynamin, to coordinate their fission (see
give birth to tiny cells). MinD is an enzyme that recruits MinC “Biogenesis of Mitochondria” in Chapter 19).

Min C/D Min E Cytokinesis in E. coli FtsZ ring


inhibitor Nucleoid

2 minutes

Zone of minimal
Min C/D

FIGURE 44.27  CYTOKINESIS IN THE BACTERIUM ESCHERICHIA COLI. See the text for details.

The MEN kinases downstream of the GTPase activate throughout cells by their specificity, determining sub-
the phosphatase Cdc14p by releasing it from sequestra- units PP2A and PP1 remove many of the phosphates
tion in the nucleolus. Cdc14p inhibits Cdk kinase activity placed on target proteins by the mitotic kinases. Targets
in two ways: (a) it inhibits the degradation of a Cdk include chromatin, where phosphorylation during
inhibitor protein, and (b) it dephosphorylates Cdh1, mitosis had displaced factors involved in both gene
which binds the APC/C and triggers the degradation of activation and repression. Removal of those phosphates
B-type cyclins and other proteins. Cdc14p also triggers allows the interphase regulation of gene expression to
other events during anaphase, including the transfer of resume. Dephosphorylation of other targets allows
chromosomal passenger proteins to the central spindle. intermediate filaments to reform, nuclear envelope reas-
In metazoans mitotic exit is triggered by the inactiva- sembly plus the resumption of RNA transcription, protein
tion of Cdk1 and other mitotic kinases. This transition translation, and membrane trafficking.
is irreversible, in part because cyclins and Aurora
(and other kinases) are degraded. PP2A and its inhibitory
ACKNOWLEDGMENTS
kinase Greatwall (see Chapter 40) replace Cdc14
in mitotic regulation in metazoans. Greatwall activity We thank David Burgess, Iain Cheeseman, Per Paolo
requires Cdks, so when Cdk activity declines, PP2A D’Avino, Arshad Desai, Tatsuo Fukagawa, Gary Gorbsky,
is released from inhibition. When directed to targets Karen Oegema, Jonathon Pines, and Graham Warren for
778 SECTION X  n  Cell Cycle

their suggestions on revisions to this chapter. We thank Jürgens G. Plant cytokinesis: Fission by fusion. Trends Cell Biol. 2005;
the Dundee Imaging Facility for access to the OMX and 15:277-283.
McIntosh JR. Mitosis. Cold Spring Harb Perspect Biol. 2016;(in press).
help with microscopy. Müller S, Jürgens G. Plant cytokinesis—no ring, no constriction but
centrifugal construction of the partitioning membrane. Semin Cell
SELECTED READINGS Dev Biol. 2016;53:10-18.
Nasmyth K, Haering CH. Cohesin: its roles and mechanisms. Annu Rev
Carmena M, Wheelock M, Funabiki H, et al. The chromosomal pas- Genet. 2009;43:525-558.
senger complex (CPC): from easy rider to the godfather of mitosis. Qian J, Winkler C, Bollen M. 4D-networking by mitotic phosphatases.
Nat Rev Mol Cell Biol. 2012;13:789-803. Curr Opin Cell Biol. 2013;25:697-703.
Collas P, Courvalin J-C. Sorting nuclear membrane proteins at mitosis. Rappaport R. Cytokinesis in Animal Cells: Developmental and Cell
Trends Cell Biol. 2000;10:5-8. Biology Series. Cambridge, England: Cambridge University Press; 1996.
Glotzer M. Cytokinesis in metazoa and fungi. Cold Spring Harb Per- Sánchez-Huertas C, Lüders J. The augmin connection in the geometry
spect Biol. 2016;(in press). of microtubule networks. Curr Biol. 2015;25:R294-R299.
Green RA, Paluch E, Oegema K. Cytokinesis in animal cells. Annu Rev Sharp DJ, Rogers GC, Scholey JM. Microtubule motors in mitosis.
Cell Dev Biol. 2012;28:29-58. Nature. 2000;407:41-47.
Haeusser DP, Margolin W. Splitsville: structural and functional insights Stukenberg PT, Burke DJ. Connecting the microtubule attachment
into the dynamic bacterial Z ring. Nat Rev Microbiol. 2016;14: status of each kinetochore to cell cycle arrest through the spindle
305-319. assembly checkpoint. Chromosoma. 2015;124:463-480.
CHAPTER 45 

Meiosis

M eiosis (from the Greek, meaning “reduction”) is a chromosomes from the two parents in each gamete,
specialized program of two coupled cell divisions used and recombination between parental chromosomes
by eukaryotes to maintain the proper chromosome produces novel chromosomes. It works like this.
number for the species during sexual reproduction. It During meiosis, one round of DNA replication and two
also generates novel combinations of genes. Meiosis is rounds of chromosome segregation reduce the number
an ancient process that occurs in virtually all eukaryotes, of chromosomes from 2n to 1n. Each haploid gamete
including the animal, fungal, and plant kingdoms, and is is endowed with a random set of the homologous
thought to have been present in the last eukaryotic chromosomes from the two parents. Prior to meiosis
common ancestor. I the chromosomes duplicate (just like mitosis) (Fig.
Sexually reproducing organisms are typically diploid, 45.1A), but during the first round of segregation (Fig.
with pairs of homologous chromosomes, the two 45.1C) the duplicated chromatids remain paired and the
highly similar but nonidentical copies of each chromo- homologous chromosomes separate randomly between
some, one inherited from each parent. The number of the two daughter cells. Thus each daughter cells ends up
chromosomes is halved during meiosis to form haploid with just one of each pair of homologous chromosomes.
gametes carrying just one set of chromosomes. The This differs from mitosis where the duplicated chroma-
subsequent fusion of male and female gametes restores tids separate, so both daughter cells get the full set of
the diploid chromosome number. This pairing and sub- homologous chromosomes from both parents. During
sequent separation of homologous chromosomes is meiosis II the duplicated chromatids separate and are
made possible by genetic recombination, which occurs partitioned equally between the two daughter cells.
during the lengthy and complex prophase of the first Equally important, homologous chromosomes exchange
meiotic division. DNA sequences during meiotic prophase I, generating
Each human somatic cell has 23 pairs of chromosomes novel chromosomes.
(46 in all). Females have 23 homologous pairs, while The unique segregational events of meiosis usually
males have 22 “autosomal” pairs and two different sex occur in the first division, termed meiosis I (Figs. 45.1
chromosomes that share a region of homology known and 45.2). Because it culminates in daughter cells carry-
as the pseudoautosomal region. One of each pair is ing just one set of chromosomes instead of two, meiosis
contributed by each parent in the egg and sperm, respec- I is also known as the reductional division. The second
tively. The number of chromosome pairs, 23, is known division, meiosis II, is similar in most respects to mitosis:
as the haploid chromosome number. In animals, the sister chromatids segregate, and the number of chromo-
only haploid cells are gametes (sperm and eggs). At fer- somes remains the same (Box 45.1; see also Chapter 44).
tilization, haploid gametes fuse to form a zygote, restor- Meiosis II is called the equational division.
ing the diploid chromosome number of 46. In plants,
the haploid phase is represented by gametophytes, Meiosis: An Essential Process for
which produce ovules and pollen. In most fungi, such
Sexual Reproduction
as yeasts, haploid and diploid forms are alternate phases
of the life cycle, and both can propagate by mitosis. Sexual reproduction is an important survival strategy
Meiosis changes the genetic makeup of offspring that offers organisms an accelerated mechanism for alter-
relative to parents in two ways: the first round of ing the genetic makeup of offspring. Without meiosis,
meiotic segregation produces novel combinations of there would be no sex, because fusion of diploid gametes

779
780 SECTION X  n  Cell Cycle

Preparation for meiosis (This step duplicates each chromatid)

A a Centromere Fused sister


centromeres
B b

Premeiotic Meiotic
Two pairs of S phase prophase
homologous
chromosomes

Meiotic prophase (Recombination drives pairing of homologous chromosomes and produces novel chromosomes)
Recombination Chiasmata
nodules
A a A
b a
Meiosis I
B
B b

Leptotene stage Bouquet Zygotene stage Pachytene stage Diplotene stage Diakinesis stage

Meiosis I (Homologous chromosomes randomly separate from one another producing haploid progeny)

Chiasmata A
A b A b
b B
a B B Meiosis I a Meiosis II
a
Interphase
(no S phase)
Metaphase I Anaphase I

Meiosis II (Sister chromatids separate producing one or more gametes)

b B
a

Prophase II Metaphase II Anaphase II Haploid gametes


FIGURE 45.1  OVERVIEW OF THE PHASES OF MEIOSIS. Shown are important structures and the outcome of each stage for a homologous
pair of metacentric chromosomes (A, a) and a homologous pair of telocentric chromosomes (B, b).

would double the number of chromosomes in the chromosomes must first find each other. They do this by
progeny at every generation. undergoing reciprocal recombination (crossover) events
Meiosis I produces random combinations of homolo- that then hold them together until anaphase of meiosis
gous maternal and paternal chromosomes. For each I. Chromosomes and the genes they carry vary hugely
pair of homologs, orientation on the spindle is random between individuals. In humans an average genome
during meiosis I (ie, each homolog has two equivalent varies from the “reference genome” (see Chapter 7) at
options for the direction to migrate). Thus, for humans 4 to 5 × 106 sites. These include not only polymorphisms
(with 23 pairs of homologous chromosomes), each (differences of single base pairs), but also thousands
gamete has one of 223 (more than 8 million) possible of longer insertions, deletions, and rearrangements.
complements of maternal and paternal chromosomes. Recombination events that result in a crossover and
This process does not create new versions of genes, exchange chromosomal segments produce new chromo-
but it guarantees the offspring will have novel combi- somes that are a patchwork of segments from the
nations of subtly different (due to polymorphisms) maternal and paternal homologs. The combined effects
chromosomes. of recombination and random assortment of homologs
Meiosis I also produces novel versions of genes and in meiosis I yields a vast number of genetically different
chromosomes by recombinational exchange of DNA gametes. This genetic diversity increases the ability of
segments between homologs. This occurs because to eukaryotic populations to adapt to changing environ-
segregate from one another, each pair of homologous mental conditions.
CHAPTER 45  n  Meiosis 781

Spindle
A. Metaphase I Spindle C. Late Spindle pole
Paired sister kinetochores
pole anaphase I pole
being pulled toward poles

X
D. Telophase I
X

Chiasma Chiasma
Spindle
pole Paired sister kinetochores
moving toward poles

B. Early Spindle
anaphase I pole

Spindle Spindle
pole pole Spindle
pole

FIGURE 45.2  FIRST MEIOTIC DIVISION STAGES FROM THE GRASSHOPPER PYRGOMORPHA CONICA (2N IN MALES = 18
AUTOSOMES + 1 X CHROMOSOME). A, Metaphase I. B, Early anaphase I. C, Late anaphase I. D, Telophase I spermatocytes stained with
lactopropionic orcein. All chromosomes are telocentric (see Fig. 7.2). Seven bivalents shown in the metaphase I spermatocyte have a single
chiasma, whereas the two bivalents at the far right and far left have two chiasmata. The sex chromosome (X) remains unpaired and moves to a
single spindle pole. (Courtesy José A. Suja and Julio S. Rufas, Universidad Autónoma de Madrid, Spain.)

as chromatin structures called chiasmata (singular:


The Language of Meiosis
chiasma, from the Greek, meaning “X-shaped cross”).
Meiosis has a language of its own, characterized by a Segregation of homologous chromosomes in meiosis
number of unusual terms, and is easiest to understand I differs from the segregation of sister chromatids during
by focusing on the essential biological processes that are mitosis (Box 45.1), because the paternal and maternal
involved. This reduces the process to only three essential homologous chromosomes segregate randomly to the
key terms: pairing, homologous recombination, and two daughter cells. When homologs orient at the meta-
segregation. This chapter discusses each step in detail, phase plate of the meiosis I spindle, centromeres belong-
so they are defined only briefly here. ing to the two sister chromatids are fused to form a single
Pairing is a two-step alignment of homologous chro- kinetochore that binds microtubules. Cohesion between
mosomes with one another in the nucleus. In alignment, chromosome arms distal to chiasmata (ie on the other
corresponding DNA sequences on the homologous side from the centromere; Figs. 45.2 and 45.10) keeps
chromosome find each other among the billions of base homologous chromosomes paired with one another
pairs of DNA in the nucleus. In many organisms, early until anaphase of meiosis I, counteracting the bipolar
events of recombination drive the homologous pairing pulling force of the spindle on the homologs (Fig. 45.2).
process. In the second stage, synapsis, the paired At anaphase I the distal cohesion is released from chro-
homologous chromosomes become intimately aligned mosomes allowing the chiasmata to separate, and the
along their entire lengths with one another separated two sister chromatids (at least one of which has under-
by approximately 100 nm. A specialized scaffolding gone a crossover exchange) move as a single unit toward
structure called the synaptonemal complex mediates the same spindle pole while the sister chromatids from
this process. other parent move to the other daughter cell. As a result,
Homologous recombination results in physical the two daughter cells produced in meiosis I have a
exchange of DNA between homologous chromosomes haploid number of chromosomes derived randomly from
(a crossover event) and is a key determinant of chromo- the two parents, each with two sister chromatids. Each
some behavior during meiotic prophase. Recombination of the four daughter cells produced in meiosis II has one
drives the pairing process in many organisms and can sister chromatid for each homologous chromosome (ie,
occur without synapsis under certain circumstances. half the number of chromatids as there are chromosomes
Crossover recombination sites are detected by microscopy in somatic cells).
782 SECTION X  n  Cell Cycle

breaks using the corresponding DNA sequence on a


BOX 45.1  Important Differences Between Meiosis
sister chromatid as a template. Meiotic cells usually use
and Mitosis
a homologous chromosome. The mechanism for this
Meiosis involves two cell divisions. The two meiotic difference in selectivity is not yet fully understood.
divisions are preceded by a round of DNA replication. Spo11, together with essential accessory proteins,
There is no DNA replication between meiosis I and generates programmed double-strand DNA breaks early
meiosis II. during meiotic prophase (Fig. 45.3). Similar to type II
The products of meiosis are haploid. The products DNA topoisomerases (see Fig. 8.16), Spo11 cleaves both
of mitosis are diploid. DNA strands in a reaction that produces a covalent
The products of meiosis are genetically different. linkage between a tyrosine of the enzyme and the cleaved
After recombination and random assortment of homologs
phosphodiester backbone. However, Spo11 does not
in meiosis I, the sister chromatids that segregate in meiosis
reseal the breaks; instead it remains attached to one
II are different from each other. In normal mitosis, sister
chromatids are identical. strand of DNA at the broken end. In mice, Spo11 creates
Prophase is longer in meiosis I. Proper orientation about 10-fold more DNA breaks than ultimately recom-
and segregation of homologous chromosomes is achieved bine to produce reciprocal DNA exchanges between
thanks to the pairing, synapsis (synaptonemal complex homologous chromosomes or crossovers. Repair of
formation), and recombination that occur in a lengthened Spo11-mediated DNA double-strand breaks can also
prophase during meiosis I. In humans, prophase in mitosis result in noncrossover events known as gene conver-
takes an hour, whereas meiotic prophase lasts many days sions (Box 45.2 and Fig. 45.3I–J).
in males and many years in females. DNA double-strand breaks generated by Spo11 are
Recombination is increased in meiosis. The required for the initial lengthwise alignment of homolo-
recombination rate is 100- to 1000-fold higher in prophase
gous chromosomes in many organisms, including mice,
I of meiosis than in mitosis. The process has two main
plants, and yeast. In mice lacking Spo11, recombination
consequences: the formation of chiasmata and the intro-
duction of genetic variation. Chiasmata are structures that is not initiated, and synapsis, if it occurs at all, is aberrant,
physically link the homologous chromosomes after cross- often involving nonhomologous chromosomes (Fig.
over and play an essential role in meiotic chromosome 45.4). Gametes in these mutant mice die by apoptosis
segregation. early in meiotic prophase. Spo11-induced double-strand
Kinetochore behavior differs in meiosis. During breaks are not required for synapsis of homologous
meiosis I, kinetochores of sister chromatids attach to chromosomes in the nematode Caenorhabditis elegans
spindle microtubules emanating from the same pole. and the fruit fly Drosophila melanogaster. How these
Homologous kinetochore pairs connect to opposite poles. organisms pair their homologs without recombination is
In mitosis and meiosis II, sister kinetochores attach to still mysterious.
spindle microtubules coming from opposite poles.
Once the DNA double-strand breaks are produced,
Chromatid cohesion differs in meiosis. Sister
the MRN (Mre11/Rad50/Nbs1) endonuclease nicks the
chromatid cohesion is essential for orientation of bivalents
(paired homologous chromosomes) on the metaphase single-stranded DNA, releasing Spo11. The DNA ends
I spindle. During anaphase of meiosis I, cohesion is that lost Spo11 then undergo further processing, as Exo1
destroyed between sister chromatid arms, and chiasmata exonuclease, chews back the 5′ strands of the double
are released to allow segregation of homologs. Cohesion helix (a process called resection) leaving single-stranded
at sister centromeres persists until the onset of anaphase DNA tails with 3′ termini (Fig. 45.3C; see also Fig. 43.14).
II, when it is lost to permit segregation of sisters. In pro- The MRN and Exo1 nucleases also function in somatic
metaphase of meiosis II, sister chromatids are joined only DNA repair.
by the centromeres, whereas at the beginning of mitotic Next, the Rad51 and Dmc1 proteins drive a search of
prometaphase, sisters are joined all along the arms. the 3′ single-stranded DNA tails for complementary DNA
sequences of the other chromosomes. Rad51 and Dmc1
are related to the Escherichia coli RecA protein used for
homologous recombination in bacteria. Rad51 and Dmc1
coassemble along 3′ single-stranded DNA tails and use
Recombination
adenosine triphosphate (ATP) hydrolysis to catalyze a
Although meiotic recombination is similar to the process strand exchange reaction with an intact homologous
of homologous recombinational repair of double-strand DNA duplex. The Rad51-and-Dmc1-decorated nucleo-
DNA breaks in somatic cells (review Box 43.1 and Fig. protein filament disrupts the targeted homologous
43.14 as a prelude to studying meiotic recombination), double helix, displacing one of the two DNA strands.
the two processes differ in two respects. First, meiotic This allows formation of new Watson-Crick base pairs
cells use a specialized enzyme called Spo11 to create between the invading 3′ single-stranded DNA and the
double-strand DNA breaks on purpose. Second, somatic complementary strand of the target DNA. Following
cells with replicated chromosomes usually repair DNA strand invasion and exchange, new DNA synthesis
5' 3'
A Cohesin
3' 5'
5' 3'
3' 5' Paired homologous
5' 3' chromosomes
3' 5'
5' 3'
3' 5'

Double-strand break Spo11 cuts DNA, remaining


covalently attached to one strand
B

Red enzymes are


meiosis specific Spo11 released by MRN complex (Rad50/
5' to 3' resection
Exo1 resects one DNA strand Mre11/Nbs1)
Green enzymes also
function in somatic C
repair

Initial strand invasion, Rad51 + Dmc1


DNA synthesis
D

Second end capture,


synthesis, ligation Strand displacement
E H

Formation of double
Holliday junction Strand annealing

F I

Holliday
junction

Holliday junction
= Resolvase cutting sites Synthesis, ligation
resolution

G J

+ +

Crossover Noncrossover

FIGURE 45.3  EVENTS OF RECOMBINATION. Recombination occurs between homologous chromosomes rather than between sister
chromatids. A, Paired homologous chromosomes. Sister chromatids are held tightly together by cohesin, shown here schematically as hoops.
B, Spo11 makes a double-strand break, remaining attached to the DNA. C, Removal of Spo11 and resection of the break. D, First strand invasion.
At this point, the pathway splits in two, one outcome leading to a crossover and the other to a noncrossover. Crossover pathway: E, The
second resected strand establishes base-pairing interactions with the displaced DNA strand of its homologous partner. New DNA synthesis fills
the gaps. F, The resulting molecule contains a double Holliday junction in which the DNA is fully base-paired (see Fig. 43.14B). If the resolvase
(nuclease) cuts the double Holliday junction asymmetrically as shown (ie, one vertical and one horizontal cut), the result is a crossover (G). If the
cuts are symmetrical, a noncrossover molecule is produced. Noncrossover pathway: H, In most cases, the invading DNA, strand is ejected
prior to stabilization and formation of a double Holliday junction. I, DNA gap-filling and ligation yield a noncrossover chromosome (J).
784 SECTION X  n  Cell Cycle

BOX 45.2  Brief Overview of Genetic Terminology

A comprehensive introduction to the field of genetics is 50% of the time as a result of the random distribution of
beyond the scope of this text. However, here are a number chromosomes to the two spindle poles. If they are on the
of terms used by geneticists that will assist in the understand- same chromosome, they will be linked to one another unless
ing of the discussion of genetic recombination and its role the chromosome undergoes a genetic recombination event
in meiosis (also see Box 6.1). between them. The greater the separation of two markers
The genotype of an organism is the combination of along one chromosome, the more likely it is for such an
genes present on the chromosomes of that organism. The intervening recombination event to occur.
phenotype is the physical manifestation of the action of Two types of recombination events occur during meiosis
these gene products (ie, the appearance and macromolecular (Fig. 45.3). The first of these—noncrossover events (fre-
composition of the organism). In discussing recombination, quently referred to as gene conversion)—may involve the
scientists typically refer to the presence or absence of spe- loss of one or more genetic markers. Noncrossover events
cific genetic markers. Each genetic marker is a particular are the most common outcome of the programmed double-
DNA sequence in or around a gene that can be monitored strand DNA breaks that occur during leptotene. They are
by examining the phenotypes of the cells that carry it. thought to involve the invasion of a double helix by a region
A genetic marker might be the presence of a functional of single-stranded DNA with complementary sequence but
gene, a mutation with altered activity, or simply a polymor- then ejection of this sequence before assembly of a Holliday
phism of DNA sequence that has no known functional junction and completion of recombination.
consequence. The second type of recombination event—crossing
A haploid organism has one copy of each chromosome. over—involves the physical breakage and reunion of DNA
A diploid organism has two homologous copies of each strands on two different chromosomes, typically producing
chromosome. A diploid organism that is homozygous for a a balanced exchange of DNA sequences. This is what most
particular genetic marker has the same sequence of that people think of as recombination. In recombination by cross-
particular region of the DNA on both the maternal and ing over, the makeup of genetic markers remains constant;
paternal homologous chromosomes. A heterozygous organ- it is the linkage between different markers that changes.
ism has different forms of the genetic marker on the two The normal separation of chromosomes or chromatids is
homologous chromosomes. Although the physical events of referred to as disjunction (disjoining). Mistakes in this sepa-
genetic recombination occur in both homozygotes and het- ration are referred to as nondisjunction. Nondisjunction in
erozygotes, they are most readily detected in the latter. meiosis I and II results in the production of gametes with
Two genetic markers located on different chromosomes either too many or too few chromosomes, a condition
will separate from one another in the anaphase of meiosis I known as aneuploidy.

restores sequences that may have been lost or damaged


A B
at the position of the original DNA double-strand break.
Mutants lacking Dmc1 are defective in homologous
chromosome pairing and interhomolog recombination.
As a result, Dmc1 is thought to facilitate the search for
homologous chromosomes as a DNA repair template,
rather than sister chromatids as in somatic DNA repair.
Rad51p and Dmc1p are found in structures called
early recombination nodules that are distributed
along the chromosome axes early in meiosis (Fig. 45.9).
Dmc1 functions only in meiosis, but Rad51 has other
essential functions.
It is now believed that noncrossover events arise
primarily from recombination intermediates that involve
FIGURE 45.4  PAIRING OF HOMOLOGOUS CHROMOSOMES
IS SEVERELY DISRUPTED IN THE SPO11 MUTANT. Pachytene
a relatively transient single strand invasion of the
chromosomes from wild-type mice (A) and mice in which the Spo11 homologous chromosome followed by restorative DNA
gene has been disrupted (B). SYC3 (axial elements) and centromeres synthesis and disassembly of the joint molecule interme-
are red. SYCP1 (in transverse filaments, which are seen only when diate. Crossovers, on the other hand, are thought to arise
synapsis has occurred) is green. (From Baudat F, Manova K, Yuen JP, predominantly through a pathway that involves stable
et al. Chromosome synapsis defects and sexually dimorphic meiotic
progression in mice lacking Spo11. Mol Cell. 2000;6:989–998.)
branched intermediates known as Holliday junctions
(Fig. 45.3F–G; see also Fig. 43.14B), which are then
cleaved by resolvases such as Gem1 to form (predomi-
nantly) crossover products.
CHAPTER 45  n  Meiosis 785

Most sexually reproducing organisms depend on Tracking the Homologous Chromosomes


recombination during meiosis to produce haploid
gametes, but fruit flies and yeast have other systems for
Through the Stages of Meiotic Prophase I
segregating homologs in meiosis I. These mechanisms, Pairing and recombination of homologous chromosomes
collectively known as achiasmate segregation, allow take place during prophase of meiosis I. Five stages of
the segregation of chromosomes that have not under- meiotic prophase are used to describe the process: lep-
gone crossover recombination. One model for the achi- totene, zygotene, pachytene, diplotene, and diakinesis
asmate segregation in flies proposes that nonrecombined (Fig. 45.1B).
chromosomes remain paired at the end of meiotic pro- Leptotene (from the Greek, meaning “thin ribbon”)
phase owing to stickiness of heterochromatin and, as a starts with the first visible condensation of the chromo-
result, segregate properly during anaphase I of meiosis. somes. Paired sister chromatids become visible as linear
In a rare but notable example, the spermatocytes of D. arrays of loops flanking a single dense protein-containing
melanogaster males do not recombine at all, yet still axis (Fig. 45.5A–B). This axis consists of proteins that
segregate their chromosomes happily during meiosis I. play a role in mitotic chromosome structure as well as
This might be regarded as a cruel joke of evolution by proteins specialized for meiotic chromosomes. For
those students who find all the Greek terms of meiotic example, the cohesin complex with several specialized
nomenclature to be daunting. However, meiosis without meiosis-specific subunits is a prominent component of
recombination is clearly the exception, and in most this axial structure (see Fig. 8.18). According to recent
species meiosis depends on recombination in both males models, recombination begins during leptotene with the
and females. formation of DNA double-strand breaks. By the end of

A. Early leptotene C. Early zygotene E. Pachytene

Sex
chromosomes

B. Late leptotene D. Late zygotene F. Diplotene


Chiasma

FIGURE 45.5  IMMUNOFLUORESCENCE IMAGES OF PROPHASE I SUBSTAGES IN MOUSE SPERMATOCYTES. These images
demonstrate the pairing and synapsis of homologous chromosomes revealed by visualizing the synaptonemal complex proteins SYCP3 (a
component of the axial elements [red]) and SYCP1 (a component of the transverse filaments that is present only when homologs are synapsed
[green]). Centromeres are blue. (Courtesy Paula Cohen, Cornell University, Ithaca, NY.)
786 SECTION X  n  Cell Cycle

A B C D E F G

0 hr 2 hr 8 hr

I Telomere

Nuclear
envelope

FIGURE 45.6  CHROMOSOMAL MOVEMENTS DURING EARLY MEIOTIC PROPHASE. A–G, Pairing of homologous chromosomes during
leptotene in the ascomycete Sordaria. Scale bar is 1 µm in A–F and 5 µm in G. A–B, In early leptotene, homologous chromosomes (visualized
in panels A–F by electron microscope reconstructions of serial-sectioned nuclei) are not yet aligned with one another. C–E, In mid-leptotene,
regions of some homologs begin to align. (In panel D, only the telomeres have aligned. In panel E, the pair of homologs is fully aligned.) F, The
alignment of homologs is complete by late leptotene. G, The alignment of homologs also can be seen by light microscopy using Spo76-GFP, a
component of the chromosome axes. H–I, Stages of formation of the bouquet arrangement in rye. H, Telomeres (green) were detected in nuclei
by in situ hybridization (see Fig. 8.10) after 0, 2, and 8 hours in culture. Chromatin is red. I, Three-dimensional models of the nuclei (nuclear
periphery [red dots], telomere position [green stars]). (A–G, Modified from Tesse S, Storlazzi A, Kleckner N, et al. Localization and roles of Ski8p
protein in Sordaria meiosis and delineation of three mechanistically distinct steps of meiotic homolog juxtaposition. Proc Natl Acad Sci U S A.
2003;100:12865–12870. Copyright 2003 National Academy of Sciences. H–I, Modified from Carlton PM, Cowan CR, Cande WZ. Directed motion
of telomeres in the formation of the meiotic bouquet revealed by time course and simulation analysis. Mol Biol Cell. 2003;14:2832–2843.)

leptotene, homologous chromosomes are aligned loosely the chromosomes parallel to each other. In C. elegans
about 400 nm apart (Fig. 45.6D–G). special chromosome regions known as “pairing centers”
During leptotene, one or both telomeres attach to the mediate chromosome movement instead of telomeres;
inner surface of the nuclear envelope and move actively in budding yeast, the telomeres are linked to actin instead
around the nuclear surface until they coalesce near the of microtubules, and D. melanogaster may have lost
centrosome (spindle pole body in yeasts [Fig. 45.6]). such a mechanism altogether.
These movements and clustering of telomeres depend During the transition from leptotene to the zygotene
on cytoplasmic microtubules. Telomeres are linked to (Greek, “yoke ribbon”) stage of prophase, clustering
microtubules through a pair of nuclear envelope proteins of chromosome ends at the nuclear envelope reaches
known as the LINC (linker of nucleoskeleton and cyto- its peak, with the “bouquet” arrangement of chromo-
skeleton) complex (see Fig. 9.8). Telomere clustering somes. During this stage homologous chromosomes
peaks at the leptotene–zygotene transition with the begin to achieve their maximal alignment as well,
chromosomes radiating into the nuclear interior like a through the initiation of synapsis (Fig. 45.5C–D). Syn-
bouquet of flowers, hence the name “bouquet stage.” apsis involves the assembly of the axial element. This
Bouquet formation is a nearly universal feature of this protein scaffold forms part of the synaptonemal
phase of meiosis and the movements of tethered telo- complex when pairing is complete.
meres help homologs find each other through physical In pachytene (from the Greek, meaning “thick
alignment. Thus, telomere clustering per se may not be ribbon”), synapsis is complete, with the homologous
the goal of this movement. The details vary among dif- chromosome axes joined together along their lengths
ferent organisms. In fission yeast dynein motors and by synaptonemal complexes (Fig. 45.5E). During pachy-
microtubule dynamics in the cytoplasm move the telo- tene, crossover-designated recombination intermediates
mere cluster from one end of the cell to the other every mature into Holliday junction-containing structures
10 minutes or so. These “horsetail movements” stretch within the context of the full-length synaptonemal
CHAPTER 45  n  Meiosis 787

complex. The final resolution of these recombination The earliest pairing events in meiosis involve a ten-
intermediates into crossovers occurs close to the time of dency of homologous chromosome territories to move
synaptonemal complex disassembly, dispersal of the together in the nucleus even before leptotene chromo-
bouquet of chromosomes and exit from pachytene. The some condensation. The mechanism is unknown. As
crossovers then mature into structures called chiasmata double-strand breaks created by Spo11 initiate the
that link homologous chromosomes through meiosis I recombination pathway during leptotene, the condensing
metaphase. homologous chromosomes align with one another at a
Early in diplotene (from the Greek, meaning “double distance of about 400 nm (Figs. 45.6 and 45.7). Genetic
ribbon”), the synaptonemal complex disassembles, analysis in budding yeast revealed that mutants defective
telomeres detach from the nuclear membrane, and chro- in the earliest stages of recombination are also defective
mosomes begin to condense in preparation for division in homolog pairing.
(Fig. 45.5F). The duplicated sister chromatids remain
closely associated, and chiasmata hold the homologous Replicating DNA
Interphase Sister Sister
chromosomes together, although their axes tend to drift chromatid 3 chromatid 4
Sister
apart in the absence of synaptonemal complex. This chromatid 1 Sister
chromatid 2 Cohesin
part of meiotic prophase may last for days or years,
depending on the sex and organism (up to 45 years or
more in female humans).
Oocytes (immature eggs) actively transcribe their Sister chromatids
linked by cohesin
chromosomes during diplotene, as they store up materials in premeiotic
for use during the first few divisions of embryonic devel- S phase
opment. Transcription can be so active that DNA loops
are massively coated with nascent RNA transcripts whose Leptotene
associated proteins are visible by light microscopy in Initiation of
recombination
oocytes of most animals (except mammals). Chromo- Pairing of Chromatid
somes at this stage are known as lampbrush chromo- homologous axis
somes (see Fig. 8.12). chromosomes
Diakinesis (from the Greek, meaning “across move- Zygotene
ment”) is the prometaphase of meiosis I. Following Assembling
central element of Synapsis
nuclear envelope breakdown, homologous chromo- synaptonemal complex
somes shorten and condense. At metaphase I, the biva-
lents (pairs of homologous chromosomes) are aligned Axial
Pachytene (lateral)
at a metaphase plate (Figs. 45.1, 45.2, and 45.12). The elements
two homologs (each a pair of tightly linked sister chro- Transverse
matids) are attached to opposite poles of the meiotic filaments
spindle, which applies force, attempting to pull them Central
apart. Cohesion of the arms distal to chiasmata resists element
these pulling forces. The homologs separate and move
to opposite spindle poles during anaphase I when the
cohesion along the chromosome arms is released. The Diplotene followed
sister chromatids move together to one pole, because by diakinesis
Chromatin
they remain linked by cohesion at their centromeres, Disassembling loops
synaptonemal
where the cohesion complex is protected by a shugoshin complex
protein (see later).
After telophase I, cells enter a brief interkinesis
Chiasma
during which there is no DNA replication. The second
Time

meiotic division is mechanistically similar to mitosis


except that the number of chromosomes is reduced by
half. Additionally, in the eggs of most female vertebrates,
meiosis is arrested at metaphase II until fertilization.

Pairing and Synapsis in More Detail


Pairing describes the side-by-side alignment of homolo-
gous chromosomes at a distance. Homologs are paired FIGURE 45.7  CHROMOSOMAL PAIRING IN MEIOTIC PRO-
in nonmeiotic (somatic) cells in a few organisms, such PHASE. Structural organization of homologous chromosomes and
as the fruit fly D. melanogaster, but not in vertebrates. synaptonemal complex during various stages of meiotic prophase.
788 SECTION X  n  Cell Cycle

The process of homolog alignment almost certainly events will mature into crossovers. By pachytene, a
involves the invasion of neighboring DNA duplexes by continuous synaptonemal complex assembles along the
single-stranded DNA complexed with Rad51 and Dmc1. full length of the aligned homologous chromosomes
Thus, recombination has important roles both in the (Fig. 45.5C–E).
exchange of genetic material and in the mechanics of It was once thought that the synaptonemal complex
chromosome behavior during meiotic prophase. Recom- aligns homologous chromosomes in preparation for
bination is probably not the only factor driving homolog recombination, but it is now clear that homolog pairing
pairing, however. Pairing is reduced but not absent in and (in many organisms) the initiation of recombination
yeast meiotic cells lacking both Rad51 and Dmc1, and precedes synapsis. Thus, synapsis is a downstream
homologous chromosomes still pair in some systems that consequence of early steps in recombination in some
lack recombination (eg, certain D. melanogaster recom- well-studied organisms including yeast and mammals.
bination mutants), synaptonemal complex formation However, under certain artificial circumstances, even
(asynaptic mutants in yeast), or both (eg, normal D. nonhomologous chromosomes can undergo synapsis.
melanogaster males). Another longstanding model proposed that the synapto-
Homolog pairing initiated during leptotene be­ nemal complex promotes the resolution of crossover-
comes much more intimate during synapsis as the designated recombination intermediates. However, analysis
chromosomes are linked by transverse fibers to form of budding yeast mutants missing certain synaptonemal
the synaptonemal complex. This structure looks complex proteins indicates that the structure per se is
roughly like railroad tracks linked by transverse bands dispensable for the formation of crossovers and that the
(Figs. 45.7 and 45.8). Each of the two outer rails, 90 resulting chiasmata can hold homologous chromosomes
to 100 nm apart, is the axis of a pair sister chromatids. paired until anaphase of meiosis I.
They are traditionally termed lateral elements, but What then is the function of the synaptonemal
for the sake of simplicity, we refer to them as axial complex? One possibility is that it may have a key role
elements. Thin transverse filaments oriented perpen- in crossover interference (see later), which ensures that
dicular to the axial elements appear to connect homolog crossovers are distributed broadly across the genome.
axes to each other and to the central element (the Another interesting possibility is that the synap-
“third rail”). Synaptonemal complex formation begins tonemal complex communicates information about
during zygotene at a limited number of sites along the meiotic chromosomes (such as homolog alignment and
paired homologous chromosomes where recombination the formation of crossover-designated recombination

A B C

CE

LE
CE

FIGURE 45.8  ELECTRON MICROGRAPHS OF THE SYNAPTONEMAL COMPLEX. A, Low-magnification view of maize synaptonemal
complexes stained with silver. The lateral (LE) and central elements (CE) are clearly seen. B, A negatively stained cricket synaptonemal complex
following treatment with deoxyribonuclease (DNase). The central element (CE) and transverse filaments (arrow) are visible. C, A whole mount of
a silk moth zygotene chromosome. Cells in meiotic prophase were swollen and then lysed under gentle conditions with detergent. The chromo-
somes were then centrifuged onto thin carbon films so that they could be examined by electron microscopy. The axial elements are easily seen
on this chromosome. Chromatin loops radiate outward from both the unpaired axial elements and the paired lateral elements (where synapsis
has occurred). (A, Modified from Gillies CB. Electron microscopy of spread maize pachytene synaptonemal complexes. Chromosoma.
1981;83:575–591. B, Modified from Solari AJ, Moses MJ. The structure of the central region in the synaptonemal complexes of hamster and
cricket spermatocytes. J Cell Biol. 1973;56:145–152, copyright the Rockefeller University Press. C, From Rattner JB, Goldsmith M, Hamkalo BA.
Chromatin organization during meiotic prophase of Bombyx mori. Chromosoma. 1980;79:215–224.)
CHAPTER 45  n  Meiosis 789

intermediates) to the cell-cycle pathways that control Several protein components of the axial elements
progression through the substages of meiosis. (sister chromatid axes) have also been identified. One of
these, SYCP3, interacts with both the cohesin complex
(see Fig. 8.18) and Rad51p and Dmc1p. In SYCP3 knock-
Synaptonemal Complex Components out mice, the axial elements are much less prominent, and
Both genetic and biochemical approaches have identi- the axis of the condensed chromosome is about twofold
fied components of the synaptonemal complex. The longer. Other proteins of the synaptonemal complex,
budding yeast protein Zip1 (mammalian SYCP1) com- including SYCP1, do not assemble properly, and as a result
prises the transverse filaments oriented perpendicular to chromosomes in male germ cells lack chiasmata, are
chromosome axes in mature synaptonemal complex, unpaired, and cells die in pachytene/diplotene. Humans
between the axial elements (Fig. 45.9). Mammalian with mutations in genes for cohesin subunits lack chias-
SYCP1 and Zip1 both consist of an extensive coiled-coil mata, fail to complete meiosis, and are infertile.
flanked by two globular domains but lack amino acid
sequence similarity. Altering the length of the Zip1
coiled-coil changes the spacing between axial elements
Chiasmata
in the synaptonemal complex. Chiasmata are specialized chromatin structures that link
homologous chromosomes together until anaphase I
(Figs. 45.1 and 45.10). They form at sites where pro-
Leptotene SYCP3 + grammed DNA breaks generated by Spo11 undergo the
Paired sister SYCP2,
chromatids Cohesin full recombination pathway to generate crossovers.
Axial element It is not known how crossover events, which repre-
(chromosome axis)
sent exchanges of DNA sequence information, are turned
Chromatin loops into chiasmata. The ultrastructure of chiasmata remains
Time

Rad51, Dmc1 a mystery, but presumably each chiasma consists of two


Early recombination
nodule (may be site of
unperturbed sister chromatid arms intertwined with two
noncrossover events) recombinant arms in which the DNA molecules and their
SYCP1
associated protein structures have been spliced. This
Zygotene (yeast Zip1) DNA complex is held in place on the chromosome by
Synaptonemal complex
Zip2/Zip4, Zip3
(yeast synapsis
cohesion of the distal sister chromatid arms between the
assembles from initiation complex) chiasma and the telomeres. Chiasmata too close to telo-
site of crossover (late Mlh1/Mlh3,
recombination nodule)
Msh4/Msh5 meres can be unstable, presumably because the short
SYCE2, length of sister chromatid arms between them and the
TEX12
telomeres is insufficient for stable cohesion. This can
FIGURE 45.9  PROTEINS OF THE SYNAPTONEMAL COMPLEX.
lead to failure of chromosome segregation in meiosis.
Homologous chromosomes and synaptonemal complex showing the A single chiasma can link homologous chromosomes
locations of some protein constituents. together during meiosis I. Humans have 39 such arms

A B C Microtubules
Fused sister
kinetochores
Paired sister
chromatids
(maternal
homolog)
Distal cohesin
and Aurora B

Chiasma
Paired sister
AIR-2 chromatids
(paternal
Arrows point to chiasmata REC-8 REC-8 homolog)

FIGURE 45.10  BIVALENTS (PAIRED HOMOLOGOUS CHROMOSOMES) ARE HELD TOGETHER BY CHIASMATA AFTER DISAS-
SEMBLY OF THE SYNAPTONEMAL COMPLEX. A, Three diplotene bivalents from the grasshopper species Chorthippus jucundus are held
together by three (left), one (middle), and four (right) chiasmata. The middle cross-shaped bivalent is telocentric; the other two longer bivalents
are submetacentric. (For an explanation of the terminology, see Fig. 7.2.) Lactopropionic orcein staining. B, Caenorhabditis elegans chromosomes
at metaphase I. Aurora B kinase AIR-2 (red) is located distal to chiasmata. Cohesin subunit REC-8 (green) is all along the chromosomes.
C, Explanatory diagram. (A, Courtesy José A. Suja and Julio S. Rufas, Universidad Autónoma de Madrid, Spain. B, Courtesy Josef Loidl, Max
Perutz Labs, Vienna Biocenter, Austria.)
790 SECTION X  n  Cell Cycle

A. Normal B. Infertile male

SB
SB

FIGURE 45.11  ABNORMAL PACHYTENE CHROMOSOMES IN AN INFERTILE MALE. A, Normal pachytene chromosome spread from
a testis biopsy showing synaptonemal complexes (red), MLH1 foci (recombination sites [green]), and centromeres (blue). B, Abnormal pachytene
spread from an infertile patient containing one synaptonemal complex with an area of asynapsis and one synaptonemal complex with a gap
(arrows). SB, sex body (the paired X and Y chromosomes). (Courtesy Renée H. Martin, University of Calgary, Alberta, Canada.)

on the 23 pairs of homologous chromosomes, if one Cohesion and Chromosomal Movements


excludes the five acrocentric short arms, which do not
normally undergo crossovers. Remarkably, there is typi-
During Meiosis I
cally only one chiasma produced for most arms; human Chromosomes in mitosis achieve a dynamic alignment at
males typically have 46 to 53 chiasmata (Fig. 45.11). metaphase as a result of a balance of forces in the spindle.
Even more remarkably, that single chiasma can hold In mitosis, the two kinetochores of the sister chromatids
homologous stably paired for over 40 years in human are attached to microtubules emanating from opposite
females, yet still be released on schedule when the spindle poles, and each chromatid is pulled toward the
oocyte matures into an egg. pole that its kinetochore faces (Fig. 45.12A).
Only a small fraction of DNA breaks formed by Spo11 In meiosis I, homologs linked by chiasmata (called
mature into full crossovers, because a mechanism called bivalents) are balanced at the metaphase plate, but the
crossover interference decreases the likelihood that organization differs in three important ways from mitosis.
DNA breaks near a crossover-designated recombination First, the two kinetochores of the sister chromatids are
event will also become crossovers. This interference fused and act as a single unit oriented toward one spindle
tends to spread crossovers apart across the genome. If pole. The structure of the meiosis I kinetochore is most
all breaks had an equal probability of forming crossovers, easily explained if the two kinetochores are each rotated
small chromosomes might be left without a crossover in 90 degrees toward one another relative to their position
a significant fraction of meiotic nuclei if large chromo- on mitotic chromosomes and then fused (Fig. 45.12A).
somes used all of the structural components necessary In yeast, this coorientation of sister kinetochores requires
to form crossovers and chiasmata. the presence of a meiosis-specific kinetochore protein—
Crossover interference has been defined genetically spo13 (meikin in vertebrates)—that associates with
for almost 100 years, but its mechanism is not certain. sister kinetochores from pachytene until anaphase of
Interference may be mediated by the synaptonemal meiosis I. Spo13/meikin recruits polo kinase to kineto-
complex. Organisms such as the fission yeast Schizosac- chores, but the critical kinase substrates are not known.
charomyces pombe and the mold Aspergillus nidulans Second, the physical connection between the fused
that naturally lack synaptonemal complex also lack kinetochores is not broken in anaphase I. As a result, at
interference. Furthermore, the frequency of meiotic anaphase the two homologs with their paired sister
recombination is directly proportional to the length of chromatids move in opposite directions.
the synapsed chromosome axis (ie, the length of the A third major difference between bivalents in meiosis
synaptonemal complex), rather than the actual length I and mitotic chromosomes is that cohesion of homolo-
of DNA in the chromosome. For example, in human gous chromosome arms distal to chiasmata (Figs. 45.2
females, the synaptonemal complex is roughly 50% and 45.10) rather than cohesion between sister chroma-
longer than in males, and females undergo recombina- tids at centromeres resists the poleward pulling of
tion about twice as frequently as males. However, other the kinetochores at the spindle midzone at metaphase I
observations suggest that interference may be established (Fig. 45.12B). This reflects specialized behavior of the
before the synaptonemal complex forms. meiotic cohesin complex in which the meiosis-specific
CHAPTER 45  n  Meiosis 791

A. Kinetochore movement
Kinetochore

90° rotation
Microtubules of kinetochores

Sister chromatids
Mitosis Meiosis Meiosis

B. Centromere behavior in meiosis

Paired sister
kinetochores
Microtubules
Chiasmata
Recombination

Arms of sisters Homolog
separate pairing
Paired Paired sister
homologs chromatids
Anaphase I Metaphase I Leptotene
Chiasmata released
Homologs separate

Sister Sisters
centromeres segregate
separate
Paired
sisters
Anaphase I Metaphase II Anaphase II
FIGURE 45.12  CHROMOSOMAL BEHAVIOR DURING MEIOSIS I AND II. During meiosis I, sister chromatids are tightly paired along their
lengths, sister kinetochores are fused, and homologs are held together at the metaphase plate by chiasmata. During anaphase I, loss of cohesion
between the arms of sister chromatids releases the chiasmata and allows homologous chromosomes to segregate to opposite spindle poles.
During metaphase of meiosis II, sister chromatids are held together only at their centromeres. Release of centromeric cohesion at meiosis II allows
the sister chromatids to segregate to opposite spindle poles.

Rec8 and Rad21L proteins replace Scc1 (see Figs. 8.18 sister chromatid centromeres tightly paired until ana-
and 44.16). After premeiotic DNA replication, the phase of meiosis II. This protection requires a class of
meiotic cohesion complex keeps sister chromatids proteins called Shugoshins (from the Japanese, meaning
together all along the arms. The cohesin complex plus “guardian spirit”), which recruit the protein phosphatase
synaptonemal complex proteins SYCP3 and SYCP2 are 2A (PP2A). Rec8 must be phosphorylated for separase to
required for assembly of the dense axial elements that cleave it efficiently, so the localized phosphatase can
extend along the length of the chromosome during block the cleavage reaction. Cleavage of centromeric
synapsis. Rec8 releases sister chromatid cohesion at the onset of
In mitosis, cohesion is released between sister chro- anaphase II similar to the release of cohesion during
matid arms during prometaphase, but in meiosis I it is mitosis (see Fig. 44.16).
retained distal to chiasmata until the onset of anaphase
(Figs. 45.7 and 45.12), when Rec8 and Rad21L along Behavior of the Sex Chromosomes
the chromosome arms are cleaved by a protease called
in Meiosis
separase (see Fig. 44.16). Separation of sister chromatid
arms allows the chiasmata to resolve (untangle), and Of the 46 human chromosomes, two sex chromosomes
the homologous chromosomes segregate to opposite carry genes that define the sex of the individual. The
spindle poles. other 22 pairs of chromosomes are called autosomes.
In the meantime, the Rec8 and Rad21L at centromeres If genetic recombination is required to stabilize
are protected from cleavage and continue to hold the homologous chromosomes at the metaphase plate in
792 SECTION X  n  Cell Cycle

recombination enzymes or if the assembly of the syn-


A C aptonemal complex (required for the completion of
recombination) is defective. When such problems are
X 2 detected, cells arrest in meiotic prophase I. In yeast, this
Y
9 has been called the pachytene checkpoint, as cells arrest
late in meiotic prophase with nuclear morphology remi-
B niscent of the pachytene stage. Apoptosis eliminates
mammalian germ cells that arrest owing to defects in
recombination.
Synapsed pseudoautosomal region
Suppression of DNA Replication Between
Y Meiosis I and Meiosis II
X Meiosis is unique in that it involves two M phases with
no intervening S phase. On exit from meiosis I, Cdk1
Condensed unpaired portion
of X and Y chromosomes kinase is reactivated immediately. This blocks assembly
of prereplication complexes (see Fig. 42.8), thereby
FIGURE 45.13  SEX CHROMOSOMES OF A CHINESE
HAMSTER AT PACHYTENE. A–B, The X and Y chromosomes are
blocking DNA replication. At least two pathways contrib-
synapsed at the pseudoautosomal region. Elsewhere, the unpaired ute to reactivation of Cdk1.
chromatin adopts a highly condensed morphology. C, In the same The first involves downregulation of translation of
preparation, autosomes are completely synapsed and show a lesser Wee1 protein kinase in meiosis. Wee1 is a mitotic inhibi-
degree of condensation. (From Dresser ME, Moses MJ. Synaptonemal tor (see Fig. 43.3) that inactivates Cdk1 by phosphoryla-
complex karyotyping in spermatocytes of the Chinese hamster (Crice-
tulus griseus). IV. Light and electron microscopy of synapsis and
tion at Tyr15. The absence of Wee1 in meiosis I was first
nucleolar development by silver staining. Chromosoma. 1980;76: observed in Xenopus laevis but this seems to be a uni-
1–22.) versally conserved way of reactivating Cdk1 without an
S phase. Ectopic expression of Wee1 in mature X. laevis
oocytes prevents reactivation of Cdk1 immediately after
meiosis I, how is this accomplished for the X and Y the meiosis I division. As a result, the oocytes reenter
chromosomes? The answer in most mammals is that the interphase and replicate their DNA. Meiotic cells also
X and Y chromosomes have a short region of homolo- express a specialized isoform of Cdc25, the phosphatase
gous sequence (approximately 2.6 million base pairs in that counteracts Wee1 (see Fig. 43.1).
humans) that does pair and undergo genetic recombina-
tion during meiosis. This pseudoautosomal region Metaphase II Arrest and
must undergo genetic recombination in every meiosis I the Mitogen-Activated Protein
cell for the X and Y chromosomes to be partitioned
correctly. Thus, the X and Y chromosomes act like short
Kinase Pathway
homologous chromosomes with large regions of unre- Following their activation and release from the ovary
lated DNA attached (Fig. 45.13). Unpaired regions of (ovulation), oocytes of many vertebrates arrest in meta-
the X and Y chromosomes acquire a distinct chromatin phase II of meiosis until they are fertilized. The activity
structure during late pachytene. that is responsible for this arrest was discovered in
X. laevis eggs arrested in metaphase of meiosis II and is
called cytostatic factor (CSF). Injection of cytoplasm
Cell-Cycle Regulation of Meiotic Events containing CSF into one blastomere of a two-cell frog
Meiosis employs the full set of functions that regulate embryo blocks the next cell cycle at metaphase, just
the division of somatic cells (see Chapters 40 to 43). like the egg (Fig. 45.14). Therefore, CSF can even
However, the peculiarities of the meiotic cell cycle block somatic cells indefinitely in mitotic metaphase.
require additional regulation. One major difference from CSF activity appears in meiosis II and disappears after
somatic cells is that meiotic chromosomes must undergo fertilization.
recombination and form chiasmata to segregate properly One active component of CSF is the X. laevis homolog
at the first meiotic division. Like somatic cells, meiotic of a well-known viral oncogene, v-mos, the transforming
cells have a DNA damage response that arrests cell-cycle gene of the Moloney murine sarcoma virus, which causes
progression in the presence of DNA breaks (see Fig. solid tumors in mice. The v-mos gene is a mutated
43.11). In addition, they have a “crossover assurance” form of the cellular c-mos gene. Vertebrates express
checkpoint that can detect the presence of stalled or c-mos (a mitogen-activated protein [MAP] kinase kinase
abnormal recombination intermediates. Such intermedi- kinase; see Fig. 27.5) exclusively in oocytes and eggs.
ates accumulate if there are problems with the core Injection of either v-Mos or c-Mos proteins into dividing
CHAPTER 45  n  Meiosis 793

modification creates a binding site for polo kinase, which


A. Two-cell embryo B then also phosphorylates Emi2. Emi2 phosphorylated by
polo kinase is recognized by SCFβTrCP, which ubiquitylates
Inject egg cytosol, it, marking it for destruction. This results in activation of
with activated
c-Mos, into one the APC/C (see Fig. 40.15), termination of the CSF meta-
blastomere phase arrest, and completion of meiosis II.

Timing of Meiosis in Humans


C The fate of cells undergoing meiosis, as well as the timing
of meiotic events, differs significantly between human
c-Mos removed
by antibody males and females.
before injection Males produce approximately 100 million sperm a day
in a process called spermatogenesis. This process
continues throughout adult life. Spermatogenesis starts
with the division of stem cells called spermatogonia
and involves eight divisions prior to meiosis. These
D. The MAP kinase cascade that arrests eggs
in metaphase of meiosis II: divisions are unusual in that cytokinesis is incomplete,
c-Mos MEK MAPK p90Rsk [Emi2] CSF and the cells remain connected by intercellular bridges.
The process could produce up to 256 cells, but usually
some cells die and others fail to divide, so a more typical
FIGURE 45.14  ROLE OF C-MOS IN THE CYTOSTATIC
FACTOR PATHWAY. A, Diagram of the experiment that identified number is around 200 cells arising from the initial
c-Mos as an essential component of cytostatic factor (CSF) required stem cell division. When these cells pass through
for arrest of eggs in meiotic metaphase. One blastomere of an Xenopus meiosis (they are then referred to as spermatocytes) the
laevis embryo at the two-cell stage was injected with cytoplasm from final result is approximately 800 postmeiotic sperma-
a metaphase-arrested egg containing CSF activity. B, This blastomere
tids. Spermatids then undergo a complex program of
(right half of the embryo) remained blocked in metaphase while the left
blastomere divided many times. C, The same experiment was per- differentiation, resulting in the production of highly
formed, but prior to injection, the c-Mos was removed from the egg specialized spermatozoa. The entire process of sper-
cytoplasm by absorption with a specific antibody. Both the injected matogenesis takes approximately 64 days, the bulk of
and uninjected blastomeres continued to divide normally. D, The which is spent in meiosis I. Approximately 16 days are
mitogen-activated protein (MAP) kinase (MAPK) pathway leading to
spent in pachytene, the longest stage of the meiosis I
metaphase II arrest in vertebrate eggs. (B–C, Micrographs courtesy
George Vande Woude, NCI, Frederick, MD. Modified from Sagata N, prophase. In contrast, only about 8 hours are spent in
Watanabe N, Vande Woude GF, et al. The c-Mos proto-oncogene meiosis II.
product is a cytostatic factor responsible for meiotic arrest in vertebrate By the twentieth week of fetal life each ovary of a
eggs. Nature. 1989;342:512–518.) human female contains approximately 3 × 106 primordial
follicles, each with an oocyte arrested in the diplotene
stage of meiosis. This lengthy arrested stage is referred
blastomeres of early frog embryos arrests the cells at to as dictyate. Thereafter, arrested primordial follicles
metaphase (Fig. 45.14). These experiments led to a are recruited continuously to mature into growing
proposal that c-Mos is CSF. primary follicles. However, successful follicular growth
CSF arrest requires the MAP kinase (MAPK) signal depends on follicle-stimulating hormone (FSH), so before
transduction pathway (see Fig. 27.5). Mos activates the puberty—when the hypothalamus-pituitary-ovarian axis
pathway by phosphorylating mitogen activated protein/ matures—all activated oocytes undergo programmed
extracellular signal-related kinase kinase (MEK), which cell death and degenerate in a process known as atresia.
then activates MAPK. MAPK then activates a downstream At birth, the ovary contains approximately 1,000,000
kinase called p90Rsk (Fig. 45.14D). Introduction of germ cells of which approximately 300,000 to 400,000
constitutively active c-Mos or p90Rsk into X. laevis eggs remain at puberty. Following puberty and in response to
induces metaphase arrest like CSF. However, this is not high levels of FSH each month, a cohort of follicles with
the whole story, because metaphase arrest is maintained their dictyate-arrested oocytes is activated to complete
in extracts depleted of p90Rsk. Thus the pathway must meiosis and grow. Only one of these activated oocytes
include at least one unidentified step beyond p90Rsk. matures fully and is shed from the ovary in response to
The extra component of CSF is an anaphase-promoting a surge of luteinizing hormone. The other follicles degen-
complex/cyclosome (APC/C) inhibitor called Emi2. A erate by atresia. As the successful oocyte is shed from
burst of cytoplasmic Ca2+ released at fertilization (see Fig. the ovary, it completes meiosis I and is arrested at meta-
26.15) activates Ca2+/calmodulin-dependent protein phase of meiosis II by CSF. It remains arrested at this
kinase II (CaMKII), which phosphorylates Emi2. This stage until fertilization occurs. By the time of menopause
794 SECTION X  n  Cell Cycle

during fetal development. (Any fetal death is a spontane-


ous abortion, commonly called a miscarriage.) It is now
First polar body thought that at least 30% of all conceptions result in
spontaneous abortions. Furthermore, more than 60% of
those spontaneous abortions are aneuploid. These figures
Metaphase II oocyte probably underestimate the frequency of meiotic abnor-
malities. Many spontaneous abortions occur very early
during pregnancy and many are never detected at all.
Few fetuses lost in the first 4 to 6 weeks of gestation are
tested in a laboratory, so their karyotypes are unknown.
Meiotic errors involving certain autosomes can
produce fetuses that survive to birth. Females with three
50 µm or more copies of the gene-rich X chromosome survive,
because all but one X chromosome are inactivated
FIGURE 45.15  POLAR BODY FORMATION. Asymmetric cleav- (silenced) in somatic cells (see Fig. 8.6). Rare individuals
age during mouse oogenesis produces a large oocyte and small polar with three copies of chromosome 13 or chromosome18
body. (Modified from Jiao ZX, Xu M, Woodruff TK. Age-associated
alteration of oocyte-specific gene expression in polar bodies: potential
survive to birth; but those who do typically die shortly
markers of oocyte competence. Fertil Steril. 2012;98:480–486.) thereafter. The exception is individuals trisomic for
chromosome 21 (a condition that is commonly known
as Down syndrome). These individuals have intellec-
the ovarian reserve is almost depleted, leaving approxi- tual disability as well as other characteristic phenotypic
mately 1000 primordial follicles in the ovary. features, including decreased life expectancy. Why do
In human females, meiosis produces only one mature individuals with Down syndrome survive whereas others
egg. Both meiotic cell divisions are asymmetrical, pro- affected by aneuploidy do not? Perhaps the very small
ducing one large and one very small and short-lived cell. number (233) of coding genes on chromosome 21
The small cells are called polar bodies (Fig. 45.15). includes none whose dosage is critical for survival.
The frequency of certain types of aneuploidy, such as
trisomy for chromosome 21, increases with the ages of
Meiotic Defects and Human Disease the mother and father. Only 0.04% of children of 20-year-
Abnormalities in meiosis are surprisingly common but are old mothers old have trisomy 21. This number rises
not widely observed in human populations because their dramatically with maternal age; nearly 5% of the concep-
consequences are extremely severe. In fact, meiotic tions in mothers 45 years old have trisomy 21 (Fig.
abnormalities are a leading cause of fetal death, particu- 45.16). This maternal age effect is a leading cause of
larly during the first trimester of pregnancy in humans. human genetic disease. Some believe that during the
The two major causes of problems are chromosome many years of arrest of oocytes in meiosis I dictyate,
nondisjunction during the meiotic divisions and the gen- chiasmata joining homologous chromosomes gradually
eration of unbalanced chromosomal rearrangements via dissociate. A mechanism to explain this might be the
faulty recombination. progressive loss of cohesion between sister chromatids
When chromosomes fail to segregate properly in one as the mother ages. There appears to be no mechanism
or both meiotic divisions (nondisjunction), the daughter to replace cohesin complexes that are gradually lost in
cells lack the normal haploid complement of chromo- dictyate oocytes. Mice with a mutation in a key subunit
somes. Embryos that have gained an entire set of chro- of the cohesin complex (Fig. 45.7; see also Fig. 8.18)
mosomes are referred to as polyploid. In human exhibit a pattern of chromosome nondisjunction with
embryos, polyploidy is a common type of chromosomal increasing maternal age that looks much like that seen
abnormality. It is estimated that 1% to 3% of all concep- in aging human mothers. Another potential source of
tions are triploid (69 chromosomes; 23 from one parent errors lies in the mechanism of spindle assembly in
and 46 from the other parent). Two-thirds of these arise oocytes, which does not involve centrosomes and
from two sperm fertilizing one egg (nothing wrong with appears to be more prone to errors than in somatic cells
meiosis there). In other cases, they come from a diploid (see Chapter 44).
gamete, the result of a defective meiotic segregation. Not all cases of human aneuploidy originate from the
Very few triploid embryos survive to birth. mother. One of the most common aneuploidies, 45,X
Most chromosomal abnormalities in human embryos (see Table 45.1 for an explanation of nomenclature)
result from the loss or gain of one or more chromosomes involves the loss of the paternal X or Y chromosome 70%
during meiosis. This condition is called aneuploidy. to 80% of the time. This aneuploidy accounts for nearly
Most zygotes that arise from aneuploid gametes die 10% of spontaneous abortions. In addition, about 7% of
CHAPTER 45  n  Meiosis 795

TABLE 45.1  Aneuploidies Involving the Sex Chromosomes in Newborn Humans


Karyotype Frequency Sex Comments
47,XXY* 1/1000 M Klinefelter syndrome. Increased height, sterile, a proportion may
have some learning difficulties.
47,XYY 1/1000 M Increased height, generally fertile, typically with chromosomally
normal offspring. A proportion may have some learning difficulties.
Other X or Y aneuploidy 1/1350
Total: 1 in 360 male births
47,XXX 1/900 F Increased height, generally fertile, typically with chromosomally
normal offspring. A proportion have serious learning difficulties.
45,X 1/4000 F Turner syndrome. Reduced height, infertile, normal intelligence.
Of 45,X embryos, 99% terminate as spontaneous abortions.
Other X or Y aneuploidy 1/2700
Total: 1 in 580 female births
*This number gives the total number of chromosomes, followed by the complement of sex chromosomes.
Modified from Nussbaum RL, McInnes RR, Willard HF. Genetics in Medicine. 6th ed. Philadelphia, PA: WB Saunders; 2001:150.

[Table 45.1]) reveal that spontaneous abortion is a highly


efficient protective mechanism for the elimination of
chromosomal imbalances that arise from errors in
meiosis.
Chromosome number
3.0

ACKNOWLEDGMENTS
16
We thank Abby Dernburg, Jim Haber, Scott Hawley, Amy
% Trisomies

MacQueen, Adele Marston, and Alberto Pendas for their


2.0 suggestions on revisions to this chapter.

21
SELECTED READINGS

1.0 de Massy B. Initiation of meiotic recombination: how and where?


Conservation and specificities among eukaryotes. Annu Rev Genet.
2013;47:563-599.
18 Duro E, Marston AL. From equator to pole: splitting chromosomes in
mitosis and meiosis. Genes Dev. 2015;29:109-122.
Fraune J, Schramm S, Alsheimer M, Benavente R. The mammalian
0 synaptonemal complex: protein components, assembly and role in
<19 20–24 25–29 30–34 35–39 40
meiotic recombination. Exp Cell Res. 2012;318:1340-1346.
Maternal age [years] Gutiérrez-Caballero C, Cebollero LR, Pendás AM. Shugoshins: from
FIGURE 45.16  RELATIONSHIP BETWEEN MATERNAL AGE protectors of cohesion to versatile adaptors at the centromere.
AND THE INCIDENCE OF HUMAN TRISOMIES. (Modified from Trends Genet. 2012;28:351-360.
Nagaoka SI, Hassold TJ, Hunt PA. Human aneuploidy: mechanisms Hiraoka Y, Dernburg AF. The SUN rises on meiotic chromosome
and new insights into an age-old problem. Nat Rev Genet. 2012; dynamics. Dev Cell. 2009;17:598-605.
13:493–504.) Lam I, Keeney S. Mechanism and regulation of meiotic recombination
initiation. Cold Spring Harb Perspect Biol. 2014;7:a016634.
instances of trisomy 21 are of paternal origin. Clearly, Nagaoka SI, Hassold TJ, Hunt PA. Human aneuploidy: mechanisms and
new insights into an age-old problem. Nat Rev Genet. 2012;13:
more than one mechanism is responsible for the genera-
493-504.
tion of aneuploid offspring. Scherthan H. A bouquet makes ends meet. Nat Rev Mol Cell Biol.
These rather sobering statistics reveal two important 2001;2:621-627.
facts about human reproduction. First, the production Schmidt A, Rauh NR, Nigg EA, Mayer TU. Cytostatic factor: an activity
of gametes is error prone. This has been confirmed by that puts the cell cycle on hold. J Cell Sci. 2006;119:1213-1218.
Subramanian VV. Hochwagen A: The meiotic checkpoint network:
direct studies, in which 20% of eggs and 3% to 4% of
step-by-step through meiotic prophase. Cold Spring Harb Perspect
sperm were found to have chromosomal abnormalities. Biol. 2014;6:a016675.
Second, the much lower rates of chromosomal abnor- Zickler D, Kleckner N. Recombination, Pairing, and Synapsis of Homo-
malities seen in live births (approximately 0.3% overall logs during Meiosis. Cold Spring Harb Perspect Biol. 2015;7(6).
This page intentionally left blank
CHAPTER 46 

Programmed Cell Death

Necessity for Cell Death in stimuli, particularly when induction of apoptosis is


inhibited. Programmed necrosis looks morphologically
Multicellular Organisms
like accidental cell death. A third pathway leading to cell
The ability to undergo programmed cell death (Box death involves autophagy (see Fig. 23.7). Although
46.1) is a built-in latent capacity in most cells of multi- usually regarded as a protective response to starvation,
cellular organisms. Cell death is important for embryonic autophagy has been implicated in certain examples of
development, maintenance of tissue homeostasis, estab- cell death, particularly during development.
lishment of immune self-tolerance, killing by immune Necrosis corresponds to what most of us naively
effector cells, and regulation of cell viability by hormones imagine cell death would be like. Owing to lack of cel-
and growth factors. Both extrinsic signals and internal lular homeostasis, water rushes into the dying cell,
imbalances can lead cells to kill themselves. Furthermore, causing it to swell until the plasma and organelle mem-
many metazoan cells will die if they fail to receive sur- branes burst. As a result, the cell undergoes a generalized
vival signals from other cells. Abnormalities of the cell
death program contribute to several important diseases,
including cancer, Alzheimer disease, and AIDS. Cell
death programs are ancient: much of the current network NEIGHBORING CELL
Fas ligand
was present in the last eumetazoan common ancestor TNF
(see Fig. 2.8). Fas receptor
TNF
receptor

Programmed Cell Death Versus Accidental Extrinsic


DISC
Cell Death: Apoptosis Versus Necrosis pathway
Complex II Complex I
Active
Although cells die in many ways, it is useful to focus on Bcl2 caspases
killer
two poles of this spectrum: apoptosis and necrosis. Fig. Necroptosis
Survival

46.1 summarizes the major pathways of programmed


Apoptosis Inflammation
cell death. (Details are filled in as we progress through
the chapter.) Apoptosis, the most widely studied MITOCHONDRION Pyroptosis
pathway for programmed cell death, is cellular suicide Apoptosome
resulting from activation of a dedicated intracellular Bcl2
killer
Signaling
Inflammasome
program. Fig. 46.2 shows a detailed description of the Intrinsic platforms
pathway
events of apoptosis. At the other end of the spectrum Pathogen
DNA damage, etc
necrosis, also called accidental cell death, occurs when attack NUCLEUS
cells sustain a structural or chemical insult that causes
FIGURE 46.1  OVERVIEW OF PROGRAMMED CELL DEATH
the cells to swell and undergo membrane lysis (Fig.
DISCUSSED IN THIS CHAPTER. All these types of death hinge on
46.3). Examples of such insults include extremes of the assembly of a signaling platform (boxed) that is often involved in
temperature and physical trauma. Cells can also initiate activating proteases called caspases. These pathways are all dis-
active programmed necrosis in response to certain cussed in greater detail in the text.

797
798 SECTION X  n  Cell Cycle

BOX 46.1  Key Terms Apoptosis

Apoptosis: Type of programmed cell death that was


identified due to a particular pattern of morphologic Junctions
changes but now is defined by the action of molecular Mitochondria
pathways involving cell surface receptors or mitochon-
dria and resulting in the activation of specialized pro- Nucleus
teases. The name comes from the Greek, referring to
shedding of the petals from flowers or leaves from
Microvilli contract
trees. Apoptotic death occurs in two phases. During Intercellular junctions break
the latent phase, the cell looks morphologically normal Chromatin begins to condense
but is committed to death. The execution phase is
characterized by a series of dramatic structural and
biochemical changes that culminate in fragmentation
of the cell into membrane-enclosed apoptotic bodies.
Activities that cause cells to undergo apoptosis are said
to be proapoptotic. Activities that protect cells from
apoptosis are said to be antiapoptotic.
Autophagic Cell Death: It is still debated how widely Cell shrinks
autophagy is used as a pathway for cell death, although Chromatin condenses around
it is accepted that the pathway (which is widely nuclear periphery
assumed to be primarily a survival pathway when cells
are starved for nutrients) can promote cell death during
development. Autophagy may also either promote or
inhibit apoptosis under specialized circumstances.
Necroptosis: Programmed necrosis that occurs when
tumor necrosis factor (TNF) and certain other cell-
surface receptors are activated. Activation of these
receptors normally leads to a proinflammatory response
and cell survival, but can lead to apoptosis. If certain Cell blebs violently
Chromatin condensation
components of the apoptotic pathway are missing, continues
cells instead undergo necroptosis, apparently as a
backup pathway.
Necrosis (Accidental Cell Death): Death that results
from irreversible injury to the cell. Cell membranes
swell and become permeable. Lytic enzymes destroy
the cellular contents, which then leak out into the inter-
cellular space, leading to an inflammatory response.
Programmed Cell Death: Any active cellular process
that culminates in cell death. This may occur in Cell fragments into membrane-
response to developmental or environmental cues or enclosed apoptotic bodies
as a response to physiological damage detected by the
cell’s internal surveillance networks.
Pyroptosis: Often in response to intracellular pathogens,
this involves activation of caspase 1. The infected
cells secrete interleukin-1β and interleukin-18, which
promote an inflammatory response, and also undergo
a form of cell death that resembles necrosis.

Apoptotic bodies phagocytosed


by neighboring cells and
roving macrophages

FIGURE 46.2  APOPTOSIS—ACTIVE SUICIDE—TYPICALLY


AFFECTS SINGLE CELLS. Neighboring cells remain healthy. Apop-
totic cell death usually does not lead to an inflammatory response.
CHAPTER 46  n  Programmed Cell Death 799

Necrosis
Trauma Dissolution of
cellular structures

Junctions

Mitochondria

Nucleus

Cells and organelles swell Cell lysis


Chromatin condenses Invasion of phagocytic cells
Membranes compromised: Inflammation
H 2O fluid rushes in H2O

FIGURE 46.3  NECROSIS USUALLY RESULTS FROM IRREVERSIBLE INJURY TO CELLS. Typically, groups of cells are affected. In most
cases, necrotic cell death leads to an inflammatory response (red “activated” macrophages).

Point of Reprieve
Insult no return (very rare)

Latent Execution
Condemned Committed
Rescue by Rescue usually Morphologic
survival factors impossible changes
FIGURE 46.4  TWO PHASES OF APOPTOSIS. The latent phase
can be subdivided into two stages: a condemned stage, during which
the cell is proceeding on a pathway toward death but can still be
rescued if it is exposed to antiapoptotic activities, and a committed
stage, beyond which rescue is usually impossible.

process of autodigestion and dissolution, until it spills its


cytoplasmic contents out into the surroundings (Fig.
46.3). This produces local inflammation as phagocytic
cells are activated, flock to the site, and ingest the debris FIGURE 46.5  SCANNING ELECTRON MICROGRAPH OF
(see Figs. 22.3 and 30.13). Because agents that damage INTACT AND APOPTOTIC MOUSE SARCOMA CELLS. Intact cells
cells act over areas that are large in comparison to the are covered with microvilli, whereas apoptotic cells have numerous
smooth blebs. These cells were stimulated to undergo apoptosis as a
size of a single cell, necrosis often involves large groups result of interference with RNA metabolism. (From Wyllie AH, Kerr JFR,
of neighboring cells. Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol.
In contrast to necrosis, apoptotic cells shrink rather 1980;68:251–306.)
than swelling and they typically do not lyse (Fig. 46.2).
Apoptosis is a two-stage process. On receipt of the Ultimately, the cells enter the execution phase
proapoptotic signal that triggers the pathway to death, of apoptosis, lasting about an hour, during which
cells enter a latent phase of apoptosis (Fig. 46.4). The they undergo dramatic morphologic and physiological
duration of the latent phase of apoptosis, during which changes. These include:
cells appear healthy, can be extremely variable, ranging • Loss of microvilli and intercellular junctions (Fig. 46.5)
from a few hours to several days. • Shrinkage of the cytoplasm
800 SECTION X  n  Cell Cycle

• Dramatic changes in cytoplasmic motility with activa-


tion of violent blebbing (Fig. 46.6)
• Loss of plasma membrane lipid asymmetry (see Fig.
13.7), so the distribution of phosphatidylserine is
randomized and it appears in the outer leaflet of the
membrane
• Hypercondensation of the chromatin and its collapse
against the nuclear periphery
• The “explosive” fragmentation of the cell into
membrane-enclosed apoptotic bodies that contain
remnants of the nucleus, mitochondria, and other 0 min 114 116
organelles.
All these changes are instigated by the action of a
specific set of death-inducing proteases and are discussed
at length later.
In tissues, surrounding cells rapidly phagocytose
apoptotic bodies in response to the phosphatidylserine
and other markers on their surfaces (Fig. 46.7). Apoptosis
can thus be considered to be the disassembly of the cell
into “bite-sized” membrane-bound vesicles, so the cel-
lular contents are not usually released into the environ- 118 120 148
ment unless phagocytosis is delayed. Surface markers on
apoptotic bodies cause cells that ingest them to secrete FIGURE 46.6  APOPTOSIS OF A TRANSFORMED PIG KIDNEY
antiinflammatory cytokines. As a result, apoptotic death CELL FOLLOWING EXPOSURE TO ETOPOSIDE, A DRUG USED
usually does not lead to an inflammatory response and IN CANCER CHEMOTHERAPY. The dramatic cytoplasmic blebbing
results in the disassembly of the cell into membrane-enclosed vesicles.
apoptotic cells seem to vanish from tissues without (Courtesy L.M. Martins and K. Samejima, Wellcome Trust Institute for
a trace. Cell Biology, University of Edinburgh, United Kingdom.)

A. Attraction of phagocytes via D


soluble “find me” signals
n
Mi
gration sign

Phagocyte Apoptotic
cell
al
s

B. Recognition and phagocytosis via


displayed “eat me” signals and
lack of “don’t eat me” signals

C. Production of antiinflammatory
cytokines

IL-10
TGF-β

FIGURE 46.7  PHAGOCYTOSIS OF APOPTOTIC CELLS. A–C, Phagocytosis that occurs when cells express “eat me” signals results in the
production of antiinflammatory cytokines. D, Electron micrograph of a phagocytosed apoptotic body containing a nuclear fragment. The nucleus
(n) of the epithelial cell that engulfed this apoptotic body is shown at top. In this case, apoptosis occurred during allograft rejection in a pig.
(A, Modified from Lauber K, Blumenthal SG, Waibel M, et al. Clearance of apoptotic cells: getting rid of the corpses. Mol Cell. 2004;12:277–287.
B, From Wyllie AH, Kerr JFR, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol. 1980;68:251–306.)
CHAPTER 46  n  Programmed Cell Death 801

Examples of Cells That Undergo properly, the T-cell receptor must recognize major his-
tocompatibility complex (MHC) glycoproteins on other
Programmed Cell Death
thymic cells during antigen presentation (see Fig. 27.8).
The following are six common causes of programmed T lymphocytes whose T-cell receptors cannot interact
cell death (Fig. 46.8). with the spectrum of MHC glycoproteins expressed in a
given individual are ineffective in the immune response.
Developmentally Defective Cells These cells die by apoptosis in a process known as posi-
During molecular maturation of T-lymphocyte antigen tive selection (Fig. 46.9).
receptors (see Figs. 27.8 and 28.9), immature T cells in Many of the newly created receptors bind to foreign
the thymus (known as thymocytes) rearrange the genes antigens, but others interact with self-antigens. The
encoding the receptor α and β chains. To function latter are potentially harmful and are eliminated through

A B C D
Up to 80% of
Epithelial cells must die neurons die in
to allow fusion of palate some ganglia

Mammary epithelium Over 95% of


cells die when deprived immature
of hormones at end of T cells die
lactation in thymus

Cells of Müllerian
ducts die in males E12.5 E13.5 E14.5

Prostate cells die Cells of


interdigital E
when deprived of
hormone webbing die Dying cells
(yellow)

FIGURE 46.8  PROGRAMMED CELL DEATH DURING DEVELOPMENT. A, Examples of cells that undergo programmed cell death.
B–D, Programmed cell death in the embryonic mouse paw. At day 12.5 of development, the digits are fully connected by webbing. By day 13.5,
the webbing has started to die, and by day 14.5, all the webbing cells are gone. E, Cells undergoing programmed cell death take up acridine
orange, whereas cells of the surrounding healthy tissue do not. (Micrographs courtesy William Wood and Paul Martin, Department of Anatomy
and Developmental Biology, University College of London, United Kingdom.)

THYMUS PERIPHERY, SPLEEN, AND LYMPH NODES

TCR-β gene Pre-TCR TCR-α gene TCR T-cell


rearrangement rearrangement stimulation

Common T-cell Pro-T4 cell Immature Mature Mature T-cell T-cell blast
precursor thymocyte thymocyte

No IL-7R signal No productive No TCR signal (no Autoreactive Lack of TCR No growth-
TCR-β gene positive selection) TCR (negative signal factor signals
rearrangement No productive TCR-α selection) Strong TCR
gene rearrangement restimulation

APOPTOSIS

FIGURE 46.9  EXAMPLES OF SIGNALS THAT PROMOTE DIFFERENTIATION OR PROGRAMMED CELL DEATH OF IMMATURE
THYMOCYTES IN THE THYMUS AND MATURE T CELLS IN THE PERIPHERY. Thymocytes that make functional T-cell receptors (TCRs)
and do not recognize self-antigens mature, provided that they receive survival signals, such as interleukin-7. Pro-T cells are also known as
double-negative thymocytes (referring to their lack of the two cell-surface markers CD4 and CD8). Immature thymocytes express CD4 and CD8
and are known as double-positive thymocytes. Thymocytes undergo apoptosis if they produce defective T-cell receptor, recognize self-antigens,
suffer DNA damage, or receive a death stimulus (glucocorticoid hormone). More than 98% of immature thymocytes die without leaving the thymus.
(Modified from Strasser A. The role of BH3-only proteins in the immune system. Nat Rev Immunol. 2005;5:189–200.)
802 SECTION X  n  Cell Cycle

apoptosis in a process known as negative selection critical threshold, these organs virtually disappear in a
(Fig. 46.9). The drug cyclosporine, which inhibits apop- relatively brief time as their constituent cells undergo
tosis in thymocytes, can cause autoimmune disease. massive apoptotic death. Should levels of circulating
Overall, defects in T-cell receptor assembly are extremely androgens rise again, the remaining prostatic stem cells
common, and up to 98% of immature T cells die by proliferate and reconstruct the gland. A similar cycle of
apoptosis without leaving the thymus. growth and involution is seen in the mammary gland of
Similar positive and negative selection steps occur female mammals, which exhibits substantial differences
during the maturation of B lymphocytes (see Fig. 28.10), in size and cellular composition in the lactating and
which is accomplished by a combination of gene rear- nonlactating states. Interference with survival signaling
rangements and facilitated mutagenesis. B lymphocytes by sex hormones is one important strategy that is com-
expressing antibodies directed against self-antigens or monly used in the treatment of breast and prostate cancer.
producing antibodies whose affinity for antigen is below Programmed cell death also eliminates certain popula-
a critical threshold are eliminated through apoptosis. tions of cells that never serve any function. The müllerian
ducts develop into the female oviduct. Male embryos
Excess Cells also develop progenitors of these ducts, which serve no
Programmed cell death is also widely used for quality purpose and are eliminated by apoptosis.
control during development. For example, in the brain,
embryonic ganglia often have many more neurons than Cells With Perturbed Cell Cycles
are required to enervate their target muscles. Production Chapters 40 to 43 describe how biochemical circuits
of excess cells is part of a Darwinian strategy to ensure called checkpoints regulate the cell cycle. Cells respond
that a sufficient number of axons reach their targets. to DNA damage by blocking cell-cycle progression while
Programmed cell death eliminates excess neurons that attempting to repair the DNA. The p53 transcription
fail to make appropriate connections. Up to 80% of factor, an important downstream checkpoint EFFECTOR
neurons in certain developing ganglia die in this way. induces the expression of genes encoding proteins that
Because of the importance of apoptosis during its devel- arrest the cell cycle. p53 also turns on genes encoding
opment, the brain is often seriously affected in mice proteins that induce cell death. It is generally thought
engineered to lack components of the apoptotic pathway. that if the damage cannot be repaired quickly, the pro-
death factors win out, and the outcome is apoptosis.
Cells That Serve No Function Double-strand DNA breaks induced by ionizing radiation
The elimination of obsolete cells whose function has and DNA damage induced by chemotherapeutic agents
been completed is most evident in organisms, such as frequently result in cell death rather than repair.
insects and amphibians that undergo metamorphosis A second important cell-cycle checkpoint regulates
during development. For example, a burst of thyroid the transition from the G1 phase to S phase. Passing the
hormone initiates programmed cell death for resorption restriction point (see Fig. 41.8) represents the commit-
of the tadpole tail. ment of the cell to undergo another cycle of DNA repli-
Mammals also use programmed cell death to eliminate cation and division. Restriction point control centers on
obsolete tissues during development. For example, the regulation of the E2F family of transcription factors.
during human embryonic development, the digits of E2F regulates genes that promote cell-cycle progression.
hands and feet are connected by a tissue webbing that E2F also regulates the expression of genes that promote
is eliminated by programmed cell death (Fig. 46.8). apoptosis. If activated too strongly, E2F can initiate
During craniofacial development, the hard palate apoptosis as, for example, after DNA damage (see Fig.
develops from two lateral precursors, each covered in a 41.14) or when restriction point control has broken
protective layer of epithelial cells. As the two halves down. Cells that die in response to inappropriate
grow together at the midline of the nasopharynx, they signals to proliferate include those infected by certain
remain separated by this epithelial covering until, in viruses or overexpressing genes that drive cell prolifera-
response to a developmental cue, the epithelial cells at tion (such as c-myc and c-fos [see Fig. 46.15]). Apoptotic
the midline undergo programmed cell death. Then the death of cells with an inappropriate stimulus to prolifer-
two halves of the palate can fuse. Failure of the epithelial ate is an important defense against cancer.
cells to die at the appropriate time can interfere with the
fusion of the bone, causing cleft palate. Virus-Infected Cells
Populations of cells that are fully functional may Cytotoxic T lymphocytes eliminate virus-infected cells
become obsolete as a result of physiological changes by causing them to undergo programmed cell death
in the status of an organism. For example, in male either by apoptosis or by a second related pathway. For
mammals, the prostate and other accessory glands of the example, programmed cell death accounts for at least
reproductive system are regulated by the levels of circu- part of the loss of mature CD4+ T-helper cells (see Fig.
lating male hormone. If hormone levels fall below a 28.9) in people infected with HIV-1. When exposed to
CHAPTER 46  n  Programmed Cell Death 803

agents that normally stimulate cell proliferation, these in the phagocytosis and subsequent processing of the
cells instead undergo apoptosis. Paradoxically, it appears cell corpses. These mutants are collectively known as
that many of the dying cells are not themselves infected “cell death abnormal” (ced) mutants. Interestingly, this
with HIV. repertoire of nematode genes is vastly simplified from
the apoptosis genes in the last eumetazoan com­mon
Chemotherapeutic Killing of Cells ancestor (see Fig. 2.8). By contrast, humans have
Exposure of cancer cells to many of the agents used in approximately 300 genes involved in apoptosis.
chemotherapy does not kill the cells outright. Instead, The three best-known worm cell death genes are ced-
they die because the drugs cause intracellular damage 3, ced-4, and ced-9. If either ced-3 or ced-4 is inactivated,
that acts as a signal for the induction of apoptotic cell all cells throughout the organism that should die by
death. Thus, the treated cancer cells kill themselves. apoptosis are reprieved. These cells remain alive and are
apparently functional. Interestingly, these worms have
normal life spans. This suggests that programmed cell
Genetic Analysis of Apoptosis death is not involved in the normal aging process, at least
Several key components that are involved in the apop- not in C. elegans. Ced-9 negatively regulates ced-3 and
totic execution of mammalian cells were discovered by ced-4. In worms with ced-9 loss-of-function mutations
genetic analysis of the nematode worm Caenorhabditis many cells die that normally stay alive. This is deleterious
elegans. Because C. elegans is optically clear, it is pos- for the organism, so ced-9 mutants die. The key gene
sible to see every cell in a developing worm using dif- triggering apoptosis is egl-1. Its regulation appears to
ferential interference contrast optics (see Fig. 6.2). This govern apoptosis in the worm. All these genes have
enabled investigators to trace the lineage of every cell in mammalian counterparts (discussed more fully later).
an adult worm back to the fertilized egg. These studies Ced-3 is a member of a specialized family of cell
led to the surprising discovery that programmed cell death proteases called caspases. Ced-4 is a scaffolding/
death is one of the most common fates for newborn C. adapter protein that plays an essential role in the activa-
elegans cells. Of the 1090 somatic cells that are produced tion of Ced-3 from its zymogen precursor. The mamma-
during embryogenesis of the C. elegans hermaphrodite, lian counterpart of Ced-4 is apoptotic protease-activating
131 undergo programmed cell death at reproducible factor-1 (Apaf-1). Ced-9 is a member of the Bcl-2 family
locations and times. of cell death regulators (Fig. 46.14). Some Bcl-2 family
Mutations in at least 14 C. elegans genes affect pro- members protect against cell death, but others actively
grammed cell death (Fig. 46.10). These are divided into promote cell death. Egl-1 is a BH3-only proapoptotic
three classes: (a) genes that mark cells for subsequent regulator.
programmed death; (b) genes that are involved in cell Phagocytosis turned out to be surprisingly complex,
killing and its regulation; and (c) genes that are involved with eight C. elegans genes encoding proteins that
are involved in the engulfment and processing of cell
corpses. Several are signaling proteins that reorganize
egl-1 Determination the cytoskeleton to permit the cell to move toward and
ced-9 ced-3 engulf its target. Another, the nuc-1 gene, encodes one of
ced-4 Killing
several nucleases that digest the DNA of the dead cell in
lysosomes of cells that ingest the corpse. In mammals, this
Dead cell digestion typically is initiated within the dying cell itself.
ced-1, -6, -7
ced-2, -5, -10, -12 Engulfment Signals and Pathways of Apoptosis
Two principal pathways lead to cell death by apoptosis.
These are introduced only briefly here and expanded
upon later. The intrinsic pathway (see Fig. 46.17) is
nuc-1 Degradation
activated by internal surveillance mechanisms or signals
sent (or not sent) by other cells. Signals that induce this
pathway include DNA damage, exposure to chemicals
Mammalian homologs
that interfere with a variety of cellular pathways and
ced-9 protein = Bcl-2 family (antiapoptotic) excessive activation of factors that promote cell-cycle
egl-1 protein = Bcl-2 family (BH3-only proapoptotic) progression. Withdrawal of nutrients or survival signals
ced-4 protein = Apaf-1
ced-3 protein = initiator caspases from the environment also activates the intrinsic
pathway. Those survival signals include lymphokines,
FIGURE 46.10  GENETIC DISSECTION OF PROGRAMMED
CELL DEATH. The ced (cell death abnormal) mutants of the nematode such as interleukin-2 and interleukin-3, which are essen-
worm Caenorhabditis elegans affect the killing, engulfment, and deg- tial for survival of thymocytes; nerve growth factor,
radation stages of programmed cell death. which is required for survival of many neurons; and
804 SECTION X  n  Cell Cycle

extracellular matrix, which is required for survival of specifically activate other factors that promote cell death.
epithelial cells. Signals that activate the intrinsic pathway In certain specialized instances caspases can participate
converge on mitochondria, which release key factors in pathways leading to activation of nuclear factor κB
that drive the apoptotic response. (NF-κB; see Figs. 10.21 and 27.8) and the immune
Signals from other cells are the primary triggers of the response and also in terminal differentiation. These
extrinsic pathway (see Fig. 46.18). Direct contact of a nonlethal roles of caspases are not discussed further here.
killer cell with the target cell activates specific receptors C. elegans has three caspases, one of which (Ced-3)
that initiate this pathway, starting at the plasma mem- is essential for cell death. In contrast, mammals have as
brane. Activation of the extrinsic pathway is one strategy many as 17 caspase-related genes (Fig. 46.11A). Analysis
that cytotoxic T lymphocytes use to kill cells that are based on sequence comparisons divides caspases into
recognized as foreign (or as harboring foreign patho- two major subfamilies. The caspase 1 subfamily encodes
gens). This pathway is also widely used to control cell enzymes that process pro–interleukin-1β to yield mature
populations in the immune system. In many cells, the interleukin-1β and pro–interleukin-18 to yield mature
extrinsic pathway activates the intrinsic pathway, which interleukin-18. Macrophages secrete these cytokines,
is then responsible for killing the cell. which cause fever and inflammation. The second caspase
3 subfamily of enzymes participates almost exclusively
Protein Regulators and Effectors of Apoptosis in apoptotic cell death.
Because the penalty for misregulation of apoptosis is Like many proteases, caspases are synthesized as
death, it is not surprising that the process is carefully inactive zymogens, known as procaspases. All living
regulated. This is essential for cells but complicates vertebrate cells synthesize procaspases constitutively.
matters for students. This section first lays out the Procaspases typically consist of three domains: an N-
over­all strategy in generic terms and then fills in some terminal prodomain followed by the large and small
important details. subunits of the mature enzyme (Fig. 46.11A–B). Aspar-
A cascade of proteases called caspases drives apop- tate residues between these three domains are targets
tosis. Each caspase is harmless until activated (usually by for cleavage by caspases. Procaspases are activated either
dimerization for initiator caspases and proteolytic cleav- by multimerization or by cleavage and release of the
age for effector caspases). Activation of a small number prodomains. Following procaspase cleavage, the two
of initiator caspases triggers a cascade that amplifies the large and two small subunits associate in a compact,
response and results in activation of numerous effector block-like heterotetrameric molecule (Fig. 46.11B–D).
caspases. The ability of effector caspases to activate Depending on the enzyme, multimerization or cleavage
further initiators and effectors further amplifies the of the procaspase permits a major conformational change
response. in the polypeptide, creating two stable active site pockets
This strategy of employing amplification and positive between the large and small subunits.
feedback has two powerful advantages. First, it can Two classes of caspases are involved in cell death.
provide a very rapid change in the state of the cytoplasm, Initiator caspases have long prodomains (Fig. 46.11A).
from pro-life to pro-death within seconds. Second, the They exist as monomers in cells and become autoactivated
relatively small number of initiator caspases that trigger when scaffolding cofactors promote their aggregation.
the cascade constitutes a feasible target for negative regu- Activation is induced by dimerization and does not require
lators that can rapidly quell responses that are initiated proteolytic cleavage (although cleavage typically follows
under borderline conditions or by mistake. This is benefi- activation). Sequences within the extended prodomains
cial but also complicates the overall system. If initiator are involved in targeting the initiator procaspases to the
caspases start apoptosis and are then inactivated by appropriate cellular locations and in interactions with
suppressers, how does the response ever take hold? The scaffolding factors.
answer is at least one more level of regulation: inhibitors Effector caspase zymogens are monomers with
of the inhibitors. This additional layer of regulation is short prodomains. These inactive enzymes are typically
thought to enable the rapid burst of caspase activation incapable of autoactivation. Instead, they are activated
that triggers the onset of apoptotic execution. through cleavage by initiator caspases or by other active
The following sections discuss the workhorses of effector caspases.
apoptosis—the caspases—followed by regulation of the Scaffolding proteins and adapters play an essential
response. role in activating initiator caspases by forming multi-
protein activation platforms. For the intrinsic pathway
Caspases of cell death, factors released from mitochondria (dis-
Caspases (cysteine aspartases) are specialized proteases cussed later) activate the scaffold protein Apaf-1, which
with a cysteine in their active site that cleave their targets is related to AAA ATPases (adenosine triphosphatases)
on the C-terminal side of aspartate residues. Caspases (see Box 36.1). Active Apaf-1 forms a ring-like structure
generally inactivate cellular survival pathways and with seven spokes called the apoptosome (Fig. 46.17).
CHAPTER 46  n  Programmed Cell Death 805

~p20 ~p10
A B. Caspase maturation
Effector caspases 0 28 175 277
Casp-3 LS SS Procaspases (zymogens) Active enzyme
303 Pro
Casp-7 LS SS Prodomain
DED DED LS SS
293
Casp-6 LS SS Caspase 8 (an initiator caspase)

Initiator caspases 479


Casp-8 DED DED LS SS LS SS

521 Caspase 3 (an effector caspase)


Casp-10 DED DED LS SS
416
Casp-9 Card LS SS
435 C. Caspase 1 D. Caspase 3
Casp-2 Card LS SS

Proinflammatory caspases 377


Casp-4 Card LS SS
418
Casp-5 Card LS SS
373
Casp-11 Card
419
Casp-12 Card
404
Casp-1 (ICE) Card LS SS
242
Casp-14

FIGURE 46.11  INTRODUCTION TO CASPASES. A, The 13 enzymatically active mammalian caspases fall into three groups. Where it has
been determined, the portions of the zymogens that give rise to the large (blue) and small (yellow) subunits are shown. B, Initiator caspases have
large prodomains that participate in subcellular targeting. Two initiator procaspases come together to form the active enzyme. C–D, Ribbon
diagrams of crystal structures of caspases 1 and 3. The catalytic residues come primarily from the large subunit (blue). The space-filling structure
(red) represents a peptide inhibitor covalently bound in the active site of the enzyme.

Binding of procaspase 9 zymogen to this platform pro- also act indirectly to cause mitochondria to release
motes dimerization and activation of the enzyme. factors that promote cell death through the intrinsic
The scaffold proteins for the extrinsic death pathway pathway. Caspase cleavage of an inhibitory chaperone is
are cytoplasmic domains of cell-surface receptors. When responsible for activation of the nuclease that ultimately
these receptors bind their ligands on the surface of other destroys the chromosomal DNA of most cells undergoing
cells, they form stable trimeric complexes that recruit apoptosis (see section on CAD nuclease).
adapter proteins to form a signaling complex called the
“death-inducing signaling complex (DISC).” Interac- Natural Caspase Inhibitors
tions involving multiple protein–protein interaction Because most healthy cells express initiator procaspases
motifs link the procaspase 8 zymogen to the DISC (Box with the potential to oligomerize and kill the cell, it is
46.2), resulting in dimerization, activation, self-cleavage, important to have a mechanism that dampens any poten-
and release of active caspase 8. tial “noise” in the pro-apoptotic pathway. The inhibitor
Caspases cleave as many as 1000 cellular proteins of apoptosis protein (IAP) family is defined by the pres-
(Fig. 46.12). Some targets are structural proteins, but ence of a motif of approximately 80 amino acids known
many are involved in cellular signaling. For example, as a baculovirus IAP repeat (BIR) domain. This is a
caspases cleave several protein kinases. Many kinases type of Zn2+ finger (see Fig. 10.14) that mediates protein–
have pseudosubstrate motifs that autoinhibit the kinase protein interactions. IAP proteins inhibit caspases in two
and can be moved out of the way in response to physi- ways. First, they bind the caspase and invade the active
ological stimuli (see Fig. 25.4). Caspase cleavage often site, thereby blocking its access to substrates. Second,
neatly clips off these regulatory domains, thereby pro- several IAPs are E3 ubiquitin ligases (see Fig. 23.3) that
ducing constitutively active enzymes. These unregulated ubiquitylate caspases and tag them for destruction by
kinases may alter signaling pathways to promote cell proteasomes.
death. Caspases also cleave and inactivate several pro- If IAP proteins inactivate caspases, then how is the
teins that normally function in the detection and repair apoptotic response ever initiated? Cells also express
of DNA damage. several molecules that can bind and inactivate IAPs. The
Caspases can also create factors that directly promote best characterized of these, is the second mitochondrial
cell death. The most obvious example is caspase activa- activator of caspases (Smac or DIABLO). Smac is nor-
tion of other caspases in the death cascade. Caspases mally sequestered in mitochondria, but is released into
806 SECTION X  n  Cell Cycle

BOX 46.2  Matchmaking for Cell Death: the Key Is in the Domains

There are so many different proteins involved in apoptosis 1. The death domain (DD) is found in many proteins that
that even experts have a difficult time keeping them all are involved in signaling pathways related to cell death.
straight. However, an understanding of the general principles These include cell death receptors, such as Fas (and
is much simplified if we recognize that most proteins others), and adapter molecules, such as FADD (Fas-
involved in apoptosis regulation are built from a relatively associated death domain).
limited number of modules, which act as sites for protein– 2. The death effector domain (DED) is found in adapters
protein interactions. The following are the most important such as FADD, the prodomains of caspases 8 and 10, and
modules for apoptosis. certain inhibitors of apoptosis.
Bcl-2 family members are defined by the presence of 3. The Pyrin domain (PYD) is found in proteins involved
short regions of conserved sequence, referred to as BH in inflammation (such as microbial sensors) and the
domains (Bcl-2 homology). One of these, the approxi- activation of caspase 1.
mately 20-residue BH3 domain, found in all Bcl-2 family 4. The caspase recruitment domain (CARD) is found in
members, promotes complex formation between Bcl-2 several adapter proteins involved in cell death, including
family members by docking into a so-called BH3 binding CED-4 and Apaf-1, and in caspases 1, 2, and 9.
groove. The domains have similar folds, but different distributions
Four domains regulate caspase targeting and activation. of surface charge. As a result, they prefer to interact with
Although the amino acid sequences of these domains have themselves (ie, DD–DD, DED–DED, PYD–PYD, and CARD–
diverged extensively, all are regions of approximately 80 to CARD). Such interactions are said to be homophilic. When
90 residues that form a characteristic bundle of six α-helices. a new apoptosis effector protein is identified, it is possible
These domains all were present in the last eumetazoan to predict from an analysis of its amino acid sequence which
common ancestor. of the known proteins it is most likely to interact with.

A. Inactivation of protective factors and activation B. Pathway of nuclear apoptosis CYTOPLASM


of proapoptotic factors by caspases
Initiator Effector Cytoplasmic
caspases caspases targets
(8, 9, etc.) (3, 6, 7, etc.)
p53 Focal
pRb Surface Ras
mdm2 receptors contact RasGAP
Raf FAK Paxillin
Actin
Cdk/cyclins Gas2
active MEK PP2A Cb1/1b CAD / Caspase-6
p27 ICAD PARP
p21 Wee1 Lamins
APC PI3K
A A
Cdk/cyclin ERK ICAD/ I CA D CAD P RP L M I NS
inactive NF-κB DFF45 Akt Domain NUCLEUS
STAT1 nuclease
CAD Death Bad
Cell-cycle ? Bcl-2 Fragmentation
arrest ? nuclease
PKCθ Bcl-XL
Survival PKCδ JNK-p38 Bid

MEKK1 PAK2
Mst1/Krs

FIGURE 46.12  SOME WAYS THAT CASPASES PROMOTE CELL DEATH. A, A few of the many proteins cleaved by caspases in apoptotic
cell death. Proteins shown in green normally have a role in keeping the cell alive and are inactivated by caspases. Proteins shown in red are
turned into active death-promoting factors as a result of caspase cleavage. Proteins shown in black are not cleaved and are included to show
the pathways that are affected by cleavage. Caspases inactivate a number of pathways that promote cell survival, thereby strongly reinforcing
the decision of the cell to die. B, Some of the roles of caspases in disassembly of the nucleus. CAD, caspase-activated deoxyribonuclease; ERK,
extracellular signal-regulated kinase; ICAD, inhibitor of caspase-activated deoxyribonuclease; JNK, c-Jun N-terminal kinase; MEK, mitogen
activated protein/extracellular signal-related kinase kinase; NF-κB, nuclear factor κB; PI3K, phosphatidylinositol 3′-kinase; PKC, protein kinase
C; STAT, signal transducer and activator of transcription.

the cytoplasm when the intrinsic pathway of apoptosis IAPs were discovered in studies of the mechanisms
is initiated as a result of permeabilization of the mito- viruses use to avoid being eliminated by cell death.
chondrial outer membrane. It then binds to the BIR When viruses infect cells and disassemble their capsids,
domain, inactivating the IAPs. Mathematical modeling they become vulnerable to suicide defense mechanisms:
shows that inactivation of IAPs by Smac is needed to If cells can kill themselves before the virus has time to
make the proapoptotic signal act like a switch rather complete its life cycle, they will take the virus with them,
than a graded response. and the organism will survive. To defend against this,
CHAPTER 46  n  Programmed Cell Death 807

viruses pilfer genes encoding cellular proteins and adapt mammalian pox viruses also make a serpin-like inhibitor
them for their own means. For example, insect baculo- of certain caspases called CrmA.
viruses make two proteins that inhibit apoptosis, keeping
the cell alive long enough for the virus to reproduce.
One of these, IAP, was derived from a cellular gene. The
CAD Nuclease and Its Chaperone ICAD
origin of the second IAP inhibitor, p35, is less clear. p35 The chromosomal DNA is destroyed during apoptosis.
is a broad-spectrum caspase inhibitor that is thought to The nucleases involved in cleaving the cellular DNA
work by a serpin-like mechanism. Serpins are special during (and after) apoptotic cell death fall into two
protease substrates that, after cleavage, form a tight classes. Cell autonomous nucleases degrade the DNA
complex with the enzyme and inactivate it. Several within the dying cell (Fig. 46.13A). The best known is

A. DNA cleavage during apoptosis B. Active CAD nuclease


DNA

Proapoptotic Execution Engulfment Clearance


stimulus
Large Nucleosomal Further digestion
fragments fragments within phagocytic cell

Cell autonomous Waste management


nucleases nucleases

Necrosis Lysis Extracellular digestion

C. ICAD regulates CAD function


D. Drug-induced DNA E. Effect of purified CAD on
Apoptotic stimuli cleavage in vivo isolated nuclei in vitro
Caspase activation
23
CAD / Inactive 9.4
ICAD ICAD 6.6

Active
CAD 2.0
ICAD Dimerized CAD
protein cleaves DNA

0.56
CAD mRNA
CYTOPLASM NUCLEUS 3 ICAD/CAD
0 1 2 3 4 6h 1 2 3 4 + Casp-3

FIGURE 46.13  NUCLEASES DIGEST THE CELLULAR DNA DURING APOPTOSIS. A, In apoptosis, the DNA is digested first to large
fragments and later to nucleosome-sized pieces (see Fig. 13.1) by cell autonomous nucleases expressed within the dying cell. Waste management
nucleases made by other cells also have an essential role in cleaning up apoptotic and necrotic debris. B, The predominant cell autonomous
nuclease (CAD) has a scissors-like structure. C, ICAD is an inhibitory chaperone for CAD, promoting its folding on the ribosome and continuing
as an inhibitor when CAD is stored in the nucleus. ICAD cleavage leads to CAD activation. D, Cleavage of the chromosomal DNA by CAD during
chemotherapy-induced apoptosis of a leukemia cell line. DNA separated according to size by electrophoresis on an agarose gel was stained with
ethidium bromide. The ladder of bands reflects cleavage between adjacent nucleosomes. E, Activated CAD causes chromatin condensation and
appearance of an apoptotic morphology in isolated cell nuclei. Cloned CAD and ICAD were expressed together in Escherichia coli and incubated
with nuclei. ICAD cleavage with caspase 3 released active CAD, which degrades the nuclear DNA (Lane 3). Other lanes: DNA gel size markers
(left); nuclei incubated with buffer or caspase 3 alone (Lanes 1 and 2, respectively); same experiment as in Lane 3 but performed by using a
mutant ICAD that could not be cleaved by caspase 3 (Lane 4). To the right is an electron micrograph of a thin section of one nucleus with
condensed chromatin at the nuclear periphery. CAD, caspase-activated deoxyribonuclease; ICAD, inhibitor of caspase-activated deoxyribonucle-
ase; mRNA, messenger RNA. (A, Modified from Samejima K, Earnshaw WC. Trashing the genome: The role of nucleases during apoptosis. Nat
Rev Mol Cell Biol. 2005;6:677–688. B, For more information, see Protein Data Bank [PDB; www.rcsb.org] file 1V0D from Woo EJ, Kim YG, Kim
MS, et al. Structural mechanism for inactivation and activation of CAD/DFF40 in the apoptotic pathway. Mol Cell. 2004;14:531–539. D, From
Kaufmann SH. Induction of endonucleolytic DNA cleavage in human acute myelogenous leukemia cells by etoposide, camptothecin, and other
cytotoxic anticancer drugs: a cautionary note. Cancer Res. 1989;49:5870–5878. E, Courtesy K. Samejima, Wellcome Trust Institute for Cell
Biology, University of Edinburgh, United Kingdom.)
808 SECTION X  n  Cell Cycle

the caspase-activated DNase (CAD; see later discussion). Bcl-2 family


A mitochondrial nuclease known as endonuclease G may
also degrade DNA in some cell types. Cell autonomous Protectors Killers Regulators
nucleases are dispensable for cell death and for the life Bcl-2 Bax Bad Bik / Nbk
Bcl-xL Bak Bid HRK
of the organism. They might have evolved to eliminate Bcl-W Bok / Mtd Bim Puma
viral DNA as part of the suicide defense response Mcl-1 Bcl-xS BmF Noxa
described in the previous section. A1 Hrk
Boo/Diva C. elegans Egl-1
Waste management nucleases clean up the debris C. elegans ced-9
after cells die. They either function within lysosomes of Adenovirus E1B19K
cells that have ingested apoptotic cell fragments or are Epstein-Barr virus BHRF1
secreted into the extracellular space. DNase II, one of
the most important waste management nucleases, is
essential for life. Mouse embryos that lack DNase II BH4 BH3 BH1 BH2 Protectors
become overwhelmed with undegraded DNA and die.
Cell autonomous nucleases act in two stages. After BH3 BH1 BH2 Killers
an initial cleavage of the chromosomes into fragments
of roughly 50,000 base pairs, DNA is usually cleaved BH3 Regulators
between nucleosomes, producing a characteristic
“ladder” of DNA fragments with a periodicity of approxi-
mately 200 base pairs. This ladder is seen when DNA FIGURE 46.14  THE BCL-2 FAMILY OF APOPTOTIC REGULA-
TORS. C. elegans, Caenorhabditis elegans.
isolated from apoptotic cells is subjected to gel electro-
phoresis. The responsible nuclease is CAD. CAD is nor-
mally present in a complex with ICAD (inhibitor of CAD normally survive into the adulthood. A Ced-9 mutation
[Fig. 46.13C]). The complex of CAD and ICAD is also kills the worm. Human Bcl-2 is functionally and structur-
known as DNA fragmentation factor (DFF). ICAD is a ally homologous to C. elegans Ced-9 and can substitute
chaperone required to fold CAD into an active conforma- for it in living worms. This ability of a human gene to
tion during translation on the ribosome. However, ICAD protect nematodes reveals that the fundamental machin-
also inhibits the nuclease activity of CAD. This dual func- ery of apoptotic cell death has been conserved over great
tion of ICAD guarantees that only inactive CAD can be evolutionary distances.
synthesized in healthy cells. During apoptosis, caspase 3 Bcl-2 family members are defined by the presence of
cleaves ICAD and releases active CAD nuclease. one to four short blocks of conserved protein sequence
called BH (Bcl-2 homology) domains. Antiapoptotic
Bcl-2 Proteins and the Intrinsic Pathway Bcl-2 protectors typically have four of the domains.
Proapoptotic Bcl-2 killers typically have three of these
of Apoptotic Cell Death
domains, while the Bcl-2 regulators have only the BH3
As mentioned previously, mitochondria are key players in domain. The BH3 domain is a short helix that fits into
a pathway to cell death that is triggered by a variety of a groove on the surface of both Bcl-2 protectors and
toxic insults (Fig. 46.17). The Bcl-2 family of proteins killers, forming complexes that regulate their activity. It is
regulates these mitochondrial events. The following sec- believed that the Bcl-2 protectors regulate the behavior of
tions describe this important protein family and the regu- Bcl-2 killers by a similar interaction. For example, the Bcl-2
lation of the intrinsic pathway of apoptosis. protein forms a complex with a proapoptotic Bcl-2 killer
called Bax, thereby interfering with the ability of Bax to
Bcl-2 Proteins kill cells. Binding of BH3-only proteins to Bcl-2 protectors
Bcl-2 proteins can be grouped into three subfamilies (Fig. can inactivate their antiapoptotic functions. Egl-1 is a
46.14). Bcl-2 protectors protect cells against apoptosis. BH3-only protein and this is how it triggers apoptosis. A
Bcl-2 killers (eg, Bax and Bak) are proapoptotic proteins new generation of BH3 mimetic drugs induces apoptosis
that actively kill cells. Bcl-2 regulators (widely known as of cancer cells by mimicking this second mechanism.
BH3-only proteins) promote cell killing by either interfer- Genetic experiments in mice revealed several differ-
ing with the protectors or activating the killers. Bcl-2 ent functions for Bcl-2 family members. Mice born
proteins primarily regulate the release of death-promoting without Bcl-2 have deficiencies of the immune system
factors from mitochondria when cells receive signals that are best understood if one role of this protein in vivo
that activate the intrinsic pathway. is to render lymphocytes resistant to proapoptotic signals
C. elegans genetics identified a gene, ced-9, that pro- during immune system maturation. Mice lacking another
tects cells against apoptosis and another gene, egl-1, pro-life family member, Bcl-xL, die during embryogenesis,
that inactivates ced-9 protein and triggers apoptosis. In apparently as a result of widespread death of neurons
ced-9 mutants, many cells die during development that in the central and peripheral nervous systems and
CHAPTER 46  n  Programmed Cell Death 809

hematopoietic cells in the liver. In contrast, loss of the


100
killers Bax plus Bak makes cells highly resistant to apop-

Cell viability (% control)


tosis by a wide variety of intrinsic pathway stimuli. 80

60 Normal levels of c-myc


Bcl-2 Family Members and Cancer 40
Overexpressed c-myc
and overexpressed Bcl-2
A gene that prevents cells from dying poses a potential 20
danger in multicellular organisms, in which rates of cell Overexpressed c-myc
proliferation and death must be balanced carefully. In 0
0 30 60 90 120
fact, the name Bcl-2 comes from the discovery that this
c-myc induction (min)
gene is the culprit responsible for certain types of B-cell
lymphoma. These particular lymphomas arise when a FIGURE 46.15  BCL-2 PROTECTS CELLS FROM ONCOGENE-
INDUCED CELL DEATH. Chinese hamster ovary cells die when they
chromosome translocation, involving chromosomes 14
are induced to express abnormally high levels of the c-myc protein,
and 18, moves the Bcl-2 gene into the immunoglobulin but simultaneous expression of the Bcl-2 protein rescues them from
heavy-chain gene cluster, a site of very active gene this effect. These cells contain many copies of the c-myc gene under
transcription in B lymphocytes. Elevated transcription of control of a promoter that is activated when the cells are briefly
Bcl-2 is thought to be directly responsible for the cancer- exposed to high temperature (43°C for 90 minutes). The curves show
the percentage of viable cells remaining at various times following the
ous phenotype in these patients, making Bcl-2 a cancer-
induction of c-myc expression. The Bcl-2 gene was introduced into
promoting oncogene (see Chapter 41). these cells on a plasmid under the control of a promoter that is always
Unlike many other oncogenes, Bcl-2 overexpression active. The blue line represents the parental cell line lacking either the
does not cause cell proliferation. Instead, it disrupts cloned c-myc or Bcl-2 genes. (Note that approximately 40% of these
the balance of regulation between life and death. Cells cells die following the heat treatment used to induce c-myc expres-
sion.) The yellow line shows that the cells that produce high levels of
that overexpress Bcl-2 protein actually grow somewhat
c-myc protein alone rapidly die (by apoptosis). The red line shows that
slower than normal but are highly resistant to many cells expressing both the c-myc and Bcl-2 proteins survive the treat-
stimuli that normally promote cell death. The net result ment almost as well as the parental cells. (From Bissonnette RP,
is an accumulation of B cells: a lymphoma. Other anti- Echeverri F, Mahboubi A, et al. Apoptotic cell death induced by c-myc
apoptotic Bcl-2 family members are amplified in a variety is inhibited by bcl-2. Nature. 1992;359:552–554.)
of solid tumors and loss of microRNAs directed at Bcl-1
is thought to play a major role in development of chronic
lymphocytic leukemia. Fig. 46.15 shows an example of pores that allow the release of pro-apoptotic factors from
Bcl-2 conferring resistance to death when the cell cycle the mitochondrial intermembrane space. Binding of
is perturbed by expression of an oncogene. antiapoptotic Bcl-2 family members to Bax/Bak somehow
prevents mitochondrial outer membrane permeabiliza-
tion. BH3-only family members can either directly
Intrinsic Pathway of Apoptotic Death promote death by facilitating Bax/Bak oligomerization or
In addition to their role in energy production, mitochon- indirectly promote it by binding and neutralizing anti-
dria have an essential role as sensors of the health of the apoptotic Bcl-2 family members.
cell. If cells sense insults from which they cannot recover, The proapoptotic factors released from the mitochon-
mitochondria trigger the intrinsic pathway of cell drial intermembrane space by Bax and Bak include the
death (Fig. 46.17). Bcl-2 family members regulate this electron transport protein cytochrome c (see Fig. 19.5),
pathway. The regulation seems straightforward in C. Smac, and endonuclease G (Fig. 46.16). These mitochon-
elegans, in which the protector CED-9 (Bcl-2–like) binds drial proteins actively promote apoptotic cell death. In
to the CED-4 scaffolding protein (Apaf-1-like) and inter- the cytoplasm, cytochrome c binds to the scaffolding
feres with its activation of the CED-3 caspase. Apoptosis protein Apaf-1, a mammalian homolog of C. elegans
is induced when the regulator BH3-only protein EGL-1 CED-4 protein, causing it to undergo a conformational
binds to CED-9 and blocks it from inactivating CED-4. change that exposes a hidden bound adenosine diphos-
In mammals, the situation is more complex, partly phate (ADP) to solvent. Exchange of this ADP for deoxy-
because Bcl-2 family members are more numerous and adenosine triphosphate (dATP) allows Apaf-1 to form a
partly because they do not interact in such a straightfor- wheel-like structure with seven spokes called the apopto-
ward fashion. In mammals, two of the killer proteins, some (Fig. 46.17). Apaf-1 in the apoptosome binds caspase
Bax and Bak, are essential for activation of the intrinsic 9 through an N-terminal caspase recruitment domain.
pathway. In healthy cells, Bak is loosely associated Binding to the apoptosome elevates the catalytic
with the mitochondrial outer membrane, and Bax is in activity of procaspase 9 approximately 2000-fold without
the cytoplasm (Fig. 46.17). On receipt of a proapoptotic the need for its cleavage. Thus, the active form of caspase
stimulus, Bax and Bak insert deeply into the mitochon- 9 is an oligomeric complex of the procaspase with
drial outer membrane and form oligomeric membrane the apoptosome. Activated caspase 9 then cleaves
810 SECTION X  n  Cell Cycle

multiple procaspase 3 zymogens, amplifying the cell


A. Life death cascade. It also cleaves itself, triggering its release
Cytochrome c
Smac, AIF from the apoptosome with a resulting loss of activity.
Intermembranous
space
Endonuclease G This autocleavage acts like a timer limiting apopto-
Others?
some activity.
Mitochondrial
outer membrane This cascade can be further amplified in at least two
permeabilization ways. First, caspase 3 cleaves other effector caspases,
directly amplifying the cascade. In addition, active cas-
B. Death pases cleave the BH3-only protein Bid, which then acti-
Bid vates more Bax and Bak in a feedback loop, thereby
Caspases
Bax promoting the release of more cytochrome c and Smac,
Other Bak
Cytochrome c
and enhancing caspase 9 activation.
Activation of signals
downstream Smac It was extremely surprising to find that an essential
caspases AIF metabolic protein such as cytochrome c has a second
Caspase 9 Endonuclease G function that is essential for death. Among the studies
activation
Other Others
supporting the Jekyll-and-Hyde–like nature of this protein
targets in life and death was the engineering of mice whose
To nucleus
Nuclear Other cytochrome c can function in electron transport but
disassembly targets cannot bind Apaf-1. These mice die as a result of brain
abnormalities caused by insufficient cell death.
FIGURE 46.16  MITOCHONDRIA INTEGRATE A CELL’S LIFE
AND DEATH DECISIONS. A, In healthy cells, a number of factors
that promote apoptosis are stored in the intermembrane space of the
mitochondria. B, In cells undergoing apoptosis, BH3-only proteins
Extrinsic Pathway of Apoptotic Death
trigger the proapoptotic Bcl-2 family members to induce the release Cells express at least six different cell surface molecules,
of these death-promoting factors; this initiates an amplifying cycle that
collectively termed death receptors, that can trigger
ultimately leads to cell death. AIF, apoptosis-inducing factor; Smac,
second mitochondrial activator of caspases. apoptotic death. Protein ligands on the surface of other

A. Life Metabolites up C. Apoptosome


to 10,000 D 3 (Apaf-1 scaffold)
9

Bak Activation of
Bid downstream
3 caspases
Autoactivation
VDAC
of caspase 9 (stays
3
6 bound to apoptosome)
MOMP
Caspases
cleave Bid
Bax B. Death
tBid 9
Proteins not to scale

Procaspase 9 9

Procaspase CARD domain

Apaf-1
CARD
domain Cytochrome c
dADP
Aggregation
Apaf-1 of Apaf-1 scaffold
* Conformational (apoptosome)
* *AIF
Smac, (ATP)
change in Apaf-1
endonuclease G (exchanges dADP for ATP) 27nm

FIGURE 46.17  INTRINSIC CELL DEATH PATHWAY. A–B, Following activation by the BH3-only protein Bid, Bax and Bak form a pore that
releases cytochrome c. Released cytochrome c binds to Apaf-1, inducing a conformational change leading to formation of the apoptosome,
which binds and activates procaspase 9 via scaffold-induced oligomerization. Activated caspase 9 subsequently activates downstream effector
caspases, leading to cell death. C, Molecular organization of the apoptosome. (C, Modified from PDB file 3JBT from Zhou M, Li Y, Hu Q, et al.
Atomic structure of the apoptosome: mechanism of cytochrome c- and dATP-mediated activation of Apaf-1. Genes Dev. 2015;29:2349–2361.)
CHAPTER 46  n  Programmed Cell Death 811

cells bind and activate these receptors, turning on path- called FADD (Fas-associated protein with a death
ways that can lead to apoptotic death. domain), assembling the DISC. The DISC is an activation
One well-characterized death receptor called Fas (also platform that binds procaspase 8 through interactions
known as Apo1 or CD95) is a member of the tumor involving another type of motif called the death effec-
necrosis factor receptor family (see Fig. 24.9). Fas is a tor domain, which is present on both FADD and
type I membrane protein whose extracellular domain the prodomain of procaspase 8. Procaspase 8 monomers
consists of three cysteine-rich domains (Fig. 24.9 shows dimerize on the DISC and acquire catalytic activity.
the atomic structure of the related trimeric tumor necro- These dimers can cleave neighboring dimers, creating
sis factor receptor with bound ligand). The cytoplasmic and releasing heterotetrameric active caspase 8, which
domain of Fas contains a death domain of approxi- can initiate the caspase cascade by activating downstream
mately 80 residues, which is shared by all the death effector caspases. Frequently, caspase 8 cleaves the BH3-
receptors (see Box 46.2). only protein Bid, thereby activating the intrinsic death
The Fas ligand is a trimeric 40-kD intrinsic membrane pathway (Fig. 46.17).
protein found on the surface of cells. Cytotoxic T lym- The extrinsic pathway poses considerable risk for the
phocytes use Fas ligand to rid the body of virally infected cell. Fas is constitutively present in the cell membrane
cells. When a cytotoxic T lymphocyte contacts a target and can form at least transient trimers in the absence of
cell, the Fas ligand on the lymphocyte surface binds to binding by its ligand. How do cells avoid the accidental
Fas on the target cell and initiates the extrinsic pathway activation of apoptosis caused by chance binding of
of apoptotic death (Fig. 46.18). Ligand binding activates procaspase 8 to naturally occurring transient Fas trimers?
signaling from the intracellular death domain of Fas, Cells express a caspase-related protein called cFLIP
possibly by stabilizing Fas trimers or by altering their (FLICE-like inhibitory protein; FLICE is another name for
conformation. Activated Fas binds an adapter protein caspase 8) that looks very much like a catalytically dead

Unfolded FasR
binding domains

TARGET
FasR CD95 APO-1 CELL
8

8 8
A. Preligation
Monomers C. Release of active caspase 8
KILLER 8
CELL

Uncleaved caspase 8
Fas active on DISC
FADD
ligand Death domain 8

Death effector 8
Released active
domain caspase 8
Trimers DISC 8
(signaling
platform)
8

Procaspase 8

B. Ligand docked on a trimerized receptor Caspase 8


Molecular cleaves Bid Procaspases 3,6,7
scaffold forms 8 7
Bid 3 6

8
E. Activation of the D. Activation
intrinsic death of effector
8
pathway (see caspases 7
Clustering of Fig. 46.17) Active caspases 3,6,7
8
caspase 8
6 3
F. Death

FIGURE 46.18  EXTRINSIC CELL DEATH PATHWAY. The pathways shown are downstream of the Fas cell death receptor. A, Preligation.
B, Ligand docked on a trimerized receptor. C, Release of active caspase. D, Activation of effector caspases. E, Activation of the intrinsic death
pathway (Fig. 46.17). F. Death (see the text for a detailed description). DISC, death-inducing signaling complex; FADD, Fas-associated death
domain.
812 SECTION X  n  Cell Cycle

version of procaspase 8. When expressed at high levels, cells. Other decoy receptors remain membrane bound
cFLIP coassembles with procaspase 8 monomers creat- but do not signal cell death when they bind ligand
ing an enzyme with altered activity that does not trigger because their intracellular domains lack functional death
cell death. This role of cFLIP may be to dampen the Fas domains.
response locally to ensure that the cascade does not get
activated by mistake. Another role of FLIP is to combine Linking Apoptosis to the Cell Cycle
with caspase 8 to ensure that TNFα (tumor necrosis
by p53
factor α) signaling, which occurs in many immune cells,
promotes inflammation rather than killing cells by No obligate link exists between particular cell-cycle
necroptosis (see later). phases and apoptosis. Noncycling G0 cells can undergo
apoptosis, and cycling cells appear able to do so from
Role of the Fas Death Receptor in Normal any cell-cycle phase. However, one link between apop-
tosis and the cell-cycle machinery is firmly established.
and Diseased Cells
This involves the p53 tumor suppresser and DNA
Fas is important for regulation of the immune system, damage.
but also has a very unexpected role in cancer cells. Mice The p53 transcription factor is one of the downstream
with mutated Fas (the lpr mutation) or Fas ligand (the EFFECTORS of the DNA damage response pathway (see
gld mutation) accumulate excessive lymphocytes. In the Fig. 43.11). When cells sense DNA damage induced by
appropriate genetic background, these mice tend to agents such as ionizing radiation, p53 levels rise dramati-
develop autoimmune disorders. cally. When stabilized and activated by phosphorylation
Fas is important in regulating the life span of activated (see Fig. 41.14) p53 upregulates the expression of a
tissue T and B lymphocytes. Normally, T cells die within number of genes, including the Cdk inhibitor p21, which
a few days of their activation during an immune response. blocks the entry into the S and M phases. p53 also can
Activation initiates the expression of Fas ligand on the T trigger an apoptotic response in instances in which the
cells themselves. This new Fas ligand interacts by an DNA damage is too severe to repair (Fig. 46.19). This
unknown mechanism with Fas already on the cell surface, tumor-suppressor protein is critical in the body’s defense
causing the cell to commit apoptotic suicide. T-cell against cancer. Mutations in the p53 gene/protein are
activation also downregulates the expression of cFLIP, found in approximately 50% of all human cancers.
thus permitting activated caspase 8 to more effectively A direct connection between p53 and apoptosis
kill the cell. A similar mechanism (export of Fas and was revealed by overexpressing the cloned p53 gene
Fas ligand to the surface of the same cell) is responsible in different cell types. In most cells, overexpression of
for some examples of p53-induced cell death and some p53 arrests the cell cycle at the G1/S boundary.
instances of cell death following exposure to chemo- However, ectopic expression of cloned p53 in certain
therapeutic agents. cancer-derived cell lines causes the cells to undergo
These features of Fas might seem to make this system apoptosis.
useful in the treatment of cancer. Unfortunately this is The role of p53 in apoptosis was confirmed in trans-
not the case, because Fas does more than signal to genic mice lacking a functional p53 gene (p53 knockout
promote cell death. It can also signal to several pathways mice). These mice develop normally but are extremely
to promote cell survival and migration. In fact, Fas signal- prone to cancer at a very young age. Thymocytes isolated
ing in cancer cells can actually promote tumor growth from p53 knockout mice are highly resistant to the
and metastasis. This serves as an example of the com- induction of apoptosis by ionizing radiation and other
plexity of cell signaling networks. agents that cause DNA breaks (Fig. 46.19B). However,
Some tissues, like the lens of the eye and the testis, p53 is not involved in all types of apoptosis. For example,
avoid immune and inflammatory responses by express- the same thymocytes isolated from p53 knockout mice
ing Fas ligand. Immune effector cells (which express show normal induction of apoptosis following exposure
Fas on their surface) that enter these tissues encounter to glucocorticoid hormone (Fig. 46.19).
Fas ligand and die by apoptosis. These tissues are known p53 promotes apoptosis by functioning as a transcrip-
as immune-privileged. Not surprisingly, certain tumor tional activator. It controls, among others, the well-
cells subvert this strategy as protection against the studied death-promoting genes Bax, Fas (CD95/APO-1),
immune system. Melanoma cells expressing Fas ligand and APAF-1. However, the key target gene is PUMA (p53
establish tumors particularly efficiently. Some tumor modulated upregulator of apoptosis), a BH3-only protein
cells, especially those from colon and lung cancers, also that promotes apoptotic cell death by activating Bax and
defend themselves against immune surveillance with Bak. PUMA knockout mice show defects in cell death
so-called decoy receptors. A secreted Fas decoy pathways that are essentially identical to those seen in
receptor blocks Fas ligand on cytotoxic T cells so that p53 knockout mice and not seen in mice that lack Bax,
it cannot engage Fas on the surface of the tumor Fas, or Apaf-1.
CHAPTER 46  n  Programmed Cell Death 813

A. Glucocorticoid B. Irradiated inflammation as this can trigger an immune response in


hormone–treated the early phases of infection.
p53 Pyroptosis (proinflammatory programmed cell
100 100 –/– death) occurs when signaling by a variety of cytoplasmic
Cell viability (%)

Cell viability (%)


80 80
receptors in response to pathogen infection leads to the
60 60 formation of the inflammasome, a cytosolic complex
40 40
p53 that contains numerous caspase 1 activation sites. Pyrin
+/–
20 20 +/+
domains (PYDs) in these receptors are linked to caspase
recruitment domains (CARDs), which in turn recruit
0 0
0 5 10 15 20 25 0 5 10 15 20 25 caspases 1, 4, and 5 (1 and 11 in mice). Despite being
Time (h) Time (h) the first caspase to be described, caspase 1 was the last
to be shown to be involved in a programmed cell death
C. Control D. Irradiated response.
The function of caspase 1 is to produce interleukin-1β
(IL-1β) and interleukin-18 (IL-18) by proteolytic process-
ing of their precursors. These proteins are secreted
together with caspase 1 by an unconventional pathway
that is still being investigated and may involve cell lysis.
IL-1β is a proinflammatory cytokine that triggers the
fever response by acting on the hypothalamus. It also
promotes the proliferation of immune cells, among other
activities. IL-18 promotes increased immune cell recruit-
ment and activation, as well as modifying local cells to
facilitate adaptive immune responses. This proinflamma-
tory signaling facilitates a ramping up of the immune
response to the pathogen.
How pyroptosis leads to cell death is still being inves-
FIGURE 46.19  LINK BETWEEN p53 AND DNA DAMAGE-
INDUCED APOPTOTIC DEATH. A–B, Survival of thymocytes from tigated, but the morphology of the dying cells resembles
three strains of mice after exposure to glucocorticoids or irradiation. that seen in necrosis. As a result, the internal components
Cell death was from apoptosis. The strains were as follows: wild-type of the cell are released into the extracellular space,
mice (yellow), heterozygous mice having one good copy of the p53 provoking a strong inflammatory response (as in necro-
gene and one defective copy (red), and mice lacking any functional
sis, above, and necroptosis, below).
copy of the p53 gene (blue). Thymocytes that lack p53 are resistant
to radiation-induced apoptosis but show normal induction of apopto- Necroptosis (programmed necrosis) is a cell death
sis following exposure to glucocorticoid hormone. C–D, Induction of pathway that may function as a backup when the extrinsic
p53 accumulation following radiation of the small intestine. Black pathway of apoptosis is not functioning. Activation of the
arrows indicate cells with increased levels of p53. Red arrows indicate CD95 (Fas) and TNFR1 (tumor necrosis factor receptor
apoptotic cells. (A–B, From Lowe SW, Schmitt EM, Smith SW, et al.
1) cell-surface receptors can have three possible out-
p53 is required for radiation-induced apoptosis in mouse thymocytes.
Nature. 1993;362:847–849. C–D, Courtesy John Hickman, Molecular comes. TNFR1 can signal via complex I (which contains
and Cellular Pharmacology Group, University of Manchester, United protective IAP proteins) to activate the canonical NF-κB
Kingdom.) pathway to promote cell survival (Fig. 46.1). Alternatively
TNFR1 can activate receptor interacting protein kinases 1
and 3 (RIPK1/RIPK3) and signal to form complex II (con-
taining FADD, TRADD [tumor necrosis factor receptor-
Other Types of Programmed Cell Death
associated death domain], caspase 8, and cFLIP). When
The terms apoptosis and programmed cell death are caspase 8 is associated with cFLIP, it inactivates RIPK3
sometimes viewed as synonymous. However, a number and cells survive. Complex II activates necroptosis if
of specific examples of programmed cell death have either caspase 8 or cFLIP is missing or inactivated and
been described that lack the features that classically if RIPK1/RIPK3 are active. RIPK1/RIPK3 activity leads
define apoptosis. to permeabilization of the cell membrane, cell lysis and
death. As in other forms of necrosis, the released cellular
Inflammatory Cell Death: Pyroptosis contents cause a local inflammatory response.
and Necroptosis Necroptosis is thought to be a backup pathway for
One of the defining features of apoptosis is that dying apoptosis during viral infection. Mouse kidney cells
cells disappear without causing an inflammatory infected with a virus that encodes a caspase 8 inhibitor
response. However, in certain instances, it is protective proceed down the necroptosis death pathway. Removal
for the body to have cell death be accompanied by of the gene from the virus restores apoptosis. Mice
814 SECTION X  n  Cell Cycle

lacking RIPK3 show increased susceptibility to viral


infection. Murine cytomegalovirus has proteins that help
it to evade both apoptosis and necroptosis, supporting Primary
lesion
this argument.

Autophagic Death
Autophagy is a catabolic, energy producing process that
allows cells to survive in adverse conditions by recycling
amino acids liberated by degradation of cellular proteins
and organelles (see Fig. 23.7). Autophagy can be acti-
vated by a wide range of stimuli, including nutrient and
oxidative stress, accumulation of unfolded proteins,
intracellular bacterial infection, and oncogenic stress. In
addition to being a response to starvation, autophagy can
also participate in antiviral responses by participating in
antigen presentation by immune cells.
Connections between autophagy and cell death are
complex and debated. In certain cases during develop-
ment, autophagic pathways can lead directly to cell Penumbra: zone of apoptotic Primary focus
death. These dying cells lack chromatin condensation death starting within 24 hours of necrotic death
of the initial lesion
and exhibit extreme vacuolization of the cytoplasm. In
some cells, autophagy can regulate apoptosis. For
FIGURE 46.20  BRAIN DAMAGE IN STROKE. Secondary pro-
example, degradation of proapoptotic BH3-only proteins grammed cell death caused by oxygen deprivation in the penumbra
by autophagy provides a mechanism protecting cells greatly increases the size of the affected area of the brain in stroke.
against apoptosis. Alternatively, degradation of protec-
tive proteins can cause autophagy to actively promote
apoptotic cell death. such as Huntington disease and Alzheimer disease, as
Connections between autophagy and cancer are well as in myocardial infarction and stroke (Fig. 46.20).
complex. It has been argued that by promoting cell At a practical level, the realization that many successful
survival under adverse conditions, autophagy might help chemotherapeutic agents act by inducing cancer cells
promote the establishment of cancers particularly in to undergo apoptosis motivated searches for newer
avascular areas and by helping cancer cells to survive and better drugs that elicit this response. The generally
chemotherapy. However, mice heterozygous for Beclin1 disappointing outcome of those studies may largely be
(a scaffolding protein that functions early in assembly of because other pathways (eg, necroptosis) kick in when
the phagophore membrane; see Fig. 23.7) develop apoptosis is inhibited. Alternatively, as more is learned
cancers, suggesting that Beclin-1 is a tumor suppressor about mechanisms of necroptosis and pyroptosis, these
and that autophagy may protect against tumorigenesis. alternative types of programmed death become attractive
Interestingly, Beclin-1 has a BH3 domain, and although therapeutic targets, as their induction may not only kill
it is not a canonical BH3-only proapoptotic protein, its the target cells, but also provoke a localized immune
function is negatively regulated by Bcl-2. Also, autophagy response that might attack cancer cells or pathogens.
is activated in oncogene-induced senescence and inhibit- With such important practical problems to be solved, pro-
ing autophagy delays senescence. Thus autophagy might grammed cell death will continue to occupy a prominent
prevent cancer formation by helping damaged cells to position in cell biology research over the coming years.
become senescent.
ACKNOWLEDGMENTS
Importance of Programmed Cell Death in
We thank Charles Earnshaw, Scott Kaufmann, Luis
Human Disease Miguel Martins, and Andrew Thorburn for their sugges-
Why have studies of programmed cell death so caught tions on revisions to this chapter.
the scientific eye? One likely answer is that cell death is
a point of intersection between cell signaling pathways, SELECTED READINGS
cell structure, the cell cycle, and, of course, human
Breckenridge DG, Xue D. Regulation of mitochondrial membrane
disease. This chapter has mentioned the roles that aber-
permeabilization by BCL-2 family proteins and caspases. Curr Opin
rations in programmed cell death play in the etiology Cell Biol. 2004;16:647-652.
of autoimmunity, AIDS, and cancer. Cell death is firmly Czabotar PE, Lessene G, Strasser A, Adams JM. Control of apoptosis by
established as a key factor in neurodegenerative diseases, the BCL-2 protein family. Nat Rev Mol Cell Biol. 2014;15:49.

You might also like