Вы находитесь на странице: 1из 19

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
cells internal surveillance networks.
Pyroptosis: Often in response to intracellular pathogens,
this involves activation of caspase 1. The infected
cells secrete interleukin-1 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 APOPTOSISACTIVE SUICIDETYPICALLY


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;68:251306.)
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 dont eat me signals

C. Production of antiinflammatory
cytokines

IL-10
TGF-

FIGURE 46.7 PHAGOCYTOSIS OF APOPTOTIC CELLS. AC, 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;12:277287.
B, From Wyllie AH, Kerr JFR, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol. 1980;68:251306.)
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 and 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 Mllerian
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.
BD, 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- gene Pre-TCR TCR- 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- gene positive selection) TCR (negative signal factor signals
rearrangement No productive TCR- 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;5:189200.)
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 mllerian
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 B
that drive the apoptotic response. (NF-B; 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 prointerleukin-1 to yield mature
populations in the immune system. In many cells, the interleukin-1 and prointerleukin-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. 46.11AB). 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. 46.11BD).
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.
apoptosisthe caspasesfollowed 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. CD, 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 proteinprotein 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, DDDD, DEDDED, PYDPYD, 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 -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-B DFF45 Akt Domain NUCLEUS
STAT1 nuclease
CAD Death Bad
Cell-cycle ? Bcl-2 Fragmentation
arrest ? nuclease
PKC Bcl-XL
Survival PKC 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-B, nuclear factor B; 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;6:677688. 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;14:531539. 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;49:58705878. 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 (43C 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;359:552554.)
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-2like) 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-Hydelike 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 CELLS 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. AB, 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;29:23492361.)
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 TNF (tumor necrosis
by p53
factor ) 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 bodys 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


hormonetreated 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-1
(IL-1) 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-1 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. AB, 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. CD, 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. (AB, 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;362:847849. CD, 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-B
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.
CHAPTER 46 n Programmed Cell Death 815

Deretic V, Saitoh T, Akira S. Autophagy in infection, inflammation and Raff MC. Social control on cell survival and cell death. Nature. 1992;
immunity. Nat Rev Immunol. 2013;13:722-737. 356:397-400.
Earnshaw WC, Martins LM, Kaufmann SH. Mammalian caspases: Struc- Savill J, Fadok V. Corpse clearance defines the meaning of cell death.
ture, activation, substrates and functions during apoptosis. Annu Nature. 2000;407:784-788.
Rev Biochem. 1999;68:383-424. Silke J, Meier P. Inhibitor of apoptosis (IAP) proteinsmodulators of
Fitzwalter BE, Thorburn A. Recent insights into cell death and cell death and inflammation. Cold Spring Harb Perspect Biol. 2013;
autophagy. FEBS J. 2015;282:4279-4288. 5:a008730.
Lamkanfi M, Festjens N, Declercq W, etal. Caspases in cell survival, Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at
proliferation and differentiation. Cell Death Differ. 2007;14:44-55. the cellular level. Nat Rev Mol Cell Biol. 2008;9:231.
Lauber K, Blumenthal SG, Waibel M, etal. Clearance of apoptotic cells: Wallach D, Kang TB, Dillon CP, etal. Programmed necrosis in inflam-
Getting rid of the corpses. Mol Cell. 2004;14:277-287. mation: Toward identification of the effector molecules. Science.
Meier P, Finch A, Evan G. Apoptosis in development. Nature. 2000; 2016;352:aaf2154.
407:796-801. Wyllie AH, Kerr JFR, Currie AR. Cell death: The significance of apop-
Metzstein MM, Stanfield GM, Horvitz HR. Genetics of programmed cell tosis. Int Rev Cytol. 1980;68:251-305.
death in C. elegans: Past, present and future. Trends Genet. 1998; Zmasek CM, Godzik A. Evolution of the animal apoptosis network.
14:410-416. Cold Spring Harb Perspect Biol. 2013;5:a008649.
Pasparakis M, Vandenabeele P. Necroptosis and its role in inflamma-
tion. Nature. 2015;517:311-320.

Вам также может понравиться