Download as pdf or txt
Download as pdf or txt
You are on page 1of 41

LIFE CYCLE OF THE CELL

Cell growth and division:


• Mitosis leads to production of cells that are genetically identical to their
parent
• Meiosis leads to production of cells with half the genetic content of the
parent.
• Mitosis serves as the basis for producing new cells, meiosis as the basis for
producing new sexually reproducing organisms.
• Together, these two types of cell division form the links in the chain between
parents and their offspring and, in a broader sense, between living species
and the earliest eukaryotic life forms present on Earth.
• each cell passes through a series of defined stages, which constitutes the
cell cycle
• The cell cycle can be divided into two major phases based on cellular
activities readily visible with a light microscope: M phase and interphase.
• M phase includes (1) the process of mitosis, during which duplicated
chromosomes are separated
• into two nuclei, and (2) cytokinesis, during which the entire cell divides into
two daughter cells.
• Interphase, the period between cell divisions, is a time when the cell grows
and engages in diverse metabolic activities.
Phases of Cell Cycle
• Interphase is divided into G1, S, and G2 phases, with S phase being
equivalent to the period of DNA synthesis.
• The division of interphase into three separate phases based on the
timing of DNA synthesis was first proposed in 1953 by Alma Howard
and Stephen Pelc of Hammersmith Hospital, London, based on their
experiments on plant meristem cells.
• G1:Cell grows and carries out normal metabolism; organelles duplicate
• S: DNA replication and chromosome duplication, S phase is also the
period when the cell synthesizes the additional histones that will be
needed as the cell doubles the number of nucleosomes in its
chromosomes
• G2: Cell grows and prepares for mitosis
• Cells that arearrested in this state—which includes the majority of cells
in the body—are said to be in the G0 state
• A cell must receive a growth-promoting signal to proceed from G0 into
G1 phase and thus reenter the cell cycle.
• M phase is the period when the contents of a cell are actually divided
CELL CYCLE CONTROL SYSTEM
• Potu Rao and Robert Johnson of the University of Colorado helped open the door to
understanding how the cell cycle is regulated. they fused mitotic cells with cells in
other stages of the cell cycle. The mitotic cell always induced compaction of the
chromatin in the nucleus of the nonmitotic cell.
• If a G1-phase and an M-phase cell were fused, the chromatin of the G1-phase
nucleus underwent premature chromosomal compaction to form a set of elongated
compacted chromosomes .
• If a G2-phase and M-phase cell were fused, the G2 chromosomes also underwent
premature chromosome compaction, but unlike those of a G1 nucleus, the
compacted G2 chromosomes were visibly doubled, reflecting the fact that
replication had already occurred .
• If a mitotic cell was fused with an S-phase cell, the S-phase chromatin also became
compacted. However, replicating DNA is especially sensitive to damage, so that
compaction in the S-phase nucleus led to the formation of “pulverized” chromosomal
fragments rather than intact, compacted chromosomes.
• The results of this experiment suggested that the cytoplasm of a mitotic cell
contained diffusible factors that could induce mitosis in a nonmitotic (i.e.,
interphase) cell. This finding suggested that the transition from G2 to M was under
positive control; that is, the transition was induced by the presence of some
stimulatory agent.
The Role of Protein Kinases
• Entry of a cell into M phase is initiated by a protein called Maturation
Promoting Factor (MPF).
• MPF consists of two subunits: (1) a subunit with kinase activity that
transfers phosphate groups from ATP to specific serine and threonine
residues of specific protein substrates and
• (2) a regulatory subunit called cyclin.
• The term cyclin was coined because the concentration of this regulatory
protein rises and falls in a predictable pattern with each cell cycle .
• When the cyclin concentration is low, the kinase lacks the cyclin subunit
and, as a result, is inactive. When the cyclin concentration rises, the
kinase is activated, causing the cell to enter M phase.
• Over the past two decades, a large number of laboratorieshave focused
on MPF-like enzymes, called cyclin-dependent kinases (Cdks). It has
been found that Cdks are not only involved in M phase but are the key
agents that orchestrate activities throughout the cell cycle.
A simplified model
for cell cycle
regulation
in fission yeast. The
cell cycle is controlled
primarily at two
points, START and
the G2–M transition.
In fission yeast,
cyclins
can be divided into
two groups, G1
cyclins and mitotic
cyclins. Passage
of a cell through
these two critical
junctures requires
the activation of the
same cdc2 kinase by
a different type of
cyclin.
A third major
transition
occurs at the end of
• The first transition point, which is called START,
occurs in late G1. Once a cell has passed
START, it is irrevocably committed to
replicating its DNA and, ultimately, completing
the cell cycle.
• Passage through START requires the activation
of cdc2 by one or more G1 cyclins, whose
levels rise during late G1 (Figure 14.5).
Activation of cdc2 by these cyclins leads to the
initiation of replication at sites where
prereplication complexes had previously
assembled
• Passage from G2 to mitosis requires activation
of cdc2 by a different group of cyclins—the
mitotic cyclins.
• Cdks containing a mitotic cyclin (e.g., MPF)
phosphorylate substrates that are required for
the cell to enter mitosis. Included among the
substrates are proteins required for the
dynamic changes in organization of both the
chromosomes and cytoskeleton that
characterize the shift from interphase to
mitosis.
• Cells make a third commitment during the
middle of mitosis, which determines
whether they will complete cell division
and reenter G1 of the next cycle.
• Exit from mitosis and entry into G1
depends on a rapid decrease in Cdk
activity that results from a plunge in
concentration of the mitotic cyclins
Regulated Protein Phosphorylation and Degradation
Control Passage Through the Cell Cycle
• When a cyclin is present in the cell, it binds to the catalytic
subunit of the Cdk, causing a major change in the
conformation of the catalytic site.
• The concentrations of the cyclins, the regulatory subunits of
the heterodimeric protein kinases that control cell-cycleevents,
increase and decrease as cells progress through the cell cycle.
The catalytic subunits of these kinases, called cyclin-
dependent kinases (CDKs), have no kinase activity unless they
are associated with a cyclin.
• Each CDK can associate with different cyclins, and the
associated cyclin determines which proteins are
phosphorylated by a particular cyclin-CDK complex.
• Three major classes of cyclin-CDK complexes control
passage through the cell cycle: the G1, S-phase, and mitotic
cyclin-CDK complexes.
• When cells are stimulated to replicate, G1 cyclin-CDK complexes are expressed
first. These prepare the cell for the S phase by activating transcription factors
that promote transcription of genes encoding enzymes required for DNA
synthesis and the genes encoding S-phase cyclins and CDKs.
• The activity of S-phase cyclin-CDK complexes is initially held in check by
inhibitors. Late in G1, the G1 cyclin-CDK complexes induce degradation of the S-
phase inhibitors by phosphorylating them and consequently stimulating their
polyubiquitination by the multiprotein SCF ubiquitin ligase.
• Subsequent degradation of the polyubiquitinated S-phase inhibitor by
proteasomes releases active S-phase cyclin-CDK complexes.
• Once activated, the S-phase cyclin-CDK complexes phosphorylate regulatory
sites in the proteins that form DNA pre-replication complexes, which are
assembled on replication origins during G1.
• Phosphorylation of these proteins not only activates initiation of DNA replication
but also prevents reassembly of new pre-replication complexes. Because of this
inhibition, each chromosome is replicated just once during passage through the
cell cycle, ensuring that the proper chromosome number is maintained in the
daughter cells.
◈ Mitotic cyclin-CDK complexes are synthesized during the S phase and G2, but their activities are held in check by
phosphorylation at inhibitory sites until DNA synthesis is completed.
◈ Once activated by dephosphorylation of the inhibitory sites, mitotic cyclin-CDK complexes phosphorylate
multiple proteins that promote chromosome condensation, retraction of the nuclear envelope, assembly of the
mitotic spindle apparatus, and alignment of condensed chromosomes at the metaphase plate.
◈ During mitosis, the anaphase promoting complex (APC), a multisubunit ubiquitin ligase, polyubiquitinates key
regulatory proteins marking them for proteasomal degradation.
◈ One important substrate of the APC is securin,( a protein that inhibits degradation of the cross-linking proteins
between sister chromatids). The polyubiquitination of securin by the APC is inhibited until the kinetochores
assembled at the centromeres of all chromosomes have become attached to spindle microtubules, causing
chromosomes to align at the metaphase plate.
◈ Once all the chromosomes are aligned, the APC polyubiquitinates securin, leading to its proteasomal
degradation and the subsequent degradation of the cross-linking proteins connecting sister chromatids. This
sequence of events initiates anaphase by freeing sister chromatids to segregate to opposite spindle poles.
◈ Late in anaphase, the APC also directs polyubiquitination and subsequent proteasomal degradation of the
mitotic cyclins.
◈ Polyubiquitination of the mitotic cyclins by APC is inhibited until the segregating chromosomes have reached
the proper location in the dividing cell . Degradation of the mitotic cyclins leads to inactivation of the protein
kinase activity of the mitotic CDKs.
◈ Decrease in mitotic CDK activity permits protein phosphatases to remove the phosphates that were added to
specific proteins by the mitotic cyclin- CDK complexes.
◈ As a result, the now separated chromosomes decondense, the nuclear envelope re-forms arounddaughter-cell
nuclei, and the Golgi apparatus reassembles during telophase; finally, the cytoplasm divides at cytokinesis,
yielding the two daughter cells.
• During early G1 of the next cell cycle, phosphatases dephosphorylate
• the proteins that form pre-replication complexes.
• These proteins had been phosphorylated by S-phase
• cyclin-CDK complexes during the previous S phase, and their
• phosphorylation was maintained during mitosis by mitotic
• cyclin-CDK complexes. As a result of their dephosphorylation
• in G1, new pre-replication complexes are able to reassemble
• at replication origins in preparation for the next S
• phase (see Figure 21-2, step ). Phosphorylation of Cdh1
• by G1 cyclin-CDK complexes in late G1 inactivates it, allowing
• accumulation of S-phase and mitotic cyclins during the
• ensuing cycle. Passage through three critical cell-cycle transitions—
• G1 → S phase, metaphase → anaphase, and anaphase →
• telophase and cytokinesis—is irreversible because these transitions
• are triggered by the regulated degradation of proteins,
• an irreversible process. As a consequence, cells are forced to
• traverse the cell cycle in one direction only.
• In higher organisms, control of the cell cycle is
achievedprimarily by regulating the synthesis and activity
of G1 cyclin-CDK complexes.
• Extracellular growth factors function as mitogens by
inducing synthesis of G1 cyclin-CDK complexes.
• The activity of G1 cyclin-CDK complexes and other
cyclin-CDK complexes is regulated by phosphorylation at
specific inhibitory and activating sites in the catalytic
subunit. Once mitogens have acted for a sufficient
period, the cell cycle continues through mitosis even
when they are removed.
• The point in late G1 where passage through the cell cycle
becomes independent of mitogens is called the
restriction point
Overview of regulation of the Eukaryotic Cell
Cycle

• Passage through the cycle is controlled by G1, S-phase, and mitotic cyclin-
dependent kinase
• complexes (green).
• These are composed of a regulatory cyclin subunit and a catalytic cyclin-
dependent kinase (CDK) subunit.
• Two ubiquitin ligase complexes (orange), SCF and APC, polyubiquitinate specific
substrates including S-phase inhibitors(step5 ), securin (step8 ), and mitotic
cyclins (step 9), marking these substrates for degradation by proteasomes.
• Proteolysis of the S-phase inhibitor activates S-phase cyclin-CDK complexes,
leading to chromosome replication.
• Proteolysis of securin results in degradation of protein complexes that connect
sister chromatids at metaphase, thereby initiating anaphase, the mitotic period
in which sister chromatids are separated and moved to the opposite spindle
poles.
• Reduction in the activity of mitotic cyclin-CDK complexes caused by proteolysis
of mitotic cyclins permits late mitotic events and cytokinesis to occur.
• These proteolytic cleavages drive the cycle in one direction because of the
irreversibility of protein degradation.
CELL CYCLE CHECKPOINTS
✓ Checkpoints are surveillance mechanisms that halt the progress of the cell cycle
if (1) any of the chromosomal DNA is damaged, or (2) certain critical processes,
such as DNA replication during S phase or chromosome alignment during M
phase, have not been properly completed.
✓ Checkpoints ensure that each of the various events that make up the cell cycle
occurs accurately and in the proper order.
✓ the genes encoding several checkpoint proteins were first identified in mutant
yeast cells
✓ Checkpoints are activated throughout the cell cycle by a system of sensors that
recognize DNA damage or cellular abnormalities and it triggers a response that
temporarily arrests further cell cycle progress.
✓ The cell can then use the delay to repair the damage or correct the defect rather
than continuing to the next stage because mammalian cells that undergo
division with genetic damage will be transformed into a cancer cell.
✓ If the DNA is damaged beyond repair, the checkpoint mechanism can transmit a
signal that leads either to (1) the death of the cell or (2) its conversion to a state
of permanent cell cycle arrest (known as senescence).
1. The Presence of Unreplicated DNA Prevents Entry
into Mitosis
• unreplicated-DNA checkpoint control involves the recognition
of unreplicated DNA and inhibition of MPF activation
• ATR and Chk1 protein kinases, which also function in the DNA-
damage checkpoint, inhibit entry into mitosis by cells that have
not completed DNA synthesis.
• The association of ATR with replication forks activate its
protein kinase activity, leading to the phosphorylation and
activation of the Chk1 kinase.
• Active Chk1 then phosphorylates and inactivates the Cdc25
phosphatase (Cdc25C in vertebrates), which normally removes
the inhibitory phosphate from CDKs that function during
mitosis.
• As a result, the cyclin A/B-CDK1 complexes remain inhibited
and cannot phosphorylate targets required to initiate mitosis.
ATR continues to initiate this protein kinase cascade until all
replication forks complete DNA replication and disassemble.
Centrosome Duplication Checkpoint
• The initiation of centrosome duplication at the
G1–S transition is normally triggered by
phosphorylation of a centrosomal protein by
Cdk2
• Centrosome duplication is a tightly controlled
process so that each mother centriole
produces only one daughter centriole during
each cell cycle.
• The formation of additional centrioles can lead
to abnormal cell division and contribute to the
development of cancer.
Improper Assembly of the Mitotic Spindle Prevents the
Initiation of Anaphase

• The spindle-assembly checkpoint prevents entry into


anaphase when just a single kinetochore of one chromatid
fails to associate properly with spindle microtubules.
• Mad2 protein associate with kinetochores that are unattached
to microtubules.
• When Mad2 associates with a kinetochore complex that is not
bound by a microtubule, it is converted to a short lived
activated form that can interact with and inhibit Cdc20.
• once all kinetochore complexes bind a microtubule
▷ generation of the activated form of Mad2 ceases,
▷ the inhibition of Cdc20 is relieved,
▷ Cdc20 is free to direct the APC to polyubiquitinate securin,
▷ thereby anaphase starts.
DNA-damage checkpoint
• Cell-cycle arrest of cells with damaged DNA depends on tumor
suppressors
• DNA-damage checkpoint blocks progression through the cell
cycle until the damage is repaired.
• Damage to DNA can result from chemical agents and from
irradiation with ultraviolet (UV) light or -rays.
• Arrest in G1 and S prevents copying of damaged bases, which
would fix mutations in the genome. Replication of damaged
DNA also promotes chromosomal rearrangements that can
lead to cancer.
• Arrest in G2 allows DNA double-stranded breaks to be repaired
before mitosis.
• The proteins encoded by several tumor-suppressor genes,
including ATM and Chk2(these genes encode protein kinases),
normally function in the DNA-damage checkpoint. Patients with
mutations in both copies of ATM or Chk2 develop cancers far
more frequently than normal.

lodish
• ATM and ATR are protein kinases that become
activated following specific types of DNA
damage. Each of these proteins acts through
checkpoint signaling pathways that lead to cell
cycle arrest.
• ATM becomes activated in response to double-
strand breaks, which are detected by the MRN
protein complex.
• ATR, becomes activated by protein-coated
ssDNA that forms when replication forks
become stalled or the DNA is being repaired
after various types of damage.
In the G2 pathway,
▷ ATR phosphorylates and
activates the checkpoint kinase
Chk1,
▷ It phosphorylates and inactivates
the phosphatase Cdc25 which
normally shuttles between the
nucleus and cytoplasm
▷ Once phosphorylated, Cdc25 is
bound by an adaptor protein in
the cytoplasm and cannot be
reimported into the nucleus,
▷ this leaves the Cdk in its
inactivated phosphorylated state
Removal of the inhibitory phosphate
from (mammalian) CDK2 by Cdc25A
is required for onset of and passage
through the S phase mediated by
cyclin E-CDK2 and cyclin A-CDK2).
▷ Degradation of Cdc25 leads to
cell cycle arrest.
In the G1 pathway shown here,
▷ ATM phosphorylates and
activates the checkpoint
kinaseChk2 (step a),
▷ it phosphorylates p53 (step
b). p53 is normally very
short-lived, but
phosphorylation by Chk2
stabilizes the protein,
enhancing its ability to
activate p21 transcription
(step c).
▷ p21 directly inhibits the Cdk
(step e) by changing its
conformation and thereby
inhibiting its kinase activity.
▷ Cell cycle arrest occurs
▷ DNA damage due to UV light activates the ATM kinase
▷ It phosphorylates Chk2, thereby activating its kinase activity.
▷ Activated Chk2 then phosphorylates the Cdc25A phosphatase, marking it for
polyubiquitination by ubiquitin ligase and subsequent proteasomal degradation.
(Removal of the inhibitory phosphate from mammalian CDK2 by Cdc25A is required for onset of
and passage through the S phase mediated by cyclin E-CDK2 and cyclin A-CDK2).
▷ Degradation of Cdc25A resulting from activation of the ATM-Chk2 pathway in G1 or S-phase
cells thus leads to G1 or S arrest.
◈ Another tumor-suppressor protein, p53, contributes to arrest of cells with damaged DNA.
Cells with functional p53 arrest in G1 and G2 when exposed to -irradiation, whereas cells
lacking functional p53 do not arrest in G1.
◈ Although the p53 protein is a transcription factor, under normal conditions it is extremely
unstable and generally does not accumulate to high enough levels to stimulate
transcription.
◈ The instability of p53 results from its polyubiquitination by a ubiquitin ligase called Mdm2
and subsequent proteasomal degradation.
◈ The rapid degradation of p53 is inhibited by ATM and probably ATR, which phosphorylate
p53 at a site that interferes with binding by Mdm2.
◈ This and other modifications of p53 in response to DNA damage greatly increase its ability
to activate transcription of specific genes that help the cell cope with DNA damage.
◈ One of these genes encodes p21CIP, a generalized CIP that binds and inhibits all
mammalian cyclin-CDK complexes.
◈ As a result, cells are arrested in G1 and G2 until the DNA damage is repaired and p53 and
subsequently p21CIP levels fall.
◈ In vertebrates, the p53 response evolved to induce apoptosis in the face of extensive DNA
damage, presumably to prevent the accumulation of multiple mutations that might convert
a normal cell into a cancer cell.

karp
Cyclins and Cyclin-dependent
kinases
• The timing of the cell cycle is controlled by a family of protein
kinases with activities that change in response to cellular
signals. By phosphorylating specific proteins at precisely timed
intervals, they control the metabolic activities of the cell to
produce orderly cell division. The kinases are heterodimers
with a regulatory subunit, cyclin, and a catalytic subunit, cyclin-
dependent protein kinase (CDK).
• In the absence of cyclin, the catalytic subunit is virtually
inactive.
• When cyclin binds, the catalytic site opens up
• Animal cells have at least ten different cyclins (designated A, B,
and so forth) and at least eight cyclin-dependent kinases
(CDK1 through CDK8), which act in various combinations at
specific points in the cell cycle. Plants also use a family of
CDKs to regulate their cell division.

leh
Regulation of CDK by phosphorylation and
proteolysis.
• (a) The cyclin-dependent protein kinase activated
at the time of mitosis (the M phase CDK) has a “T
loop” that can fold into the substrate-binding site.
When Thr160 in the T loop is phosphorylated, the
loop moves out of the substrate-binding site,
activating the CDK manyfold.
• (b) The active cyclin-CDK complex triggers its own
inactivation by phosphorylation of DBRP
(Destruction Box Recognizing Protein). DBRP and
ubiquitin ligase then attach several molecules of
ubiquitin (U) to cyclin, targeting it for destruction
by proteasomes, proteolytic enzyme complexes.
Regulation of Plant Cell Cycle
• The CDKBs are present in higher plants in two sub-types called
CDKB1 and CDKB2
• The remarkable feature of the CDKB genes is that they are
expressed only in mitotic cells, from the S-phase until the M-
phase.
• The CDKB1 genes are expressed from S phase and peak in G2,
• the CDKB2 genes are expressed somewhat later from G2–M.
• Such cell-cycle-regulated expression is conventionally
associated with the cyclin subunit of CDK–cyclin complexes
rather than CDKs, and this cell cycle regulation is a unique
feature of the plant-specific CDKB class of CDKs.
• CDKA is rather broadly expressed, both in plant tissues and
during the cycle.
• In plants, the G1 cyclins are represented by a much larger group of genes than
the three present in mammals. All higher plants have six conserved sub-groups
of cyclin D genes, named CYCD1–CYCD7
• CDKA is activated by the binding of D-type cyclins (CYCD).
• Progression from G1 phase is initiated through the phosphorylation of the RBR
protein (retinoblastoma-related proteins:similar tomammalian tumour
suppressor protein RB,suppress cell division) by activated CDKA.
• A-type cyclins associate with CDKA during S-phase.
• Mitosis-specific CDKB (of two types CDKB1 and CDKB2) along with B-type
cyclins (CYCB) are required for mitosis.
• KRP(Kip-related proteins) proteins can bind to both CYCD and CDKA subunits
and inhibit the kinase activity of the complex.
• KRP proteins can be phosphorylated and inactivated by CDKB kinase activity,
further increasing CDK activity levels in mitosis
• Cytokinin has been shown to act in the cell cycle primarily through transcriptional
regulation of the CYCD3 genes
• Auxin is known to act, in part, through stabilization of the E2FB protein which
promotes mitotic cycles
Regulation of Plant Cell Cycle
Why do plants possess a mitotic-specific CDK showing highly
regulated expression?
The most likely explanation is connected to the prevalence of an alternative cell cycle in plant
development, known as the endocycle which is characterized by endoreduplication (or
endoreplication) of DNA. Endoreduplication results from S-phase without a following mitosis (i.e. a
Gap phase–S-phase cycle) and, therefore, gives rise to a doubling of nuclear DNA content through the
complete replication of chromosomes which can be repeated several times. Endoreduplication is a
widespread feature associated with tissue and organ growth in most plants (de Veylder et al., 2011).
Plant growth is continuous and associated with the repeated formation of new organs, such as
leaves and flowers. This occurs on the flanks of the shoot meristem, where the initiation of a new
organ is accompanied by rapid cell division which generates the majority of the cells that will
comprise the organ (Carraro et al., 2006).
Mitotic cycles giving rise to cell division are therefore primarily concentrated in meristematic
regions, such as at the tips of growing shoots and roots. As the organ becomes more distant from
the meristem due to its further growth, division reduces and cell expansion and differentiation occurs
(Andriankaja et al., 2012). These processes actually account for most of the growth of the organ and
are accompanied in many cell types by endoreduplication of one or more rounds (generating nuclei of
>4C). This process is of particular importance in fruit growth, as reviewed by Chevalier et al. (2014,
this volume) for the tomato model system.
As a consequence of the importance of endoreduplication, the switch from the mitotic to the
endocycle is an important feature of cell cycle control in plants. At the molecular level, this occurs
through the inactivation of mitosis-promoting factors from the cell cycle, and CDKB1 appears to play
the most important role: in its presence, the cycle leads to mitosis whereas, in its absence, an
endocycle results (Boudolf et al., 2004, 2009). One view is thus that the endocycle is the default cycle
and that mitosis requires the addition of the mitotic factors represented by CDKB. Alternatively, the
control of cell cycle phase transitions can be viewed in terms of different thresholds of CDK activity
as proposed by Coudreuse and Nurse (2010) who showed that, in yeast, there was a lower threshold
for DNA synthesis (G1/S) and a higher threshold for mitosis (G2/M). Hence de Veylder et al. (2011)
proposed that, in a mitotically active cell, CDK activity must pass the required threshold level for DNA
replication and the higher level need for mitosis. For endocycles, however, CDK activity must exceed
the lower threshold required for DNA synthesis, but must not be so high that mitosis is triggered (de
Veylder et al., 2011). In this view, the main role of CDKB is to provide additional kinase activity rather
than acting as a specific mitotic factor.
• Caspases A family of cysteine proteases
• that are activated at an early stage of
• apoptosis and are responsible for the
• degradative events observed during cell
• death.
E
x
t
r
i
n
s
i
c
The extrinsic (receptor-mediated)
pathway of apoptosis.
• When TNF binds to a TNF receptor (TNFR1), the activated receptor
binds two different cytoplasmic adaptor proteins (TRADD and FADD)
and procaspase-8 to form a multiprotein complex at the inner surface
of the plasma membrane.
• The cytoplasmic domains of the TNF receptor, FADD, and TRADD
interact with one another by homologous regions called death domains
that are present in each protein (indicated as green boxes.
• Procaspase-8 and FADD interact by means of homologous regions
called death effector domains (indicated as brown boxes).
• Once assembled in the complex, the two procaspase molecules cleave
one another to generate an active caspase-8 molecule containing four
polypeptide segments.
• Caspase-8 is an initiator complex that activates downstream
(executioner) caspases that carry out the death sentence.
• interaction between TNF and TNFR1 also activates other signaling
pathways, one of which leads to cell survival rather than self-
destruction.
Intrinsic
The intrinsic (mitochondria-mediated)
pathway of apoptosis.
• Various types of cellular stress cause proapoptotic members
of the Bcl-2 family of proteins, such as Bax, to become
inserted into the outer mitochondrial membrane.
• Insertion of these proteins leads to the release of cytochrome
c molecules from the intermembrane space of the
mitochondria.
• Release is thought to be mediated by pores in the
mitochondrial membrane that are formed by Bax oligomers.
• Once in the cytosol, the cytochrome c molecules form a
multisubunit complex with a cytosolic protein called Apaf-1
and procaspase-9 molecules.
• Procaspase-9 molecules are apparently activated to their full
proteolytic capacity as the result of a conformational change
induced by association with Apaf-1.
• Caspase-9 molecules cleave and activate executioner
caspases, which carry out the apoptotic response.
• The tumor-suppressor gene most often implicated in human
cancer is TP53, whose product (p53) may be able to suppress
cancer formation by several different mechanisms.
• In one of its actions,p53 acts as a transcription factor that
activates the expression of a protein (p21) that inhibits the
cyclin-dependent kinase that moves a cell through the cell
cycle.
• Damage to DNA triggers the phosphorylation and stabilization
of p53, leading to the arrest of the cell cycle until the damage
can be repaired.
• p53 can also redirect cells that are on the path toward
malignancy onto an alternate path leading to either apoptosis
or senescence.
• TP53 knockout mice begin to develop tumors several weeks
after birth.
• cancer cells that have sustained
• DNA damage are more likely to become apoptotic—as long
• as they possess a functioning TP53 gene. If cancer cells lose
• p53 function, they often cannot be directed into apoptosis
• and they become highly resistant to further treatment (Figure
• 16.14). This may be the primary reason why tumors that
• typically lack a functional TP53 gene (e.g., colon cancer,
prostate cancer, and pancreatic cancer) respond much more
• poorly to radiation and chemotherapy than tumors that
possess
• a wild-type copy of the gene (e.g., testicular cancer and
• childhood acute lymphoblastic leukemias).
function of p53

You might also like