You are on page 1of 45

Cell Cycle

I Made Winarsa Ruma, M.D., Ph.D


Department of Biochemistry
Faculty of Medicine Udayana University
Cell Cycle
• The cell cycle is the series of events in which cellular
components are doubled, and then accurately segregated
into daughter cells.
• In eukaryotes, DNA replication is confined to a discrete
Synthesis or S-phase, and chromosome segregation
occurs at Mitosis or M-phase.
• Two Gap phases separate S phase and mitosis, known as
G1 and G2. These are not periods of inactivity, but
rather periods where cells obtain mass, integrate growth
signals, organize a replicated genome, and prepare for
chromosome segregation.
• It serve as regulatory windows to ensure that cell cycle
events occur at the correct time and in the right order.
• The gap phase separating M phase from S phase is
known as G1 phase and marks the time when cells
make the decision to exit the cell cycle or continue
through further rounds of division.
• This decision is classically thought of as being controlled
through the phosphorylation of retinoblastoma
(RB) family proteins by CDKs, thereby establishing a
binary switch mechanism known as the restriction
(R-) point that gates cell cycle progression in G1 phase.
• The cell division cycle plays a crucial role in the growth,
development, repair and reproduction of living
organisms in both normal and pathological conditions.
• Progression through the cell cycle requires faithful
replication of the genome during S phase (DNA synthesis)
and equal partitioning of the replicated chromosomes to
the two daughter cells during mitosis and cell division (M
phase).
• Strict alternation of S and M phases is essential
for successful cell proliferation, the mechanisms
responsible for the temporal ordering of these
two events are of fundamental importance to all
eukaryotic cell life
Cell Cycles
as the
central
governor of
growth and
proliferatio
n
1 hr

3-5 hrs

12-15 hrs

6-8 hrs
Responsiveness to Extracellular
Signals during Cell Cycle
Cyclin-dependent kinases
(CDKs)
• The central machines that drive cell cycle progression.
• These are serine/threonine protein kinases that phosphorylate
key substrates to promote DNA synthesis and mitotic progression.
• The catalytic subunits are in molar excess, but lack activity until
bound by their cognate cyclin subunits, which are tightly
regulated at both the levels of synthesis/ transcriptional
and posttranscriptional mechanisms; ubiquitin-
dependent proteolysis.
• Cyclin-binding allows inactive CDKs to adopt an active
configuration akin to monomeric and active kinases.
• CDK activity can also be negatively regulated by the binding of
small inhibitory proteins, the CKIs, or by inhibitory tyrosine
phosphorylation which blocks phosphate transfer to substrates.
Cyclin Dependent Kinases
(CDKs)
• CDKs coordinate the initiation of the two key cell cycle events,
replication of DNA and its segregation at mitosis.
• The DNA replication licensing system ensures that
chromosomal DNA is replicated precisely once before cell
division occurs.
• Replication of eukaryotic chromosomal DNA is initiated from
several sites on each chromosome, called replication origins.
• The origins thus modified are defined as licensed origins and
can be activated for initiation by S-phase CDKs as cells enter S-
phase.
• Checkpoint controls regulate cell cycle progression, so that
initiation of replication is coordinated with chromosome
segregation.
Checkpoint Control
Loss of Checkpoint Control

Hiperploidy
Cell Cycle Regulation by Checkpoints
• Cell cycle checkpoints are surveillance
mechanisms that monitor the order,
integrity, and fidelity of the major events of
the cell cycle which including:
• growth to the appropriate cell size,
• the replication and integrity of the
chromosomes, and
• their accurate segregation at mitosis.
Checkpoints emerged as a series of cell cycle
dependencies associated to:

a) Cell Size Control


b) DNA Damage Responses
c) Monitoring DNA Replication
d) S–M Dependency
e) The Mitotic Spindle Checkpoint
Cell Size Control
• Control of cell size is critical for regulating nutrient
distribution for the cell and for regulating organ size and
function in multicellular organisms.
• The existence of cell size checkpoints has been proposed
for allowing cells to coordinate cell size with cell cycle
progression.
• Cell size checkpoints have been observed in G1 and G2.
• One proposed mechanism for control of cell size is via the
monitoring of protein translation which including
ribosomal mass, and thus translational activity.
DNA Damage Responses
• The source of DNA damage may be intrinsic, such as
a) intermediates of metabolism,
b) attrition of telomeres,
c) oncogene overexpression, and
d) DNA replication errors
• extrinsic sources of DNA damage:
a) sunlight,
b) carcinogens,
c) ionizing radiation or
d) anticancer therapeutics.
• DNA damage checkpoints can be controlled by
1) the tumor suppressor and transcription factor p53,
2) the checkpoint kinase Chk1
Checkpoint-dependent cell cycle arrest and exit
Tumor suppressor and transcription factor p53
• In higher organisms, p53 is a critical component of DNA damage
checkpoints, particularly in G1 phase.
• p53 is regulated by a plethora of posttranslational
modifications, including N-terminal phosphorylation on serine-15.
• Activated p53 is stabilized through protection from its E3 ubiquitin
ligase Mdm2, and as a tetramer transactivates the expression of a
large number of genes, including the cyclin-dependent kinase
inhibitor (CKI) p21.
• Thus G1 CDKs are inhibited, and DNA damage is repaired prior to DNA
replication.
• p53 is required for prolonged G2 arrest in the face of persistent
DNA damage.
• p53 can direct the alternative cell fates of apoptosis or
senescence.
Monitoring DNA Replication
• DNA replication is initiated at specific sites, the
replication origins.
• These are epigenetically defined by a number of proteins
that ensure they fire (start replicating) once and only
once per cell cycle.
• Replication origin firing is controlled by the
phosphorylation of two proteins, Cdt1 and Cdc6 ,
which is catalyzed by both CDKs and the Dbf4-dependent
protein kinase (DDK) Cdc7.
• Such phosphorylation not only initiates replication but
also leads to degradation of these proteins, and hence
the origin cannot refire.
S–M Dependency
• When the polymerase and its associated proteins (the
replisome) encounter a blockade to progression by
modified dNTPs, abasic sites, protein–DNA
complexes, or result from the depletion of dNTPs.
• This replisome must be stabilized as the function of
the intra-S-phase checkpoint.
• Mitotic entry is blocked via Y15 phosphorylation of
Cdc2.
• In order to maintain ploidy, there is an equally
important dependency relationship to ensure one
round of replication per cell cycle.
The Mitotic Spindle Checkpoint
• The segregation of sister chromatids at anaphase is
under the mechanical control of the mitotic spindle.
• A large E3 ubiquitin ligase known as the Anaphase-
Promoting Complex or Cyclosome (APC/C) targets
the mitotic cyclins, which abolishes CDK activity.
• The formation of the spindle and the detection and
correction of spindle defects are under the control of the
Polo, Aurora, and NIMA-related (Nek) kinases.
• The spindle checkpoint controls DNA integrity by
preventing a cell cycle transition while other effectors
correct a genome-altering defect.
Pairing of Cyclins
with CDKs
Fluctuation of cyclin levels
during cell cycle
Control of Cyclin D1 Level
Control of cyclins level during
the cell cycle
Cyclin-CDK complexes are regulated
by CDK inhibitors
Control of cell cycle by TGF-β
Control of cell cycle by
extracellular signal
Cell cycle-dependent
phosphorylation
of pRb Protein Phosphatase type I
(PP1)
Control of R-point Transition by
Mitogens

pRb as the guardian of restriction-point gate


pRb
targets
Transcription
Factors
E2Fs
Control of Trancription by pRb
Positive-feedback loops and the irreversibility of cell cycle
Cell Fate Decision Associated to Cell
Cycle
• A strong connection exists between the cell cycle and
mechanisms required for executing cell fate decisions in a
wide-range of developmental contexts.
• Terminal differentiation is often associated with cell
cycle exit, whereas cell fate switches are frequently
linked to cell cycle transitions in dividing cells.
• The cell cycle machinery impactschromosome
architecture, the epigenome and
transcriptional programs required for cell identity
in multiple contexts including differentiation,
reprogramming and trans-differentiation.
the expression of cell fate ‘decision’ genes is
often coupled to cell cycle regulatory
mechanisms
Multiple roles
of cell cycle
regulators
in the
regulation of
cell cycle
signaling,
metabolism,
and growth.
Participation
of cell cycle
regulators in
metabolic
pathways.
Overview on cell cycle regulators’ modulation of mitochondrial
dynamics and the endolysosomal pathway during the cell cycle.

You might also like