Download 350-Cell Cycle-DF - Department Of Biological Sciences Hunter

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Signal transduction wikipedia , lookup

Extracellular matrix wikipedia , lookup

Apoptosis wikipedia , lookup

Tissue engineering wikipedia , lookup

Cytokinesis wikipedia , lookup

Cell encapsulation wikipedia , lookup

Cell growth wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Mitosis wikipedia , lookup

Cellular differentiation wikipedia , lookup

Cell culture wikipedia , lookup

Biochemical switches in the cell cycle wikipedia , lookup

JADE1 wikipedia , lookup

Programmed cell death wikipedia , lookup

Amitosis wikipedia , lookup

Cell cycle wikipedia , lookup

List of types of proteins wikipedia , lookup

MTOR wikipedia , lookup

MTOR inhibitors wikipedia , lookup

Transcript
TGF- and Cell Cycle Progression
G0
Restriction
Point
G1-pm
G1-ps
S
Cell Growth
Checkpoint
Cyclin D
CDK4/6
Cyclin E
CDK2
TGF-
G2
M
Effect of rapamycin on cell cycle progression in
MDA-MB-231 cells
G1 S G2/M
G1 S G2/M
G1 S G2/M
Sub genomic
Rapamycin induces primarily G1 arrest in the presence of
serum - and apoptosis in the absence of serum
Can TGF- suppress rapamycin-induced apoptosis?
TGF- is sufficient
to suppress
rapamycin-induced
apoptosis
Is TGF- necessary
for serum to
suppress rapamycininduced apoptosis?
Is TGF- in serum necessary for serum
to suppress rapamycin-induced apoptosis
% Non-Viable Cells
Figure 3A
100
% Non-Viable
50
Cells
Series1
0
1
-
2
-
+
3
+
Rap
-
+
+
+
TGF- -Ab
-
-
-
+
Serum
4
Cl PARP
actin
TGF- is necessary for serum to
suppress rapamycin-induced apoptosis
Summary:
•Rapamycin induces apoptosis in MDA-MB-231 cells in the
absence of serum
•In the presence of serum, rapamycin induces G1 arrest
•TGF- is sufficient to suppress rapamycin-induced apoptosis
in the absence of serum
•TGF- present in serum is necessary for serum to suppress
rapamycin-induced apoptosis
Question:
Why does rapamycin induce apoptosis when TGF- is
absent?
G0
Restriction
Point
G1-pm
G1-ps
S
G2
Cell Growth
Checkpoint
Cyclin D
CDK4/6
Cyclin E
CDK2
TGF-
TGF- suppresses G1 Cell Cycle Progression
M
G0
Restriction
Point
G1-pm
G1-ps
S
G2
M
Cell Growth
Checkpoint
Cyclin E
CDK2
Cyclin D
CDK4/6
TGF-
mTOR
mTOR suppresses TGF--induced G1 Cell Cycle Arrest
Nutrients
G0
Restriction
Point
G1-pm
G1-ps
S
G2
M
Cell Growth
Checkpoint
Cyclin E
CDK2
Cyclin D
CDK4/6
TGF-
mTOR
Rapamycin
Rapamycin reverses the mTOR suppression of TGF- signaling
and cells arrest in G1 in a TGF--dependent mechanism
G0
Restriction
Point
G1-pm
G1-ps
S
G2
M
Cell Growth
Checkpoint
Cyclin D
CDK4/6
Cyclin E
CDK2
X
TGF-
mTOR
Rapamycin
If TGF- signaling is suppressed or defective, there is no G1
arrest with rapamycin treatment - and now the cells die in the
presence of rapamycin - Why?
G0
Restriction
Point
G1-pm
G1-ps
S
G2
M
Cell Growth
Checkpoint
Cyclin E
CDK2
Cyclin D
CDK4/6
TGF-
mTOR
Rapamycin
Hypothesis: There is a critical requirement for mTOR in Sphase. Therefore, allowing cells into S-phase in the presence of
rapamycin (ie w/o mTOR) could result in apoptosis
G0
Aphidicolin
Restriction
Point
G1-pm
Synchronizes
Cells in early S
G1-ps
S
G2
M
Cell Growth
Checkpoint
Cyclin E
CDK2
Cyclin D
CDK4/6
TGF-
mTOR
Rapamycin
If hypothesis is correct, then blocking cells in S-phase - in the presence of serum/TGF-
- should result in apoptosis. This is because cells have passed the putative “Cell
Growth Checkpoint” and need mTOR signals to facilitate cell cycle progression through
S
Blocking cells in S-phase with aphidicolin
sensitizes cells to rapamycin
In the presence of
serum/TGF- - if cells are
allowed to enter S-phase,
then the lack of mTORC1
signals to 4E-BP1 tells the
cell that nutrients are in
short supply and that
replicating the genome is
probably a bad career move!
% Non-Viable Cells
Figure 6A
100
% Non-Viable
50
Cells
Series1
0
Rap
Aph
Cl PARP
actin
1
-
2
+
3
-
4
-
+
+
+
The cells then do the
honorable thing – and
commit suicide
IMPLICATION:
Cancer cells with defective TGF- signaling could be
selectively killed by rapamycin in the presence of either
serum or TGF-
Importantly:
Many cancers have defects in TGF- signaling –
especially Smad4 - that is critical for suppression of
G1 cell cycle progression
Cancer cells with defective TGF- signaling are
Selectively killed by rapamycin in the presence of serum
Breast (Smad4)
Breast (No TGF- defect)
MDA-MB-231
Breast (PKCδ)
100
SW480
50
Series1
% Non Viable Cells
% Non Viable Cells
Colon (Smad4)
100
BT-549
50
Series1
0
0
Serum
+
-
-
+
-
Serum
+
-
-
+
-
Rap
-
-
+
+
+
Rap
-
-
+
+
+
TGF-
-
-
-
-
+
TGF-
-
-
-
-
+
Cl PARP
actin
Cl PARP
actin
Summary:
1)
If TGF- is present, rapamycin induces cell cycle arrest in G1 - by
increasing TGF- signaling
2)
In the absence of TGF- signaling, rapamycin does not arrest cells in late
G1 and they progress through the remainder of G1 into S-phase
3)
However, if cells progress into S-phase in the presence of rapamycin, they
undergo apoptosis rather than arrest - because of an apparent stringent
requirement for mTOR during S-phase
Cell Growth
Checkpoint
Rapamycin induces arrest
Rapamycin induces apoptosis
G1
S
Nutrients
Cyclin D-CDK4/6
Cyclin E-CDK2
p27
Rapamycin
PLD
mTOR
TGF-
Survival
Signals
PI3K
Growth
Factors
Implications:
In addition to the rapamycin concentration issues – another
complication is that in most cancers, rapamycin is cytostatic
rather than cytotoxic
Therefore - Defective TGF- signaling may be an Achilles heel
for strategies that target mTOR - especially in colon and
pancreatic cancers where defects in TGF- signaling are
common
Defective TGF- signaling creates a “Synthetic Lethality” for
strategies that suppress the phosphorylation of 4E-BP1 by
mTORC1
Alternatively
Strategies that suppress mTOR could be combined with
strategies that suppress TGF- signaling – creating a synthetic
lethal situation
Complementary Signals promote G1
Cell Cycle Progression
G0
(Quiescence)
Restriction
Point
G1-pm
G1-ps
Cell Growth
Checkpoint
(mTOR)
Growth Factor Signals
Tyrosine kinases
Ras/Raf/MEK/MAPK
S
G2
Gatekeepers
Myc
SV40 Early Region
(Suppression of
p53, Rb and PP2A)
M
Growth Factor
Signals
Restriction
Point
G1-pm
G1-ps
Nutritional
Sufficiency
Cyclin D-CDK4/6
Amino acids
Fatty acids
Energy
ATP
RalA
O2
Vps34
Cell
Size
Nutritional
Sufficiency
Cell Growth
Checkpoint
(START)
S
Cyclin E-CDK2
G0
Cyclin A-CDK2
Rheb
PLD
mTOR
TGF-
Commitment
Cell Growth
Conventional View of Cell Cycle
Points:
The Restriction Point, originally characterized
by Arthur Pardee, is a point in G1 where
cells no longer require growth factors and
commit to completing the cell cycle
In the absence of growth factors, cells exit
the cell cycle into quiescence or G0
Zetterberg and colleagues have mapped the
Restriction Point to a site ~ 3.5 hr after
mitosis - where cyclin D is elevated
Figure 8.8 The Biology of Cancer (© Garland Science 2007)
From: Weinberg, The Biology of Cancer, 2007
Leland Hartwell described a site in the Yeast
cell cycle called START that is late in G1 where cells evaluate whether there is
sufficient nutrition to complete cell division
In some texts, the Restricition Point is referred to as the mammalian
equivalent of START - and located near the site where cyclin E is activated
Rapamycin treatment results in the activation of TGF- signaling and arrest at
the cyclin E site - that can be clearly distinguished both temporally and
genetically from the growth factor-dependent Restriction Point
Genetic requirements for the transformation of human cells (I)
(Hahn et al., Nature 400:464, 1999; MCB 22;2111, 2002)
Genetic effect
Molecular Target
Cell cycle target
Ras
Growth factor signals
Restriction point
SV40 Large T
p53
Rb
G1/S checkpoint
All G1 checkpoints
SV40 small t
PP2A
Cell Growth checkpoint (?)
Genetic requirements for the transformation of human cells (II)
(Boehm et al., MCB 25:6464, 2005)
Genetic effect
Molecular Target
Cell cycle target
Ras
Growth factor signals
Restriction point
p53 KO
Rb KO
p53
Rb
G1/S checkpoint
All G1 checkpoints
Myc
PTEN KO
Gene expression
mTORC1
Cell Growth checkpoint (?)
Cell Growth checkpoint (?)
Restriction Point
Cell Growth Checkpoint
Growth Factor Signals
Ras
mTOR Signals
PI3K
Insulin/IGF1
PIP3
PIP2
PTEN
mTORC2
PDK1
Raf
Ser473
Akt
T308
TSC1/2
Mek
Amino
acids
Rheb
PLD1
MAPK
FKBP38
TGF-
Myc
Cyclin D
LKB1
AMPK
Energy
status
mTORC1
S6K
AMP
Cyclin E
Growth factor, essential amino acid, glutamine, and suppression of
mTOR block G1 cells cycle progression at distinguishable sites in G1
A temporal relationship
can be established
whereby the GFdependent R is upstream
from sites that are
sensitive to EAA, Q, and
mTOR suppression
Temporal mapping of G1 cell cycle checkpoints
Cells require 12- 16 hr to enter S-phase
after GF deprivation or mTOR suppression
Cells require 20+ hr to enter S-phase after
amino acid deprivation
For cells arrested in G0 by GF deprivation,
amino acid deprivation blocks entry into Sphase for 12-14 hr
For cells arrested in G0 by GF deprivation,
mTOR suppression blocks entry into Sphase for 16-18 hr
Blocking G1 cell cycle progression by growth factor
and nutrient deprivation, and suppression of mTOR
A
B
P-Rb S807/811
D
Cyclin D
CM (hrs.)
P-AktT308
-GF
P-AktS473
-EAA
P-S6KT389
P-4EBP1
LC3-II
p85
p70
D2/D3
D1
-Q
+Rapa.
C
Akt
Pan-Rb
E
p21
CM (hrs.)
S6K
-GF
4EBP1
-EAA
Actin
-Q
+Rapa.
Different blocking conditions have differential effects on cell cycle
regulators and on autophagy – notably for EAA and Q
Summary
Data supports a model where there is GF-dependent R where multi-cellular
organisms determine whether it is appropriate for a cell to divide
During G1-ps, cells that have been given the green light to divide, determine
whether they have the means/raw materials to double the mass of a cell,
Replicate its genome, and divide into two daughter cells
The late G1 “Metabolic Checkpoints” in late G1 collectively represent a
“Cell Growth” checkpoint that responds to nutrients that is evolutionarily
equivalent to START in the yeast cell cycle
TOR/mTOR is likely the ultimate arbiter for determining nutrient sufficiency
Complementing oncogenic alterations dysregulate Restriction Point
and Cell Growth checkpoints
Restriction Point
Cell Growth Checkpoint
Growth Factor Signals
mTOR Signals
Ras
PI3K
Insulin/IGF1
PIP3
PIP2
PTEN
mTORC2
PDK1
Raf
Ser473
Akt
T308
TSC1/2
Mek
Amino
acids
Rheb
PLD1
MAPK
FKBP38
TGF-
Myc
Cyclin D
LKB1
AMPK
Energy
status
mTORC1
S6K
AMP
Cyclin E
Dysregulated metabolic checkpoints in
cancer cells
In response to amino acid deprivation, MDA-MB-231 breast and Panc1
pancreatic cells arrest in S-phase and G2/M
Conclusions
The GF-dependent R can be distinguished from late G1 metabolic
checkpoints and mTOR
The G1 metabolic checkpoints – like R – are dysregulated in
human cancer cells
Cooperating genetic alterations in cancer cells disable both R and
the late metabolic checkpoints that collectively may represent a
“Cell Growth” checkpoint with mTOR as the final arbitor
Surprisingly, mTOR, which is widely known to be regulated by amino
acids, blocked cell cycle progression well downstream of the amino
acid sites
It is hypothesized that other nutrient inputs – such as glucose and
phosphatidic acid (lipids) may be required for complete activation of
mTOR and progression into S-phase