ArticlePDF AvailableLiterature Review

Levine AJ, Hu W, Feng Z.. The P53 pathway: what questions remain to be explored? Cell Death Differ 13: 1027-1036

Authors:
  • Rutgers Cancer Institute of New Jersey

Abstract

The p53 pathway is composed of hundreds of genes and their products that respond to a wide variety of stress signals. These responses to stress include apoptosis, cellular senescence or cell cycle arrest. In addition the p53-regulated genes produce proteins that communicate these stress signals to adjacent cells, prevent and repair damaged DNA and create feedback loops that enhance or attenuate p53 activity and communicate with other signal transduction pathways. Many questions remain to be explored in our understanding of how this network of genes plays a role in protection from cancers, therapy and integrating the homeostatic mechanisms of stress management and fidelity in a cell and organism. The goal of this chapter is to elucidate some of those questions and suggest new directions for this area of research.
Review
The P53 pathway: what questions remain to be
explored?
AJ Levine*
,1
,WHu
1
and Z Feng
1
1
Institute for Advanced Study and The Cancer Institute of New Jersey,
Princeton, NJ 08540, USA
* Corresponding author: AJ Levine, Institute for Advanced Study and The
Cancer Institute of New Jersey, Princeton, NJ 08540, USA.
Fax: þ 1-609-924-7592; E-mail: alevine@ias.edu
Received 22.12.05; revised 15.2.06; accepted 22.2.06; published online 24.3.06
Edited by A Braithwaite
Abstract
The p53 pathway is composed of hundreds of genes and their
products that respond to a wide variety of stress signals.
These responses to stress include apoptosis, cellular
senescence or cell cycle arrest. In addition the p53-regulated
genes produce proteins that communicate these stress
signals to adjacent cells, prevent and repair damaged DNA
and create feedback loops that enhance or attenuate p53
activity and communicate with other signal transduction
pathways. Many questions remain to be explored in our
understanding of how this network of genes plays a role in
protection from cancers, therapy and integrating the homeo-
static mechanisms of stress management and fidelity in a cell
and organism. The goal of this chapter is to elucidate some of
those questions and suggest new directions for this area of
research.
Cell Death and Differentiation (2006) 13, 1027–1036.
doi:10.1038/sj.cdd.4401910; published online 24 March 2006
Keywords: The p53 pathway; feedback loops; apoptosis; stress
responses; DNA damage
Abbreviations: MDM-2, murine double minute 2; COP-1, coat
protein complex 1
Introduction
The goals for this chapter
The p53 protein was first described in 1979
1–4
and since that
time there have been more than 35 000 papers published
on this topic. The description of this protein and its gene
has changed from a virus-associated tumor antigen to an
oncogene to a tumor suppressor gene.
5
The major functions
of the p53 protein have been elucidated and reasonable
explanations are in hand that describe why it is an important
tumor suppressor gene in humans and animals.
6
This
progress has been reported at 12 different meetings
dedicated solely to research with the p53 protein, which have
been held every other year starting in 1981, with the most
recent meeting being held in New Zealand (2004) and the next
one will be in New York City, USA (2006). As each meeting
results in hundreds of presentations and posters they provide
the latest concepts of how the p53 protein functions and what
its many roles are in the cell and the organism. From this a
consensus forms and new questions emerge that change the
concepts, ideas and directions of this area of research. That is
just what happened at the last meeting and the goal of this
chapter is then to help guide and formulate the questions that
remain to be explored over the next years. Some of these
questions address very specific observations and are quite
narrow, but important to answer. Other questions are global
and address large conceptual issues in the field. The format of
this chapter will be to describe the functioning of the p53
pathway and at each specific step to identify those narrow and
focused questions that require the attention of experimental-
ists and need answers. By asking these questions we may
uncover contradictions in the literature, point out results that
remain unclear or bring up issues that remain to be tested and
are unresolved. As we move through the descriptions of each
step in the p53 pathway and note these focused issues they
will collectively point to holes in our understanding and lead to
the bigger issues. In the final section of this chapter the larger
or global questions can be explored in the context of
understanding the details of the p53 network and its functions.
The p53 pathway may conveniently be divided up into five
parts (Figures 1 and 2); (1) The input signals that trigger or
induce the network into a functional state. (2) The upstream
mediators that detect and interpret those signals that initiate
the functional pathway and relay the inputs to the p53 protein
or molecules that most immediately (in minutes to an hour)
regulate the concentration and activity of the p53 protein. (3)
The core set of proteins, including the p53 protein itself, which
regulates p53 activity and function. (4) The downstream
events which are composed of a set of genes and their
proteins that are regulated by the p53 protein, most commonly
by transcriptional activation but in some cases by protein–
protein interactions. (5) The cellular outputs of these down-
stream events which include cell cycle arrest, cellular
senescence or apoptosis and often result in extensive
communication with other signal transduction pathways in
the cell. Employing this format we can identify the open
questions that remain to be explored. Then we can turn our
attention to more global questions about these processes in a
larger context of the cell and organism.
The P53 Pathway
The input signals that trigger or induce the p53
pathway
The p53 protein and its signal transduction pathway are
composed of a set of genes and their protein products that are
Cell Death and Differentiation (2006) 13, 1027–1036
&
2006 Nature Publishing Group All rights reserved 1350-9047/06
$
30.00
www.nature.com/cdd
designed to respond to a wide variety of intrinsic and extrinsic
stress signals. These stress signals all impact upon the
cellular homeostatic mechanisms that monitor and control the
fidelity of DNA replication, chromosome segregation and cell
division.
7
Among the stresses that activate the p53 protein are
damage to the integrity of DNA in a cell. There are many
physical and chemical causes of DNA damage including
gamma or UV irradiation, alkylation of bases, DNA cross-
linking, depurination of DNA, alteration of the deoxyribose
sugar moiety, reaction with oxidative free radicals and more.
DNA
Damage
Hypoxia
rNTP
Depletion
Oncogene
Activatio
n
Spindle
Damage
Nitric
Oxide
(NO)
Activation of p53
?
?
?
Stress Signals
(input)
Mediators
Core Regulation
MDM2
p53
Nutrition
deprivation
E2F-1
P14/p19
ARF
ROS UV
γ-irradiation
REDD-1
ATR
CHK1
ATM
CHK2
AMPK
Heat/cold
shock
MDM-X
Kinases (ATM, ATR, CHK1, CHK2); Phosphatases (PP2A)
Acetyltransferases (p300, CBP, PCAF); Deacetylases (HDAC, SIRT-1)
Unbiquitin Ligases (UbcH5B/C); Deunbiquitinases (HAUSP)
Methylases (Set9); Sumoylases (PIAS-1, Ubc9, Topors)
Neddylation (NEDD8); Werner helicase (WRN); PML; HMG1
?
Figure 1 The input signals, mediators and core p53 functions of the p53 pathway. Several types of stress signals are detected by the cell and communicated to the p53
protein and its core constituents by the mediators. Several stress signals result in the degradation of the MDM-2 protein and the increase in the levels and activity of the
p53 protein. The MDM-2 protein levels are then restored via the p53-mediated transcription of the MDM-2 gene
Downstream Events of p53
Core Regulation
Inhibition of
angiogenesis
and metastasis
PAI
BAI-1
KAI
TSP1
Maspin
GD-AiF
A
p
o
p
tosis
IGF-BP3
PAG608
Siah-1
Scotin
PERP
Fas
PIDD
Killer/DR5
p53AIP
PIGs
Noxa
PUMA
Bax
Apaf-1
Cell cycle
arrest
G1-S
p21
14-3-3-σ
Reprimo
Gadd45
B99
G2-M
?
Cellular
senescenc
e
DNA Repair
and damage
prevention
p48
p53R2
PTEN
TSC2
IGF-BP3
Inhibition
of IGF-1
/mTOR
pathway
Siah-1
Wip1
Cyclin G
Cop-1
PIRH-2
deltaNp73
P53
negative
feedback
TSAP6
Exosome
mediated
secretion
Sestrins
Gadd45
MDM2
MDM2
p53
E2F-1
P14/p19
ARF
MDM-X
Cdc2
Cyclin B
Cdc25C
Cyclin E
Cdk2
Casp 8
Bid
Cyto C
Casp 9
Casp 3
Response
(output)
Figure 2 The core p53 functions, the downstream genes regulated by p53 and the outputs of the p53 pathway. The p53 protein is employed as a transcriptional
activator of p53-regulated genes. This results in three major outputs; cell cycle arrest, cellular senescence or apoptosis. Other p53-regulated gene functions
communicate with adjacent cells, repair the damaged DNA or set up positive and negative feedback loops that enhance or attenuate the functions of the p53 protein and
integrate these stress responses with other signal transduction pathways
The P53 pathway
AJ Levine et al
1028
Cell Death and Differentiation
Each of these types of DNA damage is different and each is
detected by a different set of proteins and then repaired by
different enzymes or activities that reverse the damage in
diverse ways. There are multiple DNA damage detection and
repair systems in the cell but every type of DNA damage
is reported to the p53 protein and its pathway.
8–10
The
duplication processes of DNA often make mistakes (i.e.
mismatch errors) that are in the main edited and repaired, but
when lesions remain they too can be detected by p53. In all of
these cases the p53 pathway functions to respond to errors (a
check on the fidelity of these processes) and eliminates those
cells with such mistakes. As cells divide and the telomeres
of chromosomes get shorter they reach a critical size that
can result in an enhanced rate of translocations or abnormal
recombination events. The presence of these shorter telo-
meres in a cell are reported to the p53 protein resulting in a
p53 response which in turn limits abnormal chromosome
translocations. In addition to DNA damage several cellular
deprivations activate the p53 response such as hypoxia,
glucose starvation or the depletion of ribonucleoside triphos-
phate pools in the cell. When ribosome biogenesis is stopped
or falls below a critical level the p53 protein is alerted and
activated. Agents that damage the cell spindle and result in
faulty chromosome segregation activate p53. Both heat and
cold shock as well as the altered or denatured protein
response in cells communicate with p53 by activating it. Nitric
oxide, which is often associated with infections and inflamma-
tion activate the p53 protein and its response. All of these
stresses will result in a loss of fidelity in the cellular duplication
process and as such their communication with the p53
pathway acts as a check point to prevent abnormal clones
of cells from arising. The mutational activation of some
oncogenes also result in the sensitization of the p53 response
and can often change the downstream output of the p53
pathway from cell cycle arrest to apoptosis. This very large
number of diverse cellular stress signals that all feed into a
central and single node to monitor and respond to those
stresses may be thought of as a poor design of a system that is
too vulnerable to the loss of a single central node.
7
Why don’t
different stress signals act upon different protein checkpoints?
Why is the pathway not duplicated or backed up so as to make
it more robust and less vulnerable to the mutational damage of
the p53 gene? The observation that demonstrates the unique
and central function of the p53 protein in this stress network is
that the p53 gene is mutated about 50% of the time in a wide
variety of cancers.
11
In a number of other cancers additional
genes in the network that encode proteins that act upon the
p53 functions are mutated. Thus, most cancers appear to
select for a loss of function of the p53 pathway.
12
The most
common answer to why evolution has designed the network to
have a single central node that receives inputs from a wide
variety of stress signals is that one entity or protein can act as
the most efficient integrator of information about the different
types of stress that act upon the cell. Multiple proteins
receiving multiple input signals would have to have a much
more complicated communication system to integrate infor-
mation about the environmental stresses. Because all of
these stresses lead to a lack of fidelity in the duplication
process, and that could lead to disease, the p53 protein is a
central player in the go-no go decision of a cell to grow or
reproduce. The question remains; is this explanation a correct
interpretation of the network architecture or is there a lot more
to learn so as to answer this enigma?
What other stress signals can induce a p53 response?
What more remains to be elucidated in the type or nature of
the input signals? Can psychological stress, in the form of
depression or nervous system activity activate the p53
protein? Do some hormonal changes result in the activation
of the p53 protein? What nutritional changes, in addition to
glucose deprivation,
13,14
can induce a p53 response? It will be
important to fill in those inputs to the p53 pathway over the
next few years. In some cases (i.e. psychological stress) it will
be a real challenge to design experiments to prove these
points. Most of the experiments in the past that test for the
activation of the p53 protein and therefore define an input
signal for this pathway have been carried out in cell culture. In
order to better define environmental stress we will need
to carry out experiments (like testing psychological stress)
in the whole organism taking into account cell type and
tissue specificity as well as the variable of time (age) and
developmental stage.
Orthologues of the p53 gene have been identified in worms,
flies, mice and humans and it will clearly be instructive to
elucidate a comparative genomic and network analysis and
follow changes in p53 function over evolutionary time scales.
Similarly two homologues of the p53 gene, p63 and p73, have
been identified
15,16
and it will be important to complete the
characterizations of their functions, their possible role in
diseases and their impact upon the p53 gene and its protein.
17
Do p63 or p73 respond to stress signals and if so which ones?
P73 is phosphorylated by the Abl kinase after DNA damage
and p73 can induce cellular apoptosis. There is a great deal to
be learned about these functions in the cell and organism.
The upstream mediators of the P53 response
In response to an input stress signal the p53 protein is
activated. We can define activation experimentally as an
increase in the concentration of the p53 protein and an
increased activity of a p53 protein for the transcription of a set
of genes that have a p53 DNA response element
18
and are
transcriptionally regulated by the p53 protein. The levels of the
p53 protein are predominantly regulated by it’s proteolytic turn
over. The p53 protein has a short half-life of 6–20 min in
several cell types and the ubiquitin ligase that confers this
short half-life is the MDM-2 protein. Recently, two other
ubiquitin ligases were shown to act upon the p53 protein,
COP-1 and PIRH-2.
19,20
Why is this redundancy employed?
What are the roles of these enzymes for the many diverse
stress inputs, in different tissue or cell types or at the different
times in development of an organism? After some types of
DNA damage (gamma radiation) the MDM-2 protein is auto-
poly-ubiquitinated resulting in its degradation and an asso-
ciated increase in p53 levels and activity.
21
While this
observation helps to explain why the p53 protein has a longer
half-life after some types of DNA damage, this mechanism is
not observed in all types of DNA damage or stress signals.
Indeed given the large number of stress signals employed as
an input to the pathway we know almost nothing about how
these diverse inputs are communicated to the p53 protein.
The P53 pathway
AJ Levine et al
1029
Cell Death and Differentiation
That is a big challenge for future experiments. We do know
that the p53 and MDM-2 proteins are extensively modified
after a stress signal.
22
The p53 protein is phosphorylated on a
large number of serine and threonine residues by many
different protein kinases. It is acetylated by histone acetyl-
transferases, methylated by methylases, ubiquitinated, sum-
molated and neddylated (at epsilon amino groups of lysine at
the carboxy-terminus of the p53 protein).
23–25
In some cases it
is clear that this is part of the process that mediates the
response from DNA damage to the p53 protein. For example,
gamma radiation activates the ATM protein kinase that then
phosphorylates MDM-2 and p53 (probably through a CHK
kinase) as well as participating in the DNA repair process. In
spite of these clear observations, mutations in many of the
p53 protein serine and threonine residues that then block
p53 phosphorylation events still result in fairly normal p53
activation and function. Similarly, the use of Nutlin (a drug
that blocks p53-MDM-2 binding) activates p53 normally with
no phosphorylation events.
26
Changes of the lysines in p53
protein, that are normally acetylated or modified by various
peptides, to arginine residues that cannot be modified resulted
in normal p53 responses in mice with these mutant proteins.
These types of experiments indicate that the protein
modifications of the p53 protein after a stress input are not
essential for the activation of the p53 protein (or are heavily
backed up by other functions not eliminated in these
experiments). This brings up the question; what are the
functions, if any, of the protein modifications in p53 or the
MDM-2 protein after exposure to a stress? Most people
believe there is a functional role for these modifications. First
of all the extent and nature of these protein alterations of the
p53 protein differs with different types of inputs. Thus, these
protein modifications form a chemical code that could inform
the p53 protein and the cell about the type of stress that is
occurring. Second the p53-regulated transcriptional output
after different stresses is different, and these protein
modifications could well dictate which p53-responsive genes
are transcribed by the cell based upon the specific protein
modifications of the p53 protein. Do p53 protein modifications
determine the genes which are transcriptionally regulated by
that p53 protein? These ideas need to be tested. P53 protein
interactions with other cellular proteins (which may be initiated
by protein modifications) impact upon gene selection and
the nature of the downstream events.
27,28
Just what protein
modifications of p53 and MDM-2 accomplish in a cell remains
a big question? In particular, the protein modifications and
protein–protein interactions (with p19 ARF, cyclin G, etc.) of
MDM-2 remain to be explored in more detail.
One of the most interesting methods of activating p53
results from the mutational inactivation of a tumor suppressor
gene like the retinoblastoma protein (which activates the
E2F-1 transcription factor) or the APC tumor suppressor
(which activates the beta catinin-TCF4 transcription factor) or
the mutational activation of oncogenes like myc or Ras. The
activities of the transcription factors that result from mutations
in these genes can transcribe the ARF gene and the ARF
protein then binds to the MDM-2 protein and inhibits its activity
as a ubiquitin ligase. This raises the p53 levels in a cell and
helps to bring about a p53 response or output that protects the
organism from the development of cancers.
29,30
This is an
excellent example of how diverse signal transduction path-
ways in a cell communicate and create feedback loops that
contribute to cellular homeostasis. Just who the other players
or modifiers of this circuit are remains to be explored? There
are some suggestions that ARF has additional functions and
may be regulated in several ways, and these questions
remain a high priority for the field.
The core control of P53
Once activated the p53 protein transcribes a number of
genes. Among these is the MDM-2 gene (and the COP-1 and
PIRH-2 genes) which is the major negative regulator of p53 in
the cell.
12
The transcriptional regulation of MDM-2 by p53 is a
slow step (hours) but once the MDM-2 protein is produced it
binds to p53 (inhibiting its activity as a transcription factor) and
ubiquitinates it enhancing its proteolytic breakdown. This
occurs as a rapid step. This auto-regulatory loop produces an
MDM-2-p53 oscillator composed of changing protein levels of
p53 and MDM-2, out of phase with one another, in the cell.
31,32
When p53 levels rise this increases MDM-2 levels which in
turn lower p53 levels, resulting in less MDM-2, etc. These
oscillations have been observed in vivo to be variable from
cell to cell but they can last a long time and the number of
oscillations is roughly proportional to the input dose of
radiation.
32
Does transcriptional activation of the downstream
genes depend upon this oscillator or does it optimize the
output of the pathway? Just what the functions of such
oscillations are, if any, and the factors that can modify these
oscillations remain to be determined? Do COP-1 and PIRH-2
participate in this process? The reason that the field has
focused upon MDM-2 as the major regulator of p53 is that it
was found first
33,34
and the gene knockout of MDM-2 in mice is
lethal early in development, just after implantation.
35
This
could be due to an hypoxia response of the p53 protein in the
absence of MDM-2, so that p53 activity is uncontrolled. This
idea needs to be tested. The double knockout mouse, with no
MDM-2 or p53 genes, is viable proving that MDM-2 acts to
block p53-mediated cell death early in development (is this
due to hypoxia?). This also demonstrates that PIRH-2 and
COP-1 do not back up MDM-2, at least early in development.
Now similar experiments should be done with COP-1 and
PIRH-2 knockouts.
The gene products that regulate MDM-2 (and p53) levels
and activities will need to be better characterized. The MDM-X
(or MDM-4) gene was identified because it is related to the
MDM-2 gene in its DNA and protein sequence. A knockout of
the MDM-X gene in mice is lethal and a MDM-X and p53
double knockout is viable.
36
Thus, MDM-X is as important as
MDM-2 in regulating p53 in vivo. It now appears that MDM-X
can negatively regulate p53 directly and positively regulates
MDM-2. After a stress signal, the poly-ubiquitination of MDM-
2 results in the degradation of MDM-X and MDM-2, increasing
p53 levels and activity. Just how this is accomplished (via
protein modification of MDM-X or 2, or protein–protein
interaction) remains to be studied. The activities that modify
MDM-X and MDM-2 (cyclin G-PP2A, ARF and its activators
and HAUSP) act only on MDM-2 and remain to be explored
in more detail. High levels of p53 appear to repress the
transcription of the ARF gene and the p53 knockout mouse
The P53 pathway
AJ Levine et al
1030
Cell Death and Differentiation
(no p53 protein) has much higher levels of ARF mRNA and
protein than a wild-type mouse. How the p53 transcription
factor acts to repress transcription remains a mystery and is in
need of study.
Several studies have suggested additional levels of the
control of the p53 protein, at the transcriptional and at
the translational level, which should now form the basis for
future research. In addition, the p53 and MDM2 proteins can
shuttle between the nucleus and the cytoplasm which
undoubtedly changes their activity and regulation.
37,38
Just
what factors control this cellular localization and therefore can
impact upon p53 function remain to be explored. The AKT-1
kinase can phosphorylate the MDM-2 protein increasing its
activity and sending it into the nucleus of the cell.
39,40
Cyclin
G, a p53-regulated gene product, combines with the catalytic
subunit of the PP2A phosphatase to remove a phosphate
residue from the MDM-2 protein and increase its activity.
41
These two modifiers of the MDM-2 protein form feedback
loops that connect the core p53 activity to other signal
transduction pathways as well as regulate the core functions
of the network. Indeed there are at least 10 negative or
positive feedback loops that start with a p53-regulated gene
product and result in increasing or decreasing the activity of
p53.
12
These feedback loops connect the core p53 activities
to other signal transduction pathways. When a positive
feedback loop is activated it results in apoptosis and cell
death, a negative feedback loop attenuates the p53 pathway.
Do these loops act in all cell types? How are they regulated or
triggered? What is the impact of the activation of another
signal transduction pathway upon the core p53 regulation?
A good example of this is the starvation of a cell for glucose.
This engages the mTOR pathway. The absence of glucose
activates the LKB kinase and the AMP kinase. The AMP
kinase phosphorylates the TSC-2 protein in a TSC-1/2
complex. TSC-1/2 is a GAP that inactivates a G-protein,
RHEB, and RHEB activity controls the kinase activity of
mTOR. Thus, glucose starvation inactivates mTOR which
leads to the activation of autophagy, a catabolic process
that breaks down proteins, fats and carbohydrates into their
momomeric components, supplying endogenous nutrients to
the starved cell. While this proceeds the AMP-kinase also
phosphorylates serine-15 on the p53 protein.
13,14
In normal
cells this does not activate the p53 protein but in transformed
cells this is a proapoptotic event. P53 serine-15 phosphory-
lation is a precursor for other phosphorylation events for the
p53 protein.
22
When glucose becomes available and mTOR
reactivates, mTOR activates a subunit of PP2A which can
then remove the phosphate residue from p53 serine-15.
42
This restores p53 to its original state and attenuates the
feedback loop.
As there are two distinct transcriptional start sites for the
p53 mRNA and several alternative splicing patterns of this
mRNA there are nine different isoforms of the p53 protein that
have been detected. The amino-terminus of the p53 protein
has two distinct transcriptional activator regions of the protein
and one or both can be present. The carboxy-terminus of the
p53 protein has a basic domain which can be spliced out or
replaced by another amino-acid sequence. All combinations
of these alternative protein isoforms have been observed
43
but many questions remain. Are these forms found preferen-
tially in different cell or tissue types or at different stages of
development? What are the functions of each alternative
domain and the resultant protein? Does a protein with its
DNA-binding domain intact but without a transactivation
domain act as a repressor? How do the three alternative
forms of the carboxy-terminus alter the functions of the p53
protein? Recently, one of these isoforms has been shown to
have a distinct and critical function in the development of the
gut of zebra fish.
44
There is now clearly more to learn.
The downstream events in the p53 pathway
Once p53 is activated in response to a stress signal it
gains the ability (and increased concentration) to bind to
p53-responsive DNA sequence elements in the genome.
The consensus DNA sequence for p53 binding is
RRRCWWGYYY, where R is a purine, W is A or T and Y is
a pyrimidine. A p53-responsive element is composed of two
of these 10 base paired sequences, separated by a spacer of
0–21 base pairs, and the sequences are often located 5
0
to the
gene or in the first or second intron of the gene regulated by
p53.
45
It is a challenge to identify all of the p53-responsive
sequences in a genome because degenerate sequences
also function in a p53-dependent fashion. An algorithm has
been described
18
that has successfully identified novel p53-
responsive genes.
13,46
It has become clear that different types
of stress signals as inputs result in different genes being
transcribed under p53 control.
47
In addition stress signals
received by different cell or tissue types also produce different
transcriptional programs regulated by the p53 protein. We
do not understand what mediates these different regulatory
responses. Both p53 protein modifications and protein–
protein interactions could play a role as well as differences
in the DNA sequences of response elements. In addition,
there are large differences in both the amounts of mRNA and
proteins produced by different p53-regulated genes and in the
kinetics of synthesis after a p53-activation event. Why this is
so remains unclear. There appear to be some p53-regulated
genes that are transcribed in response to many different types
of stress signals and in all tissues responding to the stress
(p21, cyclin G, MDM-2, GADD-45) and others that are either
stress- or tissue specific (PTEN, TSC-2). What regulates
these differences remains unclear? The functions of the p53-
response genes fall into several categories. A set of genes are
clearly involved in cell cycle arrest (p21, 14-3-3 sigma, GADD-
45). A second set of p53-regulated genes are involved in
apoptosis. These can be divided into the intrinsic and extrinsic
apoptotic pathways. In the extrinsic pathway p53 regulates
Fas production (a secreted protein), as well as killer/DR5, the
trail receptor and a membrane protein. These proteins, along
with PIDD, activate caspase 8 and Bid to release cytochrome
c which acts with APAF-1 (a p53-regulated gene) to activate
caspase 9 and then 3 resulting in apoptosis. The intrinsic
pathway is populated with many p53-regulated genes of which
bax, noxa and puma may work in different cell types to
promote cytochrome c release. Several other p53-regulated
genes have been implicated in the enhancement of apoptosis
(Perp, scotin, some PIGS, p53 AIP) but their mechanism of
action remain to be elucidated. Elucidating the details of how
p53 initiates apoptosis employing its transcriptional program
The P53 pathway
AJ Levine et al
1031
Cell Death and Differentiation
is a high priority for the field. More recently several groups
48,49
have provided good evidence that the p53 protein itself can
move out of the nucleus and act upon the mitochondria or
proteins in the mitochondria (BCl-2, BCL-XL) to promote
cytochome c release and apoptosis. A large challenge
remains to prove these mechanisms are functional in an
animal and augment or replace, under some circumstances,
the transcriptional-mediated apoptosis. This is presently a
confusing and challenging area of research and needs to be
resolved with a consensus being formed in the field. The third
output of the p53 response is senescence of cells. There are
some suggestions that senescence is as important as
apoptosis in mediating the tumor suppressor functions of
p53. In spite of this we have no idea which p53-regulated
genes contribute to senescence and what gene products
populate the pathway to cellular senescence. As normal cells
divide and age in culture and telomeres shorten, p53 levels
rise, but what transcriptional program this triggers remains
to be elucidated? Mice with an overactive p53 protein, where
the threshold for stress responses is lowered, die prematurely
and have compromised stem cell capabilities (senescence of
the stem cells).
50
These mice are more resistant to developing
cancers initiated with carcinogens. These mice also die at
younger ages than wild-type mice with a normal p53 protein.
We need to design new experimental approaches to explore
these questions of cancer resistance and longevity in more
detail. These experiments also introduce the question; what
are the functions, if any, of the p53 protein in regulating
longevity in animals and cells? The next few years will see
these questions addressed in worms, flies, mice and even
humans.
While cell cycle arrest, apoptosis and senescence are
traditionally thought of as the major outputs of the p53
pathway several other p53-responsive genes are beginning
to define additional functions of the p53 pathway. The p48
protein and p53R2 subunit of ribonucleotide reductase are
p53-responsive genes that aid in DNA repair. The sestrins are
a set of p53-regulated genes that counter the presence of
reactive oxygen species in the cell.
51
Clearly some p53
pathway functions help to protect the cell from exogenous or
endogenous stresses while others enhance the cellular repair
process. A second p53-regulated function is to communicate
with cells in its environment that a cell has DNA damage or
senses a stress signal. A number of p53-responsive or p53-
regulated genes produce secreted proteins. These proteins
fall into several functional categories. The IGF-BP3 binds the
IGF-1 hormone and prevents it from activating a growth
response signal transduction pathway (IGF-1, IGF-1R, PI3 K,
PDK, AKT-1, forkhead transcription factors).
12,52
In this case
p53 is negatively regulating cell growth, mitogen signaling and
preventing cell division in surrounding cells after a stress
signal. The p53-regulated and -secreted genes PAI and
maspin are protease inhibitors that result in alterations to the
extracellular matrix and cell surface.
53,54
Similarly the p53-
regulated thrombospondin gene produces a secreted protein
that alters the cellular matrix and is anti-angiogenic.
55
Secreted proteins permit communication between cells. More
recently the p53-regulated TSAP-6 gene was shown to
enhance the rate of exosome production from cells under-
going a p53 response to stress.
56
Exosomes also can
communicate with other cells both in the immune system
and cells in its own vicinity. Cells in stress will want to
coordinate their responses with the surrounding cell layer
in vivo and this is a new field opening up to future research
efforts. Finally, a large number of p53-responsive genes (p21,
WIP-1, SIAH-1, PTEN, TSC-2, IGF-BP-3, cyclin G, p73delta
N, MDM-2, COP-1 and PIRH-2) initiate positive or negative
feedback loops with the p53 protein and the core gene
products of the pathway.
12
This results in the integration of a
stress signal with other pathways in the cell that regulate cell
growth, division or cell death. These p53-regulated gene
products either reinforce or inhibit the p53 response and
network. This then results in a coordination of cellular
processes that need to be explored in future experiments.
The cellular outputs of the downstream events
This brief review has discussed at least six different outputs
of the p53 pathway which fall into two distinct categories,
primary response and secondary responses. The three
primary responses to a stress input signal by the p53 pathway
are; either cell cycle arrest, cellular senescence or apoptosis.
The cell cycle arrest may be reversible or in other cases
irreversible (which might be related to senescence). At this
time we do not know all of the gene products (p53 regulated or
not) that bring about cell cycle arrest. While p21 clearly plays
some role in a G-1 arrest, it is not the entire story (based upon
knockout experiments).
57,58
There is some controversy about
whether p53 mediates a G-2 arrest, when it does so and how?
There are several examples of a role for p53 in blocking the
reinitiation of a second S-phase in cells that can not enter
cytokinesis because of treatments with spindle poisons.
59
This is one of the ways in which p53 prevents karyotypic
instability in cells, which is a major function or output of the p53
pathway. Whether the p53 pathway has a method to ‘count’
the number of chromosomes that segregate into a daughter
cell, and if this number is not correct it kills the cell, or it
monitors segregation in another fashion remains an important
line of enquiry? There is good evidence that the p53 protein
and its functions regulate or monitor the number of centro-
somes produced at each cell cycle. The p53 knockout mouse
produces cells with abnormal numbers of centrosomes.
60
The way in which the p53 pathway brings about cellular
senescence is completely unknown. Just which p53-regulated
genes initiate this state remain unclear. Indeed, there are very
few reliable biomarkers for the senescent state and we do
not know what mediates this process of irreversible division
leaving the cell in a metabolically active state? There are good
reasons to suspect that senescence occurs in vivo as well
as in cell culture (cells from older animals undergo fewer
divisions in culture) and plays a role in cancer responses.
As such this is clearly an important line of research for the
future. It appears that the majority of the p53-regulated genes
play a role in promoting apoptosis. This pathway seems to
contain examples of redundant functions so experiments
where genes are knocked out are less informative. It also
appears that tissue specific p53-regulated gene products
play a role here. The transcriptional program that initiates
apoptosis triggers both the intrinsic and extrinsic apoptotic
pathways. The proposed p53 transcription-independent
The P53 pathway
AJ Levine et al
1032
Cell Death and Differentiation
pathway to apoptosis needs additional evidence and several
lines of proof that it is employed in vivo, and not only in cell
culture under abnormal circumstances. It is an important
potential mechanism and proof of its validity should be held to a
high standard. An important question that remains in examining
the output of the p53 pathway is how a cell which has received a
stress signal chooses which of these three outputs (apoptosis,
cell cycle arrest or cellular senescence) to implement? We do
know that normal fibroblasts taken from an animal and
irradiated undergo G-1 arrest while their transformed counter-
parts undergo apoptosis. Normal irradiated T cells undergo
apoptosis, so both the state of cellular differentiation and the
input of other signal transduction pathways influence this
decision. In other experimental studies, hormones or other
mediators which turn on signal transduction pathways altered
the p53 output response. Now we need to understand the rules
which regulate the p53-mediated output.
The secondary outputs of the p53 pathway come from p53-
regulated gene products that either (1) prevent DNA damage
(sestrins) or aid in DNA repair, (2) mediate communication
between the cell under stress and its neighbors, the
extracellular matrix shared by these cells or even other more
distant cells in the body, and (3) create intracellular or
extracellular p53 feedback loops that modulate p53 activity
and the pathway and simultaneously communicate with other
networks in the cell.
12
Prevention of DNA damage by
removing free radicals or other chemical insults needs to be
explored in more detail. This concept brings up the question
of whether the p53 pathway is solely inductive, responding to
stress and then preventing more damage, or is also active
before a stress signal and is therefore truly preventing the
initiation of DNA damage? The p53 functions that aid in DNA
repair suggest that the cell cycle arrest that is preventing
errors is reversible and the cell lives to divide again. The
nature of the communication between a cell undergoing a p53
response (apoptosis, senescence or cell cycle arrest) and its
neighbors is a poorly explored area of research. The soluble
secreted p53-responsive proteins can change the extracel-
lular matrix (maspin, PAI-1), block hormonal signaling (IGF-
BP3) to another pathway, and alter angiogenesis in the vicinity
of the alarmed cells (thrombospondin). In addition p53
activation can enhance the rate of exosome production by
the damaged cell by inducing the p53-regulated TSAP-6
gene.
56,61
Exosomes can communicate with adjacent cells
by cell fusion and with T cells by presenting antigens to the
immune system. Could p53-mediated apoptosis enhance
immunization of the host in this fashion? The activation of p53
(and its inactivation) was first detected in virus-infected
cells.
1,2
Could the p53 pathway be playing an important role
in infectious diseases and immunity? Is it possible that a
cancer cell undergoing a p53 response is also immunizing its
host to cancer antigens and preventing or slowing the disease
in this fashion? If this was the case then cancer cells with
mutant p53 would fail to immunize their host and selection for
a mutant p53 could be due to an immunoselective process.
These are provocative ideas that will need to be tested in the
future. The negative- and positive-feedback loops that are
initiated by p53-regulated genes and then act upon p53
activity and functions have been reviewed elsewhere
12
and in
this chapter. They serve two functions; (1) to modulate up or
down p53 activity; and (2) to communicate with other signal
transduction pathways in the cell. For example, after a
commitment to apoptosis, a PTEN-mediated feedback loop
modulates up p53 activity (a positive feedback) and shuts
down the IGF-1 and mTOR pathways (growth and cell division
pathways). This activates other processes such as autophagy
which provides nutrients for adjacent cells and exosome
production. Both autophagy and exosome production are
functions of the lysosomal compartment and pathway. Could
this indicate some additional fundamental roles of this cellular
compartment in the p53 pathway and its control of cellular
processes?
Questions about the p53 pathway in a larger
context
Several observations have demonstrated that the p53 gene
and its protein play a central role in the origins of cancers: (1)
the role of the p53 protein and its gene in viral and cellular
transformation, (2) the cancers in the p53 gene knockout
mice, (3) cancers observed with the Li-Fraumeni families and
(4) the high incidence of p53 gene mutations in many human
cancers. However, just what does p53 do to prevent cancers
from arising in murine and human populations? The presence
of hundreds of genes in the p53 pathway and genetic
polymorphisms in those genes suggests that individuals
may have more or less efficient p53 pathways. The p53 gene
itself has a codon 72 polymorphism (proline–arginine change)
and the arginine allele appears to be more efficient in
apoptosis in cell culture.
62
The observation that the ratio of
proline to arginine alleles in individuals changes as the latitude
changes from the equator to the north pole already suggests
that selection is playing a role upon these alleles.
63
There are
several studies that suggest that one or the other allele could
influence the occurrence or progression of cancer.
64,65
At
present however the results are confusing and a better
understanding of the role of these two alleles in the pathway
will be important. Recently, a polymorphism in the promoter of
the human MDM-2 gene has been identified (SNP309).
66
The
minor allele enhances the levels of the MDM-2 protein in cells,
lowers the levels of p53 after a stress response and lowers the
frequency of apoptosis in these cells. This same allele lowers
the age of onset of the development of several types of
cancer.
66
More recently, a separate study demonstrated that
the minor allele of SNP309 increased the frequency of
esophageal cancer in individuals and that the codon 72
polymorphism further increased the odds ratio of developing
this cancer (in an additive fashion) and that smoking further
increased this odds ratio (in a more than additive fashion).
67
Clearly this is one way to relate the participation of the p53
pathway to the development of cancers, the progression
of cancers and the treatment of cancers. There will be more
p53 pathway polymorphisms, more environmental variables
tested and more studies that will uncover the functions of the
p53 network in humans.
Several lines of evidence suggest that the p53 pathway
contains a sexual dimorphism that distinguishes gender
responses. The p53 gene knockout mice have a sex ratio
distortion, the severity of which depends upon the genetic
The P53 pathway
AJ Levine et al
1033
Cell Death and Differentiation
background of the inbred strain.
68–70
This is due to the fact
that many female mice are born with an exencephalic
condition, where the brain case never closes and the brain
is outside of the head.
69
This is seen only in females. Whether
the cranium fails to close or be developed or the brain is too
large (failure of neuronal apoptosis in development?) remains
unclear and will be important to explore. The genetic back-
ground determines the penetrance of this phenotype so some
female mice are born. Female heterozygous p53 knockout
mice develop more osteogenic sarcomas than do their
male counterparts indicating that sexual dimorphism is
operative.
71,72
In humans with Li-Fraumeni Syndrome (also
heterozygotes) the females develop cancers at a higher
frequency and earlier in life
73
and this is the case even if breast
and prostate cancers are not considered. The MDM-2 gene,
can under certain circumstances be regulated by the estrogen
receptor.
74
The IGF-1 pathway (which interfaces with the
p53 pathway via the activation of MDM-2 by AKT-1) is also
influenced by sexual hormones and sensory inputs.
39,40
The
SIR-2 or SIRT-1 gene product, an NAD-dependent histone
deacetylase, acts to remove acetyl groups from both the
forkhead and the p53 proteins and can modulate the activity
of the IGF-1 pathway.
75,76
Certain types of mutations in the
IGF-1 pathway and SIR2/SIRT-1 play a central role in
extending the longevity of an organism (yeast, worms, flies
and mice).
77,78
Will the p53 pathway play a role in determining
longevity in worms, flies, mice and humans? Longevity is also
sexually dimorphic in some of these species. In mice (2–3
years life span) and dogs (8–16 years life span) and humans
(75 years life span) most cancers arise in the last quarter
of their life spans rather than a specific number of years
after birth. In fact the frequency of cancer in these species
increases exponentially (over 1- to 10 000-fold) during the last
quarter of their life span. Thus evolutionary selection has fixed
in each of these species the age of sexual maturity, the age of
producing offspring, the age of developing the majority of
cancers in the population, and in relation to that, the longevity
of the species (there would be no species if you died before
reproducing). The longevity of a species may be set in part by
alterations in the IGF-1 pathway and the age of onset of
cancers by tumor suppressor genes like p53 and their
pathways. The reproductive age is fixed by a related and
interactive set of genes and their networks. Future experi-
ments will explore these variables and their relationships.
In this evolutionary context, how did a gene function like p53
arise and what does (did) it do for the animal? Worms and flies
are born with a body plan that is largely (except for the germ
line) postmitotic. No more divisions occur in an adult animal.
In these cases the major job of p53 is the surveillance of
the germ line for environmental insults that would result in
poor offspring. The body plan of adult vertebrates, however,
contains many tissues that continue to divide throughout life
and renew themselves. Here p53 has been employed in the
surveillance of somatic cells to prevent cancers from arising in
the organism. The exploration of the functions of the p53
pathway throughout evolution and how it has adapted to novel
situations and organisms will be a profitable avenue of
research.
We now know enough about the p53 pathway to move in
some new directions. One of the input signals that activates
p53 is nitric oxide which is produced and employed for
signaling after inflammatory responses. Indeed inflammatory
syndromes have been shown to cause cells in the vicinity of
lymphocyte invasions to undergo a p53 response.
79,80
Communication may occur in the other direction with p53-
regulated exosomes signaling to T cells and immunizing the
organism with parts of damaged cells. These ideas suggest
the exploration of whether p53 plays a role in the recovery
from infectious diseases, inflammation or the initiation of
autoimmunity or the establishment of tolerance? The apopto-
tic functions of the p53 pathway may normally be employed by
the immune system for optimal function. These same
apoptotic pathways could play a role in neurodegenerative
diseases. There is a growing body of evidence for a p53-
regulated program cell death in neurons, glial cells or
Schwann cells. What role does the p53 pathway and its
response to environmental insults play in the origins or
propagation of neurodegenerative diseases? With an increas-
ing age of the host does the p53 pathway become more active
(is there a pathological condition that makes it more active?)
or does it decay in its efficiency? Could it be the case that the
large and rapid increase in cancers in a population during the
last quarter of our life span is due to accumulated mutations
in oncogenes and tumor suppressor genes in a cell and a
declining efficiency of the p53 response which increases the
rate of these fixed mutations in that cell? These are more
complicated questions to answer and more difficult experi-
ments to perform but the answers will open up new avenues of
research and understanding.
One thing we can be sure of, some of the answers to the
questions posed in this chapter will be answered. Other
questions will be rephrased or changed completely before it is
productive to test these ideas. However, all of these questions
will be discussed, probed and eventually explored. We have
uncovered and explored a process central to life, how a cell
responds to stress or perturbations in its environment.
Understanding these homeostatic mechanisms central to all
life processes ensures a productive future of questioning.
References
1. Lane DP and Crawford LV (1979) T antigen is bound to a host protein in SV40-
transformed cells. Nature 278: 261–263.
2. Linzer DI and Levine AJ (1979) Characterization of a 54 K dalton cellular SV40
tumor antigen present in SV40-transformed cells and uninfected embryonal
carcinoma cells. Cell 17: 43–52.
3. Kress M, May E, Cassingena R and May P (1979) Simian virus 40-transformed
cells express new species of proteins precipitable by anti-simian virus 40 tumor
serum. J. Virol. 31: 472–483.
4. DeLeo AB, Jay G, Appella E, Dubois GC, Law LW and Old LJ (1979) Detection
of a transformation-related antigen in chemically induced sarcomas and other
transformed cells of the mouse. Proc. Natl. Acad. Sci. USA 76: 2420–2424.
5. Levine AJ, Finlay CA and Hinds PW (2004) P53 is a tumor suppressor gene.
Cell 116 (2 Suppl): S67–S69 1 p following S69.
6. Robins H, Alexe G, Harris SL and Levine AJ (2005) The first twenty-five years
of p53 research In 25 Years of p53 Research Hainaut P, Wiman KG (eds)
Dordrecht: Springer pp. 1–26.
7. Vogelstein B, Lane D and Levine AJ (2000) Surfing the p53 network. Nature
408: 307–310.
8. Giaccia AJ and Kastan MB (1998) The complexity of p53 modulation: emerging
patterns from divergent signals. Genes Dev. 12: 2973–2983.
The P53 pathway
AJ Levine et al
1034
Cell Death and Differentiation
9. Gudkov AV and Komarova EA (2003) The role of p53 in determining sensitivity
to radiotherapy. Nat. Rev. Cancer 3: 117–129.
10. Oren M (2003) Decision making by p53: life, death and cancer. Cell Death
Differ. 10: 431–442.
11. Soussi T (2005) The p53 pathway and human cancer. Br. J. Surg. 92: 1331–1332.
12. Harris SL and Levine AJ (2005) The p53 pathway: positive and negative
feedback loops. Oncogene 24: 2899–2908.
13. Feng Z, Zhang H, Levine AJ and Jin S (2005) The coordinate regulation of the
p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. USA 102: 8204–8209.
14. Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, Birnbaum MJ and
Thompson CB (2005) AMP-activated protein kinase induces a p53-dependent
metabolic checkpoint. Mol. Cell 18: 283–293.
15. Yang A, Kaghad M, Caput D and McKeon F (2002) On the shoulders of giants:
p63, p73 and the rise of p53. Trends Genet. 18: 90–95.
16. McKeon F and Yang A (2005) p53, p63, and p73: internecine relations? In 25
Years of p53 Research Hainaut P, Wiman KG (eds) Dordrecht: Springer
pp. 209–222.
17. Monti O, Damalas A, Strano S and Blandino G (2005) p73, p63 and mutant
p53: members of protein complexs floating in cancer cells In 25 Years for p53
Research Hainaut P, Wiman KG (eds) Dordrecht: Springer pp. 223–232.
18. Hoh J, Jin S, Parrado T, Edington J, Levine AJ and Ott J (2002) The p53MH
algorithm and its application in detecting p53-responsive genes. Proc. Natl.
Acad. Sci. USA 99: 8467–8472.
19. Leng RP, Lin Y, Ma W, Wu H, Lemmers B, Chung S, Parant JM, Lozano G,
Hakem R and Benchimol S (2003) Pirh2, a p53-induced ubiquitin–protein
ligase, promotes p53 degradation. Cell 112: 779–791.
20. Dornan D, Wertz I, Shimizu H, Arnott D, Frantz GD, Dowd P, O’Rourke K,
Koeppen H and Dixit VM (2004) The ubiquitin ligase COP1 is a critical negative
regulator of p53. Nature 429: 86–92.
21. Stommel JM and Wahl GM (2004) Accelerated MDM2 auto-degradation
induced by DNA-damage kinases is required for p53 activation. EMBO J. 23:
1547–1556.
22. Appella E and Anderson CW (2001) Post-translational modifications and
activation of p53 by genotoxic stresses. Eur. J. Biochem. 268: 2764–2772.
23. Xirodimas DP, Saville MK, Bourdon JC, Hay RT and Lane DP (2004) Mdm2-
mediated NEDD8 conjugation of p53 inhibits its transcriptional activity. Cell 118:
83–97.
24. Brooks CL and Gu W (2003) Ubiquitination, phosphorylation and acetylation:
the molecular basis for p53 regulation. Curr. Opin. Cell Biol. 15: 164–171.
25. Xu Y (2003) Regulation of p53 responses by post-translational modifications.
Cell Death Differ. 10: 400–403.
26. Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, Kong N,
Kammlott U, Lukacs C, Klein C, Fotouhi N and Liu EA (2004) In vivo activation
of the p53 pathway by small-molecule antagonists of MDM2. Science 303:
844–848.
27. McKinney K, Mattia M, Gottifredi V and Prives C (2004) p53 linear diffusion
along DNA requires its C terminus. Mol. Cell 16: 413–424.
28. McKinney K and Prives C (2005) Regulation of p53 DNA binding In 25 Years of
p53 Research Hainaut P, Wiman KG (eds) Dordrecht: Springer pp. 27–52.
29. Lowe SW and Sherr CJ (2003) Tumor suppression by Ink4a-Arf: progress and
puzzles. Curr. Opin. Genet. Dev. 13: 77–83.
30. Hemann MT and Lowe S (2005) p53 links tumor development to cancer therapy
In 25 Years of p53 Research Hainaut P, Wiman KG (eds) Dordrecht: Springer
pp. 339–352.
31. Lev Bar-Or R, Maya R, Segel LA, Alon U, Levine AJ and Oren M (2000)
Generation of oscillations by the p53-Mdm2 feedback loop: a theoretical and
experimental study. Proc. Natl. Acad. Sci. USA 97: 11250–11255.
32. Lahav G, Rosenfeld N, Sigal A, Geva-Zatorsky N, Levine AJ, Elowitz MB and
Alon U (2004) Dynamics of the p53-Mdm2 feedback loop in individual cells. Nat.
Genet. 36: 147–150.
33. Momand J, Zambetti GP, Olson DC, George D and Levine AJ (1992) The
mdm-2 oncogene product forms a complex with the p53 protein and inhibits
p53-mediated transactivation. Cell 69: 1237–1245.
34. Oliner JD, Kinzler KW, Meltzer PS, George DL and Vogelstein B (1992)
Amplification of a gene encoding a p53-associated protein in human sarcomas.
Nature 358: 80–83.
35. Montes de Oca Luna R, Wagner DS and Lozano G (1995) Rescue of early
embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 378:
203–206.
36. Parant J, Chavez-Reyes A, Little NA, Yan W, Reinke V, Jochemsen AG and
Lozano G (2001) Rescue of embryonic lethality in Mdm4-null mice by loss of
Trp53 suggests a nonoverlapping pathway with MDM2 to regulate p53. Nat.
Genet. 29: 92–95.
37. Tao W and Levine AJ (1999) Nucleocytoplasmic shuttling of oncoprotein Hdm2
is required for Hdm2-mediated degradation of p53. Proc. Natl. Acad. Sci. USA
96: 3077–3080.
38. Boyd SD, Tsai KY and Jacks T (2000) An intact HDM2 RING-finger domain is
required for nuclear exclusion of p53. Nat. Cell Biol. 2: 563–568.
39. Mayo LD and Donner DB (2001) A phosphatidylinositol 3-kinase/Akt pathway
promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc. Natl.
Acad. Sci. USA 98: 11598–11603.
40. Zhou BP, Liao Y, Xia W, Zou Y, Spohn B and Hung MC (2001) HER-2/neu
induces p53 ubiquitination via Akt-mediated MDM2 phosphorylation. Nat. Cell
Biol. 3: 973–982.
41. Okamoto K, Li H, Jensen MR, Zhang T, Taya Y, Thorgeirsson SS and
Prives C (2002) Cyclin G recruits PP2A to dephosphorylate Mdm2. Mol. Cell 9:
761–771.
42. Levine AJ, Feng Z, Mak T, You H and Jin S (2006) Coordination and
communication between the p53 and IGF-1-AKT-TOR signal transduction
pathways. Genes Dev. 20: 267–275.
43. Bourdon JC, Fernandes K, Murray-Zmijewski F, Liu G, Diot A, Xirodimas DP,
Saville MK and Lane DP (2005) p53 isoforms can regulate p53 transcriptional
activity. Genes Dev. 19: 2122–2137.
44. Chen J, Ruan H, Ng SM, Gao C, Soo HM, Wu W, Zhang Z, Wen Z, Lane DP
and Peng J (2005) Loss of function of def selectively up-regulates
\{Delta\}113p53 expression to arrest expansion growth of digestive organs in
zebrafish. Genes Dev. 19: 2900–2911.
45. el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW and Vogelstein B (1992)
Definition of a consensus binding site for p53. Nat. Genet. 1: 45–49.
46. Feng Z, Jin S, Zupnick A, Hoh J, de Stanchina E, Lowe S, , Prives C and Levine
AJ (2005) p53 tumor suppressor protein regulates the levels of huntingtin gene
expression. Oncogene 24: 1–7.
47. Zhao R, Gish K, Murphy M, Yin Y, Notterman D, Hoffman WH, Tom E, Mack DH
and Levine AJ (2000) The transcriptional program following p53 activation.
Cold Spring Harb. Symp. Quant. Biol. 65: 475–482.
48. Moll UM, Wolff S, Speidel D and Deppert W (2005) Transcription-independent
pro-apoptotic functions of p53. Curr. Opin. Cell Biol. 17: 631–636.
49. Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD,
Schuler M and Green DR (2004) Direct activation of Bax by p53
mediates mitochondrial membrane permeabilization and apoptosis. Science
303: 1010–1014.
50. Donehower LA (2002) Does p53 affect organismal aging? J. Cell Physiol. 192:
23–33.
51. Budanov AV, Sablina AA, Feinstein E, Koonin EV and Chumakov PM (2004)
Regeneration of peroxiredoxins by p53-regulated sestrins, homologs of
bacterial AhpD. Science 304: 596–600.
52. Buckbinder L, Talbott R, Velasco-Miguel S, Takenaka I, Faha B, Seizinger BR
and Kley N (1995) Induction of the growth inhibitor IGF-binding protein 3 by
p53. Nature 377: 646–649.
53. Kunz C, Pebler S, Otte J and von der Ahe D (1995) Differential regulation of
plasminogen activator and inhibitor gene transcription by the tumor suppressor
p53. Nucl. Acids Res. 23: 3710–3717.
54. Zou Z, Gao C, Nagaich AK, Connell T, Saito S, Moul JW, Seth P, Appella E and
Srivastava S (2000) p53 regulates the expression of the tumor suppressor gene
maspin. J. Biol. Chem. 275: 6051–6054.
55. Dameron KM, Volpert OV, Tainsky MA and Bouck N (1994) Control of
angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science
265: 1582–1584.
56. Amzallag N, Passer BJ, Allanic D, Segura E, Thery C, Goud B, Amson R and
Telerman A (2004) TSAP6 facilitates the secretion of translationally controlled
tumor protein/histamine-releasing factor via a nonclassical pathway. J. Biol.
Chem. 279: 46104–46112.
57. Deng C, Zhang P, Harper JW, Elledge SJ and Leder P (1995) Mice lacking
p21CIP1/WAF1 undergo normal development, but are defective in G1
checkpoint control. Cell 82: 675–684.
58. Brugarolas J, Chandrasekaran C, Gordon JI, Beach D, Jacks T and Hannon GJ
(1995) Radiation-induced cell cycle arrest compromised by p21 deficiency.
Nature 377: 552–557.
The P53 pathway
AJ Levine et al
1035
Cell Death and Differentiation
59. Vaziri C, Saxena S, Jeon Y, Lee C, Murata K, Machida Y, Wagle N, Hwang DS
and Dutta A (2003) A p53-dependent checkpoint pathway prevents
rereplication. Mol. Cell 11: 997–1008.
60. Wang XJ, Greenhalgh DA, Jiang A, He D, Zhong L, Brinkley BR and Roop DR
(1998) Analysis of centrosome abnormalities and angiogenesis in epidermal-
targeted p53172 H mutant and p53-knockout mice after chemical
carcinogenesis: evidence for a gain of function. Mol. Carcinog. 23: 185–192.
61. Passer BJ, Nancy-Portebois V, Amzallag N, Prieur S, Cans C, Roborel de
Climens A, Fiucci G, Bouvard V, Tuynder M, Susini L, Morchoisne S, Crible V,
Lespagnol A, Dausset J, Oren M, Amson R and Telerman A (2003) The
p53-inducible TSAP6 gene product regulates apoptosis and the cell cycle
and interacts with Nix and the Myt1 kinase. Proc. Natl. Acad. Sci. USA 100:
2284–2289.
62. Dumont P, Leu JI, Della Pietra III AC, George DL and Murphy M (2003) The
codon 72 polymorphic variants of p53 have markedly different apoptotic
potential. Nat. Genet. 33: 357–365.
63. Khrunin AV, Tarskaia LA, Spitsyn VA, Lylova OI, Bebyakova NA, Mikulich AI
and Limborska SA (2005) p53 polymorphisms in Russia and Belarus:
correlation of the 2-1-1 haplotype frequency with longitude. Mol. Genet.
Genomics 272: 666–672.
64. Ohayon T, Gershoni-Baruch R, Papa MZ, Distelman Menachem T, Eisenberg
Barzilai S and Friedman E (2005) The R72P P53 mutation is associated with
familial breast cancer in Jewish women. Br. J. Cancer 92: 1144–1148.
65. van Heemst D, Mooijaart SP, Beekman M, Schreuder J, de Craen AJ, Brandt
BW, Slagboom PE and Westendorp RG (2005) Variation in the human TP53
gene affects old age survival and cancer mortality. Exp. Gerontol. 40: 11–15.
66. Bond GL, Hu W, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel
F, Taubert H, Wuerl P, Onel K, Yip L, Hwang SJ, Strong LC, Lozano G and
Levine AJ (2004) A single nucleotide polymorphism in the MDM2 promoter
attenuates the p53 tumor suppressor pathway and accelerates tumor formation
in humans. Cell 119: 591–602.
67. Hong Y, Miao X, Zhang X, Ding F, Luo A, Guo Y, Tan W, Liu Z and Lin D (2005)
The role of P53 and MDM2 polymorphisms in the risk of esophageal squamous
cell carcinoma. Cancer Res. 65: 9582–9587.
68. Rotter V, Schwartz D, Almon E, Goldfinger N, Kapon A, Meshorer A,
Donehower LA and Levine AJ (1993) Mice with reduced levels of p53 protein
exhibit the testicular giant-cell degenerative syndrome. Proc. Natl. Acad. Sci.
USA 90: 9075–9079.
69. Sah VP, Attardi LD, Mulligan GJ, Williams BO, Bronson RT and Jacks T
(1995) A subset of p53-deficient embryos exhibit exencephaly. Nat. Genet. 10:
175–180.
70. Armstrong JF, Kaufman MH, Harrison DJ and Clarke AR (1995) High-
frequency developmental abnormalities in p53-deficient mice. Curr. Biol. 5:
931–936.
71. Mitsumori K, Shimo T, Onodera H, Takagi H, Yasuhara K, Tamura T, Aoki Y,
Nagata O and Hirose M (2000) Modifying effects of ethinylestradiol but not
methoxychlor on N-ethyl-N-nitrosourea-induced uterine carcinogenesis in
heterozygous p53-deficient CBA mice. Toxicol. Sci. 58: 43–49.
72. Baatout S, Jacquet P, Michaux A, Buset J, Vankerkom J, Derradji H, Yan J, Von
Suchodoletz H, De Saint-Georges L, Desaintes C and Mergeay M (2002)
Developmental abnormalities induced by X-irradiation in p53 deficient mice.
In Vivo 16: 215–221.
73. Strong LC (2003) General keynote: hereditary cancer: lessons from
Li-Fraumeni syndrome. Gynecol. Oncol. 88 (1 Part 2): S4–S7; discussion
S11–S13.
74. Kinyamu HK and Archer TK (2003) Estrogen receptor-dependent proteasomal
degradation of the glucocorticoid receptor is coupled to an increase in mdm2
protein expression. Mol. Cell. Biol. 23: 5867–5881.
75. Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, Guarente L and Gu W
(2001) Negative control of p53 by Sir2alpha promotes cell survival under stress.
Cell 107: 137–148.
76. Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y,
McBurney M and Guarente L (2004) Mammalian SIRT1 represses forkhead
transcription factors. Cell 116: 551–563.
77. Kenyon C (2005) The plasticity of aging: insights from long-lived mutants. Cell
120: 449–460.
78. Guarente L and Picard F (2005) Calorie restriction – the SIR2 connection. Cell
120: 473–482.
79. Hofseth LJ, Saito S, Hussain SP, Espey MG, Miranda KM, Araki Y, Jhappan C,
Higashimoto Y, He P, Linke SP, Quezado MM, Zurer I, Rotter V, Wink DA,
Appella E and Harris CC (2003) Nitric oxide-induced cellular stress and p53
activation in chronic inflammation. Proc. Natl. Acad. Sci. USA 100: 143–148.
80. Valenti LM, Mathieu J, Chancerelle Y, De Sousa M, Levacher M, Dinh-Xuan AT
and Florentin I (2005) High levels of endogenous nitric oxide produced after
burn injury in rats arrest activated T lymphocytes in the first G1 phase of the cell
cycle and then induce their apoptosis. Exp. Cell Res. 306: 150–167.
The P53 pathway
AJ Levine et al
1036
Cell Death and Differentiation
... Such protective activity of Parkin may be associated with its regulatory action toward p53. Inhibition of p53-mediated apoptosis is a mechanism that causes the neuroprotective effect of Parkin [118]. ...
Article
Full-text available
Epidemiological evidence points to an inverse association between Parkinson’s disease (PD) and almost all cancers except melanoma, for which this association is positive. The results of multiple studies have demonstrated that patients with PD are at reduced risk for the majority of neoplasms. Several potential biological explanations exist for the inverse relationship between cancer and PD. Recent results identified several PD-associated proteins and factors mediating cancer development and cancer-associated factors affecting PD. Accumulating data point to the role of genetic traits, members of the synuclein family, neurotrophic factors, the ubiquitin–proteasome system, circulating melatonin, and transcription factors as mediators. Here, we present recent data about shared pathogenetic factors and mediators that might be involved in the association between these two diseases. We discuss how these factors, individually or in combination, may be involved in pathology, serve as links between PD and cancer, and affect the prevalence of these disorders. Identification of these factors and investigation of their mechanisms of action would lead to the discovery of new targets for the treatment of both diseases.
... It has previously been reported that Ercc1-deficient mice accumulate p53 in the brain and have a range of neurodegenerative changes 7,8 . Indeed, p53 has been implicated in the neurodegenerative changes associated with DNA damage, aging and to mediate neuronal degeneration 49,50,[54][55][56][57] . We therefore tested if the activation of p53 was responsible for the histophatological changes previously reported in Ercc1 −/− mice. ...
Article
Full-text available
DNA repair deficiency can lead to segmental phenotypes in humans and mice, in which certain tissues lose homeostasis while others remain seemingly unaffected. This may be due to different tissues facing varying levels of damage or having different reliance on specific DNA repair pathways. However, we find that the cellular response to DNA damage determines different tissue-specific outcomes. Here, we use a mouse model of the human XPF-ERCC1 progeroid syndrome (XFE) caused by loss of DNA repair. We find that p53, a central regulator of the cellular response to DNA damage, regulates tissue dysfunction in Ercc1-/- mice in different ways. We show that ablation of p53 rescues the loss of hematopoietic stem cells, and has no effect on kidney, germ cell or brain dysfunction, but exacerbates liver pathology and polyploidisation. Mechanistically, we find that p53 ablation led to the loss of cell-cycle regulation in the liver, with reduced p21 expression. Eventually, p16/Cdkn2a expression is induced, serving as a fail-safe brake to proliferation in the absence of the p53-p21 axis. Taken together, our data show that distinct and tissue-specific functions of p53, in response to DNA damage, play a crucial role in regulating tissue-specific phenotypes.
... In the absence of external pressure, the level of the wild-type p53 remains in a quiescent state [1]. When the cell is subjected to a certain external pressure, such as ionizing radiation, p53 will be stabilized and activated [2][3][4]. In the experimental study, we know that after receiving ionizing radiation, the concentration level of p53 in MCF-7 breast cancer cells is activated from the resting state to the oscillating state [5]. ...
Article
Full-text available
This paper focuses on the analysis of multiple types of time delays and fractional-order proportional-derivative (PD\(^\alpha \)) controller on the dynamics of fractional-order p53 model. The multiple types of time delay schemes are dexterously designed during the bifurcation control for the proposed system, and the comparative study is elaborately performed on bifurcation control theoretically. By analyzing the corresponding characteristic equation, some explicit conditions for the local asymptotical stability of the trivial equilibrium can be given for the fractional-order controlled system. Analysis reveals that the negative feedback delay restrains the delay threshold, while the positive feedback delay promotes the delay threshold and makes the system more robust. System dynamics is affected by fractional order, which is more conducive to the generation of system oscillations. We have adapted control system techniques and designed a PD\(^\alpha \) controller, which is used to activate p53 protein. Numerical results display that the addition of PD\(^\alpha \) controller can increase the richness of the dynamic system. It is worth noting that the differential gain parameter \(k_d\) of PD\(^\alpha \) controller has a significant impact on the system period, and the proportional gain parameter \(k_p\) can amplify the system amplitude. In addition, \(k_p\) can significantly advance the time threshold of system compared to \(k_d\). The PD controller is a good strategy to control the oscillation behaviors of the system.
... Positive staining for mutant p53 encoding protein using a specific antibody indicated abnormal expression (Fig. 3). Dysregulation or mutations in the p53 gene can lead to loss of its tumor suppressor function, contributing to tumor development and progression (15,16). ...
Article
Full-text available
Gastric-type endocervical adenocarcinoma (GEA) is an uncommon form of uterine cervical adenocarcinoma with an unfavorable prognosis. The tumor consists of glands exhibiting a morphological resemblance to gastric cells and occasionally manifests features akin to pancreaticobiliary mucinous adenocarcinoma. GEA differs from the typical cervical cancer, particularly in its lack of association with the human papillomavirus. Immunophenotypic analysis suggests intestinal differentiation. The present study reports two cases of GEA occurring in postmenopausal individuals who were diagnosed in Lishui Central Hospital (Lishui, China) between January 2015 and January 2023. Microscopic examination revealed cysts lined with mucinous cells within the tumors. Immunohistochemical assays confirmed the positivity of the tumors for cytokeratin 7, mucin (MUC)5AC, and mutant tumor protein p53, while the results were negative for tumor suppressor p16, and in one case for paired box protein 8, consistent with characteristics of mucinous adenocarcinoma originating from the gastrointestinal tract. Programmed death-ligand 1 expression was also negative. The proto-oncogene K-ras was identified using amplification refractory mutation system polymerase chain reaction. Both cases were negative for mutations in codons 12 and 13 of exon 2, codon 61 of exon 3 and codon 146 of exon 4, but were positive for wild-type K-ras. Clinical follow-up revealed a potential association between histopathological features and resistance to chemotherapeutic drugs. The infrequency of this tumor type may contribute to diagnostic challenges.
... Positive staining for mutant p53 encoding protein using a specific antibody indicated abnormal expression (Fig. 3). Dysregulation or mutations in the p53 gene can lead to loss of its tumor suppressor function, contributing to tumor development and progression (15,16). ...
Article
Full-text available
Gastric-type endocervical adenocarcinoma (GEA) is an uncommon form of uterine cervical adenocarcinoma with an unfavorable prognosis. The tumor consists of glands exhibiting a morphological resemblance to gastric cells and occasionally manifests features akin to pancreaticobiliary mucinous adenocarcinoma. GEA differs from the typical cervical cancer, particularly in its lack of association with the human papillomavirus. Immunophenotypic analysis suggests intestinal differentiation. The present study reports two cases of GEA occurring in postmenopausal individuals who were diagnosed in Lishui Central Hospital (Lishui, China) between January 2015 and January 2023. Microscopic examination revealed cysts lined with mucinous cells within the tumors. Immunohistochemical assays confirmed the positivity of the tumors for cytokeratin 7, mucin (MUC)5AC, and mutant tumor protein p53, while the results were negative for tumor suppressor p16, and in one case for paired box protein 8, consistent with characteristics of mucinous adenocarcinoma originating from the gastrointestinal tract. Programmed death-ligand 1 expression was also negative. The proto-oncogene K-ras was identified using amplification refractory mutation system polymerase chain reaction. Both cases were negative for mutations in codons 12 and 13 of exon 2, codon 61 of exon 3 and codon 146 of exon 4, but were positive for wild-type K-ras. Clinical follow-up revealed a potential association between histopathological features and resistance to chemotherapeutic drugs. The infrequency of this tumor type may contribute to diagnostic challenges.
... Since both the gain-and loss-of-its-function mutations have been observed in various tumors, the tumor suppressor gene, p53 has revealed a complex participation in the process of carcinogenesis. 90 The p53 gene is known to directly enhance the Bax gene transcription and hence, the production of Bax protein. 91 Cumulative studies have pointed to the ability of both Cr 86 and CuO NPs 87,88 treatments to produce a signicant increase in the p53 gene expression, promoting that of the Bax gene in a variety of tumors. ...
Article
Full-text available
In the current study, we biosynthesized copper oxide NPs (CuO NPs) utilizing the essential oils extracted from Boswellia carterii oleogum resin, which served as a bioreductant and capping agent with the help of microwave energy. Afterwards, the platinum(II) based anticancer drug, carboplatin (Cr), was loaded onto the CuO NPs, exploiting the electrostatic interactions forming Cr@CuO NPs. The produced biogenic NPs were then characterized using zeta potential (ZP), high-resolution transmission electron microscopy (HRTEM), X-ray diffraction spectroscopy (XRD), and Fourier transform infrared spectroscopy (FTIR) techniques. In addition, the entrapment efficiency and release profile of the loaded Cr were evaluated. Thereafter, SRB assay was performed, where Cr@CuO NPs demonstrated the highest cytotoxic activity against human colon cancer cells (HCT-116) with an IC 50 of 5.17 mg mL −1 , which was about 1.6 and 2.2 folds more than that of Cr and CuO NPs. Moreover, the greenly synthesized nanoparticles (Cr@CuO NPs) displayed a satisfactory selectivity index (SI = 6.82), which was far better than the free Cr treatment (SI = 2.23). Regarding the apoptosis assay, the advent of Cr@CuO NPs resulted in an immense increase in the cellular population percentage of HCT-116 cells undergoing both early (16.02%) and late apoptosis (35.66%), significantly surpassing free Cr and CuO NPs. A study of HCT-116 cell cycle kinetics revealed the powerful ability of Cr@CuO NPs to trap cells in the Sub-G1 and G2 phases and impede the G2/M transition. RT-qPCR was utilized for molecular investigations of the pro-apoptotic (Bax and p53) and antiapoptotic genes (Bcl-2). The novel Cr@CuO NPs treatment rose above single Cr or CuO NPs therapy in stimulating the p53-Bax mediated mitochondrial apoptosis. The cellular and molecular biology investigations presented substantial proof of the potentiated anticancer activity of Cr@CuO NPs and the extra benefits that could be obtained from their use.
... Inhibition of p53-mediated apoptosis is a mechanism that causes the neuroprotective effect of Parkin [100]. ...
Preprint
Full-text available
Epidemiological evidence points to an inverse association between Parkinson's disease (PD) and almost all cancers except melanoma, for which this association is positive. The results of many studies have found that patients with PD are at reduced risk of the majority of neoplasm occurrence and death. Several potential biological explanations exist for the inverse relationship between cancer and PD. Recent results identified several PD-associated proteins and factors mediating cancer development and cancer-associated factors affecting PD. Accumulating data points to the role of genetic traits, members of the synuclein family, neurotrophic factors, ubiquitin-proteasome system, circulating melatonin, and transcription factors as such mediators. We present recent data about shared pathogenetic factors and mediators that might be involved in the association between these two diseases. We also discuss how these factors, individually or in combination, may be involved in pathology, serve as links between PD and cancer, and affect the prevalence of these disorders. Identification of these factors and investigations of their mechanisms will lead to the discovery of new targets for the treatment of both diseases
Chapter
Despite more than 200 approved anticancer agents, cancer remains a leading cause of death worldwide due to disease complexity, tumour heterogeneity, drug toxicity, and the emergence of drug resistance. Accordingly, the development of chemotherapeutic agents with higher efficacy, a better safety profile, and the capability of bypassing drug resistance would be a cornerstone in cancer therapy. Natural products have played a pivotal role in the field of drug discovery, especially for the pharmacotherapy of cancer, infectious, and chronic diseases. Owing to their distinctive structures and multiple mechanistic activities, natural products and their derivatives have been utilized for decades in cancer treatment protocols. In this review, we delve into the potential of natural products as anticancer agents by targeting cancer’s hallmarks, including sustained proliferative signalling, evading growth suppression, resisting apoptosis and cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. We highlight the molecular mechanisms of some natural products, in vivo studies, and promising clinical trials. This review emphasizes the significance of natural products in fighting cancer and the need for further studies to uncover their fully therapeutic potential.
Article
Oncogenic intercellular signaling is regulated by extracellular vesicles (EVs), but the underlying mechanisms remain mostly unclear. Since TCTP (translationally controlled tumor protein) is an EV component, we investigated whether it has a role in genotoxic stress signaling and malignant transformation. By generating a Tctp -inducible knockout mouse model ( Tctp –/f– ) , we report that Tctp is required for genotoxic stress-induced apoptosis signaling via small EVs (sEVs). Human breast cancer cells knocked-down for TCTP show impaired spontaneous EV secretion, thereby reducing sEV-dependent malignant growth. Since Trp53 –/– mice are prone to tumor formation, we derived tumor cells from Trp53 –/– ; Tctp –/f– double mutant mice and describe a drastic decrease in tumori-genicity with concomitant decrease in sEV secretion and content. Remarkably, Trp53 –/– ; Tctp –/f– mice show highly prolonged survival. Treatment of Trp53 –/– mice with sertraline, which inhibits TCTP function, increases their survival. Mechanistically, TCTP binds DDX3, recruiting RNAs, including miRNAs, to sEVs. Our findings establish TCTP as an essential protagonist in the regulation of sEV-signaling in the context of apoptosis and tumorigenicity.
Preprint
Full-text available
Iron is an irreplaceable co-factor for metabolism. Iron deficiency affects >1 billion people, causing symptoms including anaemia and impaired immunity. Nevertheless, precisely how iron deprivation impacts immune cell function remains poorly characterised. We therefore interrogated how physiologically low iron availability affected activated CD8+ T cell metabolism and function, using multi-omic and metabolic labelling approaches. Iron limitation profoundly stalled proliferation without influencing cell viability, altered histone methylation status and disrupted mitochondrial membrane potential. Consistently, metabolism of glucose and glutamine in the TCA cycle was limited, indeed TCA cycle activity was partially reversed to a reductive trajectory. Previous studies have shown mitochondria-derived aspartate is crucial for proliferation of transformed cells. Surprisingly, we found aspartate was increased in stalled iron deficient CD8+ T-cells, but was not utilised cytosolically for nucleotide synthesis, likely due to trapping within depolarised mitochondria. Conversely, exogenous aspartate, which directly accesses the cytosol, markedly rescued the clonal expansion of even severely iron-deficient CD8+ T-cells. Overall, iron scarcity creates a mitochondrial-located metabolic bottleneck impairing T-cells, which can be bypassed by resupplying inhibited biochemical processes with aspartate. These findings reveal molecular consequences of iron deficiency for CD8+ T cell function, providing mechanistic insight into the basis for immune impairment during iron deficiency.
Article
Full-text available
The p53 tumor suppressor exerts anti-proliferative effects in response to various types of stress including DNA damage and abnormal proliferative signals. Tight regulation of p53 is essential for maintaining normal cell growth and this occurs primarily through posttranslational modifications of p53. Here, we describe Pirh2, a gene regulated by p53 that encodes a RING-H2 domain-containing protein with intrinsic ubiquitin-protein ligase activity. Pirh2 physically interacts with p53 and promotes ubiquitination of p53 independently of Mdm2. Expression of Pirh2 decreases the level of p53 protein and abrogation of endogenous Pirh2 expression increases the level of p53. Furthermore, Pirh2 represses p53 functions including p53-dependent transactivation and growth inhibition. We propose that Pirh2 is involved in the negative regulation of p53 function through physical interaction and ubiquitin-mediated proteolysis. Hence, Pirh2, like Mdm2, participates in an autoregulatory feedback loop that controls p53 function.
Article
We previously developed a transgenic mouse model that expresses in the epidermis a murine p53172R→H mutant (p53m) under the control of a human keratin-1–based vector (HK1.p53m). In contrast to mice with wild-type p53 and p53-knockout mice, HK1.p53m mice exhibit increased susceptibility to chemical carcinogenesis, with greatly accelerated benign papilloma formation, malignant conversion, and metastasis. In the study presented here, we examined the expression pattern of several differentiation markers and observed that p53m tumors exhibited a less differentiated phenotype than tumors elicited in non-transgenic mice. Metastasis in p53m tumors was also associated with a poorly differentiated phenotype. To determine whether genomic instability was associated with a putative gain-of-function role for this p53m, in situ examination of centrosomes was performed in HK1.p53m and equivalent p53-null papillomas. In contrast to HK1.p53m papillomas, which had centrosome abnormalities at high frequencies (75% of cells contained more than three centrosomes/cell), p53-null tumors exhibited few abnormal centrosomes (4% of cells contained more than three centrosomes/cell). To determine whether angiogenesis played a role in the rapid progression of p53m tumors, the expression of vascular endothelial growth factor, a promoter of angiogenesis, and thrombospondin-1, an inhibitor of angiogenesis, was examined in tumors derived from either p53m or p53-knockout mice. Regardless of their p53 status (wild type, p53m, p53–/–), all of the papillomas exhibited similar levels of vascular endothelial growth factor expression and decreased expression of thrombospondin-1 as did normal epidermis. In addition, tumors from different p53 genotypes showed a similar density of blood vessels. Because p53 status did not appear to play an overt role in angiogenesis, these data suggest that p53m accelerates tumorigenesis primarily by exerting a gain of function associated with genomic instability. Mol. Carcinog. 23:185–192, 1998. © 1998 Wiley-Liss, Inc.
Article
Cottus kanawhae, a member of the Cottus carolinae complex, is described from the New River System of Virginia and West Virginia.
Article
We previously developed a transgenic mouse model that expresses in the epidermis a murine p53172R→H mutant (p53m) under the control of a human keratin-1–based vector (HK1.p53m). In contrast to mice with wild-type p53 and p53-knockout mice, HK1.p53m mice exhibit increased susceptibility to chemical carcinogenesis, with greatly accelerated benign papilloma formation, malignant conversion, and metastasis. In the study presented here, we examined the expression pattern of several differentiation markers and observed that p53m tumors exhibited a less differentiated phenotype than tumors elicited in non-transgenic mice. Metastasis in p53m tumors was also associated with a poorly differentiated phenotype. To determine whether genomic instability was associated with a putative gain-of-function role for this p53m, in situ examination of centrosomes was performed in HK1.p53m and equivalent p53-null papillomas. In contrast to HK1.p53m papillomas, which had centrosome abnormalities at high frequencies (75% of cells contained more than three centrosomes/cell), p53-null tumors exhibited few abnormal centrosomes (4% of cells contained more than three centrosomes/cell). To determine whether angiogenesis played a role in the rapid progression of p53m tumors, the expression of vascular endothelial growth factor, a promoter of angiogenesis, and thrombospondin-1, an inhibitor of angiogenesis, was examined in tumors derived from either p53m or p53-knockout mice. Regardless of their p53 status (wild type, p53m, p53–/–), all of the papillomas exhibited similar levels of vascular endothelial growth factor expression and decreased expression of thrombospondin-1 as did normal epidermis. In addition, tumors from different p53 genotypes showed a similar density of blood vessels. Because p53 status did not appear to play an overt role in angiogenesis, these data suggest that p53m accelerates tumorigenesis primarily by exerting a gain of function associated with genomic instability. Mol. Carcinog. 23:185–192, 1998.
Article
The intracellular activity of the p53 tumor suppressor protein is regulated through a feedback loop involving its transcriptional target, mdm2. We present a simple mathematical model suggesting that, under certain circumstances, oscillations in p53 and Mdm2 protein levels can emerge in response to a stress signal. A delay in p53-dependent induction of Mdm2 is predicted to be required, albeit not sufficient, for this oscillatory behavior. In line with the predictions of the model, oscillations of both p53 and Mdm2 indeed occur on exposure of various cell types to ionizing radiation. Such oscillations may allow cells to repair their DNA without risking the irreversible consequences of continuous excessive p53 activation.
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.
Chapter
During the 1960s, the field of cancer research lacked clear direction. Several facts appeared to be well-established and correct, but the relationships among these observations were not apparent. Fifty years of research had demonstrated that viruses with both DNA and RNA genomes could cause cancer in animals. Over the next 45 years six new viruses were to be discovered that were able to initiate cancers in humans (Epstein-Barr Virus, Human T-Cell Leukemia Virus, Hepatitis B and C Viruses, Kaposi Sarcoma Virus and the Papilloma Viruses) (McKinnel et al., 1998). It was equally clear from the perspective of the 1960s that certain chemicals, when applied to animals, were able to initiate cancers (Yamagawa et al., 1918). Chemical carcinogenesis was a field both separate and distinct (both in the experiments one did and the experimentalists who did them) from viral carcinogenesis and very few scientists thought to find a common ground between concepts generated in each field. Thirdly, the study of mouse genetics demonstrated that some cancers were clearly inherited and these observations confirmed many prior publications that suggested a role for cancer causing genes in humans and other animals (DeOme, 1965). Finally epidemiologists, studying a variety of important variables that predispose humans to developing cancers, had made the very striking observation that the rates of cancer incidence increase exponentially with age and begin to rise dramatically by the fifth and sixth decade of life (Miller, 1991). While these four observations were all accepted facts the relationship between these concepts was not clear and researchers who studied viruses hardly ever discussed chemicals and those who thought about genes and viruses didn’t know what to make of aging as an important variable. Literally researchers from each of these fields, virology, chemical carcinogenesis, genetics and epidemiology never got together to discuss these issues.
Chapter
Approximately half of human tumors bear p53 mutations (Hollestein et al., 1997). The most prevalent type consists of missense mutations that are frequently accompanied by loss of the remaining wild-type p53 (wt-p53) allele (Hainaut et al., 1997; Levine, 1997). The major site of the p53 mutations is the highly conserved DNA binding core domain (Hussain et al., 1998; Prives et al., 1999). Thus, mutant p53 (mt-p53) proteins are unable to specifically bind DNA and to activate specific wt-p53 target genes. Unlike wt-p53, whose half-life is short, mutant p53 proteins are quite stable and abundantly present in cancer cells. One certain outcome of p53 mutations is the loss of wild type activities such as growth arrest, apoptosis, and differentiation (Michalovitz et al., 1990; Yonish-Rouach et al., 1991; Soddu et al., 1996; Almog et al., 1997). However, at variance with other tumor suppressor genes, cells with p53 mutations maintain expression of the fulllength protein. This may suggest that, at least certain mutant forms of p53 can gain additional functions through which actively contribute to cancer progression (Prives et al., 1999; Sigal et al., 2000; Strano et al., 2001; Bullock et al., 2001). Such evidence is provided by several in vitro and in vivo studies (Haley et al., 1990; Dittmer et al., 1993; Gualberto et al., 1998; Frazier et al., 1998; Li et al., 1998; Blandino et al., 1999; Aas et al., 1996; Irwin et al., 2003; Strano et al., 2003)