The intersection between DNA damage response and cell death pathways
Apoptosis is a finely regulated process that serves to determine the fate of cells in response to various stresses. One such stress is DNA damage, which not only can signal repair processes but is also intimately involved in regulating cell fate. In this review we examine the relationship between th...
Збережено в:
Дата: | 2012 |
---|---|
Автори: | , |
Формат: | Стаття |
Мова: | English |
Опубліковано: |
Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України
2012
|
Назва видання: | Experimental Oncology |
Теми: | |
Онлайн доступ: | http://dspace.nbuv.gov.ua/handle/123456789/139048 |
Теги: |
Додати тег
Немає тегів, Будьте першим, хто поставить тег для цього запису!
|
Назва журналу: | Digital Library of Periodicals of National Academy of Sciences of Ukraine |
Цитувати: | The intersection between DNA damage response and cell death pathways / S. Howsheen, E.S. Yang // Experimental Oncology. — 2012. — Т. 34, № 3. — С. 243-254. — Бібліогр.: 128 назв. — англ. |
Репозитарії
Digital Library of Periodicals of National Academy of Sciences of Ukraineid |
irk-123456789-139048 |
---|---|
record_format |
dspace |
spelling |
irk-123456789-1390482018-06-20T03:06:01Z The intersection between DNA damage response and cell death pathways Nowsheen, S. Yang, E.S. Reviews Apoptosis is a finely regulated process that serves to determine the fate of cells in response to various stresses. One such stress is DNA damage, which not only can signal repair processes but is also intimately involved in regulating cell fate. In this review we examine the relationship between the DNA damage/repair response in cell survival and apoptosis following insults to the DNA. Elucidating these pathways and the crosstalk between them is of great importance, as they eventually contribute to the etiology of human disease such as cancer and may play key roles in determining therapeutic response. This article is part of a Special Issue entitled “Apoptosis: Four Decades Later”. 2012 Article The intersection between DNA damage response and cell death pathways / S. Howsheen, E.S. Yang // Experimental Oncology. — 2012. — Т. 34, № 3. — С. 243-254. — Бібліогр.: 128 назв. — англ. 1812-9269 http://dspace.nbuv.gov.ua/handle/123456789/139048 en Experimental Oncology Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України |
institution |
Digital Library of Periodicals of National Academy of Sciences of Ukraine |
collection |
DSpace DC |
language |
English |
topic |
Reviews Reviews |
spellingShingle |
Reviews Reviews Nowsheen, S. Yang, E.S. The intersection between DNA damage response and cell death pathways Experimental Oncology |
description |
Apoptosis is a finely regulated process that serves to determine the fate of cells in response to various stresses. One such stress is DNA damage, which not only can signal repair processes but is also intimately involved in regulating cell fate. In this review we examine the relationship between the DNA damage/repair response in cell survival and apoptosis following insults to the DNA. Elucidating these pathways and the crosstalk between them is of great importance, as they eventually contribute to the etiology of human disease such as cancer and may play key roles in determining therapeutic response. This article is part of a Special Issue entitled “Apoptosis: Four Decades Later”. |
format |
Article |
author |
Nowsheen, S. Yang, E.S. |
author_facet |
Nowsheen, S. Yang, E.S. |
author_sort |
Nowsheen, S. |
title |
The intersection between DNA damage response and cell death pathways |
title_short |
The intersection between DNA damage response and cell death pathways |
title_full |
The intersection between DNA damage response and cell death pathways |
title_fullStr |
The intersection between DNA damage response and cell death pathways |
title_full_unstemmed |
The intersection between DNA damage response and cell death pathways |
title_sort |
intersection between dna damage response and cell death pathways |
publisher |
Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України |
publishDate |
2012 |
topic_facet |
Reviews |
url |
http://dspace.nbuv.gov.ua/handle/123456789/139048 |
citation_txt |
The intersection between DNA damage response and cell death pathways / S. Howsheen, E.S. Yang // Experimental Oncology. — 2012. — Т. 34, № 3. — С. 243-254. — Бібліогр.: 128 назв. — англ. |
series |
Experimental Oncology |
work_keys_str_mv |
AT nowsheens theintersectionbetweendnadamageresponseandcelldeathpathways AT yanges theintersectionbetweendnadamageresponseandcelldeathpathways AT nowsheens intersectionbetweendnadamageresponseandcelldeathpathways AT yanges intersectionbetweendnadamageresponseandcelldeathpathways |
first_indexed |
2025-07-10T07:31:31Z |
last_indexed |
2025-07-10T07:31:31Z |
_version_ |
1837244321429979136 |
fulltext |
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
THE INTERSECTION BETWEEN DNA DAMAGE RESPONSE
AND CELL DEATH PATHWAYS
S. Nowsheen1, E.S. Yang1,2,3,*
Departments of 1Radiation Oncology, 2Cell, Developmental, and Integrative Biology, and 3Pharmacology
and Toxicology,
Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Alabama, USA
Apoptosis is a finely regulated process that serves to determine the fate of cells in response to various stresses. One such stress
is DNA damage, which not only can signal repair processes but is also intimately involved in regulating cell fate. In this review
we examine the relationship between the DNA damage/repair response in cell survival and apoptosis following insults to the
DNA. Elucidating these pathways and the crosstalk between them is of great importance, as they eventually contribute to the etio
logy of human disease such as cancer and may play key roles in determining therapeutic response. This article is part of a Special
Issue entitled “Apoptosis: Four Decades Later”.
Key Words: poly(ADPribose) polymerase, EGFR, GSK3, BRCA1, apoptosis, p53, DNA damage and repair, balance between
cell survival and death.
Cell death is a fundamental cellular response that
has a pivotal role in development as well as maintain-
ing tissue homeostasis �y eliminating unwanted cells.
It is composed of �oth controlled and uncontrolled
mechanisms� including apoptosis� autophagy� and
necrosis. Apoptosis is a regulated cell death process
that reflects the cellular decision to die in response
to cues from the environment and is executed �y in-
trinsic cellular machinery [�� �]. In contrast� necrosis
is uncontrolled cell death �rought upon �y overwhelm-
ing stress. Lastly� autophagy is characterized �y self-
destruction starting with engulfment of cytoplasmic
material �y the phagophore and sequestration of ma-
terial to the autophagic vacuoles� where they are even-
tually destroyed [�]. The type and strength of stimuli�
tissue type� developmental stage of the tissue� and
the physiologic cellular microenvironment determines
which cell death process is undertaken [�].
The human �ody is continuously exposed to vari-
ous external and internal stresses� such as hypoxia�
toxins� oxidative stress� and many others [����]. The
a�ility of individual cells to adapt to these stresses
is crucial for their survival. Alternatively� if too much
damage has �een sustained� coordinated activation
of cell death processes must occur to rid the �ody
of cells that contain potential disease initiating muta-
tions. Thus� complex adaptation strategies such as cell
cycle checkpoints� DNA damage response pathways�
and programmed cell death have evolved to com�at
these environmental and physiological threats [�].
In this review� we will focus on one of these stresses�
DNA damage� as it relates to the cell death processes.
Ultimately� im�alance �etween DNA damage/repair
and activation/inactivation of these cell death pro-
cesses leads to carcinogenesis and may even alter
tumor response to therapy.
APOPTOSIS
Apoptosis is a vital process of programmed
cell death characterized �y distinct morphological
characteristics and energy-dependent �iochemical
mechanisms [�� �]. It is an integral component of vari-
ous homeostatic and defense processes including
normal cell turnover� aging� proper development
and functioning of the immune system� hormone
dependent atrophy� em�ryonic development� and
chemical-induced cell death [�]. Either too much
or too little apoptosis leads to various disease condi-
tions including autoimmune and neurodegenerative
disorders� ischemic damage� and cancer [�� 9���].
Thus� the a�ility to modulate the life and death of a cell
has immense therapeutic potential and has �een the
su�ject of intense research over the years.
Apoptosis ultimately leads to a series of coordi-
nated and energy-dependent activation of a group
of cysteine proteases — caspases [�� ����8]. This
Received: June 22, 2012.
*Correspondence: Fax: 205-975-0784
E-mail: eyang@uab.edu
Abbreviations used: 8-OHdG — 8-hydroxydeoxyguanosine; AP — apu-
rinic/apyrimidinic; APAF-1 — apoptotic protease activating factor-1;
ATM — ataxia telangiectasia mutated; ATR — ataxia telangiectasia
and Rad3 related; BARD — BRCA1-associated RING domain protein;
BER — base excision repair; BRCT — BRCA1 C-terminus; CDK — cy-
clin dependent kinase; CREB — cyclic AMP response element binding
protein; DD — death domain; DED — death effector domain; DISC —
death inducing signaling complex; DNA PK — DNA-dependent protein
kinase; DSB — double strand break; EGFR — epidermal growth factor
receptor; FADD — Fas-associated death domain protein; FOXO —
Forkhead box class O; GSK3 — glycogen synthase kinase 3; HR — ho-
mologous recombination; HSF-1 — heat shock factor-1; MMR — mis-
match repair; MRN — Mre11–Rad50–Nbs1; NEMO — NF-κB essential
modulator; NER — nucleotide excision repair; NF-κB — nuclear factor-
κB, NHEJ — non-homologous end joining, PAR — poly(ADP-ribose),
PARP — poly(ADP-ribose) polymerase; PARylation — poly(ADP-
ribosyl)ation; PUMA — p53 upregulated modulator of apoptosis;
RIP1 — receptor-interacting protein 1; RPA — replication protein A;
SIRT — sirtuins; SSB — single strand break; SUMO — small ubiquitin-
like modifier; TNFR — tumor necrosis factor receptor; TRADD — tumor
necrosis factor receptor type 1-associated DEATH domain.
Exp Oncol ����
��� �� �������
INVITED REVIEW
��� Experimental Oncology ��� �������� ���� ��eptem�er�
leads to a cascade of events that link the initiating
stimuli to cellular death �Fig. ��. Early apoptosis is cha-
racterized �y cell shrinkage� dense cytoplasm� tightly
packed organelles� and pyknosis due to chromatin
condensation [�� ��� �8� �9]. This is followed �y �ud-
ding which involves extensive plasma mem�rane �le�-
�ing� karyorrhexis and separation of cell fragments
into apoptotic �odies [�� �9]. The apoptotic �odies
are su�sequently phagocytosed �y macrophages�
parenchymal cells� or neoplastic cells and degraded
within phagolysosomes [�� ��� �9]. �ince apoptotic
cells do not release their cellular content into the
interstitial tissue and there are no inflammatory cyto-
kines produced� there are no inflammatory reactions
associated with apoptosis [�� ��� �9].
The major apoptotic pathways include the extrinsic
or death receptor pathway� the intrinsic or mitochon-
drial pathway� and the perforin/granzyme pathway that
involves T-cell mediated cytotoxicity �Fig. ��. For this
review we will focus �riefly on the extrinsic and intrinsic
pathways. For a more in depth discussion� please refer
to these excellent reviews [�� �6].
Extrinsic pathway
As mentioned a�ove� the extrinsic apoptotic signal-
ing is mediated �y the activation of death receptors [��
��� ��]. The death receptors are cell surface recep-
tors that transmit apoptotic signals after �inding with
specific activating ligands. Death receptors �elong
to the tumor necrosis factor receptor �TNFR� gene
superfamily� including TNFR-�� Fas/CD9�� and the
TRAIL receptors DR-� and DR-�. They are characte-
rized �y cysteine rich extracellular su�domains which
allow highly specific ligand recognition� su�sequent
trimerization� and activation of the death receptor
[��]. �u�sequent signaling is mediated �y the cy-
toplasmic part of the death receptor which contains
a conserved sequence termed the death domain �DD�.
Adapter molecules like Fas-associated death domain
protein �FADD� or Tumor necrosis factor receptor
type �-associated DEATH domain �TRADD� possess
the same sequence which allows them to form the
death inducing signaling complex �DI�C� and further
propagate the signal [��� ��]. Another domain of the
FADD� the death effector domain �DED�� sequesters
procaspase-8 to the DI�C. Accumulation of procas-
pase-8 at the DI�C leads to autocatalytic activation
due to autopreoteolysis. This su�sequently releases
active caspase-8 which activates effector caspases
resulting in cell death [�� ��� ��].
Intrinsic pathway
On the other hand� the intrinsic apoptosis pathways
involve procaspase-9 which is activated downstream
of mitochondrial proapoptotic events at the cytosolic
death signaling protein complex� the apoptosome [��
��]. Disruption of the inner mitochondrial transmem-
�rane potential and permea�ility releases proapoptotic
proteins from the mitochondrial intermem�rane space
into the cytoplasm. The released proteins include
Fig. 1. �ignaling events characteristic of apoptosis
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
cytochrome c� which activates the apoptosome and
therefore the caspase cascade [�� ��]. Dimerization
of procaspase-9 molecules at the Apaf-� scaffold
induces caspase-9 activation and su�sequent proteo-
lytic activation of the effector procaspases-�� -6� and
-7 [�� ��]. These cleave protein su�strates� including
procaspases� resulting in the mediation and amplifica-
tion of the death signal and eventually in the execution
of cell death [�� ��].
There is significant crosstalk �etween the pathways
and molecules in one pathway can influence the other
[�� ��]. The pathways converge on the same execu-
tion route which is initiated �y the cleavage of caspase
� and results in DNA fragmentation� degradation
of cytoskeletal and nuclear proteins� cross-linking
of various proteins� formation of apoptotic �odies�
expression of ligands for phagocytic cell receptors and
phagocytosis [�]. Ultimately� activation of caspases
leads to an irreversi�le cascade of events progressing
towards cell death.
As mentioned earlier� many cellular stresses can
impact survival versus death pathways. In the next
section� we focus on one particular cell stress� that
is� DNA damage.
THE DNA DAMAGE RESPONSE
The human genome is under constant attack which
leads to thousands of DNA lesions per day. The cellular
response to DNA damage is critical for maintenance
of genomic integrity [�� �� ��]. Dysregulation of this
DNA damage response leads to genomic insta�ility�
which can result in the inactivation of pro-apoptotic
pathways and the survival of cells that are polyploid�
contain damaged DNA� and have dysregulated telomere
maintenance [�� �� ��� ��]. �uppression of the tightly
regulated apoptotic process may thus play a critical
role in the development of some cancers [�� �� 8� ��].
Com�ating this malignant transformation process
is the DNA damage response� a complex mechanism
to detect the a�ove mentioned lesions� signal their
presence� and promote their repair. Additionally�
mechanisms are in place such that if the damage
is too great or repair is ineffective� activation of cell
death pathways such as apoptosis or necrosis ensues
�Fig. �� [�� 8� ��]. These steps are in place to com�at
the threats posed �y excess or unrepaired DNA da-
mage [�� 8� ��]. In this next section� we �riefly discuss
the different types of DNA damage and the cellular
responses to such damage to ultimately regulate cell
fate.
DNA can �e damaged �y exogenous agents such
as radiation� x-ray� UV� alkylating agents� as well as �y the
�y-products from endogenous processes such as reac-
tive oxygen and nitrogen species from meta�olism and
errors from DNA replication [��8� ��]. The resultant
DNA damage either involves one or �oth strands of the
DNA �single strand vs. dou�le strand �reaks� respec-
tively�. While unresolved single strand �reaks ���Bs�
can �e converted to dou�le strand �reaks �D�Bs� and
repaired� unrepaired D�Bs can lead to severe conse-
quences in cells. D�Bs can �e mutagenic� since they can
potentially affect the expression of multiple genes. Most
importantly� as little as one unrepaired D�B can �e lethal
to the cell [��8� ��].
Fig. 2. The complex relationship �etween DNA damage� repair
and apoptosis. DNA damage triggers cellular responses such
as cell cycle arrest� post-translational protein modifications� DNA
repair� and transcription of pro/anti survival genes. The �alance
�etween these processes ultimately determines cell fate
Exogenously� ultraviolet and other types of high en-
ergy radiation can induce ��Bs and D�Bs. UV-induced
damage can also result in the production of pyrimidine
dimers� where covalent cross-links occur in cytosine
and thymine residues� disrupting DNA polymerases
and preventing DNA replication [��8]. Other agents
can form DNA adducts and inter/intra strand crosslinks
which� if left unrepaired� can lead to permanent mu-
tations resulting in cell transformation and ultimately
tumor development [��8].
Endogenously� oxidative DNA damage can occur
and involves oxidation of specific �ases. 8-hydroxy-
deoxyguanosine �8-OHdG� is the most common marker
for oxidative DNA damage [��8]. Oxidative stress plays
a central role in the pathophysiology of age-induced
apoptosis via accumulated free radical-induced dam-
age to the mitochondria. On the other hand� hydrolytic
DNA damage involves deamination or the total removal
of individual �ases. Loss of DNA �ases� known as AP
�apurinic/apyrimidinic� sites� can �e particularly muta-
genic and if left unrepaired they can inhi�it transcription
[7� ��]. Interestingly� hydrolytic damage may result
from the overa�undance of reactive oxygen species�
often a �yproduct of respiration. Of course� the type
and severity of damage to the DNA dictate the cellular
response and ultimately cell fate [��].
Regardless of the type of DNA damage� the re-
sponse to this insult involves sensing the damage�
activating the checkpoints� and repairing/resolving the
DNA lesions. These processes will �e discussed �elow.
DNA damage sensors
In order to initiate the DNA damage response� DNA
damage sensors must first detect the a�errant DNA
lesions. The Mre���Rad���N�s� �MRN� complex acts
as the sensor of DNA damage and maintains genomic
��6 Experimental Oncology ��� �������� ���� ��eptem�er�
sta�ility �y processing DNA ends and recruiting/�ridg-
ing other mem�ers of the DNA damage response [��
��� ��� �6��8]. �pecifically� Rad�� recognizes the
DNA� N�s� recruits other DNA repair proteins to D�B
lesions� and Mre�� processes the DNA ends with its
DNA nuclease activity [�7]. This leads to activation
of Ataxia Telangiectasia Mutated �ATM� or Ataxia Telan-
giectasia and Rad� Related �ATR� depending on where
the damage resides �ATM: DNA D�Bs on chromatin vs.
ATR: stalled replication forks� [�6� �9� ��]. It is at this
point where a cascade of signaling events is orches-
trated to activate checkpoints and assem�le the re-
maining mem�ers of the DNA repair complex.
Checkpoint activation
Upon sensing the DNA damage� a coordinated
activation of DNA damage checkpoints as well as DNA
repair proteins is required to arrest the cell cycle� thus
allowing time for repair processes [��]. Checkpoints
also induce changes in telomeric chromatin and
recruitment of DNA repair proteins to sites of DNA
damage� activation of transcription� telomere length�
and induction of cell death �y apoptosis [��]. Not
surprisingly� several checkpoint genes are essential
for cell and organism survival.
Chk�� a serine/threonine-protein kinase is required
for checkpoint-mediated cell cycle arrest and activa-
tion of DNA repair in response to the presence of DNA
damage or unreplicated DNA. Chk� �inds to and
phosphorylates CDC�� which creates �inding sites for
��-�-� proteins and triggers degradation of CDC�� via
u�iquitination pathway and proteosomal degradation
[��� ��]. This leads to increased inhi�itory tyrosine
phosphorylation of CDK-cyclin complexes and �locks
cell cycle progression. Chk� also �inds to Rad�� which
promotes the release of Rad�� from BRCA�� increas-
ing the chromatin association of Rad�� and su�se-
quent HR-mediated DNA repair [��]. Chk� also pro-
motes repair of DNA cross-links �y phosphorylating
FANCE which is required for the nuclear accumulation
of FANCC and provides a critical �ridge �etween
the FA complex and FANCD� [��]. Chk� also plays
an essential role in maintenance of replication fork
�y regulating PCNA [��]. Besides� it also plays a role
�y modulating transcription of genes involved in cell-
cycle progression through phosphorylation of his-
tones and su�sequent epigenetic silencing of genes.
Chk� phosphorylates R�� to promote its interaction
with the E�F family of transcription factors trigger-
ing su�sequent cell cycle arrest and phosphorylates
p�� activating the protein and promoting cell cycle
arrest as well [��� ��].
Chk� functions similar to Chk�� regulating cell
cycle checkpoint arrest through phosphorylation
of CDC��� inhi�iting their activity [��� ����7]. In-
hi�ition of CDC�� phosphatase activity leads to in-
creased inhi�itory tyrosine phosphorylation of CDK-
cyclin complexes and �locks cell cycle progression.
Chk� also phosphorylates NEK6 which is involved
in G�/M cell cycle arrest [��]. �imilar to Chk��
Chk� regulates HR-mediated DNA repair through
phosphorylation of BRCA�� enhancing the chromatin
association of RAD��. Moreover� Chk� promotes the
transcription of genes involved in DNA repair �including
BRCA�� through the phosphorylation and activation
of the transcription factor FOXM� [��]. Chk� also
regulates apoptosis through the phosphorylation
of p��� MDM� and PML [��� �6� �8]. Chk� mediated
phosphorylation of p�� reverses inhi�ition �y MDM��
leading to accumulation of active p��. Chk� depen-
dent phosphorylation of MDM� also functions to re-
duce degradation of p��. The kinase also controls
the transcription of pro-apoptotic genes through
phosphorylation of the transcription factor E�F�. Fi-
nally� Chk� has a tumor suppressor role as well in that
it functions in mitotic spindle assem�ly �y phosphory-
lating BRCA� and a�sence of Chk� has �een o�served
in some cancers [��� ����8].
Cyclin dependent kinase �CDK� family of serine/
threonine kinases regulate cell cycle progression
through phosphorylation of proteins that function
at specific phases of the cell cycle [�6� �9]. Differ-
ent CDKs act at different phases of the cell cycle and
their activity is each dependent on association with
a mem�er of the cyclin family of proteins. Cdk�-cyclin
B is important for the M phase transition while Cdk�-
cyclin E association is critical for G�/� transition.
Cdk�-cyclin E also functions in the � and G� phases
while CDK�-cyclin E and CDK6-cyclin D control
progression through the G� phase of the cell cycle
�y phosphorylation of the tumor suppressor protein�
R� [�6� �9]. These proteins have thus �een actively
studied for cancer therapy.
DNA repair pathways
Once the DNA damage has �een sensed and
checkpoints activated� the process of repairing this
damage is initiated. We will first focus on the D�B
repair pathways� of which there are � major path-
ways: the homologous recom�ination �HR� and
non-homologous end joining �NHEJ� [6�8� ��� ��].
HR relies on the presence of a sister chromatid and
the cell cycle-regulated �`-to-�` resection of DNA ends
that generates stretches of single stranded DNA. This
single stranded DNA is �ound �y replication protein
A �RPA�. BRCA� su�sequently �inds Rad�� and pro-
motes its loading onto RPA coated single stranded
DNA to produce a RAD��-single stranded DNA nu-
cleoprotein filament. RPA-coated ssDNA also leads
to recruitment and activation of the checkpoint kinase
ATR� which phosphorylates various targets� includ-
ing the downstream checkpoint kinase CHK�. This
cascade of events promotes DNA strand invasion and
su�sequent HR events. Because of the use of the ho-
mologous sister chromatid as template� HR is an error
free repair pathway and the major mechanism utilized
�y cells for repairing D�Bs and restarting stalled rep-
lication forks [8� �6].
On the contrary� the error-prone NHEJ involves
connecting and resealing the two ends of DNA D�B
without the need for sequence homology �etween the
ends. Thus� this process is not dependent on the cell
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ��7
cycle and is� in fact� active throughout all phases of the
cell cycle. It involves among others� Ku 7�/8�� DNA
protein kinase family of proteins [8� ��� ��].
It is not yet clear what dictates the choice of repair
pathway. Research suggests that the choice �etween
NHEJ or HR pathways depends on cell cycle stage;
NHEJ is active throughout the cell cycle� and its activity
increases as cells progress from G� to G�/M �G� < �
< G�/M�. HR is nearly a�sent in G�� most active in the
� phase� and declines in G�/M [7� 8� ��� ��]. The over-
all efficiency of NHEJ is higher than HR at all cell cycle
stages. Cells usually utilize error-prone NHEJ as the
major D�B repair pathway at all cell cycle stages� while
HR is used primarily in the � phase. Reports also sug-
gest key repair players such as CtIP� BRCA�� Ku� and
others to impact the choice of D�B repair �y control-
ling the initial events of D�B repair such as D�B end
processing/resection [�����].
For ��Bs� different repair processes are utilized.
Usually� the intact complementary strand can �e used
as a template to repair the damaged strand via a variety
of repair mechanisms like �ase excision repair �BER�
to repair damage to a single �ase caused �y oxida-
tion� alkylation� hydrolysis� or deamination� nucleotide
excision repair �NER� to repair �ulky� helix-distorting
lesions such as pyrimidine dimers and photo-adducts�
and mismatch repair �MMR� to corrects errors of DNA
replication and recom�ination which may have resulted
in mispaired ��ut undamaged� nucleotides [�� 7� 8�
��]. Formation of ��Bs is closely linked to damaged
�ases and their attempted repair. It is worth noting that
D�Bs may form during the attempted repair of ��Bs
[�� 7� 8� ��]. Interestingly� the chromatin structure may
�e modulated to facilitate protein recruitment during
repair [��]. Modifications of DNA-associated histone
proteins maintain genomic sta�ility. Upon the induction
of DNA damage� phosphorylation of histones dictates
if repair is justified or apoptosis is warranted [��� �6].
It is truly amazing how a cell decides whether to pursue
repair and when to a�ort repair to favor apoptosis.
In this next section� we will discuss various key
players in the DNA damage response who also play
vital roles in regulating apoptosis.
INTERSECTION BETWEEN DNA DAMAGE
RESPONSE AND APOPTOSIS
DNA damage sustained from normal DNA repli-
cation/cell processes� stress� mitotic catastrophe�
agents such as radiation� toxins� hormones� growth
factors� cytokines� and drugs� and reactive oxygen
species can induce apoptosis if left unrepaired [�� ��
7� 8� ��]. Additionally� current DNA damaging agents
used in therapies act to overwhelm cellular DNA repair
capacity to activate cell death processes. For example�
irradiation� a standard treatment modality for a num�er
of cancers such as �rain� �reast� and prostate� as well
as a variety of chemotherapeutic agents induce DNA
damage leading to apoptosis [�� �� 7� 8� ��� �7]. Below�
we will discuss several key players that intersect the
DNA repair pathways with apoptotic pathways.
P53 and apoptosis
The tumor suppressor protein p�� has �een shown
to mediate cellular stress responses in that p�� can
initiate DNA repair� cell-cycle arrest� senescence and�
importantly� apoptosis [��� �6� �8� ��� �8���]. These
responses suppress tumor formation. Thus� it is not
surprising that most tumors have p�� mutations [��].
p�� mediates DNA damage response �y stimulating
the nuclear release of histone H�. Phosphorylation
is one of the primary post-translational modifications
of p�� and this increases its sta�ility. Various kinases
such as ATM� Chk� and Chk� are responsi�le for the
phosphorylation of p��. Other post-translational modi-
fications such as acetylation� u�iquitination� methy-
lation� sumoylation� and neddylation also regulates
p�� protein sta�ility and transcriptional activation [�6�
�8� ��� �8� ��� ��]. The E� u�iquitin ligase MDM� regu-
lates p�� activity �y �inding to its N-terminal transac-
tivation domain� thus preventing its interaction with
other transcriptional factors. MDM� also induces
nuclear export of p�� and targets it for proteasomal
degradation [�6� �8� ��� �8� ��� ��]. In event of failed
DNA repair p�� initiates apoptosis �y transactivating
pro-apoptotic proteins such as BAX� BID� PUMA and
NOXA which permea�ilizes the mitochondrial mem-
�rane and leaks the pro-apoptotic factors [�6� �8� ���
�8� ��� ��]. p�� stimulates the extrinsic and/or the
intrinsic pathway depending on the DNA damage.
p�� has �een reported to �ind to the outer mitochon-
drial mem�rane and antagonize the anti-apoptotic
function of BCL� and BCL-XL. p�� represses the
activity of BCL�� an anti-apoptotic protein involved
in retaining the mitochondrial permea�ility� as well
as survivin. Moreover� p�� also initiates apoptosis via
proteins localized on the endoplasmic reticulum and
plasma mem�rane such as DR�. p�� can also exert
transcription independent effects on mitochondria
mem�rane permea�ility �y activating the pro-apop-
totic protein BAX or �y neutralizing the anti-apoptotic
proteins BCL� or BCL-XL [��� �6� �8� ��� �8� ��� ��].
Tumor cells possessing wild type p�� undergo
apoptosis to a greater extent following DNA damage
than cells that possess mutations in p��. DNA damage
can also activate p��-independent apoptosis which
may �e called upon especially in cases in which p�� are
mutated. The p�� homologs p6� and p7� are involved
in this response. Unlike mutations in p��� mutations
in p7� do not predispose to tumor formation �ut do have
an impact on the DNA damage response. p7� is also
often overexpressed in cancer. As mentioned a�ove�
in response to DNA lesions� ATM and/or ATR activate
CHK� and CHK�� which in turn activate E�F� [�8]. This
in turn stimulates transcription of the p7� gene� increas-
ing the levels of p7� protein. While p�� requires p6� and
p7� to activate apoptosis� p7� is pro-apoptotic even
in the a�sence of p��. p7�-induced apoptosis is me-
diated �y transcriptional upregulation of PUMA� which
in turn induces mitochondrial translocation of BAX and
cytochrome c release �discussed a�ove�. p7� also in-
duces mitochondrial dysfunction via NOXA. On the other
��8 Experimental Oncology ��� �������� ���� ��eptem�er�
hand� p6� has the a�ility to suppress p7�-mediated
apoptosis [��� �6� �8� ��� �8� ��� ��].
Nuclear factor-κB �NF-κB� also has a role in p��-
independent apoptosis. This transcription factor is gen-
erally anti-apoptotic and promotes survival. Activation
of NF-κB in response to DNA damage is mediated
�y �UMOylation and ATM-dependent phosphorylation
of NEMO �NF-κB essential modulator�. Under some
circumstances� however� NF-κB exhi�its pro-apoptotic
activity. For instance� presence of excess reactive oxygen
species can induce NFκB-mediated transcription of the
FA� ligand� there�y stimulating apoptosis [��]. NF-κB in-
duces TNF-α production and su�sequent receptor-inter-
acting protein � �RIP�� autophosphorylation. In associa-
tion with NEMO� RIP� kinase promotes JNK�-mediated
induction of IL-8 and recruits FADD to activate caspase
8 which then induces apoptosis [��]. p��-independent
apoptosis can also �e triggered �y BCL-� degradation
and G�K� which is detailed in a later section [��].
BRCA1 and apoptosis
The tumor suppressor BRCA� plays an integral
role in the maintenance of genomic sta�ility and
modulates cellular response to DNA damage [�6��8].
It is involved in �oth the major DNA dou�le strand �reak
repair pathways — HR and NHEJ [�6]. BRCA� func-
tions in a variety of cellular processes including chro-
matin remodeling� protein u�iquitination� DNA rep-
lication� DNA repair� regulation of transcription� cell
cycle checkpoint control� and apoptosis [�6��8].
BRCA� function is regulated through diverse mecha-
nisms including transcription control� protein-protein
interaction� and post-translational modification [�9�
�6� �8� �9]. BRCA� is a nuclear-cytoplasmic shuttling
protein and its function may �e regulated via active
shuttling �etween the cellular compartments [�9� �6�
�8� �9]. When nuclear� BRCA� controls high fidelity
repair of damaged DNA. In contrast� BRCA� has �een
shown to enhance p��-independent apoptosis when
cytoplasmic [�9� �6� �8� �9]. BRCA� contains two nu-
clear localization signals which target it to the nucleus
via importin and two nuclear export sequences which
transport it to the cytoplasm via the CRM�/exportin
pathway [�9� �6� �8�6�].
As mentioned a�ove� BRCA� shuttling can also
�e regulated via protein-protein interaction. The
BRCA�-associated RING domain protein �BARD��
has �een shown to prevent CRM� dependent nuclear
export of BRCA� �y �inding to and masking the
BRCA� NE� located at the N-terminal RING domain.
On the contrary� the BRCA� C-terminus �BRCT� do-
main has �een shown to play a crucial role in DNA
damage-induced nuclear import of BRCA� through
association with numerous other proteins� including
p��� CtIP and BACH [��� �9� �6� 6�]. p�� seems
to �e an important player in DNA damage induced
BRCA� nuclear export since human �reast cancer
cells with deficiency in p�� function exhi�it a�er-
rant BRCA� shuttling [�9]. Mutations that target the
BRCT region of BRCA� have �een shown to exclude
BRCA� from the nucleus �y �locking nuclear import.
Thus� the critical region responsi�le for regulating
the location of BRCA� appears to reside in the BRCT
domain. Nuclear exclusion of BRCA� can �e thera-
peutically exploited with poly�ADP-ri�ose� polymerase
�PARP� inhi�itors as well as other DNA damaging
agents such as cisplatin [�9].
In addition to the repair of damaged DNA�
BRCA� plays a role in apoptosis [6��6�]. �pecifically�
overexpression of BRCA� induces apoptosis and the
process has �een linked to DNA damage-induced
BRCA� nuclear export and the c-Jun N-terminal ki-
nase pathway [6�]. BARD�� which �inds and masks
the BRCA� nuclear export sequence to prevent
BRCA� nuclear export� inhi�its this BRCA�-mediated
apoptosis. Moreover� the apoptotic pathway stimu-
lated �y BRCA� is independent of p�� [�6� �9].
BRCA� has also �een reported to �e present
in the mitochondria where it promotes BCL� mediated
apoptosis. BCL�-mediated targeting of BRCA� to the
endomem�ranes depletes BRCA� from the nucleus
resulting in decreased HR-mediated repair [66� 67].
In addition� BCL� expression is low in BRCA�-asso-
ciated tumors [68].
DNA-dependent protein kinase (DNA PK) and
apoptosis
The DNA-dependent protein kinase �DNA PK� plays
a critical role in D�B repair and V�D�J recom�ination
[69]. DNA PK plays a central role in the NHEJ pathway
for D�B repair in mammalian cells via autophosphory-
lation events as well as its association with other DNA
repair proteins such as BRCA� and Ku. Ku �inds to the
DNA end and recruits DNA PK� sta�ilizing its �inding
to DNA. This is followed �y �ridging of the �roken ends
�y DNA PK to facilitate rejoining. DNA-PK also recruits
and activates proteins involved in DNA end-processing
and ligation. The coordinated assem�ly of Ku and
DNA-PKcs on DNA ends is followed �y recruitment
of the DNA ligase IV�XRCC� complex that is respon-
si�le for the rejoining step [�� 7� 8� ��� 69].
DNA PK is present at the telomere and cap chromo-
some ends� protecting them telomere and preventing
chromosome end-to-end fusions. This interaction with
telomerase helps to maintain telomere length as well
[7�]. Recently� it was reported that poly�ADP-ri�ose�
polymerase � �PARP�� interacts genetically with the
DNA PK catalytic su�unit to prevent cancer �lympho-
ma� �y suppressing p�� mutation and telomere fusions
[7�]. The role of PARP in DNA repair and apoptosis
is discussed in a su�sequent section.
DNA PK also plays a crucial role in triggering apop-
tosis in response to severe DNA damage or critically
shortened telomeres [7�� 7�� 7�]. The a�ility to trigger
apoptosis in the presence of unresolved DNA dam-
age is critical for preventing progression to cancer.
In response to DNA damage� DNA PK phosphorylates
p�� and triggers p��-dependent apoptosis. Converse-
ly� DNA PK undergoes proteasomal degradation later
on in the apoptotic process which aids to suppress pro-
survival signals [7�]. The Ku7� su�unit of DNA-PK has
�een shown to suppress apoptosis �y sequestering
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ��9
Bax from mitochondria [7�]. Increased acetylation
of cytoplasmic Ku7� disrupts the Ku7�-Bax interaction
augmenting apoptosis [7�].
PARP and apoptosis
PARP is a family of proteins involved in a num-
�er of cellular processes including DNA repair and
apoptosis [76]. PARP is predominantly located in the
nucleus where it promotes BER-mediated DNA single
strand �reak repair �y �inding to the DNA and induc-
ing a structural modification [77]. It also induces the
synthesis of poly�ADP-ri�ose� �PAR� chains which
acts as a signal for other DNA repair proteins. An early
transient �urst of poly�ADP-ri�osyl�ation �PARylation�
of nuclear proteins followed �y caspase-� mediated
cleavage of PARP is required for apoptosis to pro-
ceed [8� 76�8�]. This inactivation of PARP prevents
depletion of NAD �a PARP su�strate� and ATP� which
are required for later events in apoptosis. PARylation
plays diverse roles in many molecular and cellular
processes� including DNA damage detection and
repair� chromatin modification� transcription� cell
death pathways� and mitotic apparatus function [76�
77� 79�8�]. These processes are critical for genome
maintenance� carcinogenesis� aging� inflammation�
and neuronal function [8� 76�8�].
PARP-� interacts physically and functionally with
various proteins involved in these DNA repair path-
ways� and recruits the repair proteins to sites of DNA
damage such as XRCC-� in BER and DNA PK in NHEJ-
mediated repair. PAR� as covalent attachment of au-
tomodified PARP-� and PARP-�� acts to recruit repair
proteins to sites of DNA damage [8� 7�� 76�78� 8�� 8�].
PARP dependent ��B repair and BRCA�- and
BRCA�-dependent D�B repair has �een exploited for
cancer therapy [6�8� 78� 8�]. As mentioned a�ove�
BRCA� and BRCA� are tumor-suppressor proteins
important for D�B repair �y HR� and mutation of the
genes encoding these proteins causes predisposition
to �reast and ovarian cancers. PARP inhi�itors have
shown promising results in BRCA deficient tumors
and other DNA repair deficient tumors in clinical trials
when com�ined with other cytotoxic agents [8��87].
We and others have recently reported that the PARP
inhi�ition induces apoptosis in a variety of cell types.
We have shown that in conjunction with EGFR inhi�i-
tors� the PARP inhi�itor ABT-888 activates the intrinsic
apoptotic pathway as evidenced �y cleavage of caspase
� and 9 [78]. PARP inhi�itor treatment also induces
phosphorylation of DNA PK and stimulates error-prone
NHEJ-mediated repair in HR-deficient cells� resulting
in cell death. PARP� catalytic activity possi�ly regulates
NHEJ in the a�sence of HR and thus� deregulated NHEJ
may explain the exquisite cytotoxicity of HR deficient
cells to PARP inhi�itors [78� 88].
PARP� inhi�itor induces caspase-independent cell
death as well. It causes mitochondrial depolarization�
mitochondrial permea�ility transition and mitochon-
drial release of AIF which� upon release from the
mitochondria� translocates into the nucleus� where
it triggers nuclear DNA fragmentation [89� 9�]. Thus�
PARP inhi�itors tilt cell death from necrosis to apop-
tosis in cancer cells [9�].
ATM/ATR and apoptosis
Defects in ATM are associated with cancers such
as T-cell pro-lymphocytic leukemia� and B-cell chronic
lymphocytic leukemia [6�8� 9�]. Defective ATM also
predisposes to sporadic colon cancer in tumors with
microsatellite insta�ility [9�]. Loss of ATM results
in hypersensitivity to radiation and defect in cell cycle
arrest [9�]. ATM is also involved in p7� mediated
apoptosis [9�]. Radiation induces ATM-dependent
c-A�l phosphorylation which then activates p7�. Upon
commitment to apoptosis� caspases �cysteine aspartic
acid proteases� are activated in a proteolytic cascade
and ATM is cleaved �y a caspase-�-like apoptotic pro-
tease. This generates a truncated ATM protein devoid
of kinase activity �ut still retaining its DNA �inding
a�ility. This functions to prevent further DNA repair
and propagation of DNA damage signaling [9�� 9�].
As mentioned �efore� ATR activates p�� in response
to DNA damage �y phosphorylating p�� [�9� ��� �6�
��]. In response to DNA damage� ATR also phosphory-
lates and activates Chk�� which in turn phosphorylates
p�� and regulates cell cycle progression. ATR also
mediates phosphorylation of BRCA� in response to UV
[�9� ��� �6� ��]. Thus� these signaling pathways can
converge on �oth p�� and BRCA� mediated apoptosis.
ATM/ATR also mediates BID phosphorylation�
which is required for DNA damage-induced intra-
� phase checkpoint [96]. An intact BH� domain is re-
quired for apoptosis �ut not BID-mediated � phase
effects. Thus the two functions may �e distinct. BID
accumulates in �oth the nucleus and mitochondria
following stress suggesting a possi�le role in DNA
damage response. Furthermore� like ATM and ATR�
BID localizes to the chromatin fraction of the nucleus
following treatment with DNA-damaging agents [96].
Sirtuins (SIRT) and apoptosis
�irtuins ��IRT� are a family of histone deacetylases
which also has a role in the maintenance of genomic
sta�ility [97]. They also possess mono-ri�osyltransferase
activity [97� 98]. �IRT have �een implicated in influencing
aging and regulating transcription� apoptosis and stress
resistance [99]. For instance� �IRT� can deacetylate vari-
ous factors linked to the repair of DNA damage� includ-
ing the Werner helicase and NB�� [�������]. A�sence
of �IRT results in increased chromosomal a�errations
and impaired DNA repair. In addition �IRTs are recruited
to sites of DNA �reaks following DNA damage to avoid
genomic insta�ility [���]. Thus� �IRTs regulate epigenetic
silencing and chromatin modification.
�IRT�� the human �ir� homolog� acts as a negative
regulator of the transactivation function of p�� �y �ind-
ing and deacetylating p��� there�y repressing apop-
tosis induced �y DNA damage [���� ���]. Persistent
lesions keep PARP in an activated state and the
nicotinamide produced �y the process inhi�its �IRT�.
This leads to hyperacetylation and enhanced trans-
activation of p�� which in turn leads to increase in the
transcription of pro-apoptotic genes and su�sequent
��� Experimental Oncology ��� �������� ���� ��eptem�er�
apoptosis. Besides p��� �IRT� can regulate other
targets linked to cell death� including Ku7�� E�F� and
TGF-β signaling [��6� ��7].
Another �IRT-mediated pro-survival pathway in-
volves the Forkhead �ox class O �FOXO� transcription
factors which control the expression of genes involved
in apoptosis such as Fas ligand� Bim� TRAIL� cyclin D�
Gadd��� p�7/Kip�� Gadd��� Mn�OD. Akt phosphory-
lates FOXO factors in the presence of growth factors
[��8� ��9]. This prevents their nuclear translocation.
However� when the growth factor signaling is switched
off� FOXOs are located in the nuclei and act as tran-
scription factors. Acetyltransferases� PCAF and p���/
CBP mediated acetylation silences the transcription
factors. The transcriptional activity of FOXO� is re-
stored �y deacetylation carried out �y �IRT� which
leads to resumption of gene transcription including
DNA damage checkpoint genes. This increases the
a�ility of FOXO to induce cell cycle arrest and resist oxi-
dative stress [97�99� ��6���9]. Thus �IRT promotes
cell survival via transcriptional regulation. Moreover�
the concerted action of �IRT� and �IRT� appear
to inhi�it cell death �y maintaining mitochondrial NAD
levels following stress [98� ���]. It should �e noted
that although �IRT predominantly antagonize stress-
induced cell death pathways� �IRT� can also deacety-
late components of the NFκB complex� leading to in-
creased cell death� primarily senescence [98� ���].
Epidermal growth factor receptor (EGFR) and
apoptosis
The epidermal growth factor receptor �EGFR� plays
an important role in the development and progres-
sion of solid tumors. In addition� EGFR activation also
mediates resistance to chemotherapy and radiation
therapy [78� 88]. EGFR inhi�ition down-modulates
survival pathways and shifts towards the proapoptotic
Bcl-� expression and/or activation [���].
There are multiple inhi�itors of EGFR that are currently
either used as a standard of care or are in clinical trials.
Inhi�itors of the tyrosine kinase activity of EGFR compete
with ATP for �inding to the tyrosine kinase pocket of the
receptor. They have significant antitumor activity since
the EGFR-TKIs �lock signaling �y �oth ERK and AKT
pathways and induce apoptosis [���]. EGFR mediated
apoptosis requires an active kinase �ut not EGFR auto-
phosphorylation sites� meaning the truncated receptor
can generate the apoptotic signal. EGFR can activate
Ras and the induction in apoptosis is due to impaired
Akt activation. EGFR inhi�ition induces BIM expression
via inhi�ition of the MEK-ERK pathway and BIM induc-
tion plays a key role in EGFR-TKI-induced apoptosis.
Research suggests that �oth the PI�K-AKT-survivin
and MEK-ERK-BIM pathways contri�ute independently
to EGFR inhi�itor-induced apoptosis [������6].
The BH�-only protein PUMA �p�� upregulated
modulator of apoptosis� plays an essential role in p��-
dependent and -independent apoptosis. PUMA medi-
ates apoptosis through the Bcl-� family proteins Bax/
Bak and the mitochondrial pathway [��7]. PUMA is also
induced �y EGFR inhi�itors independent of p��. EGFR
inhi�itors �lock phosphorylation of EGFR and inhi�it the
PI�K/AKT pathway� which leads to increased expres-
sion of p7� and its �inding to the PUMA promoter and
su�sequent transactivation [��8]. Thus� PUMA func-
tions as a critical mediator of EGFR inhi�itor-induced
apoptosis� especially in head and neck cancer cells
where EGFR inhi�itors are widely used. Moreover� p7��
p6�� and the PI�K/AKT pathway serve as key regulators
of PUMA induction after EGFR inhi�ition [��9].
Recent evidence also suggests a key role of EGFR
in �oth major DNA D�B repair pathways. �pecifically�
EGFR �inds and activates DNA PK for NHEJ [����
���]. Additionally� EGFR inhi�itors have �een shown
to attenuate DNA repair pathways. Interestingly�
we recently reported that the EGFR inhi�itor cetux-
ima� reduced �oth NHEJ and HR in head and neck
cancer cells and su�sequently induced a synthetic
lethality with the PARP inhi�itor ABT-888 [78]. This
enhanced cytotoxicity was associated with activation
of the intrinsic apoptotic pathway [78]. This �rings
forth the possi�ility that other DNA repair proteins may
�e involved in the apoptotic response. We are actively
investigating this avenue.
Glycogen synthase kinase 3 (GSK3) and apop-
tosis
Another important player in linking extracellular sig-
nals to DNA damage/repair and ultimately apoptosis
is the glycogen synthase kinase � �G�K��. Phosphory-
lation of su�strates �y G�K� allows it to modulate key
processes including cell structure� meta�olism� gene
expression and apoptosis [���]. G�K� has the unique
capacity to either increase or decrease the apoptotic
threshold due to its opposing regulation of the two
major apoptotic signaling pathways. G�K� promotes
cell death caused �y the mitochondrial intrinsic
apoptotic pathway� �ut inhi�its the death receptor-
mediated extrinsic apoptotic signaling pathway [���].
G�K� is involved in the apoptotic response following
growth factor withdrawal� inhi�ition of the PI�K/Akt
signaling pathway� DNA damage� ER stress� hypoxia/
ischemia� and oxidative stress [���]. Intrinsic apop-
totic signaling which is activated �y cell damage
is promoted �y G�K� �y facilitation of signals that
cause disruption of mitochondria and �y regulation
of transcription factors that control the expression
of anti- or pro-apoptotic proteins. These transcription
factors include p�� which was discussed a�ove and
cyclic AMP response element �inding protein �CREB�
[���� ���]. G�K�β activity in the nucleus promotes
p��-induced expression of Bax in response to DNA
damage and inhi�its CREB� which can �lock the
CREB-dependent expression of the anti-apoptotic
protein Bcl-� [���]. G�K� can regulate p�� levels
through the phosphorylation of the p��-regulating
protein MDM� as well as �y directly interacting with
p�� [���]. G�K� promotes p��-mediated transcrip-
tion of specific genes and regulates the intracellular
localization of p�� [���]. p�� is also a�le to activate
apoptosis independently of its transcription function
�y acting directly on mitochondrial proteins� and
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
G�K�β �inds p�� in the mitochondria� which may
contri�ute to p��-induced apoptosis [��6]. In the
canonical Wnt signaling pathway� the transcriptional
co-activator β-catenin promotes growth and survival�
�ut phosphorylation of β-catenin �y G�K� targets it for
proteosomal degradation there�y promoting apopto-
sis [���]. Activation of Wnt signaling inhi�its G�K� se-
lectively in the Wnt signaling protein complex� causing
accumulation of β-catenin and its translocation to the
nucleus where it interacts with the TCF/LEF transcrip-
tion factors to induce expression of pro-survival genes.
G�K� phosphorylates heat shock factor-� �H�F-���
a pro-survival transcription factor to inhi�it its activity�
there�y reducing expression of heat shock proteins�
an action that can facilitate apoptosis [��7]. �ince
G�K� is present in the mitochondria as well and there
is a spike in G�K� levels following DNA damage�
G�K� is uniquely positioned to regulate apoptosis.
Pro-apoptotic mem�ers of the Bcl-� family of pro-
teins such as Bax transmit the apoptotic signal to the
mitochondria following phosphorylation �y G�K�. �tress
such as DNA damage induces a conformational change
in Bax that promotes its translocation from the cyto-
plasm to the mitochondria where it can �oth sequester
anti-apoptotic Bcl-� family proteins and oligomerize
within the mitochondrial mem�rane. This as well as phos-
phorlylation of the voltage-dependent anion channels
�y G�K� disrupts the mitochondrial mem�rane potential
and releases apoptotic proteins such as cytochrome
c from the mitochondrial intermem�rane space into the
cytoplasm. Cytochrome c in turn �inds to the protein
apoptotic protease activating factor-� �APAF-��� ATP/
dATP� and procaspase-9 to form the apoptosome in the
cytoplasm. This causes the activation of caspase-9�
there�y triggering the activation of the caspase cascade
as discussed a�ove [���� ���].
The extrinsic apoptotic pathway entails extracel-
lular ligands stimulating cell-surface death receptors
that initiate apoptosis �y activating caspase-8� and this
early step in extrinsic apoptotic signaling is inhi�ited
�y G�K�. Examples of death receptors include p���
Fas� DR� and DR�. Cellular insults induce recep-
tor homo-trimerization followed �y the recruitment
of cytoplasmic adaptor and effector proteins which
activates the receptor. This complex su�sequently
�inds to the cytoplasmic proteins FADD and procas-
pase-8 �or procaspase-��� to form the DI�C. DI�C
formation can allow autoactivation of caspase-8� which
then leads to the activation of effector caspases� pri-
marily caspases-�� -6� and -7 [���� ���]. This pathway
is discussed in detail a�ove. Thus� G�K� modulates
key steps in each of the two major pathways of apop-
tosis� �ut in opposite directions.
G�K�β knockout mice are em�ryonically lethal due
to massive hepatocyte apoptosis� which demonstrates
that G�K�β is an important inhi�itor of apoptosis [�7�
���� ��8]. G�K� inhi�itors promote apoptosis induced
�y stimulation of DD-containing receptors �ut provide
protection from many other insults that induce apoptosis.
For instance� we have previously reported that inhi�ition
of G�K� using lithium and other chemical inhi�itors se-
lectively kill tumor cells such as gliomas and leukemia �ut
protect normal tissues from radiation-induced toxicity.
The mechanism involved enhanced NHEJ mediated D�B
repair following IR in normal tissues �ut not cancer. �ince
radiation cannot �e selectively delivered to cancer cells�
it leads to many destructive cellular processes including
apoptosis� genomic insta�ility� and autophagy. Cranial
irradiation therapy is a standard method for treatment
of �rain cancer �ut results in long term neurocognitive
deficits� especially in children. Thus� treatment with
G�K� inhi�itors may potentially improve the quality of life
of cancer patients undergoing radiation treatment. �eve-
ral clinical trials have �een initiated to test the efficacy
of lithium in neuroprotection during the treatment of �rain
tumors �ut the trials are still at their infancy [�7� ���� ��8].
It is perplexing that inhi�ition of G�K� upregulates
DNA repair exclusively in normal cells. A possi�le
explanation may �e that G�K� is already maximally
inhi�ited in the majority of cancer. p�� status may
also play a role in determining cellular response
to G�K� inhi�ition �ut further research is warranted
in this avenue. Our la� is currently investigating the role
of G�K� in DNA damage/repair and how this relates
to G�K�-induced neuroprotection.
CONCLUSION
A wide array of key players in the DNA damage re-
sponse also is also involved in the interplay �etween cell
survival and apoptosis. Further research is necessary
to decipher the mechanisms �y which cell fate is de-
termined. In this complex network� uncovering these
mechanisms may allow for the understanding of certain
diseases and the generation of more effective therapies.
ACKNOWLEDGEMENT
�ome of the work descri�ed in this review was
supported �y the IMPACT Award from the Department
of Radiation Oncology� University of Ala�ama-Birming-
ham Comprehensive Cancer Center� a translational
scholar award from the �idney Kimmel Foundation
for cancer research� a pilot award from the Center
for Clinical and Translational �cience �CCT�� and the
Council of Center Directors �COCD� Translational Re-
search Intramural Pilot Grant Program �grant num�er
�UL� RR���777-��� from the NIH National Center
for Research Resources� and a medical research
award from the Ga�rielle’s Angel Foundation �to E�Y�.
We recognize that we were una�le to cover all aspects
of DNA damage response and cell death pathways
in this review. We apologize to those whom we have
�een una�le to cite owing to space constraints.
REFERENCES
1. Chittenden T, Harrington EA, O’Connor R, et al. Induc-
tion of apoptosis by the Bcl-2 homologue Bak. Nature 1995;
374: 733–6.
2. Elmore S. Apoptosis: a review of programmed cell death.
Toxicol Pathol 2007; 35: 495–516.
3. Degenhardt K, Mathew R, Beaudoin B, et al. Autophagy
promotes tumor cell survival and restricts necrosis, inflamma-
tion, and tumorigenesis. Cancer Cell 2006; 10: 51–64.
��� Experimental Oncology ��� �������� ���� ��eptem�er�
4. Bartek J, Lukas C, Lukas J. Checking on DNA damage
in S phase. Nat Rev Mol Cell Biol 2004; 5: 792–804.
5. Jackson SP, Bartek J. The DNA-damage response
in human biology and disease. Nature 2009; 461: 1071–8.
6. Aziz K, Nowsheen S, Georgakilas AG. Nanotechnology
in cancer therapy: targeting the inhibition of key DNA repair
pathways. Curr Mol Med 2010; 10: 626–39.
7. Aziz K, Nowsheen S, Pantelias G, et al. Targeting DNA
damage and repair: Embracing the pharmacological era for
successful cancer therapy. Pharmacol Ther 2011; 133: 334–50.
8. Nowsheen S, Whitley AC, Yang ES. Biomarkers to as-
sess the targeting of DNA repair pathways to augment tumor
response to therapy. Curr Mol Med 2012; 12: 788–803.
9. Carson DA, Ribeiro JM. Apoptosis and disease. Lancet
1993; 341: 1251–4.
10. Savill J. Apoptosis in disease. Eur J Clin Invest 1994;
24: 715–23.
11. Brown JM, Attardi LD. The role of apoptosis in cancer
development and treatment response. Nat Rev Cancer 2005;
5: 231–7.
12. Kerr JFR, Wyllie AH, Currie AR. Apoptosis: a basic
biological phenomenon with wide-ranging implications in tis-
sue kinetics. Br J Cancer 1972; 26: 239–57.
13. Reed JC, Green DR. Apoptosis: Physiology and Pa-
thology. Cambridge: Cambridge University Press, 2011.
14. Kroemer G, Martin SJ. Caspase-independent cell
death. Nat Med 2005; 11: 725–30.
15. Roos WP, Kaina B. DNA damage-induced apopto-
sis: From specific DNA lesions to the DNA damage response
and apoptosis. Cancer Lett 2012 [Epub ahead of print].
16. Zhivotovsky B, Kroemer G. Apoptosis and genomic
instability. Nat Rev Mol Cell Biol 2004; 5: 752–62.
17. Fan TJ, Han LH, Cong RS, et al. Caspase family pro-
teases and apoptosis. Acta Biochim Biophys Sin (Shanghai)
2005; 37: 719–27.
18. Trisciuoglio L, Bianchi ME. Several nuclear events
during apoptosis depend on caspase-3 activation but do not
constitute a common pathway. PloS One 2009; 4: e6234.
19. Kroemer G, Dallaporta B, RescheRigon M. The
mitochondrial death/life regulator in apoptosis and necrosis.
Ann Rev Physiol 1998; 60: 619–42.
20. Fulda S, Debatin KM. Extrinsic versus intrinsic apop-
tosis pathways in anticancer chemotherapy. Oncogene 2006;
25: 4798–811.
21. Wajant H. The Fas signaling pathway: more than
a paradigm. Science 2002; 296: 1635–6.
22. Wang S, ElDeiry WS. TRAIL and apoptosis induction
by TNF-family death receptors. Oncogene 2003; 22: 8628–33.
23. Polo SE, Jackson SP. Dynamics of DNA damage
response proteins at DNA breaks: a focus on protein modifica-
tions. Genes Dev 2011; 25: 409–33.
24. Zhou BBS, Elledge SJ. The DNA damage response: put-
ting checkpoints in perspective. Nature 2000; 408: 433–9.
25. Robertson A, Klungland A, Rognes T, et al. DNA repair
in mammalian cells. Cell Mol Life Sci 2009; 66: 981–93.
26. Falck J, Forment JV, Coates J, et al. CDK targeting
of NBS1 promotes DNA-end resection, replication restart
and homologous recombination. EMBO Rep 2012; 13: 561–8.
27. Lee JH, Paull TT. ATM activation by DNA double-
strand breaks through the Mre11-Rad50-Nbs1 complex.
Science 2005; 308: 551–4.
28. Uziel T, Lerenthal Y, Moyal L, et al. Requirement of the
MRN complex for ATM activation by DNA damage. EMBO
J 2003; 22: 5612–21.
29. Flynn RL, Zou L. ATR: a master conductor of cellular
responses to DNA replication stress. Trends Biochem Sci
2011; 36: 133–40.
30. Smith J, Tho LM, Xu N, et al. The ATM-Chk2 and
ATR-Chk1 pathways in DNA damage signaling and cancer.
Adv Cancer Res 2010; 108: 73–112.
31. Vermeulen K, van Bockstaele DR, Berneman ZN. The
cell cycle: a review of regulation, deregulation and therapeutic
targets in cancer. Cell Prolif 2003; 36: 131–49.
32. Bahassi EM, Ovesen JL, Riesenberg AL, et al. The
checkpoint kinases Chk1 and Chk2 regulate the functional
associations between hBRCA2 and Rad51 in response to DNA
damage. Oncogene 2008; 27: 3977–85.
33. Stracker TH, Usui T, Petrini JHJ. Taking the time
to make important decisions: the checkpoint effector kinases
Chk1 and Chk2 and the DNA damage response. DNA Repair
(Amst) 2009; 8: 1047–54.
34. Shieh SY, Ahn J, Tamai K, et al. The human homologs
of checkpoint kinases Chk1 and Cds1 (Chk2) phosphorylate
p53 at multiple DNA damage-inducible sites. Genes Dev
2000; 14: 289–300.
35. Bartek J, Lukas J. Chk1 and Chk2 kinases in check-
point control and cancer. Cancer Cell 2003; 3: 421–9.
36. Pabla N, Huang S, Mi QS, et al. ATR-Chk2 signaling
in p53 activation and DNA damage response during cisplatin-
induced apoptosis. J Biol Chem 2008; 283: 6572–83.
37. Stracker TH, Couto SS, CordonCardo C, et al.
Chk2 suppresses the oncogenic potential of DNA replication-
associated DNA damage. Mol Cell 2008; 31: 21–32.
38. Polager S, Ginsberg D. p53 and E2f: partners in life
and death. Nat Rev Cancer 2009; 9: 738–48.
39. Malumbres M, Barbacid M. Cell cycle, CDKs and
cancer: a changing paradigm. Nat Rev Cancer 2009; 9: 153–66.
40. Bernstein C, Bernstein H, Payne CM, et al. DNA
repair/pro-apoptotic dual-role proteins in five major DNA
repair pathways: fail-safe protection against carcinogenesis.
Mutat Res 2002; 511: 145–78.
41. Fattah F, Lee EH, Weisensel N, et al. Ku regulates the
non-homologous end joining pathway choice of DNA double-
strand break repair in human somatic cells. PLoS Genet 2010;
6: e1000855.
42. Symington LS, Gautier J. Double-strand break end
resection and repair pathway choice. Annu Rev Genet 2011;
45: 247–71.
43. Shibata A, Conrad S, Birraux J, et al. Factors de-
termining DNA double-strand break repair pathway choice
in G2 phase. EMBO J 2011; 30: 1079–92.
44. Yun MH, Hiom K. CtIP-BRCA1 modulates the choice
of DNA double-strand-break repair pathway throughout the
cell cycle. Nature 2009; 459: 460–3.
45. Nakamura K, Kogame T, Oshiumi H, et al. Collabora-
tive Action of Brca1 and CtIP in elimination of covalent modi-
fications from double-strand breaks to facilitate subsequent
break repair. PLoS Genet 2010; 6: e1000828.
46. Ikura T, Ogryzko VV, Grigoriev M, et al. Involvement
of the TIP60 histone acetylase complex in DNA repair and
apoptosis. Cell 2000; 102: 463–73.
47. Yang ES, Wang H, Jiang G, et al. Lithium-mediated
protection of hippocampal cells involves enhancement of DNA-
PKdependent repair in mice. J Clin Invest 2009; 119: 1124–35.
48. Dai C, Tang Y, Jung SY, et al. Differential effects
on p53-mediated cell cycle arrest vs. apoptosis by p90. Proc
Natl Acad Sci USA 2011; 108: 18937–42.
49. Jiang J, Yang ES, Jiang G, et al. p53-dependent
BRCA1 nuclear export controls cellular susceptibility to DNA
damage. Cancer Res 2011; 71: 5546–57.
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
50. Pauklin S, Kristjuhan A, Maimets T, et al. ARF and
ATM/ATR cooperate in p53-mediated apoptosis upon
oncogenic stress. Biochem Biophys Res Commun 2005;
334: 386–94.
51. Thangaraju M, Kaufmann SH, Couch FJ. BRCA1 fa-
cilitates stress-induced apoptosis in breast and ovarian cancer
cell lines. J Biol Chem 2000; 275: 33487–96.
52. Levine AJ. p53, the cellular gatekeeper review for
growth and division. Cell 1997; 88: 323–31.
53. Ryan KM, Ernst MK, Rice NR, et al. Role of NF-
kB in p53-mediated programmed cell death. Nature 2000;
404: 892–6.
54. Biton S, Ashkenazi A. NEMO and RIP1 control cell
fate in response to extensive DNA damage via TNF-α feed-
forward signaling. Cell 2011; 145: 92–103.
55. Bock FJ, Villunger A. GSK3 TIPping off p53 to unleash
PUMA. Mol Cell 2011; 42: 555–6.
56. Yang ES, Xia F. BRCA1 16 years later: DNA damage-
induced BRCA1 shuttling. FEBS J 2010; 277: 3079–85.
57. Nowsheen S, Aziz K, Tran PT, et al. Epigenetic inactiva-
tion of DNA repair in breast cancer. Cancer Lett 2012 [Epub
ahead of print].
58. Zhang J, Powell SN. The role of the BRCA1 tumor
suppressor in DNA double-strand break repair. Mol Cancer
Res 2005; 3: 531–9.
59. Wang H, Yang ES, Jiang J, et al. DNA damage-
induced cytotoxicity is dissociated from BRCA1’s DNA re-
pair function but is dependent on its cytosolic accumulation.
Cancer Res 2010; 70: 6258–267.
60. Feng Z, Kachnic L, Zhang J, et al. DNA damage
induces p53-dependent BRCA1 nuclear export. J Biol Chem
2004; 279: 28574–84.
61. Fitzgerald LD, Bailey CK, Brandt SJ, et al. BRCA1 ac-
cumulates in the nucleus in response to hypoxia and TRAIL
and enhances TRAIL-induced apoptosis in breast cancer cells.
FEBS J 2007; 274: 5137–46.
62. Gu M, Li H, Shen C, et al. Cloning and character-
ization of a new BRCA1 variant: A role for BRCT domains
in apoptosis. Cancer Lett 2010; 295: 205–13.
63. Martin SA, Ouchi T. BRCA1 phosphorylation regulates
caspase-3 activation in UV-induced apoptosis. Cancer Res
2005; 65: 10657–62.
64. Shao N, Chai YL, Shyam E, et al. Induction of apop-
tosis by the tumor suppressor protein BRCA1. Oncogene
1996; 13: 1–7.
65. Harkin DP, Bean JM, Miklos D, et al. Induction
of GADD45 and JNK/SAPK-dependent apoptosis following
inducible expression of BRCA1. Cell 1999; 97: 575–86.
66. Wang YL, Wang B, Zhang H, et al. BRCA1 involved in regu-
lation of Bcl-2 expression and apoptosis susceptibility to ionizing
radiation. Sci China Phys Mech Astron 2011; 54; 916–22.
67. Laulier C, Barascu A, GuirouilhBarbat J, et al.
Bcl-2 inhibits nuclear homologous recombination by localizing
BRCA1 to the endomembranes. Cancer Res 2011; 71: 3590–602.
68. Freneaux P, StoppaLyonnet D, Mouret E, et al.
Low expression of bcl-2 in Brca1-associated breast cancers.
Br J Cancer 2000; 83: 1318–22.
69. Mandal PK, Blanpain C, Rossi DJ. DNA damage
response in adult stem cells: pathways and consequences. Nat
Rev Mol Cell Biol 2011; 12: 198–202.
70. Burma S, Chen DJ. Role of DNA PK in the cellular
response to DNA double-strand breaks. DNA Repair (Amst)
2004; 3: 909–18.
71. Rybanska I, Ishaq O, Chou J, et al. PARP1 and DNA-
PKcs synergize to suppress p53 mutation and telomere fusions
during T-lineage lymphomagenesis. Oncogene 2012 [Epub
ahead of print].
72. Espejel S, Franco S, Sgura A, et al. Functional interac-
tion between DNA-PKcs and telomerase in telomere length
maintenance. EMBO J 2002; 21: 6275–87.
73. Espejel S, Martí n M, Klatt P, et al. Shorter telomeres,
accelerated ageing and increased lymphoma in DNA-PKcs-
deficient mice. EMBO Rep 2004; 5: 503–9.
74. Sawada M, Sun W, Hayes P, et al. Ku70 suppresses
the apoptotic translocation of Bax to mitochondria. Nat Cell
Biol 2003; 5: 320–9.
75. Cohen HY, Lavu S, Bitterman KJ, et al. Acetylation
of the C terminus of Ku70 by CBP and PCAF controls Bax-
mediated apoptosis. Mol Cell 2004; 13: 627–38.
76. Luo X, Kraus WL. On PAR with PARP: cellular stress
signaling through poly (ADP-ribose) and PARP-1. Genes
Dev 2012; 26: 417–32.
77. Krishnakumar R, Kraus WL. The PARP side of the
nucleus: molecular actions, physiological outcomes, and
clinical targets. Mol Cell 2010; 39: 8–24.
78. Nowsheen S, Bonner JA, LoBuglio AF, et al. Cetuximab
augments cytotoxicity with poly (ADP-ribose) polymerase
inhibition in head and neck cancer. PloS One 2011; 6: e24148.
79. Yu SW, Andrabi SA, Wang H, et al. Apoptosis-inducing
factor mediates poly(ADP-ribose) (PAR) polymer-induced
cell death. Proc Natl Acad Sci USA 2006; 103: 18314–9.
80. Andrabi SA, Kim NS, Yu SW, et al. Poly(ADP-ribose)
(PAR) polymer is a death signal. Proc Natl Acad Sci USA
2006; 103: 18308–13.
81. Kim MY, Zhang T, Kraus WL. Poly(ADP-ribosyl)ation
by PARP-1: “PAR-laying” NAD+ into a nuclear signal. Genes
Dev 2005; 19: 1951–67.
82. Kraus WL, Lis JT. PARP goes transcription. Cell 2003;
113: 677–83.
83. O’Shaughnessy J, Osborne C, Pippen JE, et al. Iniparib
plus chemotherapy in metastatic triple-negative breast cancer.
N Engl J Med 2011; 364: 205–14.
84. Lord CJ, Ashworth A. The DNA damage response and
cancer therapy. Nature 2012; 481: 287–94.
85. Tutt A, Robson M, Garber JE, et al. Oral poly(ADP-
ribose) polymerase inhibitor olaparib in patients with
BRCA1 or BRCA2 mutations and advanced breast can-
cer: a proof-of-concept trial. Lancet 2010; 376: 235–44.
86. Audeh MW, Carmichael J, Penson RT, et al. Oral
poly(ADP-ribose) polymerase inhibitor olaparib in patients
with BRCA1 or BRCA2 mutations and recurrent ovarian
cancer: a proof-of-concept trial. Lancet 2010; 376: 245–51.
87. Fong PC, Yap TA, Boss DS, et al. Poly(ADP)-ribose
polymerase inhibition: frequent durable responses in BRCA
carrier ovarian cancer correlating with platinum-free interval.
J Clin Oncol 2010; 28: 2512–9.
88. Nowsheen S, Bonner JA, Yang ES. The poly(ADP-
ribose) polymerase inhibitor ABT-888 reduces radiation-
induced nuclear EGFR and augments head and neck tumor
response to radiotherapy. Radiother Oncol 2011; 99: 331–8.
89. Yu SW, Wang H, Poitras MF, et al. Mediation of poly
(ADP-ribose) polymerase-1-dependent cell death by apopto-
sis-inducing factor. Science 2002; 297: 259.
90. Cregan SP, Dawson VL, Slack RS. Role of AIF
in caspase-dependent and caspase-independent cell death.
Oncogene 2004; 23: 2785–96.
91. Ye K. PARP inhibitor tilts cell death from necrosis
to apoptosis in cancer cells. Cancer Biol Ther 2008; 7: 942–4.
92. Derheimer FA, Kastan MB. Multiple roles of ATM
in monitoring and maintaining DNA integrity. FEBS Lett
2010; 584: 3675–81.
��� Experimental Oncology ��� �������� ���� ��eptem�er�
93. Yoshida K, Ozaki T, Furuya K, et al. ATM-dependent
nuclear accumulation of IKK-alpha plays an important role
in the regulation of p73-mediated apoptosis in response to cis-
platin. Oncogene 2007; 27: 1183–8.
94. Smith GCM, d’Adda di Fagagna F, Lakin ND, et al.
Cleavage and inactivation of ATM during apoptosis. Mol Cell
Biol 1999; 19: 6076–84.
95. Wang J, Pabla N, Wang CY, et al. Caspase-mediated
cleavage of ATM during cisplatin-induced tubular cell apopto-
sis: inactivation of its kinase activity toward p53. Am J Physiol
Renal Physiol 2006; 291: F1300–7.
96. Zinkel SS, Hurov KE, Ong C, et al. A role for proapoptotic
BID in the DNA-damage response. Cell 2005; 122: 579–91.
97. Kelly GS. A review of the sirtuin system, its clinical
implications, and the potential role of dietary activators like
resveratrol: part 2. Altern Med Rev 2010; 15: 313–28.
98. Finkel T, Deng CX, Mostoslavsky R. Recent progress in the
biology and physiology of sirtuins. Nature 2009; 460: 587–91.
99. Longo VD, Kennedy BK. Sirtuins in aging and age-
related disease. Cell 2006; 126: 257–68.
100. Yuan Z, Zhang X, Sengupta N, et al. SIRT1 regulates
the function of the Nijmegen breakage syndrome protein. Mol
Cell 2007; 27: 149–62.
101. Vaitiekunaite R, Butkiewicz D, Krzesniak M, et al. Ex-
pression and localization of Werner syndrome protein is modu-
lated by SIRT1 and PML. Mech Ageing Dev 2007; 128: 650–61.
102. Uhl M, Csernok A, Aydin S, et al. Role of SIRT1 in ho-
mologous recombination. DNA Repair (Amst) 2009; 9: 383–93.
103. Wang RH, Sengupta K, Li C, et al. Impaired DNA
damage response, genome instability, and tumorigenesis
in SIRT1 mutant mice. Cancer Cell 2008; 14: 312–23.
104. Chen WY, Wang DH, Yen RWC, et al. Tumor suppres-
sor HIC1 directly regulates SIRT1 to modulate p53-dependent
DNA-damage responses. Cell 2005; 123: 437–48.
105. Kim EJ, Kho JH, Kang MR, et al. Active regulator
of SIRT1 cooperates with SIRT1 and facilitates suppression
of p53 activity. Mol Cell 2007; 28: 277–90.
106. Brooks CL, Gu W. How does SIRT1 affect metabo-
lism, senescence and cancer? Nat Rev Cancer 2008; 9: 123–8.
107. Michan S, Sinclair D. Sirtuins in mammals: insights
into their biological function. Biochem J 2007; 404: 1–13.
108. Giannakou ME, Partridge L. The interaction between
FOXO and SIRT1: tipping the balance towards survival. Trends
Cell Biol 2004; 14: 408–12.
109. Yang Y, Hou H, Haller EM, et al. Suppression
of FOXO1 activity by FHL2 through SIRT1-mediated deacety-
lation. EMBO J 2005; 24: 1021–32.
110. Ahn BH, Kim HS, Song S, et al. A role for the mito-
chondrial deacetylase Sirt3 in regulating energy homeostasis.
Proc Natl Acad Sci USA 2008; 105: 14447–52.
111. Kawahara TLA, Michishita E, Adler AS, et al.
SIRT6 links histone H3 lysine 9 deacetylation to NF-kB-
dependent gene expression and organismal life span. Cell
2009; 136: 62–74.
112. Huang SM, Bock JM, Harari PM. Epidermal growth
factor receptor blockade with C225 modulates proliferation,
apoptosis, and radiosensitivity in squamous cell carcinomas
of the head and neck. Cancer Res 1999; 59: 1935–40.
113. Sarkar S, Mazumdar A, Dash R, et al. ZD6474, a dual
tyrosine kinase inhibitor of EGFR and VEGFR-2, inhibits
MAPK/ERK and AKT/PI3-K and induces apoptosis in breast
cancer cells. Cancer Biol Ther 2010; 9: 592–602.
114. Okamoto K, Okamoto I, Okamoto W, et al. Role
of survivin in EGFR inhibitor induced apoptosis in non-small
cell lung cancers positive for EGFR mutations. Cancer Res
2010; 70: 10402–10.
115. Costa DB, Halmos B, Kumar A, et al. BIM mediates
EGFR tyrosine kinase inhibitor-induced apoptosis in lung can-
cers with oncogenic EGFR mutations. PLoS Med 2007; 4: e315.
116. Gong Y, Somwar R, Politi K, et al. Induction of BIM
is essential for apoptosis triggered by EGFR kinase inhibitors
in mutant EGFR-dependent lung adenocarcinomas. PLoS
Med 2007; 4: e294.
117. Youle RJ, Strasser A. The BCL-2 protein family: op-
posing activities that mediate cell death. Nat Rev Mol Cell
Biol 2008; 9: 47–59.
118. Allocati N, Di Ilio C, De Laurenzi V. p63/p73 in the con-
trol of cell cycle and cell death. Exp Cell Res 2012; 318: 1285–90.
119. Sun Q, Ming L, Thomas SM, et al. PUMA mediates
EGFR tyrosine kinase inhibitor-induced apoptosis in head and
neck cancer cells. Oncogene 2009; 28: 2348–57.
120. Dittmann K, Mayer C, Kehlbach R, Rodemann HP. Ra-
diation-induced caveolin-1 associated EGFR internalization
is linked with nuclear EGFR transport and activation of DNA-
PK. Mol Cancer 2008; 7: 69.
121. Bandyopadhyay D, Mandal M, Adam L, et al. Physi-
cal interaction between epidermal growth factor receptor and
DNA-dependent protein kinase in mammalian cells. J Biol
Chem 1998; 273: 1568–73.
122. Mills CN, Nowsheen S, Bonner JA, et al. Emerging
roles of glycogen synthase kinase 3 in the treatment of brain
tumors. Front Mol Neurosci 2011; 4: 47.
123. Beurel E, Jope RS. The paradoxical pro- and anti-
apoptotic actions of GSK3 in the intrinsic and extrinsic apop-
tosis signaling pathways. Prog Neurobiol 2006; 79: 173–89.
124. Gotschel F, Kern C, Lang S, et al. Inhibition
of GSK3 differentially modulates NF-kB, CREB, AP-1 and
B-catenin signaling in hepatocytes, but fails to promote TNF-
alpha-induced apoptosis. Exp Cell Res 2008; 314: 1351–66.
125. Charvet C, Wissler M, BraunsSchubert P, et al.
Phosphorylation of Tip60 by GSK-3 determines the induction
of PUMA and apoptosis by p53. Mol Cell 2011; 42: 584–96.
126. Watcharasit P, Bijur GN, Song L, et al. Glycogen
synthase kinase-3B (GSK3B) binds to and promotes the ac-
tions of p53. J Biol Chem 2003; 278: 48872–9.
127. Bijur GN, Jope RS. Opposing actions of phosphati-
dylinositol 3’kinase and glycogen synthase kinase 3B in the
regulation of HSF1 activity. J Neurochem 2000; 75: 2401–8.
128. Yang ES, Nowsheen S, Wang T, et al. Glycogen syn-
thase kinase 3B inhibition enhances repair of DNA double-
strand breaks in irradiated hippocampal neurons. Neuro-
Oncology 2011; 13: 459–70.
Copyright © Experimental Oncology, 2012
|