ArticlePDF Available

Abstract and Figures

Knowledge of factors and mechanisms contributing to the inherent radioresistance of pancreatic cancer may improve cancer treatment. Irradiation inhibits glycogen synthase kinase 3beta (GSK3beta) by phosphorylation at serine 9. In turn, release of cytosolic membrane beta-catenin with subsequent nuclear translocation promotes survival. Both GSK3beta and beta-catenin have been implicated in cancer cell proliferation and resistance to death. We investigated pancreatic cancer cell survival after radiation in vitro and in vivo, with a particular focus on the role of the function of the GSK3beta/beta-catenin axis. Lithium chloride, RNAi-medicated silencing of GSK3beta, or the expression of a kinase dead mutant GSK3beta resulted in radioresistance of Panc1 and BxPC3 pancreatic cancer cells. Conversely, ectopic expression of a constitutively active form of GSK3beta resulted in radiosensitization of Panc1 cells. GSK3beta silencing increased radiation-induced beta-catenin target gene expression as measured by studies of AXIN2 and LEF1 transcript levels. Western blot analysis of total and phosphorylated levels of GSK3beta and beta-catenin showed that GSK3beta inhibition resulted in stabilization of beta-catenin. Xenografts of both BxPC3 and Panc1 with targeted silencing of GSK3beta exhibited radioresistance in vivo. Silencing of beta-catenin resulted in radiosensitization, whereas a nondegradable beta-catenin construct induced radioresistance. These data support the hypothesis that GSK3beta modulates the cellular response to radiation in a beta-catenin-dependent mechanism. Further understanding of this pathway may enhance the development of clinical trials combining drugs inhibiting beta-catenin activation with radiation and chemotherapy in locally advanced pancreatic cancer.
Content may be subject to copyright.
GSK3βand β-Catenin Modulate
Radiation Cytotoxicity in
Pancreatic Cancer
1
Richard L. Watson*
,2
, Aaron C. Spalding*
,,2
,
Steven P. Zielske*, Meredith Morgan*,
Alex C. Kim
, Guido T. Bommer
§,¶
,
Hagit Eldar-Finkelman
#
, Thomas Giordano**,
Eric R. Fearon
,§,¶,#
,GaryD.Hammer
,
Theodore S. Lawrence*and Edgar Ben-Josef*
*Department of Radiation Oncology, University of
Michigan Medical School, Ann Arbor, MI, USA;
The Norton
Cancer Institute Radiation Center and Kosair Childrens
Hospital, Louisville, KY, USA;
Department of Internal
MedicineMetabolism, Endocrinology & Diabetes,
University of Michigan Medical School, Ann Arbor, MI, USA;
§
Department of Internal Medicine, University of Michigan
Medical School, Ann Arbor, MI, USA;
Department of
Internal MedicineMolecular Medicine and Genetics,
University of Michigan Medical School, Ann Arbor, MI, USA;
#
Department of Human Genetics, Tel Aviv University, Ramat
Aviv, Israel; **Department of Pathology, University of
Michigan Medical School, Ann Arbor, MI, USA
Abstract
BACKGROUND: Knowledge of factors and mechanisms contributing to the inherent radioresistance of pancreatic cancer may
improve cancer treatment. Irradiation inhibits glycogen synthase kinase 3β(GSK3β) by phosphorylation at serine 9. In turn, release
of cytosolic membrane β-catenin with subsequent nuclear translocation promotes survival. Both GSK3βand β-catenin have been
implicated in cancer cell proliferation and resistance to death. METHODS: We investigated pancreatic cancer cell survival after
radiation in vitro and in vivo, with a particular focus on the role of the function of the GSK3β/β-catenin axis. RESULTS: Lithium
chloride, RNAi-medicated silencing of GSK3β, or the expression of a kinase dead mutant GSK3βresulted in radioresistance of
Panc1 and BxPC3 pancreatic cancer cells. Conversely, ectopic expression of a constitutively active form of GSK3βresulted in radio-
sensitization of Panc1 cells. GSK3βsilencing increased radiation-induced β-catenin target gene expression as measured by studies
of AXIN2 and LEF1 transcript levels. Western blot analysis of total and phosphorylated levels of GSK3βand β-catenin showed that
GSK3βinhibition resulted in stabilization of β-catenin. Xenografts of both BxPC3 and Panc1 with targeted silencing of GSK3β
exhibited radioresistance in vivo.Silencingofβ-catenin resulted in radiosensitization, whereas a nondegradable β-catenin construct
induced radioresistance. CONCLUSIONS: These data support the hypothesis that GSK3βmodulates the cellular response to
radiation in a β-catenindependent mechanism. Further understanding of this pathway may enhance the development of clinical
trials combining drugs inhibiting β-catenin activation with radiation and chemotherapy in locally advanced pancreatic cancer.
Neoplasia (2010) 12, 357365
Address all correspondence to: Aaron C. Spalding, MD, PhD, The Norton Cancer Institute Radiation Center, 4001 Dutchmans Ln, Louisville, KY 40207.
E-mail: acspalding1@gmail.com
1
These studies were supported by an American Society of Therapeutic Radiology and Oncology Resident Seed Grant (A.S.), and National Institutes of Health R03 CA127050-01
(E.B.J.). These studies were conducted in partial fulfillment for the requirements (R.W.) for graduationwith Honors, College of Literature, Science, and the Arts, from the University of
Michigan. A.Spalding has been designated a B. Leonard Holman Pathway Fellow by the American Board of Radiology, and this work was presented in part at the 2008Gastrointestinal
Malignancies Symposium.
2
These authors contributed equally to this work.
Received 30 December 2009; Revised 15 February 2010; Accepted 16 February 2010
Copyright © 2010 Neoplasia Press, Inc. All rights reserved 1522-8002/10/$25.00
DOI 10.1593/neo.92112
www.neoplasia.com
Volume 12 Number 5 May 2010 pp. 357365 357
Introduction
Roughly 37,000 US patients are diagnosed annually with pancreatic
cancer, with the annual death rate from pancreatic cancer approaching
its incidence. Aggressive treatment of locally advanced pancreatic can-
cer patients with highly conformal radiation and chemotherapy pro-
duces a moderate beneficial effect on local control of the cancer, albeit
with possible increases in toxicity and a minor impact on patient sur-
vival. Unfortunately, the addition of agents targeting either Ras [1]
(zarnestra, tipifarnib), matrix metalloproteases [2] (marimistat), or
EGFR [3] (erlotinib) has not dramatically increased overall survival
either. Thus, elucidation of the mechanisms underlying pancreatic
cancer radioresistance may lead to improved targeted therapies, which
may improve clinical outcomes.
Depending on the context and cell type under study, glycogen
synthase kinase 3β(GSK3β) can promote cell survival or apoptosis
after cytotoxic insults [4]. Active GSK3βcan induce mitochondrial
release of cytochrome c, leading to the activation of the intrinsic apop-
tosis pathway [5]. Active GSK3βphosphorylates β-catenin, a tran-
scription factor involved in survival and proliferation, at serine 33,
which primes β-catenin for ubiquitination and subsequent proteosome-
mediated degradation [6]. Survival signals such as binding of Wnt ligands
or growth factors to their respective receptors leads to inhibition of GSK3β
through phosphorylation at serine 9. Inhibition of GSK3βthen leads to
the stabilization of β-catenin resulting in nuclear translocation and hetero-
dimerization of β-catenin with T-cell factor family members thus pro-
moting transcription of β-catenin target genes and cell survival [7].
We previously conducted a study that showed that the protein kinase
Cβinhibitor enzastaurin prevents radiation-induced phosphorylation
of protein kinase Cβand leads to radiosensitization through persistent
activation of GSK3βin pancreatic cancer cells. In addition, we demon-
strated that radiation induces phosphorylation of GSK3βat serine 9, a
site known to inhibit GSK3βactivity [8,9]. Our current study expands
on that observation to test the hypothesis that GSK3βmediates the
radiation resistance of pancreatic cancer by suppression of β-catenin.
Our findings establish that the GSK3β/β-catenin pathway modulates
radiation resistance of pancreatic cancer and suggest potential targets
to increase efficacy of radiation therapy in pancreatic cancer.
Materials and Methods
Cell Line Generation
Panc-1 and BxPc3 human pancreatic cancer cells were obtained from
the American Type Culture Collection (Manassas, VA) and were main-
tained according to standard tissue culture conditions. We generated
lentivirus particles for transduction of shRNA to silence GSK3βor
β-catenin. Mission short hairpin RNA (shRNA) lentiviral plasmids
(Sigma, St Louis, MO) contain a U6 promoter transcribing nonspecific
(NS), GSK3β,orβ-catenin shRNA along with a puromyocin resistance
gene for selection. We collected supernatants after cotransfection of
HEK293T cells with mission shRNA and packaging plasmids [10].
BxPC3 or Panc1 cells were then transduced with NS, GSK3β,orβ-
catenin shRNA particles and selected under 2 μg/ml puromycin.
We used lentivirus transduction to express kinase inactive GSK3β
[11]. We subcloned the GSK3β
KK(85,86)MA
KKMA insert using KpnI
(5end) and XbaI(3end) from the pCMV4 vector to pLentilox-
IRES-GFP. The resulting pLLGSK3β
KK(85,86)MA-IGFP
lentiviral
plasmid uses a CMV promoter to drive expression of a single messenger
RNA with both insert and GFP for the identification of transduced
cells. We collected supernatants after cotransfection of HEK293T cells
with empty vector control or pLLGSK3β
KK(85,86)MA-IGFP
and pack-
aging plasmids. BxPC3 or Panc1 cells were thentransduced with empty
vector control or pLLGSK3β
KK(85,86)MA-IGFP
particles and analyzed
by flow cytometry. Only cell populations with more than 90% GFP
expression were used.
We used stable transfection to generate cells expressing nondegrad-
able β-catenin [12]. We subcloned the β-catenin
S33Y-FLAG
insert using
BamHI (5end) and XbaI(3end) from the pCMV4 vector to pcDNA3.1
(+). Cells were transfected with 1 μg of empty vector control or pcDNA3.1
(+)β-catenin
S33Y-FLAG
and then selected with G418. Stable pooled pop-
ulations of individual clones were verified by Western blot analysis
for FLAG.
Colony Formation Assays
After irradiation, cells were trypsinized, counted, and plated at pre-
determined clonal densities. Two weeks later, cells were fixed with a
methanol/acetic acid mixture (7:1) and stained with crystal violet.
Colony counting was done using an automated counter. Data were
then analyzed by determining the surviving fraction at each dose of
radiation. Cell survival curves were fit using the linear-quadratic
equation. Radiation sensitivity is expressed in mean inactivation dose
(MID), which represents the area under the cell survival curve [13].
MID was calculated for control and each experimental manipulation,
and the enhancement ratio was calculated as the MID in the control
curve divided by the MID in the experimental curve.
Reverse TranscriptionPolymerase Chain Reaction
A Qiagen RNeasy RNA extraction kit was used to collect RNA for
reverse transcriptionpolymerase chain reaction (RT-PCR). RT-PCR
was performed in duplicate using a Qiagen Quantitect Syber Green RT-
PCR kit on GAPDH[14], AXIN2 [15], and Lef1 [16]using previously
published primer sequences. C
T
values for each unknown were com-
pared with a standard curve made of serially diluted RNA from wild-
type BxPC3 and Panc1 cells in logarithmic phase growth. AXIN2 and
Lef1 values were normalized to the level of GAPDH in each sample.
Antibodies and Immunoblot Analysis
Antibodies to GSK3β(Cell Signaling, Danvers, MA), phospho-
Ser9 GSK3β(Cell Signaling), β-catenin (Cell Signaling), phospho-
Ser33 β-catenin (Cell Signaling), and β-actin (Sigma) were used at
dilutions per the manufacturer. Cell lysate production with RIPA buffer
and immunoblot analysis were performed using detailed protocols from
Cell Signaling. Xenograph samples were taken after treatment and
frozen in a dry ice bath. A mortar and pestle was then used to grind
the xenograph samples. β-Actin was used as a control to show that total
protein quantities were equal among the groups. Each Western blot was
performed three independent times from unique lysates; representative
films are shown in Figures 1A,2A,and3A.
Xenografts
Animals used in this study were maintained in facilities approved by
the American Association for Accreditation of Laboratory Animal Care
in accordance with current regulations and standards of the United
States Department of Agriculture and Department of Health and Hu-
man Services. Under an institutionally approved protocol, 4-week-old
female athymic nude mice were implanted with 5 × 10
7
BxPC3 or
358 Wnt Pathway Modulates Radiosensitivity Watson et al. Neoplasia Vol. 12, No. 5, 2010
Panc1 cells subcutaneously. Tumor volume (TV) was calculated accord-
ing to the following equation: TV = Π/6 × a×b
2
,whereaand bare the
longer and shorter dimensions of the tumor, respectively. When the
average tumor volume achieved 100 mm
3
,micewererandomizedto
treatment groups.
Irradiation
Cells or xenografts were irradiated using a Phillips 250 orthovoltage
unit at approximately 2 Gy/min for cells or 1.4 Gy/min for mice in the
Irradiation Core of the University of Michigan Cancer Center. Dosim-
etry is carried out using an ionization chamber connected to an elec-
trometer system, which is directly traceable to a National Institute of
Standards and Technology calibration. Mice were anesthetized with a
mixture of ketamine 60 mg/kg and xylazine 3 mg/kg and positioned
such that the apex of each flank tumor was at the center of a 2.4-cm
aperture in the secondary collimator and irradiated with the rest of
the mouse being shielded from radiation.
Statistical Analysis
The clonogenic assays were conducted on three independent occa-
sions in triplicate. Mean and SD from the three independent experi-
ments are displayed in Figures 1A,2,Band C, and 6. A two-tailed
t-test was used to analyze differences between mean values of in vitro
assays, with αvalues less than 0.05 considered significant. The radia-
tion enhancement factor (REF) was calculated as previously described
[17], with numbers less than 1 indicating radioprotection and numbers
greater than 1 indicating radiosensitization.
The RT-PCR data in Figure 5Arepresent the mean and SD values
of three independent experiments performed in triplicate after irradi-
ation. A two-tailed t-test was used to analyze differences between
mean values at each time point, with αvalues less than 0.05 consid-
ered significant.
The in vivo experiments were designed with a power of 80% to
detect a 20% difference in tumor growth delay between the control
versus irradiated tumors, resulting in a sample size of 10 tumors per
group. Tumor volumes are plotted relative to the pretreatment volume
in Figure 3, Band C. A two-tailed t-test was used to analyze differences
between mean values at each measurement, with αvalues less than 0.05
considered significant.
Results
GSK3βSignaling Modulates Radiation Resistance In Vitro
Inhibition of GSK3βby phosphorylation at Ser9 has been pre-
viously observed after irradiation of pancreatic cancer cells [17], po-
tentially underscoring their observed radioresistance. We examined
the phenotypic effects of GSK3βmodulation on radiation response
in vitro. Previous studies have shown that lithium chloride (LiCl) is a
pharmacological inhibitor of GSK3β, with inhibition correlating with
increased phosphorylation of GSK3βat serine 9 [18]. We determined
the concentration of LiCl needed to increase GSK3βphosphorylation
and found that 30 mM was associated with phosphorylation in BxPC3
and Panc1 cells (Figure 1A). Inhibition of GSK3βby a 6-hour exposure
to LiCl before radiation led to an increase in survival in response to
radiation in both BxPC3 and Panc1 cells (REFs: 0.78 and 0.79, respec-
tively, P< .05; Figure 1B). Because pharmacologic inhibition such as
LiCl treatment may have unintended off-target effects, we also used
genetic approaches to test our hypothesis that GSK3βinhibition pro-
motes radioresistance in pancreatic cancer.
To further characterize the role of GSK3βin radiation survival, we
transduced BxPC3 and Panc1 cells with a lentivirus construct expressing
Figure 1. (A) BxPC3 and Panc1 cells were treated with LiCl for 6 hours, and Western blot analysis for total and phosphorylated GSK3β
was performed. (B) Clonogenic survival of control () or LiCl-pretreated () BxPC3 and Panc1 cells. *P0.05. Error bars are SD of three
independent experiments performed in triplicate and are smaller than the symbols at some data points.
Neoplasia Vol. 12, No. 5, 2010 Wnt Pathway Modulates Radiosensitivity Watson et al. 359
an shRNA designed to inhibit GSK3βexpression. We generated poly-
clonal populations of BxPC3 and Panc1 cells expressing the shRNA
construct and then determined the effect of GSK3βknockdown on
survival after radiation. Radiation delivered to pancreatic cancer cells ex-
pressing an NS shRNA construct resulted in serine 9 phosphorylation,
similar to wild-type cells. Ser9 GSK3βphosphorylation was increased
with a peak at 1 hour after a 2-Gy radiation (Figure 2A). Silencing of
GSK3βprevented radiation-induced GSK3βserine phosphorylation in
response to a 2-Gy radiation and produced radioresistance (REFs of
0.82 in BxPC3 and 0.60 in Panc1, P< .05; Figure 2B) similarly to
pharmacological inhibition of GSK3β. These data indicate that inhibi-
tion of GSK3βpromotes survival in response to irradiation.
As a second genetic approach, we generated polyclonal populations
of cells stably expressing GSK3β
KK(85,86)MA
, which has an inactive
substrate phosphorylation domain. Expression of the kinase dead
GSK3β
KK(85,86)MA
inhibited radiation cytotoxicity compared with
cells transduced with empty vector (REFs of 0.76 in BxPC3 and
0.70 in Panc1; Figure 2C). These data show that radiation resistance
of pancreatic cancer cells in vitro can be modulated through manipu-
lation of GSK3β.
GSK3βSignaling Modulates Radiation Resistance In Vivo
After observing the radioprotective effect of GSK3βinhibition in vitro,
we studied the consequences of GSK3βinhibition in vivo. Polyclonal
populations of BxPC3 and Panc1 cells expressing GSK3βshRNA
maintained knockdown of GSK3 10 weeks after subcutaneous implan-
tation, whereas those with NS shRNA retained the expression of
GSK3β(Figure 3A). Control BxPC3 xenografts expressing NS shRNA
exhibited a 26-day growth delay with ten 2-Gy fractions (Figure 3B)
and a 61-day growth delay with ten 3-Gy fractions (Figure 3C). Si-
lencing of GSK3βleads to shortened growth delay from both the 2-Gy
(17 days, REF of 0.65) and the 3-Gy (25 days, REF of 0.40) treatment
courses (P< .05 for both). Similarly, control Panc1 xenografts expressing
NS shRNA exhibited a 24-day growth delay with five 2-Gy fractions
Figure 2. (A) BxPC3 and Panc1 cells expressing NS or GSK3βshRNA were treated with 2 Gy, and Western blot analysis for total and
phosphorylated GSK3βwas performed. The blots were confirmed in at least three independent experiments. (B) Clonogenic survival of
NS () or GSK3βshRNA () BxPC3 and Panc1 cells. (C) Clonogenic survival of empty vector control () or GSK3β
KK(85,86)MA
() BxPC3 and
Panc1 cells. *P0.05. Error bars are SD of three independent experiments performed in triplicate and are smaller than the symbols at
some data points.
360 Wnt Pathway Modulates Radiosensitivity Watson et al. Neoplasia Vol. 12, No. 5, 2010
(Figure 3D) and a 43-day growth delay with five 3-Gy fractions
(Figure 3E). Silencing of GSK3βleads to shortened growth delay from
the 2-Gy (16 days, REF of 0.64) and the 3-Gy (23 days, REF of 0.53)
treatment courses (P< .05 for both). Thus, tumors without GSK3βwere
less sensitive to radiation, similar to the results from the in vitro clono-
genic assays.
To determine changes in vivo induced by radiation, a separate ex-
periment with identical arms was conducted; tumors were collected
immediately after the last fraction of radiation, and staining for hema-
toxylin and eosin (H&E) and Ki67 was performed (Figure 4). H&E
staining revealed that knock down of GSK3βresulted in increased
nuclear-to-cytoplasmic ratio and decreased production of mucin, sug-
gesting cellular dedifferentiation, a phenotype consistent with β-catenin
activation. Radiation reduced the proliferation index from 95% to 30%
in NS shRNA tumors, whereas GSK3βshRNA tumors had a less pro-
nounced reduction from 98% to 65% (Figure 4B).
The decreased tumor growth delay, increased tumor cell density,
and increased proliferation in the GSK3βknockdown tumors all cor-
relate with the in vitro observation that inhibition of GSK3βpro-
motes radiation resistance.
Modulation of the Radiation Response through β-Catenin
We hypothesized that GSK3βmodulates the radiation response
through a β-catenindependent gene transcription. We first tested
whether radiation induced β-catenin activity. In BxPC3 and Panc1 cells
expressing NS shRNA, radiation induced the transcription of Lef1 and
Axin2 [19], two well-characterized β-catenin target genes, in a time-
dependent manner as measured by quantitative RT-PCR. Targeted
Figure 3. (A) Xenografts from BxPC3 and Panc1 cells expressing NS or GSK3βshRNA were analyzed for expression of GSK3β. The blots
were confirmed in at least three independent experiments. BxPC3 NS shRNA and GSK3βknockdown xenografts were treated with ten
2-Gy fractions (B) or ten 3-Gy fractions (C) and were compared with unirradiated controls. Panc1 NS shRNA and GSK3βknockdown
xenografts were treated with five 2-Gy fractions (D) or five 3-Gy fractions (E) and were compared with unirradiated controls. *P
0.05 between the NS versus GSK3βknockdown. Error bars are SEM of the 10 tumors per treatment arm. The dashed line indicates
a four-fold increase in tumor size, used to determine the enhancement ratio.
Neoplasia Vol. 12, No. 5, 2010 Wnt Pathway Modulates Radiosensitivity Watson et al. 361
silencing of GSK3βresulted in both higher basal and radiation-induced
levels of Lef1 or Axin2 gene transcription (Figure 5A). Because we ob-
served that radiation affects β-catenin transcriptional activity in vitro
through GSK3β, we hypothesized that radiation would have similar
effects in vivo (Figure 5B). Before irradiation, β-catenin localized to the
cytosolic membrane. After radiation of xenografts, β-catenin translocated
to the nucleus, suggesting induction of β-catenin signaling. Radiation
induction of β-catenin nuclear translocation correlates with the obser-
vation in vitro that GSK3βphosphorylation modulates β-catenin
dependent gene transcription.
If GSK3βmodulates pancreatic cancer cell response to radiation
through β-catenin, then modulation of β-catenin activity may influ-
ence cell survival after radiation. Therefore, we transduced BxPC3
and Panc1 cells with lentivirus encoding shRNA targeting β-catenin.
Compared with cells transduced with NS shRNA, cells with silenced
β-catenin were more sensitive to radiation as shown by reduced clono-
genic survival (REFs of 1.4 in BxPC3 and 1.25 in Panc1; Figure 6).
On the basis of these experiments, constitutive activation of β-
catenin would be predicted to render pancreatic cancer cells resistant
to radiation. Therefore, a β-catenin
S33Y-FLAG
vector was used to create
cells expressing constituently active β-catenin. The S33Y mutation
prevents GSK3β-mediated phosphorylation at Ser33, thus preventing
ubiquitination and subsequent degradation [20]. Cells expressing con-
stituently active β-catenin
S33Y
showed increased clonogenic survival
(REF for Panc1 cells was 0.8; Figure 6C). The effects of nondegradable
β-catenin
S33Y
were analogous to those resulting from GSK3βinhibition
or silencing because both showed an increased resistance to radiation
together with an increased level of β-catenin activity. Thus, increased
β-catenin activity results in greater radiation resistance of pancreatic
cancer cells, whereas loss of β-catenin through RNAi-mediated silenc-
ing results in increased radiation sensitivity.
Discussion
In this study, we found that inhibition of GSK3β, by either genetic
or pharmacological methods, induces radiation resistance of pancre-
atic cancer cells in vitro, reduces the duration of radiation-induced
tumor growth delay, and leads to increased cell proliferation in vivo.
Similarly, the expression of a constituently active β-catenin in pancreatic
cancer cells increases the resistance to radiation. Our results reinforce
and expand on previous studies of radiation effects on GSK3β.
We have previously demonstrated that radiation induces phos-
phorylation of GSK3βat Ser9, an event known to inhibit GSK3β
kinase activity [17], and that abrogation of this phosphorylation re-
sulted in radiosensitization. Radiation was also shown to inhibit
GSK3βactivity in SAOS-2 cells [21], although the phenotypic con-
sequences in sensitivity to radiation were not investigated. Irradiation
of A549 cells induced phosphorylation of GSK3βat Ser9, and this
effect was reduced when cells were plated on fibronectin [22]. The
authors suggested that GSK3βis involved in the interaction of cells
with the extracellular matrix after radiation to modulate the cytotox-
icity of radiation. These studies implicate GSK3βas a mediator of
radiation sensitivity.
We hypothesized that GSK3βmodulates radiation cytotoxicity, at
least in part, through its downstream effector β-catenin. Herein, we
show that radiation induces the transcription of Lef1 and Axin2, two
well-characterized β-catenin target genes, and targeted silencing of
GSK3βresults in both higher basal and radiation-induced levels of
Figure 4. Xenografts from BxPC3 and Panc1 cells expressing NS or GSK3βshRNA were analyzed by H&E (A). Black arrows indicate
glandular structures present in the NS shRNA xenografts, which are absent in GSK3βshRNA xenografts. Panc1 xenografts with and
without radiation were analyzed for proliferation by Ki67 (B). Original magnification, ×400.
362 Wnt Pathway Modulates Radiosensitivity Watson et al. Neoplasia Vol. 12, No. 5, 2010
Lef1 or Axin2 gene transcription. Furthermore, we show that radia-
tion induces translocation of cytosolic β-catenin to the nucleus in
Panc1 and BxPC3 xenographs, an observation consistent with the
in vitro induction of transcription of Lef1 and Axin2. Finally, we show
that cells with silenced β-catenin are more sensitive, whereas cells expres-
sing constituently active β-catenin
S33Y
are more resistant to radia-
tion. β-Catenin has been shown to prevent epithelial cell death
after radiation or anoikis [23]. These findings suggest that β-catenin
is involved in determining clonogenic survival of pancreatic cancer
cells after irradiation.
Our studies potentially explain the relationship between Wnt sig-
naling and radiation cytotoxicity in other tumor sites. Activation of the
Wnt signaling pathway resulted in β-catenin cytoplasmic accumulation
with translocation to the nucleus in head and neck cancer cell lines
expressing COX-2 [24]. In turn, up-regulation of Ku expression leads
to increased radioresistance. Blocking COX-2 signaling led to the sup-
pression of β-catenininduced Ku expression and consequent radiation
sensitivity. Others have suggested that the radioresistance observed clin-
ically in glioblastoma depends in part on the activation of β-catenin in
putative cancer stem cells [25]. In a mouse model of breast develop-
ment, radiation selectively enriched for mammary epithelial progenitors
isolated from transgenic mice with activated Wnt/β-catenin signaling
but not for background-matched controls [26]. We conversely showed
that suppressing β-catenin using shRNA correlated with an increase in
radiation sensitivity.
Our data reinforce observations from others that GSK3βinhibition
protects normal tissue from radiation toxicity. Radiation-induced
GSK3βactivation results in mouse hippocampal neuronal apoptosis
and subsequent neurocognitive decline. The expression of kinase-inactive
GSK3βor pharmacologic inhibition before irradiation significantly
attenuated radiation-induced apoptosis in hippocampal neurons, lead-
ing to improved cognitive function in irradiated animals [27]. Mice
treated with lithium chloride, a known GSK3βinhibitor, had decreased
neurocognitive impairment after irradiation as well [28]. Akt serves to
inhibit GSK3βafter irradiation in normal vascular endothelium [29],
and administration of recombinant growth factors known to activate
Figure 5. (A) Time course of Lef1 and Axin2 levels in NS () or GSK3βshRNA () BxPC3 and Panc1 cells subjected to 2-Gy radiation.
Mean of three experiments with SDs, *P0.05. (B) BxPC3 or Panc1 xenografts were treated with 2-Gy radiation and were stained for
β-catenin (green) and propidium idodide (red). Yellow indicates overlap of red and green, consistent with nuclear β-catenin.
Neoplasia Vol. 12, No. 5, 2010 Wnt Pathway Modulates Radiosensitivity Watson et al. 363
Akt may prevent normal tissue toxicity. However, any pharmacologic
strategy to reduce normal tissue damage must be carefully weighed
against the risk of tumor protection.
Our results are consistent with radioprotection caused by active
β-catenin. A reporter mouse model demonstrated that ionizing radia-
tion activates β-cateninmediated, T-cell factordependent transcrip-
tion both in vitro and in vivo. Mouse-derived fibroblast cultures
expressing stabilized β-catenin formed more colony-forming units than
wild-type or null cells after irradiation. β-Catenin levels in irradiated
wounds correlated with tensile strength of the wound, and lithium
chloride treatment also increased β-catenin levels and increased wound
strength [30]. The newly identified R-Spondin1 augments canonical
Wnt/β-catenin signaling and causes nuclear translocation of β-catenin.
R-Spondin1 reduced mucosal ulceration after whole-body or snout-
only irradiation in mouse models [31]. Therefore, in normal cells, GSK3β
inhibition with β-catenin activation may be a radioprotective mecha-
nism. Pancreatic cancer cells potentially invoke a similar mechanism
to evade the cytotoxic effects of radiation.
Our results help explain an apparent contradiction present in the
literature regarding pancreatic cancer and β-catenin. Mutations in
APC leading to β-catenin nuclear accumulation have been well char-
acterized to play a role in colon cancer. However, mutations in APC
[32] or β-catenin [33] have not been found in pancreatic cancer. The
published literature suggests that constitutive activation of β-catenin
does not play a role in pancreatic cancer development. In fact, our
results also demonstrate similar findings, as unirradiated tumors
lacked nuclear β-catenin, and we did not find evidence of increased
β-catenin target gene expression without irradiation. However, we
did find that pancreatic cancer cells activate β-catenin in response
to radiation to promote survival. Our results may therefore explain
in part the clinically observed radioresistance of pancreatic cancer;
specifically, it may not be the basal level of β-catenin but rather the in-
duction of β-catenin by radiation that promotes pancreatic cancer cell
survival. We plan to test this hypothesis by immunoflorescence of pan-
creatic cancer specimens treated with neoadjuvant radiation to deter-
mine whether activation of β-catenin occurs in patients.
The implications of this work identify a link between radiation
and a pathway central to tumor growth, invasion, and metastasis
of pancreatic cancer. By further discovering the molecular signaling
cascades upstream and downstream of GSK3β, we will also start to
gain insight into the potential interactions with other signaling path-
ways that are known to be involved in radioresistance. Further under-
standing of this pathway will also help develop clinical trials combining
drugs inhibiting β-catenin activation with radiation and cytotoxic
agents in locally advanced pancreatic cancer.
Acknowledgment
The authors thank Steven Kronenberg for his graphical expertise.
References
[1] Macdonald JS, McCoy S, Whitehead RP, Iqbal S, Wade JL III, Giguere JK, and
Abbruzzese JL (2005). A phase II study of farnesyl transferase inhibitor
R115777 in pancreatic cancer: a Southwest oncology group (SWOG 9924)
study. Invest New Drugs 23, 485487.
[2] Jones L, Ghaneh P, Humphreys M, and Neoptolemos JP (1999). The matrix
metalloproteinases and their inhibitors in the treatment of pancreatic cancer.
Ann N Y Acad Sci 880, 288307.
[3] Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ,
Murawa P, Walde D, Wolff RA, et al. (2007). Erlotinib plus gemcitabine com-
pared with gemcitabine alone in patients with advanced pancreatic cancer: a
phase III trial of the National Cancer Institute of Canada Clinical Trials Group.
J Clin Oncol 25, 19601966.
[4] Beurel E and Jope RS (2006). The paradoxical pro- and anti-apoptotic actions of
GSK3 in the intrinsic and extrinsic apoptosis signaling pathways. Prog Neurobiol
79, 173189.
[5] Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, Ziman BD,
Wang S, Ytrehus K, Antos CL, et al. (2004). Glycogen synthase kinase-3βmedi-
ates convergence of protection signaling to inhibit the mitochondrial permeabil-
ity transition pore. J Clin Invest 113, 15351549.
[6] KorinekV,BarkerN,MorinPJ,vanWichenD,deWegerR,KinzlerKW,
Vogelstein B, and Clevers H (1997). Constitutive transcriptional activation by a
β-cateninTcf complex in APC/colon carcinoma. Science 275,17841787.
[7] Chan TA, Wang Z, Dang LH, Vogelstein B, and Kinzler KW (2002). Targeted
inactivation of CTNNB1 reveals unexpected effects of β-catenin mutation. Proc
Natl Acad Sci USA 99, 82658270.
Figure 6. Clonogenic survival of NS ()orβ-catenin shRNA ()
BxPC3 (A) and Panc1 (B) cells. Clonogenic survival of empty vector
control ()orβ-catenin
S33YFLAG
() Panc1 cells (C). Error bars are
SD of three independent experiments performed in triplicate and
are smaller than the symbols at some data points.
364 Wnt Pathway Modulates Radiosensitivity Watson et al. Neoplasia Vol. 12, No. 5, 2010
[8] Sutherland C, Leighton IA, and Cohen P (1993). Inactivation of glycogen
synthase kinase-3 βby phosphorylation: new kinase connections in insulin
and growth-factor signalling. Biochem J 296(pt 1), 1519.
[9] Welsh GI and Proud CG (1993). Glycogen synthase kinase-3 is rapidly inacti-
vated in response to insulin and phosphorylates eukaryotic initiation factor eIF-
2B. Biochem J 294(pt 3), 625629.
[10] Zufferey R, Dull T, Mandel RJ, Bukovsky A, Quiroz D, Naldini L, and Trono D
(1998). Self-inactivating lentivirus vector for safe and efficient in vivo gene delivery.
JVirol72,98739880.
[11] Eldar-FinkelmanH, Argast GM, Foord O, Fischer EH,and Krebs EG(1996).Expres-
sion and characterization of glycogen synthase kinase-3 mutants and their effect on
glycogen synthase activity in intact cells. Proc Natl Acad Sci USA 93, 1022810233.
[12] Kolligs FT, Hu G, Dang CV, and Fearon ER (1999). Neoplastic transformation
of RK3E by mutant β-catenin requires deregulation of Tcf/Lef transcription but
not activation of c-myc expression. Mol Cell Biol 19, 56965706.
[13] Fertil B, Dertinger H, Courdi A, and Malaise EP (1984). Mean inactivation
dose: a useful concept for intercomparison of human cell survival curves. Radiat
Res 99,7384.
[14] Simpson DA, Feeney S, Boyle C, and Stitt AW (2000). Retinal VEGF mRNA
measured by SYBR green I fluorescence: a versatile approach to quantitative
PCR. Mol Vis 6, 178183.
[15] Koinuma K, Yamashita Y, Liu W, Hatanaka H, Kurashina K, Wada T, Takada S,
Kaneda R, Choi YL, Fujiwara SI, et al. (2006). Epigenetic silencing of AXIN2
in colorectal carcinoma with microsatellite instability. Oncogene 25, 139146.
[16] Kahler RA, Galindo M, Lian J, Stein GS, van Wijnen AJ, and Westendorf JJ
(2006). Lymphocyte enhancer-binding factor 1 (Lef1) inhibits terminal differ-
entiation of osteoblasts. J Cell Biochem 97, 969983.
[17] Spalding AC, Watson R, Davis ME, Kim AC, Lawrence TS, and Ben-Josef E
(2007). Inhibition of protein kinase C{beta} by enzastaurin enhances radiation
cytotoxicity in pancreatic cancer. Clin Cancer Res 13, 68276833.
[18] De Sarno P, Li X, and Jope RS (2002). Regulation of Akt and glycogen synthase
kinase-3 βphosphorylation by sodium valproate and lithium. Neuropharmacology
43,11581164.
[19] Leung JY, Kolligs FT, Wu R, Zhai Y, Kuick R, Hanash S, Cho KR, and Fearon ER
(2002). Activation of AXIN2 expression by β-cateninT cell factor. A feedback
repressor pathway regulating Wnt signaling. JBiolChem277,2165721665.
[20] Sun Y, Kolligs FT, Hottiger MO, Mosavin R, Fearon ER, and Nabel GJ (2000).
Regulation of β-catenin transformation by the p300 transcriptional coactivator.
Proc Natl Acad Sci USA 97, 1261312618.
[21] Turenne GA and Price BD (2001). Glycogen synthase kinase3 βphosphorylates
serine 33 of p53 and activates p53s transcriptional activity. BMC Cell Biol 2, 12.
[22] Cordes N, Beinke C, Plasswilm L, and van Beuningen D (2004). Irradiation
and various cytotoxic drugs enhance tyrosine phosphorylation and β(1)-integrin
clustering in human A549 lung cancer cells in a substratum-dependent manner
in vitro.Strahlenther Onkol 180, 157164.
[23] Orford K, Orford CC, and Byers SW (1999). Exogenous expression of β-catenin
regulates contact inhibition, anchorage-independent growth, anoikis, and
radiation-induced cell cycle arrest. J Cell Biol 146,855868.
[24] Chang HW, Roh JL, Jeong EJ, Lee SW, Kim SW, Choi SH, Park SK, and Kim SY
(2008). Wnt signaling controls radiosensitivity via cyclooxygenase-2mediated Ku
expression in head and neck cancer. Int J Cancer 122,100107.
[25] Rich JN (2007). Cancer stem cells in radiation resistance. Cancer Res 67,
89808984.
[26] Woodward WA, Chen MS, Behbod F, Alfaro MP, Buchholz TA, and Rosen JM
(2007). WNT/β-catenin mediates radiation resistance of mouse mammary pro-
genitor cells. Proc Natl Acad Sci USA 104, 618623.
[27] Thotala DK, Hallahan DE, and Yazlovitskaya EM (2008). Inhibition of glyco-
gen synthase kinase 3 βattenuates neurocognitive dysfunction resulting from
cranial irradiation. Cancer Res 68, 58595868.
[28] Yazlovitskaya EM, Edwards E, Thotala D, Fu A, Osusky KL, Whetsell WO Jr,
Boone B, Shinohara ET, and Hallahan DE (2006). Lithium treatment pre-
vents neurocognitive deficit resulting from cranial irradiation. Cancer Res 66,
1117911186.
[29] Tan J, Geng L, Yazlovitskaya EM, and Hallahan DE (2006). Protein kinase B/
Aktdependent phosphorylation of glycogen synthase kinase-3βin irradiated
vascular endothelium. Cancer Res 66, 23202327.
[30] Gurung A, Uddin F, Hill RP, Ferguson PC, and Alman BA (2009). β-Catenin is a
mediator of the response of fibroblasts to irradiation. Am J Pathol 174,248255.
[31] Zhao J, Kim KA, De Vera J, Palencia S, Wagle M, and Abo A (2009). R-Spondin1
protects mice from chemotherapy or radiation-induced oral mucositis through the
canonical Wnt/β-catenin pathway. Proc Natl Acad Sci USA 106,23312336.
[32] Caca K, Kolligs FT, Ji X, Hayes M, Qian J, Yahanda A, Rimm DL, Costa J, and
Fearon ER (1999). β- and γ-catenin mutations, but not E-cadherin inactivation,
underlie T-cell factor/lymphoid enhancer factor transcriptional deregulation in
gastric and pancreatic cancer. Cell Growth Differ 10, 369376.
[33] Gerdes B, Ramaswamy A, Simon B, Pietsch T, Bastian D, Kersting M, Moll R,
and Bartsch D (1999). Analysis of β-catenin gene mutations in pancreatic tu-
mors. Digestion 60, 544548.
Neoplasia Vol. 12, No. 5, 2010 Wnt Pathway Modulates Radiosensitivity Watson et al. 365
... The accumulation of DNA damage, improper repair of DNA breaks, and failure to remove cells with damaged DNA have been shown to contribute to oncogenesis (Bartek et al., 2007;Bartkova et al., 2005;Gorgoulis et al., 2005). This is especially true in adult stem cells, which accumulate cancer-driving mutations with age (Bartek et al., 2007;Liu et al., 2014;Sperka et al., 2012;Welch et al., 2012;Xie et al., 2014). Thus, insights into the sensitivity of cells to DNA damage have direct implications in understanding the molecular basis of cellular transformation as well as DNA damage-targeted chemotherapy. ...
... This is in agreement with numerous studies showing that gain of function mutations in β-catenin signaling and loss of α-catenin regulation are prevalent in cancer (Aaltomaa et al., 1999;Anttila et al., 1998;Clevers and Nusse, 2012;Gofuku et al., 1999;Lifschitz-Mercer et al., 2001;Matsui et al., 1994;Nakopoulou et al., 2002;Polakis, 2000;Richmond et al., 1997;Rimm et al., 1995;Shiozaki et al., 1994;Silvis et al., 2011;Tanaka et al., 2003;van Oort et al., 2007;Yang et al., 2006). This additional level of regulation by α-catenin may help explain why WNT stimulation has been reported to decrease the sensitivity of cells to DNA damage despite increased nuclear βcatenin levels (Chandra et al., 2015;Chen et al., 2007;Jun et al., 2016;Woodward et al., 2007), and why different experimental systems have had confounding results (Chevillard-Briet et al., 2014;Orford et al., 1999;Tao et al., 2015;Watson et al., 2010). Intriguingly, p53 is able to regulate WNT ligand production in a cell type-dependent manner (Lee et al., 2010) as well as β-catenin levels (Kim et al., 2011;Sadot et al., 2001), suggesting a complicated interplay between the DNA damage response and WNT signaling. ...
Article
?-Catenin is an F-actin-binding protein widely recognized for its role in cell-cell adhesion. However, a growing body of literature indicates ?-catenin is also a nuclear protein. In this study, we show that ?-catenin is able to modulate the sensitivity of cells to DNA damage and toxicity. Furthermore, nuclear ?-catenin is actively recruited to sites of DNA damage. This recruitment occurs in a ?-catenin-dependent manner and requires nuclear actin polymerization. These findings provide mechanistic insight into the WNT-mediated regulation of the DNA damage response and suggest a novel role for the ?/?-catenin complex in the nucleus.
... In NF-κB cell signaling, GSK-3α inhibition leads to suppression of cell growth in PnCa [35], while GSK-3β up-regulates NF-κB activity which stimulates cell proliferation, protumorigenic cytokine production, resistance to apoptosis, and chemoresistance in PnCa [12,[36][37][38]. GSK-3β is also involved in the WNT/βcatenin pathway by modulating resistance to radiation therapy [39]. Future studies investigating GSK-3 in PnCa with a PET radiotracer that is selective for GSK-3α or -β would also be worthwhile to investigate in oncology as newer generations of GSK-3 PET radiotracers are discovered [28]. ...
Article
Glycogen synthase kinase-3 (GSK-3) contributes to tumorigenesis in pancreatic cancer by modulating cell proliferation and survival. This study evaluated the lead GSK-3 targeted PET radiotracers for neuro-PET imaging, [11C]PF-367 and [11C]OCM-44, in pancreatic cancer xenograft mice. Immunohistochemistry showed that GSK-3α and GSK-3β were overexpressed in PANC-1 xenografts. In autoradiography studies, higher specific binding was observed for [3H]PF-367 compared to [3H]OCM-44 when co-incubated with unlabeled PF-367 (59.2±1.8% vs 22.6±3.75%, respectively). Co-incubation of [11C]OCM-44 with OCM-44 did not improve the specific binding (25.5±30.2%). In dynamic PET imaging of PANC-1 xenograft mouse models, tumors were not visualized with [11C]PF-367 but were well visualized with [11C]OCM-44. Time-activity curves revealed no difference in accumulation in PANC-1 tumor tissue compared to muscle tissue in [11C]PF-367 baseline studies, while a significant difference was observed for [11C]OCM-44 with a tumor-to-muscle ratio of 1.6. Tumor radioactivity accumulation following injection with [11C]OCM-44 was not displaced by pre-treatment with unlabeled PF-367. Radiometabolite analysis showed that intact [11C]PF-367 accounted for 7.5% of tumor radioactivity, with >30% in plasma, at 40 min post-injection of the radiotracer, and that intact [11C]OCM-44 accounted for 20% of tumor radioactivity, with >60% in plasma. [11C]OCM-44 is superior to [11C]PF-367 for detecting lesions in preclinical mouse models of pancreatic cancer, however, both radiotracers undergo rapid metabolism in vivo. GSK-3 PET radiotracers with improved in vivo stability are needed for clinical translation. To our knowledge this work represents the first PET imaging study of GSK-3 in oncology.
... In HCC xenografted nude mice, increased GSK-3β expression downregulated the nuclear and cytosolic β-catenin levels by facilitating the proteosomal degradation of β-catenin [8]. In addition, GSK-3β knockdown enhanced cell survival and proliferation in HCC [13], increased cisplatin resistance via activation of Wnt/βcatenin signaling in lung cancer [14], and resulted in radioresistance of pancreatic cancer [15]. Moreover, the repressive function of GSK-3β in rRNA biogenesis, Wnt, and TGF-β pathways supported its role as a tumor suppressor [16,17]. ...
Article
Hepatocellular carcinoma (HCC) is the second most common cause of cancer-related death worldwide. The role of GSK-3β in cancer progression is considered critical. However, the prognostic value of total GSK-3β protein levels in HCC remains undetermined. In this study, the expression and biologic significance of total GSK-3β in HCC were evaluated at mRNA and protein levels. We showed that GSK-3β mRNA levels were significantly upregulated in HCC tissues relative to the levels in the adjacent non-tumor tissues as recorded on the TCGA database (P < 0.001). Notably, GSK-3β protein levels were significantly downregulated in HCC tissues relative to those in the adjacent non-tumor tissues by immunohistochemistry (P < 0.001). We found that GSK-3β was negatively associated with the American Joint Committee on Cancer (AJCC) stage (P = 0.030) and positively correlated with good prognosis for HCC patients (P = 0.036). The data further indicated that GSK3β expression tended to be an independent prognostic marker for HCC after surgical resection (HR = 1.658, 95% CI 0.945-2.909, P = 0.078) and can potentially serve as a biomarker for the clinical diagnosis and prognosis of HCC.
... Chemotherapy or radiation can up-regulate Wnt signaling, and up-regulation of Wnt signaling protects cancer cells from cell cycle arrest or apoptosis. While this protective effect has been attributed to the downstream effectors of Wnt/b-catenin signaling such as survivin in one study, more generally the underlining mechanism still remains unclear (Watson et al., 2010;Gao et al., 2013;Nagano et al., 2013;Emons et al., 2017). Two recent studies found that elevated Wnt signaling enhanced the DNA damage repair and thereby conferred resistance to the PARP inhibitor olaparib in ovarian cancers (Fukumoto et al., 2019;Yamamoto et al., Chartier et al., 2016;Trillsch et al., 2017;. ...
Article
Wnts are secreted proteins that bind to cell surface receptors to activate downstream signaling cascades. Normal Wnt signaling plays key roles in embryonic development and adult tissue homeostasis. The secretion of Wnt ligands, the turnover of Wnt receptors, and the signaling transduction are tightly regulated and fine-tuned to keep the signaling output "just right". Hyperactivated Wnt signaling due to recurrent genetic alterations drives several human cancers. Elevated Wnt signaling also confers resistance to multiple conventional and targeted cancer therapies through diverse mechanisms including maintaining the cancer stem cell population, enhancing DNA damage repair, facilitating transcriptional plasticity, and promoting immune evasion. Different classes of Wnt signaling inhibitors targeting key nodes of the pathway have been developed and show efficacy in treating Wnt-driven cancers and subverting Wnt-mediated therapy resistance in preclinical studies. Several of these inhibitors have advanced to clinical trials, both singly and in combination with other existing FDA- approved anti-cancer modalities. In the near future, pharmacological inhibition of Wnt signaling may be a real choice for cancer patients. SIGNIFICANCE STATEMENT: We review the latest insights in Wnt signaling, ranging from basic biology to therapeutic implications in cancer. Recent studies extend our understanding of this ancient signaling pathway and describe the development and improvement of anti-Wnt therapeutic modalities for cancer.
... However, silencing of β-catenin resulted in sensitization to cell death. 46 In the current study, we observed that MiaPaCa-2 cells expressed higher GSK-3 than Panc-1 cells. Therefore, PD-0332991 exerted the opposite effect on GSK-3 phosphorylation. ...
Article
Full-text available
The mortality rate of pancreatic cancer has close parallels to its incidence rate because of limited therapeutics and lack of effective prognosis. Despite various novel chemotherapeutics combinations, the 5‐year survival rate is still under 5%. In the current study, we aimed to modulate the aberrantly activated PI3K/AKT pathway and epithelial‐mesenchymal transition (EMT) signaling with the treatment of CDK4/6 inhibitor PD‐0332991 (palbociclib) in Panc‐1 and MiaPaCa‐2 pancreatic cancer cells. It was found that PD‐0332991 effectively reduced cell viability and proliferation dose‐dependently within 24 hours. In addition, PD‐0332991 induced cell cycle arrest at the G1 phase by downregulation of aberrant expression of CDK4/6 through the dephosphorylation of Rb in each cell lines. Although PD‐0332991 treatment increased epithelial markers and decreased mesenchymal markers, the nuclear translocation of β‐catenin was not prevented by PD‐0332991 treatment, especially in MiaPaCa‐2 cells. Effects of PD‐0332991 on the regulation of PI3K/AKT signaling and its downstream targets such as GSK‐3 were cell type‐dependent. Although the activity of AKT was inhibited in both cell lines, the phosphorylation of GSK‐3β at Ser9 increased only in Panc‐1. In conclusion, PD‐0332991 induced cell cycle arrest and reduced the cell viability of Panc‐1 and MiaPaCa‐2 cells. However, PD‐0332991 differentially affects the regulation of the PI3K/AKT pathway and EMT process in cells due to its distinct influence on Rb and GSK‐3/β‐catenin signaling. Understanding the effect of PD‐0332991 on the aberrantly activated signaling axis may put forward a new therapeutic strategy to reduce the cell viability and metastatic process of pancreatic cancer.
... Previous studies had reported that DHA can increase GSK3β activity and reduce TCF/LEF activity in cells such as Hep3B hepatocarcinoma cells [43], cholangiocarcinoma cells [17], and pancreatic cancer PANC-1 cells [44]. Interestingly, GSK3β activation increased the radiosensitization of PANC-1 cells, whereas GSK3β silencing increased the expression of β-catenin target genes, measured as the activity levels of the transcription factors AXIN2 and LEF1 [45]. Moreover, in the CRC cell lines HCT-116 and SW480, DHA was demonstrated to induce β-catenin degradation, inhibit nuclear translocation of β-catenin, and reduce the expression of proteins such as Peroxisome proliferator-activated receptor gamma (PPAR-γ), membrane type-1 matrix metalloproteinase-7, and vascular endothelial growth factor [16]. ...
Article
Full-text available
The effects of radiation are known to be potentiated by N‐3 polyunsaturated fatty acids, which modulate several signaling pathways, but the molecular mechanisms through which these fatty acids enhance the anticancer effects of irradiation in colorectal cancer (CRC) treatment remain poorly elucidated. Here, we aimed to ascertain whether the fatty acid docosahexaenoic acid (DHA) exerts a modulating effect on the response elicited by radiation treatment (RT). Two CRC cell lines, Caco‐2 and HT‐29, were exposed to RT, DHA, or both (DHA + RT) for various times, and then cell viability, proliferation, and clonogenicity were assessed. Moreover, cell cycle, apoptosis, and necrosis were analyzed using flow cytometry, and the involvement of WNT/β‐catenin signaling was investigated by immunofluorescence to determine nuclear β‐catenin, GSK3β phosphorylation status, and TCF/LEF‐activity reporter. DHA and RT applied separately diminished the viability of both HT‐29 and Caco‐2 cells, and DHA + RT caused a further reduction in proliferation mainly in HT‐29 cells, particularly in terms of colony formation. Concomitantly, our results verified cell cycle arrest in G0/G1 phase, a reduction of cyclin D1 expression, and a decrease in GSK3β phosphorylation after the combined treatment. Furthermore, immunofluorescence quantification revealed that nuclear β‐catenin was increased in RT‐exposed cells, but this effect was abrogated in cells exposed to DHA + RT, and the results of TCF/LEF‐activity assays confirmed that DHA attenuated the increase in nuclear β‐catenin activity induced by irradiation. Our finding shows that DHA applied in combination with RT enhanced the antitumor effects of irradiation on CRC cells, and that the underlying mechanism involved the WNT/β‐catenin pathway. © 2018 BioFactors, 9999(9999):1–11, 2018
... Despite advancements in multimodality treatments, the current therapies often result in chemoresistance [5]. Thus, identifying new targets and elucidating mechanisms are highly needed for the treatment of osteosarcoma [6,7]. ...
Article
Osteosarcoma is the most common primary malignant bone tumor among adolescents worldwide with high mortality rate. Glycogen synthase kinase 3β (GSK3β) is a serine/threonine kinase and is considered as a validated target in osteosarcoma therapy. Therefore, the study of GSK3β inhibitors is one of the most popular fields in anti-osteosarcoma drug development. Here, the tools of bioinformatics were used to screen novel effective inhibitors of GSK3β from ZINC Drug Database. The molecular docking, molecular dynamic simulations, MM/GBSA, and energy decomposition analysis were performed to identify the inhibitors. Finally, ZINC08383479 and ZINC08441251 were selected as potential GSK3β inhibitors. These two inhibitors were evaluated by GSK3β kinase inhibition assay in vitro. The inhibition of cell proliferation was tested in osteosarcoma cell lines U2OS and MG63 in vitro. The result showed that ZINC08383479 and ZINC08441251 had high inhibition activity against GSK3β. We found that CHIR99021 (a known GSK3β inhibitor), ZINC08383479, and ZINC08441251 had significant inhibition activity in U2OS cells and MG63 cells. These findings may provide new ideas for the design of more potent GSK3β inhibitors and therapeutic targets for osteosarcoma.
... Inhibition of GSK3-beta by lithium chloride, shRNA knockdown, or expression of a kinase dead mutant GSK3-beta has been shown to increase radioresistance of Panc-1 and BxPC3 cells. This GSK3-beta inhibition resulted in stabilization of beta-catenin and shRNA knockdown of beta-catenin radiosensitized both of these cell lines, suggesting beta-catenin could be another potential target for radiosensitization [285]. ...
Article
Full-text available
Despite recent advances in radiotherapy, a majority of patients diagnosed with pancreatic cancer (PC) do not achieve objective responses due to the existence of intrinsic and acquired radioresistance. Identification of molecular mechanisms that compromise the efficacy of radiation therapy and targeting these pathways is paramount for improving radiation response in PC patients. In this review, we have summarized molecular mechanisms associated with the radio-resistant phenotype of PC. Briefly, we discuss the reversible and irreversible biological consequences of radiotherapy, such as DNA damage and DNA repair, mechanisms of cancer cell survival and radiation-induced apoptosis following radiotherapy. We further describe various small molecule inhibitors and molecular targeting agents currently being tested in preclinical and clinical studies as potential radiosensitizers for PC. Notably, we draw attention towards the confounding effects of cancer stem cells, immune system, and the tumor microenvironment in the context of PC radioresistance and radiosensitization. Finally, we discuss the need for examining selective radioprotectors in light of the emerging evidence on radiation toxicity to non-target tissue associated with PC radiotherapy.
... Previous studies have demonstrated that the activation of the Wnt signaling pathway is a key radioprotective mechanism in irradiated cancer cells (37)(38)(39)(40). Woodward et al reported that Wnt and β-catenin signaling may contribute to the radioresistance of breast cancer stem cells (41), and Watson et al reported that cells with silenced β-catenin are more sensitive to radiation compared with the parental cell line (42). The present report is the first to demonstrate an association between miRNA-320 downregulation and radioresistance through a negative C regulation of β-catenin. ...
Article
Full-text available
Cervical cancer is the second most common malignancy in women worldwide and always has recurrence owing to radioresistance. MicroRNA (miRNA or miR) has been identified to relate to the sensitivity of cancer radiotherapy. Here, we investigated the potential of miRNA-320 as a biomarker for radiosensitivity by targeting β-catenin in cervical cancer. A radioresistant cervical cancer cell line, C33AR, was established, and the radioresistance of C33AR cells was confirmed by a colony-formation assay. The expression of miRNA-320 was detected by reverse transcription-quantitative polymerase chain reaction, and compared between C33A and C33AR. β-catenin, the target of miRNA-320, was determined at the protein level by western blotting after transfecting the inhibitor of miRNA-320. The expression of miRNA-320 was markedly decreased in C33AR cells, which appeared to be more radioresistant, compared with its parental cell line C33A. Target prediction suggested that miRNA-320 negatively regulated the expression of β-catenin. Knockdown of β-catenin increased C33AR radiosensitivity, which revealed that the inhibition of β-catenin could rescue the miRNA-320-mediated cell radioresistance. On the other hand, overexpressing miRNA-320 increased C33AR radiosensitivity. In conclusion, miRNA-320 regulated the radiosensitivity of C33AR cells by targeting β-catenin. This finding provides evidence that miRNA-320 may be a potential biomarker of radiosensitivity in cervical cancer.
... In addition to Mcl-1, USP9x also stabilizes β-Catenin and ubiquitin ligase Itch. 27,28 High β-catenin levels were correlated with increased radioresistance in pancreatic cancer cells, 29 whereas Itch regulates the internalization of epidermal growth factor receptor, a growth receptor that mediates radioresistance in glioblastoma tumors. 30,31 Moreover, stabilization of Foxo3A by USP9x resulted in decreased cyclin D1 levels and cell cycle arrest. ...
Article
Full-text available
Glioblastoma is a very aggressive form of brain tumor with limited therapeutic options. Usually, glioblastoma is treated with ionizing radiation (IR) and chemotherapy after surgical removal. However, radiotherapy is frequently unsuccessful, among others owing to resistance mechanisms the tumor cells have developed. Antiapoptotic B-cell leukemia (Bcl)-2 family members can contribute to radioresistance by interfering with apoptosis induction in response to IR. Bcl-2 and the closely related Bcl-xL and Mcl-1 are often overexpressed in glioblastoma cells. In contrast to Bcl-2 and Bcl-xL, Mcl-1 is a short-lived protein whose stability is closely regulated by ubiquitylation-dependent proteasomal degradation. Although ubiquitin ligases facilitate degradation, the deubiquitylating enzyme ubiquitin-specific protease 9x (USP9x) interferes with degradation by removing polyubiquitin chains from Mcl-1, thereby stabilizing this protein. Thus, an inability to downregulate Mcl-1 by enhanced USP9x activity might contribute to radioresistance. Here we analyzed the impact of USP9x on Mcl-1 levels and radiosensitivity in glioblastoma cells. Correlating Mcl-1 and USP9x expressions were significantly higher in human glioblastoma than in astrocytoma. Downregulation of Mcl-1 correlated with apoptosis induction in established glioblastoma cell lines. Although Mcl-1 knockdown by siRNA increased apoptosis induction after irradiation in all glioblastoma cell lines, USP9x knockdown significantly improved radiation-induced apoptosis in one of four cell lines and slightly increased apoptosis in another cell line. In the latter two cell lines, USP9x knockdown also increased radiation-induced clonogenic death. The massive downregulation of Mcl-1 and apoptosis induction in A172 cells transfected with USP9x siRNA shows that the deubiquitinase regulates cell survival by regulating Mcl-1 levels. In contrast, USP9x regulated radiosensitivity in Ln229 cells without affecting Mcl-1 levels. We conclude that USP9x can control survival and radiosensitivity in glioblastoma cells by Mcl-1-dependent and Mcl-1-independent mechanisms.
Article
Full-text available
There are now more than 10 million cancer survivors in the United States. With these numbers, chronic sequelae that result from cancer therapy have become a major health care problem. Although radiation therapy of the brain has improved cancer cure rates, learning disorders and memory deficits are a common consequence of this therapy. Here we show that glycogen synthase kinase 3beta (GSK-3beta) is required for radiation-induced hippocampal neuronal apoptosis and subsequent neurocognitive decline. Inhibition of GSK-3beta either by small molecules (SB216763 or SB415286) or by ectopic expression of kinase-inactive GSK-3beta before irradiation significantly attenuated radiation-induced apoptosis in hippocampal neurons. GSK-3beta inhibition with SB216763 or SB415286 also decreased apoptosis in the subgranular zone of the hippocampus in irradiated mice, leading to improved cognitive function in irradiated animals. Studies of the molecular mechanisms of the cytoprotective effect showed that GSK-3beta activity in hippocampal neurons was not significantly altered by radiation, pointing to the indirect involvement of this enzyme in radiation-induced apoptosis. At the same time, radiation led to increased accumulation of p53, whereas inhibition of the basal level of GSK-3beta activity before radiation prevented p53 accumulation, suggesting a possible mechanism of cytoprotection by GSK-3beta inhibitors. These findings identify GSK-3beta signaling as a key regulator of radiation-induced damage in hippocampal neurons and suggest that GSK-3beta inhibitors may have a therapeutic role in protecting both pediatric and adult cancer patients and may help to improve quality of life in cancer survivors.
Article
Mutations of the adenomatous polyposis coli (APC) tumor suppressor gene have been described in a subset of pancreatic carcinomas. The APC gene modulates the beta-catenin-Tcf pathway. The major player in this pathway is the beta-catenin protein encoded by the beta-catenin gene. A variety of different tumors, including colon, prostate, endometrial, and hepatocellular carcinomas, carry mutations in exon 3 of the beta-catenin gene. The aim of this study was to determine the role of the beta-catenin gene in the genesis of exocrine and endocrine tumors of the pancreas. 78 ductal pancreatic adenocarcinomas, 14 ductal pancreatic cancer cell lines, and 33 endocrine pancreatic tumors were evaluated for mutations in exon 3 of the beta-catenin gene by single-strand conformation polymorphism analysis and direct DNA sequencing. In addition, 40 ductal pancreatic adenocarcinomas were analyzed for intracellular beta-catenin accumulation by immunohistochemistry, indicating alterations of the beta-catenin gene. Neither the 111 exocrine and endocrine pancreatic tumors nor the 14 pancreatic cancer cell lines carried mutations in exon 3 of the beta-catenin gene. Intracellular beta-catenin accumulation was not identified in any of the 40 pancreatic adenocarcinomas. These data suggest that the beta-catenin gene as the major player of the beta-catenin-Tcf pathway does not play an important role in the genesis of pancreatic tumors.
Article
Matrix metalloproteinases (MMPs) are a family of zinc-containing proteolytic enzymes that break down extracellular matrix proteins (ECM) in physiological and pathological conditions. Disruption in the tight control of MMP metabolism occurs in cancer, resulting in excessive destruction of theECM, neovascularization, tumor spread and metastases. Recent studies have shown that overexpression of MMPs is associated with poor prognosis. Several MMP inhibitors have been developed and preclinical trials have confirmed a reduction in tumor spread and metastases. Marimastat is a broad spectrum inhibitor, and recent published results shows the drug is well tolerated in patients with advanced cancer. Phase II studies which have used marimistat alone or in combination with other cytotoxic agents, have produced encouraging results with improved survival. Phase III trials are now underway for the use of marimastat in advanced pancreatic cancer and as an adjuvant therapy in patients following resection of pancreatic cancer.
Article
R-Spondin1 (RSpo1) is a novel secreted protein that augments canonical Wnt/beta-catenin signaling. We injected recombinant RSpo1 protein into transgenic Wnt reporter TOPGAL mice and have identified the oral mucosa as a target tissue for RSpo1. Administration of RSpo1 into normal mice triggered nuclear translocation of beta-catenin and resulted in increased basal layer cellularity, thickened mucosa, and elevated epithelial cell proliferation in tongue. We herein evaluated the therapeutic potential of RSpo1 in treating chemotherapy or radiotherapy-induced oral mucositis in several mouse models. Prophylactic treatment with RSpo1 dose-dependently overcame the reduction of basal layer epithelial cellularity, mucosal thickness, and epithelial cell proliferation in tongues of mice exposed to whole-body irradiation. RSpo1 administration also substantially alleviated tongue mucositis in the oral cavity of mice receiving concomitant 5-fluorouracil and x-ray radiation. Furthermore, RSpo1 significantly reduced the extent of tongue ulceration in mice receiving a single fraction, high dose head-only radiation in a dose-dependent manner. Moreover, combined therapy of RSpo1 and keratinocyte growth factor resulted in complete healing of tongue ulcers in mice subjected to snout-only irradiation. In conclusion, our results demonstrate RSpo1 to be a potent therapeutic agent for oral mucositis by enhancing basal layer epithelial regeneration and accelerating mucosal repair through up-regulation of Wnt/beta-catenin pathway.
Article
Radiation causes soft tissue complications that include fibrosis and deficient wound healing. beta-Catenin, a key component in the canonical Wnt-signaling pathway, is activated in fibrotic processes and wound repair and, as such, could play a role in mediating cellular responses to irradiation. beta-Catenin can form a transcriptionally active complex with members of the Tcf family. A reporter mouse model, in addition to human cell cultures, was used to demonstrate that ionizing radiation activates beta-catenin-mediated, Tcf-dependent transcription both in vitro and in vivo. Furthermore, radiation activates beta-catenin via a Wnt-mediated mechanism, as in the presence of dickkopf-1, an inhibitor of Wnt receptor activation, beta-catenin levels did not increase after irradiation. Fibroblast cell cultures were derived from mice expressing either null or stabilized beta-catenin alleles. Cells expressing stabilized beta-catenin alleles had a higher proliferation rate and formed more colony-forming units than wild-type or null cells after irradiation. Wound healing was studied in these same mice after irradiation. There was a positive correlation between the tensile strength of the wound, the expression levels of type 1 collagen in the skin, and beta-catenin levels. Mice treated with lithium showed increased beta-catenin levels and increased wound strength. beta-Catenin mediates the effects of ionizing radiation in fibroblasts, and its modulation has the potential to decrease the severity of radiation-induced soft tissue complications.
Article
The mean inactivation dose (D) is calculated for published in vitro survival curves obtained from cell lines of both normal and neoplastic human tissues. Cells belonging to different histological categories (melanomas, carcinomas, etc.) are shown to be characterized by distinct values of D which are related to the clinical radiosensitivity of tumors from these categories. Compared to other ways of representing in vitro radiosensitivity, e.g., by the multitarget parameters D0 and n, the parameter D has several specific advantages: (i) D is representative for the whole cell population rather than for a fraction of it; (ii) it minimizes the fluctuations of the survival curves of a given cell line investigated by different authors; (iii) there is low variability of D within each histological category; (iv) significant differences in radiosensitivity between the categories emerge when using D. D appears to be a useful concept for specifying intrinsic radiosensitivity of human cell lines.
Article
The beta-isoform of glycogen synthase kinase-3 (GSK3 beta) isolated from rabbit skeletal muscle was inactivated 90-95% following incubation with MgATP and either MAP kinase-activated protein kinase-1 (MAPKAP kinase-1, also termed RSK-2) or p70 S6 kinase (p70S6K), and re-activated with protein phosphatase 2A. MAPKAP kinase-1 and p70S6K phosphorylated the same tryptic peptide on GSK3 beta, and the site of phosphorylation was identified as the serine located nine residues from the N-terminus of the protein. The inhibitory effect of Ser-9 phosphorylation on GSK3 beta activity was observed with three substrates, (inhibitor-2, c-jun and a synthetic peptide), and also with glycogen synthase provided that 0.15 M KCl was added to the assays. The results suggest that Ser-9 phosphorylation underlies the reported inhibition of GSK3 beta by insulin and that GSK3 may represent a point of convergence of two major growth-factor-stimulated protein kinase cascades.
Article
We have studied the control of insulin-regulated protein kinases in Chinese hamster ovary cells transfected with the human insulin receptor (CHO.T cells). Among these enzymes is one that is obtained after chromatography of cell extracts on Mono-S, whose activity is decreased (7.3 +/- 1.9-fold) within 10 min of insulin treatment. This enzyme phosphorylates glycogen synthase and the largest subunit of protein synthesis eukaryotic initiation factor (eIF)-2B (the guanine nucleotide exchange factor). The kinase appears to be glycogen synthase kinase-3 (GSK-3), on the basis of: (1) its ability to phosphorylate a peptide based on the phosphorylation sites for GSK-3 in glycogen synthase, and (2) the finding that the fractions possessing this activity contain immunoreactive GSK-3, whose peak is coincident with that of kinase activity, as judged by immunoblotting using antibodies specific for the alpha- and beta-isoforms of GSK-3. The decrease in kinase activity induced by insulin was reversed by treatment of the column fractions with protein phosphatase-2A. These data indicate that insulin rapidly causes inactivation of GSK-3 and that this is due to phosphorylation of GSK-3. The implications of these findings for the control of glycogen and protein metabolism are discussed.