ArticlePDF Available

Resveratrol is pro-apoptotic and thyroid hormone is anti-apoptotic in glioma cells: Both actions are integrin and ERK mediated

Authors:

Abstract and Figures

The stilbene resveratrol (RV) initiates p53-dependent apoptosis via plasma membrane integrin alphaVbeta3 in human cancer cells. A thyroid hormone (L-thyroxine, T(4)) membrane receptor also exists on alphaVbeta3. Stilbene and T(4) signals are both transduced by extracellular-regulated kinases 1 and 2 (ERK1/2); however, T(4) promotes cell proliferation in cancer cells, whereas RV is pro-apoptotic. Thyroid hormone has been shown to interfere with RV-induced apoptosis. However, the mechanisms involved are not fully understood. In this study, we examined the mechanism whereby T(4) inhibits RV-induced apoptosis in glioma cells. RV activated conventional protein kinase C and ERK1/2 and caused nuclear localization of cyclooxygenase-2 (COX-2), consequent p53 phosphorylation and apoptosis. RV-induced ERK1/2 activation is involved in not only COX-2 expression but also nuclear COX-2 accumulation. NS-398, a COX-2 inhibitor, did not affect ERK1/2 activation, but reduced the nuclear abundance of COX-2 protein and the formation of complexes of nuclear COX-2 and activated ERK1/2 that are required for p53-dependent apoptosis in RV-treated cells. T(4) inhibited RV-induced nuclear COX-2 and cytosolic pro-apoptotic protein, BcLx-s, accumulation. Furthermore, T(4) inhibited RV-induced apoptosis by interfering with the interaction of nuclear COX-2 and ERK1/2. This effect of T(4) was prevented by tetraiodothyroacetic acid (tetrac), an inhibitor of the binding of thyroid hormone to its integrin receptor. Tetrac did not, in the absence of T(4), affect induction of apoptosis by RV. Thus, the receptor sites on alphaVbeta3 for RV and thyroid hormone are discrete and activate ERK1/2-dependent downstream effects on apoptosis that are distinctive.
Content may be subject to copyright.
Carcinogenesis vol.29 no.1 pp.62–69, 2008
doi:10.1093/carcin/bgm239
Advance Access publication November 4, 2007
Resveratrol is pro-apoptotic and thyroid hormone is anti-apoptotic in glioma cells:
both actions are integrin and ERK mediated
Hung-Yun Lin
1,2,
, Heng-Yuan Tang
1
, Travis Keating
1
,
Yun-Hsuan Wu
1
, Ai Shih
2
, Douglas Hammond
2
, Mingzeng
Sun
1
, Aleck Hercbergs
3
, Faith B.Davis
1
and Paul J.Davis
1,2
1
Signal Transduction Laboratory, Ordway Research Institute, 150 New
Scotland Avenue, Albany, NY 12208, USA,
2
Research Service, Stratton
Veterans Affairs Medical Center, Albany, NY, USA and
3
Department of
Radiation Oncology, The Cleveland Clinic, Cleveland, OH, USA
To whom correspondence should be addressed. Tel: þ1 518 641 6428;
Email: hlin@ordwayresearch.org
The stilbene resveratrol (RV) initiates p53-dependent apoptosis
via plasma membrane integrin aVb3 in human cancer cells.
A thyroid hormone (L-thyroxine, T
4
) membrane receptor also
exists on aVb3. Stilbene and T
4
signals are both transduced by
extracellular-regulated kinases 1 and 2 (ERK1/2); however, T
4
promotes cell proliferation in cancer cells, whereas RV is pro-
apoptotic. Thyroid hormone has been shown to interfere with
RV-induced apoptosis. However, the mechanisms involved are
not fully understood. In this study, we examined the mechanism
whereby T
4
inhibits RV-induced apoptosis in glioma cells. RV
activated conventional protein kinase C and ERK1/2 and caused
nuclear localization of cyclooxygenase-2 (COX-2), consequent p53
phosphorylation and apoptosis. RV-induced ERK1/2 activation is
involved in not only COX-2 expression but also nuclear COX-2
accumulation. NS-398, a COX-2 inhibitor, did not affect ERK1/2
activation, but reduced the nuclear abundance of COX-2 protein
and the formation of complexes of nuclear COX-2 and activated
ERK1/2 that are required for p53-dependent apoptosis in RV-
treated cells. T
4
inhibited RV-induced nuclear COX-2 and cyto-
solic pro-apoptotic protein, BcLx-s, accumulation. Furthermore,
T
4
inhibited RV-induced apoptosis by interfering with the inter-
action of nuclear COX-2 and ERK1/2. This effect of T
4
was pre-
vented by tetraiodothyroacetic acid (tetrac), an inhibitor of the
binding of thyroid hormone to its integrin receptor. Tetrac did
not, in the absence of T
4
, affect induction of apoptosis by RV.
Thus, the receptor sites on aVb3 for RV and thyroid hormone
are discrete and activate ERK1/2-dependent downstream effects
on apoptosis that are distinctive.
Introduction
Thyroid hormone (L-thyroxine, T
4
; 3,5,3#-triiodo-L-thyronine) is
a proliferation factor in vitro for a rat glioblastoma cell line (C6)
via a cell-surface receptor for the hormone on integrin aVb3 (1). This
receptor is at or near the arginine–glycine–aspartate (RGD) recogni-
tion site on the integrin that is involved in the interactions of the
integrin with extracellular matrix proteins. The intracellular domain
of the integrin may activate extracellular-regulated kinases 1 and 2
(ERK1/2) (2–4) and we have shown that T
4
rapidly increases cellular
ERK1/2 activity via the integrin (5). Short integrin antagonist peptides
that contain the RGD sequence have been designed as tools to dem-
onstrate the role of integrins in transducing the signals of a number of
extracellular matrix proteins. RGD peptides block thyroid hormone
actions mediated by the integrin (1).
Resveratrol (RV), a polyphenolic antioxidant enriched in grape
skin, has been shown to cause apoptosis in a number of different
cancer cell lines (6,7) and to suppress cancer growth in vivo in animal
models (8). We have recently described a RV receptor on integrin
aVb3 using probes such as antibody to aVb3 and an siRNA of
aVb3 construct to interfere with integrin-mediated actions of the
stilbene (9). RV (1–10 lM) induces p53-dependent apoptosis in breast
(10), prostate (11,12) and thyroid cancer (13) cells in culture. RV
stimulates Ser-15 phosphorylation of p53 by a mitogen-activated pro-
tein kinase-dependent pathway (ERK1/2), and this step in RV-treated
cells is required for apoptosis to occur (13). Other kinases such as
conventional protein kinase C (cPKC), p38 and c-jun N-terminal
kinase are also activated in RV-treated cells (12,14). On the other
hand, nuclear factor-jB is inhibited by RV (15). Growth factors such
as epidermal growth factor (12) and hormones such as estrogen have
also been shown to activate ERK1/2 in selected human cancer cells
(16), but cancer cells treated with these agents may no longer undergo
apoptosis when treated with RV (10). We have shown that thyroid
hormone (T
4
,10
7
M total hormone concentration, 10
10
M free T
4
)
induces ERK1/2 activation in the CV-1 green monkey kidney epithe-
lial cell (17) and RV did not activate ERK1/2 in the same cell line until
the stilbene concentration reached 100 lM (Lin HY et al., unpub-
lished results). On the other hand, thyroid hormone, specifically T
4
,
blocks the pro-apoptotic effect of RV in thyroid cancer cells (18).
However, the mechanisms underlying the inhibitory effect of thyroid
hormone on RV-induced apoptosis are not fully understood.
We show here in glioma cells that while RV and thyroid hormone
both bind to the plasma membrane integrin aVb3 and activate ERK1/2,
they do so via different ligand-binding sites, leading to the disparate
effects seen in these cells. While T
4
stimulates glial cell proliferation,
the p53-dependent action of RV leads to apoptosis. We have shown
elsewhere (19) that RV-associated apoptosis requires a pool of induc-
ible cyclooxygenase-2 (COX-2) in the nucleus, upstream of p53. The
inhibition of apoptosis by thyroid hormone in RV-treated cells is
initiated at the plasma membrane integrin receptor for the hormone;
this leads to suppression of the nuclear interaction of COX-2 protein
and activated ERK1/2, which is essential to the pro-apoptotic action
of RV. That is, this nuclear complex is formed in glioma cells exposed
to RV, alone, but does not occur in T
4
-treated cells or in cells in-
cubated with both RV and T
4
.
Materials and methods
Materials
T
4
, tetraiodothyroacetic acid (tetrac) and RGD and RGE peptides were ob-
tained from Sigma Chemical Co. (St Louis, MO). T
4
and tetrac were prepared
as a stock 10
4
M solution in 0.04 N KOH and 4% propylene glycol. RV was
purchased from Calbiochem (San Diego, CA) and dissolved in ethanol as a 100
mM stock solution. The PKC agonist phorbol 12-myristate 13-acetate (PMA)
was obtained from Sigma Chemical Co. and prepared as a 100 lg/ml PMA in
dimethyl sulfoxide. A specific PKC inhibitor, CGP41251 (CGP) obtained as
a gift from Novartis Pharma (Basel, Switzerland) was prepared as a stock 100
mM solution in dimethyl sulfoxide. The specific ERK1/2 kinase inhibitor,
PD98059, purchased from Calbiochem was prepared as a stock 30 mM solu-
tion in dimethyl sulfoxide. The final concentrations of solvents in which re-
agents were dissolved were tested for activity and did not affect the
experimental outcomes. Polyclonal rabbit anti-phospho-ERK1/2 (pERK12)
was obtained from Cell Signaling (Beverly, MA), monoclonal mouse anti-
proliferating cell nuclear antigen and anti-BcLx-s from Santa Cruz Biotech-
nology (Santa Cruz, CA) and polyclonal rabbit anti-mouse COX-2 from
Cayman (Ann Arbor, MI). Goat anti-rabbit IgG and rabbit anti-mouse IgG
were obtained from Dako (Carpinteria, CA), and the chemiluminescence
reagents (ECL kit) and [
3
H]-thymidine from Amersham (Piscataway, NJ).
Nucleosome enzyme-linked immunoadsorbent assay (ELISA) kit was pur-
chased from Calbiochem.
Abbreviations: COX-2, cyclooxygenase-2; cPKC, conventional protein
kinase C; ELISA, enzyme-linked immunoadsorbent assay; ERK1/2,
extracellular-regulated kinases 1 and 2; FBS, fetal bovine serum; pERK12,
phospho-ERK1/2; PMA, phorbol 12-myristate 13-acetate; RGD, arginine–
glycine–aspartate; RGE, arginine-glycine-glutamate; RV, resveratrol; TCA,
trichloroacetic acid; tetrac, tetraiodothyroacetic acid; T
4
,L-thyroxine.
ÓThe Author 2007. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org 62
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
Cell culture
The rat glioma C6 cell line and human glioblastoma U87MG cell line were
purchased from American Type Culture Collection (Manassas, VA). The rat
glioma GL261 cells were obtained from the Roswell Park Cancer Institute,
Buffalo, NY. C6 cells were maintained in F12K medium supplemented with
18% fetal bovine serum (FBS), U87MG cells were maintained in Minimal
essential medium supplemented with 10% FBS and GL261 cells were main-
tained in Dulbecco’s modified Eagle’s medium supplemented with 10% FBS.
All cell cultures were maintained in a 5% CO
2
/95% air incubator at 37°C. Prior
to treatment, cells were placed in 0.25% hormone-stripped FBS-containing
medium for 2 days. The free T
4
concentration was 0.7 10
10
M, as directly
measured in aliquots of representative culture media of hormone-treated cells
by the Clinical Chemistry Laboratory of Albany Medical Center Hospital,
Albany, NY (20).
Apoptosis/nucleosomes
An early event in apoptosis is DNA fragmentation followed by release of
nucleosomes into the cytoplasm. The nucleosome is the basic unit of chromatin
and results from the ordered association of histones and DNA (21). The double-
antibody sandwich ELISA is based upon the specific recognition of
nucleosomes by a pair of monoclonal antibodies and detects cytoplasmic
nucleosomes onto the ELISA plate. Cells were treated with different reagents
for 48 h. The medium was harvested, spun down and pellets were washed twice
with phosphate-buffered saline. Pelleted cells were lysed. Supernatants were
collected and stored for at least 18 h at 20°C. From each appropriately diluted
sample, 100 ll were added to a 96-well plate coated with a DNA-binding
protein and incubated at room temperature for 3 h. After three washes with
wash buffer, detector antibody was added for 1 h. Streptavidin conjugate was
added and incubated for 0.5 h before adding substrate. Plates were read at 450 nm.
Immunoblotting and immunoprecipitation
Nuclear and cytosol protein extracts were prepared, quantitated and separated
on discontinuous sodium dodecyl sulfate–polyacrylamide gel electrophoresis,
and then transferred by electroblotting to nitrocellulose membranes (Millipore,
Bedford, MA), as we have described previously (9–13). For immunoprecipi-
tation, 200 lg of each nucleoprotein sample was exposed to anti-pERK1/2
and immunoprecipitated proteins separated by sodium dodecyl sulfate–
polyacrylamide gel electrophoresis, and then transferred by electroblotting to
nitrocellulose membranes (Millipore). Membranes were blocked with 5% milk
in Tris-buffered saline containing 0.1% Tween, and then incubated with se-
lected antibodies overnight. Secondary antibodies were either goat anti-rabbit
IgG or rabbit anti-mouse IgG (1:1000), depending on the origin of the primary
antibody. Immunoreactive proteins were detected by chemiluminescence
(ECL, Amersham Life Science, Arlington Heights, IL) and integrated optical
density of bands was quantitated by phosphoimaging and with ImageQuant
software on a Storm 860 Phosphorimager (Molecular Dynamics, Sunnyvale,
CA) as described previously (19).
Confocal microscopy
Exponentially growing C6 glioma cells were seeded in slide chambers. After
exposure to 0.25% stripped FBS-containing medium for 2 days, cells were
either treated with 20 lM RVin the presence or absence of the cPKC inhibitor,
CGP or the MEK inhibitor, PD for 24 h. In other sets of experiments, U87MG
cells and GL261 cells were treated with 20 lMRV,10
7
MT
4
or both for 24 h.
Cells were fixed with 4% formaldehyde in acetone for 30 min and then per-
meabilized in 0.06% Triton X-100 for 30 min. The cells were incubated with
monoclonal antibody to COX-2 followed by Alexa-488-labeled goat anti-
mouse antibody and the signal revealed using the Histostain SP kit as recom-
mended by the manufacturer (Zymed–Invitrogen, Carlsbad, CA). Nuclear
staining with propidium iodide was also employed. Cells were examined under
250 magnification.
Radiolabeled thymidine incorporation
Aliquots of cells were incubated with 1 lCi [
3
H]-thymidine (final concentra-
tion, 13 nM) in a 24-well culture tray for 16 h in the presence or absence of
reagents as indicated. Cells were then washed twice with cold phosphate-
buffered saline, after which 5% trichloroacetic acid (1 ml) was added and
the plate was kept at 4°C for 30 min. The precipitate was then washed twice
with cold ethanol, and 1 ml of 2% sodium dodecyl sulfate was added to each
well. The trichloroacetic acid-precipitable radioactivity was quantitated in
a liquid scintillation counter.
Quantitation of results and statistical analysis
Immunoblot densities were measured with a Storm 860 Phosphorimager fol-
lowed by analysis with ImageQuant software (Molecular Dynamics). Student’s
t-test, with P,0.05 as the threshold for significance, was used to evaluate
results from three or more experiments.
Results
Thyroid hormone and RV, both ligands of integrin aVb3, have
competitive actions in C6 glial cells
In studies of rat glioma C6 cell proliferation, T
4
treatment increased
radiolabeled thymidine incorporation (Figure 1A). In contrast, RV
Fig. 1. Effects of RV, T
4
and tetrac on cell proliferation and apoptosis. (A)The
effect of 10
7
MT
4
and 1–50 lMRVon[
3
H]-thymidine incorporation was
measured as described in Materials and Methods. T
4
increased thymidine
incorporation by C6 glial cells. This effect was inhibited by RV in
a concentration-dependent manner (P,0.05 with 10 lMandP,0.01 with
50 lM). (B) C6 cells were treated with 10 lM RV in the presence or absence
of T
4
(10
8
and 10
7
M). RV-induced apoptosis (nucleosome ELISA) was
inhibited significantly by co-incubation with 10
8
to 10
7
MT
4
(P,0.01).
(C) The effect of T
4
on RV-induced apoptosis was examined in the presence
and absence of tetrac. The inhibition by T
4
of apoptosis induced by RV (lane 7
compared with lane 5, P,0.05) was blocked by the addition of tetrac (lane 8
versus lane 7, P,0.01), indicating that tetrac blocks the inhibitory effect of
T
4
, but does not block the effect of the stilbene.
Apoptosis in glioma cells
63
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
Fig. 2. RV activates ERK1/2 and cPKC via binding to integrin aVb3 in C6 glioma cells. (A) Cells were treated with 10 lM RV for 4 h, in the presence or absence
of 5–500 nM RGD peptide or 500 nM arginine–glycine–glutamate (RGE) peptide (24 h). Concentration-dependent inhibition of RV-induced cPKC and ERK1/2
activation was seen with the RGD peptide (P,0.01), but no inhibition was seen with arginine–glycine–glutamate peptide. Lamin-B and b-actin immunoblots are
provided in this and subsequent figures as controls for gel loading of nucleoproteins and cytosolic proteins, respectively. The graphs in this and subsequent figures
represent the mean ± SD of band intensities normalized to a value of 1 in untreated cells, in three separate experiments. (B) C6 cells were treated with PMA
(100 ng/ml) for 24 h prior to addition of RV (10 lM) for 4 h. PMA treatment, alone, caused a significant increase in nuclear accumulation of nuclear factor-jB
subunits, both p65 and p50 (P,0.01). RV did not cause nuclear factor-jB (NFjB) accumulation, but did cause a significant increase in ERK1/2 activation and
nuclear translocation (P,0.01), which was reduced by 24 h pre-treatment with PMA (P,0.01). The levels of cytosolic phosphorylated PKCa/bin cells treated
with RV were not seen in cells depleted of PKC with PMA treatment. (C) Cells were treated with RV (10 lM, 4 h), in the presence or absence of the ERK1/2
activation inhibitor, PD98059 (PD, 3 or 30 lM, 4.5 h) or the PKCa/binhibitor, CGP41251 (CGP, 10 or 100 nM, 4.5 h). ERK1/2 activation associated with RV
treatment, shown in nuclear fractions, was inhibited by both PD and CGP. RV-induced phosphorylation of PKCa/bwas inhibited by CGP but not by PD (P,0.05,
comparing RV-treated cell fractions with corresponding samples treated with RVand either inhibitor).
H.-Y.Lin et al.
64
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
(1–50 lM) caused apoptosis and thus suppressed thymidine uptake.
Further, the growth-stimulatory effect of T
4
on C6 cells was pro-
gressively diminished by increased concentrations of RV (Figure
1A). In studies of C6 cell apoptosis, T
4
alone had no appreciable
effect, whereas RV treatment produced apoptosis as expected
(nucleosome ELISA, Figure 1B). The deaminated T
4
analog tetrac
did not inhibit the effect on apoptosis of RV, alone (Figure 1C, lane 6),
but did inhibit T
4
suppression of RV-induced apoptosis (comparing
lanes 7 and 8). This finding suggests that T
4
and RV interact with the
integrin at discrete sites and that tetrac only binds to the T
4
-binding
site.
RV-stimulated ERK1/2 and cPKC activation in C6 cells is inhibited by
an integrin aVb3 recognition site RGD peptide
To illustrate the role of integrin aVb3 and consequent intracellular
signaling in RVaction, we measured the effect of RGD and arginine–
glycine–glutamate peptides on the activation of ERK1/2 and of cPKC
by RV. While control arginine–glycine–glutamate peptide did not
alter ERK1/2 or cPKC phosphorylation by the stilbene, increasing
concentrations of RGD peptide (5–500 nM) suppressed these effects
(Figure 2A). This finding is consistent with displacement of RV from
an integrin-binding site at or near the RGD recognition domain.
Fig. 3. Activation of the ERK1/2 pathway is required for RV-induced COX-2 nuclear accumulation in C6 cells. (A) Cells were treated with RV (10 lM) for 24 h,
with or without PD98059 (PD, 30 lM) or with PD for the last 12 h of RV treatment. Nuclear content of COX-2 increased with RV treatment, and this effect was
inhibited by PD (P,0.05), particularly after 24 h of inhibitor treatment. On the other hand, the accumulation of cytosolic COX-2 in RV-treated cells was
significantly enhanced by PD treatment (P,0.05) for 12 or 24 h. (B) C6 cells were treated with 10 lM RV in the presence or absence of CGP41251 (CGP) or
PD98059 (PD) for 24 h and then examined by confocal microscopy. RV caused nuclear accumulation of COX-2, indicated by the appearance of a yellow color due
to superimposition (co-localization) of COX-2 (green) and nuclear (red) images. In the presence of CGP (two middle panels) and PD (two panels at right), nuclear
COX-2 was not evident, although cytosolic COX-2 is still seen in some cells. Cells were viewed at 250 magnification. Images by differential interference contrast
are shown at the lower left of each group of four images, in order to clarify outlines of cells and nuclei.
Apoptosis in glioma cells
65
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
In cells depleted of PKC by treatment with PMA (100 mg/ml) for 24 h,
RV-induced ERK1/2 activation and phosphorylation of PKCa/bwere
both inhibited (Figure 2B). There was no nuclear factor-jB activation
in RV-treated cells, although PMA alone increased both p50 and p65
subunit accumulation in nuclei (Figure 2B). RV-induced ERK1/2 ac-
tivation and nuclear translocation were inhibited by the ERK1/2 ac-
tivation inhibitor PD (Figure 2C). Treatment of cells with the cPKC
activation inhibitor, CGP, resulted in similar suppression of ERK1/2
phosphorylation; this inhibitor also partially inhibited phosphoryla-
tion of PKCa/b, although PD did not inhibit this latter effect. These
results indicate that ERK1/2 activation in RV-treated C6 cells is de-
pendent on cPKC activation as well as on MEK activation.
Activated ERK is associated in nuclei with COX-2 in C6 cells treated
with RV; this effect is required for apoptosis
We have shown that activated ERK1/2 is essential for RV-induced
COX-2 expression (19). Treatment of C6 glioma cells with RV caused
nuclear accumulation of COX-2, and inhibition of ERK1/2 activation
with PD for 24 h blocked this RV effect (Figure 3A). However, treat-
ment of C6 cells with PD for only the last 12 h of a 24-h RV exposure
did not inhibit COX-2 expression but partially inhibited nuclear ac-
cumulation of COX-2 and there was increased accumulation of COX-
2 in the cytosol. COX-2 levels increased in the cytosol of cells treated
with PD alone and in cells exposed to RV and PD; this suggests that
activation of ERK1/2 is required for nuclear accumulation of COX-2
in the presence of RV.
In confocal microscopy studies, we have also demonstrated ERK1/2
activation-dependent nuclear accumulation of COX-2 (green) in
RV-treated cells (Figure 3B). Untreated cells demonstrate a minimal
amount of COX-2 in cytosol. Cells treated with RV show increased
cellular COX-2, and in particular increased nuclear COX-2, indicated
by a yellow color formed by superimposition of the COX-2 green
color on a red nucleoprotein stain. Inhibitors of cPKC activation
and pERK1/2 activation both blocked nuclear COX-2 accumulation
in the presence of RV. These results confirm that RV-induced cPKC
activation is upstream of ERK1/2 activation and that inhibition of
either cPKC or ERK1/2 activation will inhibit ERK1/2-dependent
nuclear COX-2 accumulation in RV-treated C6 cells.
Demonstration of the interrelationships between COX-2 activity,
pERK1/2 accumulation and Ser-15 phosphorylation of p53
In the presence of the specific COX-2 inhibitor, NS-398 (0.1–10 nM),
RV-induced nuclear pERK1/2 content was unaffected (Figure 4).
However, the formation of co-immunoprecipitated nuclear complexes
of COX-2 and pERK1/2 in RV-treated cells was inhibited by NS-398,
but not by the non-specific COX inhibitor, indomethacin. NS-398 did
not affect pERK1/2 activation per se confirming that RV-induced
ERK1/2 activation is upstream COX-2 expression. On the other hand,
NS-398 reduced RV-induced Ser-15–p53 phosphorylation (Figure 4).
Apoptosis induced by RV was also inhibited by NS-398, but not
affected by indomethacin (Figure 4). These results also appear to
implicate RV-inducible nuclear COX-2 accumulation in the ERK1/2-
dependent activation (phosphorylation) of p53 that leads to cancer cell
apoptosis.
Thyroid hormone blocks RV action by disrupting COX-2–pERK1/2
complexing in C6 glial cells
Thyroid hormone inhibits RV-induced apoptosis in C6 cells (Figure 1),
although the two agonists share the same plasma membrane receptor
and some of the same signaling pathways. At what point in the sig-
naling sequence does the hormone block the action of RV? We have
demonstrated that T
4
stimulates ERK1/2 activation and cell prolifer-
ation in different glioma cell lines (1). In contrast, RV (10 lM) also
stimulates ERK1/2 activation (Figures 2 and 4), but causes nuclear
COX-2 accumulation and apoptosis. We therefore examined whether
T
4
blocks RV-induced nuclear COX-2 accumulation. Studies of
confocal microscopy using two different glioma cell lines, human
glioblastoma U87MG cells (Figure 5A, left panel) and rat glioma
GL261 cells (Figure 5A, right panel) indicate that RV caused nuclear
COX-2 accumulation and that this effect was blocked by T
4
. This
inhibitory effect of T
4
on RV-induced nuclear COX-2 accumulation
is also shown in immunoblots of nuclear fractions with a parallel
decrease in the pre-apoptotic protein, BcLx-s (Figure 5B).
Fig. 4. Inhibition of COX-2 activity suppresses nuclear accumulation of
COX-2, complexing of pERK1/2 and COX-2, nuclearSer-15-phosphorylated
p53 and apoptosis in RV-treated cells. C6 cells were treated with the COX-2
inhibitor NS-398 (0.1–10 lM) and 10 lM RV for 24 h. Aliquots of nuclear
extracts were immunoblotted with antibodies to COX-2, pSer15–p53,
pERK1/2 or ERK1/2, whereas additional nuclear extracts were
immunoprecipitated with anti-pERK1/2 and these solubilized
immunoprecipitates (IP) then separated by sodium dodecyl sulfate–
polyacrylamide gel electrophoresis and immunoblotted with anti-COX-2 to
demonstrate co-immunoprecipitation of pERK1/2 and COX-2. RV increased
nuclear content of COX-2, pERK1/2 and pSer15–p53, as well as co-
immunoprecipitation of pERK1/2 and COX-2 (lane 4). NS-398 had no effect
alone (lane 2) and did not inhibit RV-induced ERK1/2 activation (lanes 5–7).
However, nuclear co-immunoprecipitation of pERK1/2 with COX-2 was
inhibited, as were nuclear accumulation of COX-2 and of Ser-15-
phosphorylated p53 (lanes 5–7 compared with lane 4). The inhibitory effect
of NS-398 on RV-induced nuclear accumulation of COX-2 was significant
(P,0.01). In contrast, indomethacin was not inhibitory (comparing lanes
4 and 8). A representative IgG-H chain immunoblot served as a loading
control for the immunoprecipitates. Apoptosis (nucleosome ELISA) due to
RV treatment was also inhibited by NS-398, but not by indomethacin.
H.-Y.Lin et al.
66
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
The thyroid hormone analog, tetrac, is known to inhibit T
4
binding
to plasma membranes and purified integrin aVb3 (5) and to inhibit
membrane-associated effects of T
4
on signal transduction pathways
(20,22). Results also indicate that tetrac reversed the inhibitory effect
of T
4
on RV-induced apoptosis (Figure 1C). We therefore examined
the actions of tetrac on the inhibitory effect of T
4
on nuclear accu-
mulation of pERK1/2, COX-2 and pSer15–p53 in C6 cells treated
with RV. T
4
treatment alone caused nuclear accumulation of
pERK1/2 and pSer15–p53, but not that of COX-2 (Figure 6, lane 3).
Tetrac partially blocked the hormone effects (lane 4). RV stimulated
nuclear accumulation of pERK1/2, COX-2 and pSer15–p53, as well
as the pro-apoptotic protein BcLx-s (Figure 6, lane 5). Tetrac did not
inhibit the action of RV on these parameters (lane 6 compared with
lane 5). When RV and T
4
were added together to cells (Figure 6, lane 7),
Fig. 5. Effect of T
4
on RV-induced nuclear COX-2 and cytosolic BcLx-s accumulation. (A) U87MG cells (left panel) and GL261 cells (right panel) were treated
with 20 lMRV,10
7
MT
4
or both for 24 h, and then examined by confocal microscopy. RV caused nuclear accumulation of COX-2, indicated by the appearance
of a yellow color due to superimposition of COX-2 (green) and nuclear (red) images. In the presence of T
4
, however, nuclear COX-2 was not evident in RV-treated
cells, although cytosolic COX-2 is still seen in some cells. Cells were viewed at 250 magnification. (B) U87MG cells (left panel) and GL261 cells (right panel)
were treated with 10 lM RV in the presence or absence of 10
7
MT
4
for 24 h. RV induced nuclear COX-2 and cytosolic pro-apoptotic protein BcLx-s
accumulation in both cell lines, and T
4
inhibited those effects of the stilbene (P,0.01).
Apoptosis in glioma cells
67
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
the stilbene effects on COX-2, pSer15–p53 and BcLx-s were in-
hibited, although pERK1/2 activation remained. In contrast, when
tetrac was added to both RV and T
4
, the inhibitory effects of T
4
on
RV action were blocked (lane 8), but the changes with RV were
similar to those shown in lane 5 with RV, alone, and with RV and
tetrac (lane 6).
Further, development of the immunoprecipitable nuclear complex of
inducible COX-2 and activated ERK1/2 in RV-treated cells (Figure 6,
lane 5) was inhibited by co-incubation with RV and T
4
(lane 7),
whereas T
4
alone did not induce this nuclear association (lane 3).
With the addition of tetrac, the inhibitory effect of T
4
on COX-2
and pERK1/2 complex formation was reversed (Figure 6, lane 8).
These results suggest that the nuclear immunoprecipitable complex
formation between pERK1/2 and COX-2 plays a crucial role in RV-
induced apoptosis. Further, this complex formation can be disrupted
by thyroid hormone, leading to inhibition of RV-induced pro-
apoptotic protein BcLx-s accumulation (Figure 6, lane 7) and apopto-
sis (Figure 1).
As indicated above, we know that both thyroid hormone and RV have
receptor sites on integrin aVb3 and that both hormone and stilbene
activate ERK1/2. The antagonism by T
4
of the action of RVon signal-
ing events that promote apoptosis and the reversal by tetrac of this
hormone effect, but not the activity of RV, point to the existence of
discrete hormone- and stilbene-binding sites on integrin aVb3.
Discussion
RV is a stilbene that has antitumor properties in glial cells (8). Using
a variety of human cancer cell lines, we have demonstrated that RV is
capable of inducing p53-dependent apoptosis via ERK1/2-mediated
serine phosphorylation at residue 15 of the oncogene suppressor pro-
tein (10–13). This is the case even when the cancer cells contain
mutated p53, as long as residue 15 remains a serine (12). We have
recently shown, however, that this clinically desirable action of
RV may be antagonized by estrogen in estrogen-responsive (ERa-
positive) tumor cells, such as MCF-7 breast cancer cells (10).
We have also determined that the growth of glioma cells in vitro is
importantly thyroid hormone dependent (1). We have shown previ-
ously that both T
4
and RVare ligands of integrin aVb3 (5,9) and both
activate ERK1/2 via the integrin. Both receptors are at or near the
RGD recognition site on the integrin, but our current findings do not
disclose direct competition between thyroid hormone and RV for
a single site on integrin aVb3. That is, tetrac, a specific inhibitor of
binding of T
4
to the integrin and of downstream actions of the hor-
mone initiated at the integrin (1,5) did not block the actions of RV.
We have identified a step downstream of ERK1/2 in the transduc-
tion of the RV and thyroid hormone signals at which a divergence
occurs that appears to account for the different biological effects of T
4
on cell growth and of RV leading to apoptosis. RV increases nuclear
abundance of COX-2, whereas T
4
, alone, has no effect on nuclear
COX-2 (Figures 5 and 6). However, the hormone decreases formation
of nuclear complexes between pERK1/2 and COX-2 in stilbene-
treated cells (Figure 6) and also inhibits RV-induced p53 phosphory-
lation. How this signal transduction step (nuclear complexing of
COX-2 and activated ERK1/2, upstream of activation of p53) is dif-
ferentially affected by T
4
and RV is not yet clear. Among the possible
explanations is that the pools of activated ERK1/2 that result from the
transduction of stilbene and thyroid hormone signals are discrete (23).
For example, the pro-apoptotic response of C6 cells to RV treatment
was blocked in vitro by physiological concentrations of thyroid hor-
mone. In the intact animal test model, normal circulating levels of
thyroid hormone may blunt an apparent response to the stilbene. Since
tetrac does not block stilbene action, however, a potential therapeutic
combination of tetrac—to block endogenous T
4
action at the
integrin—and RV would allow an unopposed pro-apoptotic effect of
the stilbene at its integrin receptor to be obtained.
Funding
Office of Research Development, Medical Research Service, Depart-
ment of Veterans Affairs Merit Award to H.-Y. Lin (5317-002) and P.J.
Davis (4999-0002); Charitable Leadership Foundation and Beltrone
Foundation.
Acknowledgements
The authors appreciate the contributions of Cassie Lin to the work described.
Conflict of Interest Statement: None declared.
References
1. Davis,F.B. et al. (2006) Acting via a cell surface receptor, thyroid hormone
is a growth factor for glioma cells. Cancer Res.,66, 7270–7275.
Fig. 6. Effects of T
4
and tetrac on RV-induced ERK1/2 activation,
complexing of pERK1/2 and COX-2, serine-15–p53 phosphorylation and
apoptosis in C6 cells. Nuclear accumulation of pERK1/2 was stimulated by
either T
4
(lane 3) or RV (lane 5) applied separately to cells. Tetrac partially
blocked accumulation of pERK1/2 in the presence of T
4
(lane 4), but did not
block the activation of ERK1/2 by RV (lane 6). Nuclear COX-2 accumulation
was not increased by T
4
, alone (lane 3), but was stimulated by RV, along with
accumulation of nuclear pSer15–p53 and pERK1/2, and cytosolic pro-
apoptotic protein BcLx-s (lane 5). These latter effects of the stilbene were
inhibited by T
4
(comparing lanes 5 and 7, P,0.01) except for pERK1/2
accumulation, which is also stimulated by T
4
. Inhibition of the RV effect by
T
4
, however, was blocked by tetrac (comparing lanes 7 and 8, P,0.05 for
BcLx-s and P,0.01 for changes in COX-2, pSer15–p53 and complexing of
pERK1/2 and COX-2), implicating the cell-surface T
4
integrin receptor in
this action of the hormone. Tetrac, in the absence of T
4
, had no effect on the
actions of RV (lanes 5 and 6). Lamin-B and b-actin immunoblots served as
gel-loading controls for nuclear and cytosol proteins, respectively.
H.-Y.Lin et al.
68
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
2. Plow,E.F. et al. (2000) Ligand binding to integrins. J. Biol. Chem.,275,
21785–21788.
3. Arnaout,M.A. et al. (2002) Coming to grips with integrin binding to li-
gands. Curr. Opin. Cell Biol.,14, 641–651.
4. Xiong,J.P. et al. (2003) Integrins, cations and ligands: making the connec-
tion. J. Thromb. Haemost.,1, 1642–1654.
5. Bergh,J.J. et al. (2005) Integrin alphavbeta3 contains a cell surface receptor
site for thyroid hormone that is linked to activation of mitogen-activated
protein kinase and induction of angiogenesis. Endocrinology,146, 2864–2871.
6. Aggarwal,B.B. et al. (2004) Role of resveratrol in prevention and therapy of
cancer: preclinical and clinical studies. Anticancer Res.,24, 2783–2840.
7. Jang,M. et al. (1997) Cancer chemopreventive activity of resveratrol, a nat-
ural product derived from grapes. Science,275, 218–220.
8. Tseng,S.H. et al. (2004) Resveratrol suppresses the angiogenesis and tumor
growth of gliomas in rats. Clin. Cancer Res.,10, 2190–2202.
9. Lin,H.Y. et al. (2006) Integrin alphavbeta3 contains a receptor site for
resveratrol. FASEB J.,20, 1742–1744.
10. Zhang,S. et al. (2004) Oestrogen inhibits resveratrol-induced post-
translational modification of p53 and apoptosis in breast cancer cells.
Br. J. Cancer,91, 178–185.
11. Lin,H.Y. et al. (2002) Resveratrol induced serine phosphorylation of p53
causes apoptosis in a mutant p53 prostate cancer cell line. J. Urol.,168,
748–755.
12. Shih,A. et al. (2004) Inhibitory effect of epidermal growth factor on
resveratrol-induced apoptosis in prostate cancer cells is mediated by protein
kinase C-alpha. Mol. Cancer Ther.,3, 1355–1364.
13. Shih,A. et al. (2002) Resveratrol induces apoptosis in thyroid cancer cell
lines via a MAPK- and p53-dependent mechanism. J. Clin. Endocrinol.
Metab.,87, 1223–1232.
14. She,Q.B. et al. (2002) Involvement of c-Jun NH2-terminal kinases in
resveratrol-induced activation of p53 and apoptosis. Mol. Carcinog.,33,
244–250.
15. Kundu,J.K. et al. (2006) Resveratrol inhibits phorbol ester-induced expres-
sion of COX-2 and activation of NF-kappaB in mouse skin by blocking
I-kappaB kinase activity. Carcinogenesis,27, 1465–1474.
16. Tang,H.Y. et al. (2004) Thyroid hormone causes mitogen-activated protein
kinase-dependent phosphorylation of the nuclear estrogen receptor. Endo-
crinology,145, 3265–3272.
17. Lin,H.Y. et al. (1999) Thyroid hormone induces activation of mitogen-
activated protein kinase in cultured cells. Am. J. Physiol.,276, C1014–
C1024.
18. Lin,H.Y. et al. (2007) Thyroid hormone is a MAPK-dependent growth
factor for thyroid cancer cells and is anti-apoptotic. Steroids,72, 180–187.
19. Tang,H.Y. et al. (2006) Resveratrol-induced cyclooxygenase-2 facilitates
p53-dependent apoptosis in human breast cancer cells. Mol. Cancer Ther.,
5, 2034–2042.
20. Shih,A. et al. (2001) Thyroid hormone promotes serine phosphoryla-
tion of p53 by mitogen-activated protein kinase. Biochemistry,40, 2870–
2878.
21. Salgame,P. et al. (1997) An ELISA for detection of apoptosis. Nucleic
Acids Res.,25, 680–681.
22. Davis,P.J. et al. (2000) Thyroxine promotes association of mitogen-
activated protein kinase and nuclear thyroid hormone receptor (TR) and
causes serine phosphorylation of TR. J. Biol. Chem.,275, 38032–38039.
23. Davis,P.J. et al. (2005) Membrane receptors mediating thyroid hormone
action. Trends Endocrinol. Metab.,16, 429–435.
Received July 16, 2007; revised October 3, 2007; accepted October 19, 2007
Apoptosis in glioma cells
69
by guest on December 28, 2010carcin.oxfordjournals.orgDownloaded from
... Yet, this have been reported to occur only at specific concentrations (1 pM−10 µM) and for short-term activation (Lin et al., 2011b). Being confirmed as the main mechanism in breast, prostate, ovarian, glial, head, and neck cancer cells (Lin et al., 2002(Lin et al., , 2008a(Lin et al., ,b, 2011aZhang et al., 2004;Yang et al., 2011), other researchers have also shown that in addition to MAPK, resveratrol also intervenes in p38 kinase and cJun N-terminal kinase (Dong, 2003). Interestingly, it was also reported that the activated form of ERK1/2 induces the phosphorylation of p53 in several human cancer cell lines (Lin et al., 2011b), with resveratrol being able to induce p53-independent apoptosis. ...
... Indeed, the p53 oncogene suppressor protein is involved in apoptosis, having the ability to be coupled to DNA in its active form. In addition, resveratrol at a dose of 10 µM is able to boost the nuclear accumulation of COX-2 by p53-dependent apoptosis in breast, prostate, ovarian, head, and neck cancer cells (Lin et al., 2008a(Lin et al., ,b, 2011a. Specifically, the inducible COX-2 associated with small ubiquitin-related modifier-1 (SUMO-1) is transported to the nucleus and forms a complex with phosphorylated p53, and phosphorylated ERK1/2 and p300. ...
Article
Full-text available
Resveratrol is a polyphenolic stilbene derivative widely present in grapes and red wine. Broadly known for its antioxidant effects, numerous studies have also indicated that it exerts anti-inflammatory and antiaging abilities and a great potential in cancer therapy. Regrettably, the oral administration of resveratrol has pharmacokinetic and physicochemical limitations such as hampering its effects so that effective administration methods are demanding to ensure its efficiency. Thus, the present review explores the published data on the application of resveratrol nanoformulations in cancer therapy, with the use of different types of nanodelivery systems. Mechanisms of action with a potential use in cancer therapy, negative effects, and the influence of resveratrol nanoformulations in different types of cancer are also highlighted. Finally, the toxicological features of nanoresveratrol are also discussed.
... The S2 domain has a higher affinity for T4 than the S1 or S2 sites have for T3. A trophic effect of T4 mediated through the membrane integrin avb3 has been shown experimentally in vitro and in vivo in cancer cells, leading to induction of proliferation (19,20,(30)(31)(32)(33)(34) and inhibition of apoptosis (32). Tetrac, the deaminated analogue of T4, blocks thyroid hormone-binding at the integrin and inhibits the ability of thyroid hormone analogues to activate MAPK (20). ...
... The S2 domain has a higher affinity for T4 than the S1 or S2 sites have for T3. A trophic effect of T4 mediated through the membrane integrin avb3 has been shown experimentally in vitro and in vivo in cancer cells, leading to induction of proliferation (19,20,(30)(31)(32)(33)(34) and inhibition of apoptosis (32). Tetrac, the deaminated analogue of T4, blocks thyroid hormone-binding at the integrin and inhibits the ability of thyroid hormone analogues to activate MAPK (20). ...
Article
Full-text available
Background Thyroid hormones (TH), T4 and T3, mediate pro-mitogenic effects in cancer cells through binding the membrane receptor αvβ3 integrin. The deaminated analogue tetrac effectively blocks TH binding to this receptor and prevents their action. While computational data on TH binding to the αvβ3 integrin was published, a comprehensive analysis of additional TH metabolites is lacking. Methods In-silico docking of 26 TH metabolites, including the biologically active thyroid hormones (T3 and T4) and an array of sulfated, deiodinated, deaminated or decarboxylated metabolites, to the αvβ3 receptor binding pocket was performed using DOCK6, based on the three-dimensional representation of the crystallographic structure of the integrin. As the TH binding site upon the integrin is at close proximity to the well-defined RGD binding site, linear and cyclic RGD were included as a reference. Binding energy was calculated for each receptor-ligand complex using Grid score and Amber score with distance movable region protocol. Results All TH molecules demonstrated negative free energy, suggesting affinity to the αvβ3 integrin. Notably, based on both Grid and Amber scores sulfated forms of 3,3’ T2 (3,3’ T2S) and T4 (T4S) demonstrated the highest binding affinity to the integrin, compared to both cyclic RGD and an array of examined TH metabolites. The major thyroid hormones, T3 and T4, showed high affinity to the integrin, which was superior to that of linear RGD. For all hormone metabolites, decarboxylation led to decreased affinity. This corresponds with the observation that the carboxylic group mediates binding to the integrin pocket via divalent cations at the metal-ion-dependent adhesion (MIDAS) motif site. A similar reduced affinity was documented for deaminated forms of T3 (triac) and T4 (tetrac). Lastly, the reverse forms of T3, T3S, and T3AM showed higher Amber scores relative to their native form, indicating that iodination at position 5 is associated with increased binding affinity compared to position 5’. Summary Three-dimensional docking of various TH metabolites uncovered a structural basis for a differential computational free energy to the αvβ3 integrin. These findings may suggest that naturally occurring endogenous TH metabolites may impact integrin-mediate intracellular pathways in physiology and cancer.
... Although resveratrol can induce apoptosis in tumoral cells [111][112][113][114][115][116], it can also protect normal cells/tissues during cancer chemotherapy via its anti-apoptotic effects [117][118][119][120]. In this regard, resveratrol upregulates Bcl-xL and Bcl-2 expressions [117,121,122] and downregulates BAX, caspase-3, p53, and PARP expressions in the normal cells [122][123][124][125][126]. The findings pre-sented in the current systematic review also showed that the combined treatment of resveratrol and cisplatin declined the apoptosis level of inner ear cells in comparison with the cisplatin-treated groups alone [43,48]. ...
Article
Introduction: Although cancer treatment with cisplatin is effective, dose-dependent adverse effects such as ototoxicity occurs often, which limits its clinical use. The use of resveratrol may alleviate the cisplatin-induced ototoxic effects. This study is aimed to review the potential otoprotective effects of resveratrol against cisplatin-induced ototoxicity. Method: According to the PRISMA guideline, a systematic search was accomplished to identify all relevant scientific papers on "the role of resveratrol against cisplatin-induced ototoxicity" in different electronic databases up to May 2021. Fifty-five articles were screened based on a pre-defined set of inclusion and exclusion criteria. Eight eligible studies were finally included in the current systematic review. The in-vitro findings revealed that cisplatin administration significantly decreased the HEI-OC1 cell viability compared to the untreated cells; however, resveratrol co-treatment (in a dose-dependent manner) could protect HEI-OC1 cells against cisplatin-induced decrease in cell viability. Results: Furthermore, the in-vivo finding showed a decreased value of DPOAE, and increased values of ABR threshold, ABR-I, ABR-IV, and ABR I-IV interval in cisplatin-treated animals; in contrast, resveratrol co-administration demonstrated an opposite pattern on these parameters. Conclusion: Thus, it can be mentioned that resveratrol co-treatment alleviates cisplatin-induced ototoxicity. Mechanically, resveratrol exerts its otoprotective effects through various mechanisms such as anti-oxidant, anti-apoptosis, and anti-inflammatory.
... By interacting with mitochondria, it induces an imbalance in the activities of endogenous antioxidants resulting in an increased accumulation of ROS and lipid peroxides in cancer cells. Increased accumulation of ROS and lipid peroxide can induce OS in glioma cells and endorses apoptosis [60,137,138]. Thus, resveratrol can serve as a potential chemotherapeutic agent to treat brain tumors [139]. ...
Article
Full-text available
Brain cancer is regarded among the deadliest forms of cancer worldwide. The distinct tumor microenvironment and inherent characteristics of brain tumor cells virtually render them resistant to the majority of conventional and advanced therapies. Oxidative stress (OS) is a key disruptor of normal brain homeostasis and is involved in carcinogenesis of different forms of brain cancers. Thus, antioxidants may inhibit tumorigenesis by preventing OS induced by various oncogenic factors. Antioxidants are hypothesized to inhibit cancer initiation by endorsing DNA repair and suppressing cancer progression by creating an energy crisis for preneoplastic cells, resulting in antiproliferative effects. These effects are referred to as chemopreventive effects mediated by an antioxidant mechanism. In addition, antioxidants minimize chemotherapy-induced nonspecific organ toxicity and prolong survival. Antioxidants also support the prooxidant chemistry that demonstrate chemotherapeutic potential, particularly at high or pharmacological doses and trigger OS by promoting free radical production, which is essential for activating cell death pathways. A growing body of evidence also revealed the roles of exogenous antioxidants as adjuvants and their ability to reverse chemoresistance. In this review, we explain the influences of different exogenous and endogenous antioxidants on brain cancers with reference to their chemopreventive and chemotherapeutic roles. The role of antioxidants on metabolic reprogramming and their influence on downstream signaling events induced by tumor suppressor gene mutations are critically discussed. Finally, the review hypothesized that both pro- and antioxidant roles are involved in the anticancer mechanisms of the antioxidant molecules by killing neoplastic cells and inhibiting tumor recurrence followed by conventional cancer treatments. The requirements of pro- and antioxidant effects of exogenous antioxidants in brain tumor treatment under different conditions are critically discussed along with the reasons behind the conflicting outcomes in different reports. Finally, we also mention the influencing factors that regulate the pharmacology of the exogenous antioxidants in brain cancer treatment. In conclusion, to achieve consistent clinical outcomes with antioxidant treatments in brain cancers, rigorous mechanistic studies are required with respect to the types, forms, and stages of brain tumors. The concomitant treatment regimens also need adequate consideration.
... Some studies have reported that resveratrol can induce apoptosis in various malignant tumor cells, such as glioma [102], prostate [103], gastric [104], breast [105], head and neck [106], and ovarian [107] cancer cells. The apoptotic activation of resveratrol is linked to induce ROS production, various caspases activation, mitochondrial membrane permeability, BAX and p53 activation, etc. [108][109][110][111][112][113]. ...
Article
Full-text available
Purpose 5-fluorouracil (5-FU), an effective chemotherapy drug, is commonly applied for colorectal cancer treatment. Nevertheless, its toxicity to normal tissues and the development of tumor resistance are the main obstacles to successful cancer chemotherapy and hence, its clinical application is limited. The use of resveratrol can increase 5-FU-induced cytotoxicity and mitigate the unwanted adverse effects. This study aimed to review the potential therapeutic effects of resveratrol in combination with 5-FU against colorectal cancer. Methods According to the PRISMA guideline, a comprehensive systematic search was carried out for the identification of relevant literature in four electronic databases of PubMed, Web of Science, Embase, and Scopus up to May 2021 using a pre-defined set of keywords in their titles and abstracts. We screened 282 studies in accordance with our inclusion and exclusion criteria. Thirteen articles were finally included in this systematic review. Results The in vitro findings showed that proliferation inhibition of colorectal cancer cells in the groups treated by 5-FU was remarkably higher than the untreated groups and the co-administration of resveratrol remarkably increased cytotoxicity induced by 5-FU. The in vivo results demonstrated a decrease in tumor growth of mice treated by 5-FU than the untreated group and a dramatic decrease was observed following combined treatment of resveratrol and 5-FU. It was also found that 5-FU alone and combined with resveratrol could regulate the cell cycle profile of colorectal cancer cells. Moreover, this chemotherapeutic agent induced the biochemical and histopathological changes in the cancerous cells/tissues and these alterations were synergized by resveratrol co-administration (for most of the cases), except for the inflammatory mediators. Conclusion The results obtained from this systematic review demonstrated that co-administration of resveratrol could sensitize the colorectal cancer cells to 5-FU treatment via various mechanisms, including regulation of cell cycle distribution, oxidant, apoptosis, anti-inflammatory effects.
... The induction of apoptosis in cancer cells by tetrac or chemically modified tetrac involves multiple intrinsic and extrinsic mechanisms (9,14,32). Specific serine phosphorylation of p53 is a critical feature of the intrinsic pathway ( Figure 4) (9), which has been studied as a biochemical site of competition between T4 and tetrac (32)(33)(34)(35). The extrinsic pathway depends on activation of caspases ( Figure 4). ...
Article
Full-text available
Apoptosis is induced in cancer cells and tumor xenografts by the thyroid hormone analogue tetraiodothyroacetic acid (tetrac) or chemically modified forms of tetrac. The effect is initiated at a hormone receptor on the extracellular domain of plasma membrane integrin αvβ3. The tumor response to tetrac includes 80% reduction in size of glioblastoma xenograft in two weeks of treatment, with absence of residual apoptotic cancer cell debris; this is consistent with efferocytosis. The molecular basis for efferocytosis linked to tetrac is incompletely understood, but several factors are proposed to play roles. Tetrac-based anticancer agents are pro-apoptotic by multiple intrinsic and extrinsic pathways and differential effects on specific gene expression, e.g., downregulation of the X-linked inhibitor of apoptosis (XIAP) gene and upregulation of pro-apoptotic chemokine gene, CXCL10. Tetrac also enhances transcription of chemokine CXCR4, which is relevant to macrophage function. Tetrac may locally control the conformation of phagocyte plasma membrane integrin αvβ3; this is a cell surface recognition system for apoptotic debris that contains phagocytosis signals. How tetrac may facilitate the catabolism of the engulfed apoptotic cell debris requires additional investigation.
Article
Full-text available
Oral cancer is a fatal disease, and its incidence in Taiwan is increasing. Thyroid hormone as L-thyroxine (T4) stimulates cancer cell proliferation via a receptor on integrin αvβ3 of plasma membranes. It also induces the expression of programmed death-ligand 1 (PD-L1) and cell proliferation in cancer cells. Thyroid hormone also activates β-catenin-dependent cell proliferation in cancer cells. However, the relationship between PD-L1 and cancer proliferation is not fully understood. In the current study, we investigated the role of inducible thyroid hormone-induced PD-L1-regulated gene expression and proliferation in oral cancer cells. Thyroxine bound to integrin αvβ3 to induce PD-L1 expressions via activation of ERK1/2 and signal transducer and activator of transcription 3 (STAT3). Inactivated STAT3 inhibited PD-L1 expression and nuclear PD-L1 accumulation. Inhibition of PD-L1 expression reduced β-catenin accumulation. Furthermore, nuclear PD-L1 formed a complex with nuclear proteins such as p300. Suppression PD-L1 expression by shRNA blocked not only expression of PD-L1 and β-catenin but also signal transduction, proliferative gene expressions, and cancer cell growth. In summary, thyroxine via integrin αvβ3 activated ERK1/2 and STAT3 to stimulate the PD-L1-dependent and β-catenin-related growth in oral cancer cells.
Article
Background: Nicotine leads to reproductive changes culminating in male infertility and subfertility. Resveratrol, a polyphenol, is a biological modulator. Sirtuin 1 (SIRT1) protein can positively act on male reproduction and its expression can be affected by nicotine and modulated by the resveratrol. Objectives: The capability of the resveratrol to reverse the reproductive damage in adult male offspring, which was nicotine-exposed during intrauterine phase and breastfeeding, was investigated. Materials and methods: Four groups were established from male offspring born from nicotine-exposed and non-exposed rat dams during pregnancy and lactation. Forty-eight male Wistar rats were distributed into four groups: Sham Control (SC), Resveratrol (R), Nicotine (N), Nicotine + Resveratrol (NR). Rat dams of the N and NR offspring were exposed to nicotine (2 mg/kg/day) during pregnancy and lactation, using a minipump subcutaneously implanted. The offspring of R and NR groups received resveratrol (300 mg/kg of body weight, gavage) for 63 days, from puberty. At 114 days of age, the male rats were euthanized . Results: Nicotine did not alter body weight, biometry of reproductive organs and quantitative sperm parameters of adult offspring but caused an evident worsening of all sperm qualitative parameters studied. Daily treatment with resveratrol from puberty up to adulthood promoted significant improvement of all qualitative sperm parameters, leading some of them close to the control values. Resveratrol also improved the morphological integrity and expression of SIRT1 in the seminiferous epithelium of nicotine-exposed offspring. Conclusion and discussion: Nicotine crosses blood-placental membrane and is present in breast milk of smoker mothers. Resveratrol restored the altered reproductive parameters in the male adult offspring that was nicotine-exposed during intrauterine life and breastfeeding. Epigenetic modulating action of resveratrol can be involved in this nicotine damage reversion. Resveratrol may be a promising candidate to be investigated regarding the adjuvant strategies in treatments of male infertility. This article is protected by copyright. All rights reserved.
Article
Acetaminophen (APAP) is a painkiller that can cause hepatotoxicity if taken in excess. We investigated the effect of pu-erh tea extract (PTE) on hepatotoxicity induced by excess APAP using physiological, metabolomic, and transcriptomic analyses. PTE decreased levels of oxidative stress, inflammatory response, and apoptosis markers induced by excess APAP. And 156 metabolites and 703 genes were identified as differentially expressed metabolites and differentially expressed genes, respectively. KEGG enrichment analysis revealed that PTE and overdose of APAP altered tyrosine, caffeine, and amino acid-related metabolism. Six differentially expressed metabolites associated with these pathways have hepatoprotective effects and were upregulated by PTE pretreatment. The expression levels of 10 vital differentially expressed genes regulating these metabolites were verified by qRT-PCR. The findings confirm the beneficial role of PTE pretreatment in alleviating the hepatotoxicity caused by overdose of APAP, indicating that PTE can be used as an effective dietary supplement for the development of functional foods.
Article
Resveratrol, a phytoalexin found in grapes and other food products, was purified and shown to have cancer chemopreventive activity in assays representing three major stages of carcinogenesis. Resveratrol was found to act as an antioxidant and antimutagen and to induce phase II drug-metabolizing enzymes (anti-initiation activity); it mediated anti-inflammatory effects and inhibited cyclooxygenase and hydroperoxidase functions (antipromotion activity); and it induced human promyelocytic leukemia cell differentiation (antiprogression activity). In addition, it inhibited the development of preneoplastic lesions in carcinogen-treated mouse mammary glands in culture and inhibited tumorigenesis in a mouse skin cancer model. These data suggest that resveratrol, a common constituent of the human diet, merits investigation as a potential cancer chemopreventive agent in humans.
Article
Diphenylhydantoin (DPH, phenytoin sodium, Dilantin) inhibited the growth of cultured human astrocytoma cells in 7 of the 10 cell lines studied. This inhibition, determined by a microtiter assay, was dose-dependent; DPH levels of 20 micrograms/ml and above produced significant depression of growth in astrocytoma cultured cells. However, normal cultured human astrocytes were not affected until DPH levels of 60 micrograms/ml and above were added to the cells; normal fibroblasts also showed no growth inhibition up to 100 micrograms/ml. We have confirmed that DPH is 1.5 times as concentrated in tumor tissue as it is in normal tissue and serum. These findings suggest that DPH has properties that inhibit the growth of human astrocytoma cells in tissue culture at levels that are achievable clinically.
Article
Cultured P388 cells derived from leukemia P388 passaged in vivo in (C57BL X DBA/2)F1 mice were approximately one-tenth less sensitive to N-trifluoroacetyladriamycin-14-valerate (AD32) and cinerubin A, inasmuch as the concentrations of these agents had to be increased tenfold to produce a reduction in cell viability of the same order of magnitude as that produced by adriamycin, daunomycin, or actinomycin D. Cultured P388 cells derived from resistant cell populations developed in vivo retained their resistance to vincristine. These resistant cells exhibited cross-resistance to actinomycin D, adriamycin, daunomycin, and AD32, but cross-resistance to cinerubin A could not be detected. Data also showed that the vincristine-resistant cells were more sensitive to AD32 (2.8 x 10(-6) M) than to adriamycin (3.4 x 10(-6) M) during a 6-hour exposure of cells to agents.
Article
5,5-Diphenylhydantoin or 5-(4-hydroxyphenyl)-5-phenylhydantoin, 3.6 X 10(-4) M, inhibited polymerization of purified pig brain tubulin 50% as measured by viscosometric assay and estimation of tubulin length by electron microscopy. 5,5-Diphenylhydantoin did not bind to the low molecular weight fraction of tubulin or to guanosinetriphosphate; it displaced colchicine weakly from its binding site. 5,5-Diphenylhydantoin did not depolymerize established microtubules, indicating that it differs from colchicine in site and mode of action.
Article
The overexpression of P-glycoprotein (PGP) and alterations in DNA topoisomerase II (TOPO II) were evaluated in mouse leukemia P388 cells selected in vivo for mitoxantrone (MTT) resistance (P388/MTT) and compared to doxorubicin (DOX) resistant (P388/DOX) or vincristine (VCR) resistant (P388/VCR) models. Among a panel of TOPO II inhibitors which included etoposide (VP-16), DOX, MTT and 4'-[(9-acridinyl)-amino]methanesulfon-m-anisidide (m-AMSA), the relative resistance compared to parental sensitive P388/S cells was: P388/DOX greater than P388/MTT greater than P388/VCR. All the resistant sublines exhibited minimal cell kill (less than 20%) at vincristine concentrations greater than 100-fold the IC50 for P388/S cells. In a soft-agar colony-forming assay, the modulation of cytotoxicity in P388/MTT cells by the calmodulin inhibitor trifluoperazine following a 3-hr drug treatment demonstrated a marked potentiation in cell kill with MTT, VP-16, DOX and m-AMSA but not VCR. Immunoblotting data revealed that while PGP was not detectable in P388/S cells, the overexpression of PGP was apparent in P388/MTT cells and the relative expression between the resistant sublines was: P388/DOX greater than P388/MTT greater than P388/VCR. Although the amount and DNA cleavage activity of TOPO II in nuclear extracts from P388/VCR cells were comparable to those in P388/S cells, they were markedly lower in both P388/DOX and P388/MTT cells. However, decatenation activity of TOPO II in nuclear extracts was comparable between the sensitive (P388/S) and resistant sublines (P388/MTT, P388/DOX, and P388/VCR). Results from the present study demonstrated that P388 cells selected for resistance to mitoxantrone exhibit changes in TOPO II and overexpression of PGP similar to P388/DOX cells, while vincristine resistant cells only overexpress PGP. Since therapeutic strategies are primarily designed to interfere with PGP-mediated drug efflux, the choice of agents for modulating resistance in tumors which overexpress PGP versus tumors which overexpress PGP with altered TOPO II could be different.
Article
Tumor cell resistance due to enhanced efflux of drugs with diverse structures and/or mechanisms of action is termed multidrug resistance (MDR), and modulation of the MDR phenotype by calcium blockers or calmodulin inhibitors is suggested to involve P-glycoprotein. In drug-sensitive (S) and 5-fold doxorubicin (DOX)-resistant (R0) L1210 mouse leukemia cells, no obvious differences in mdr mRNA or P-glycoprotein expression or alterations in cellular uptake, retention, or cytotoxicity of vincristine (VCR) were observed. However, in the 10-fold (R1) and 40-fold (R2) DOX-resistant sublines, expression of P-glycoprotein was correlated with the level of resistance (R2 greater than R1). An RNase protection assay revealed that elevated levels of mdr1 and mdr2 mRNA were detected in R1 and R2 cells, with an additional increase in mdr3 mRNA in the R2 subline. Further, in the R1 and R2 sublines, no VCR dose-dependent cytotoxicity was apparent, and cell kill of greater than 40% was not achievable following a 3-hr drug exposure. Cellular uptake and retention of VCR were 2- to 4-fold lower in the R1 and R2 sublines, compared with similarly treated S or R0 cells. Potentiation of VCR cytotoxicity by a noncytotoxic concentration of 5 microM trifluoperazine (TFP) was greater than 2-fold in S and R0 cells and less than 1.3-fold in the R1 and R2 sublines. Modulation of VCR uptake by 5 microM TFP in the S and R0 cells was 2-fold and it was 4- to 7-fold in the R1 and R2 sublines. The presence of 5 microM TFP, by competing for efflux, enhanced VCR retention 1.5-fold in S and R0 cells and 2- to 4-fold in the R1 and R2 sublines. In contrast to these results with VCR, dose-dependent cytotoxicity of DOX was apparent in all the resistant sublines, and modulation of DOX cytotoxicity by 5 microM TFP was dependent on the level of resistance. Cellular accumulation of DOX was 20 and 50% lower in the R1 and R2 sublines, respectively, compared with similarly treated S or R0 cells. Marked increases (greater than 1.5-fold) in cellular accumulation of DOX by TFP were apparent only in the R2 subline. Results suggest that a relationship between overexpression of P-glycoprotein isoforms and their role in affecting cellular drug levels and consequent cytotoxicity in MDR L1210 cells determines resistance to VCR but not DOX.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
The results from the present study using the sensitive and progressively DOX resistant L1210 model system demonstrated that the effects of TFP are not due to redistribution of DOX to the nucleus, and modulation of cytotoxicity is related to effects on DOX-induced DNA strand breaks. Although TFP affects phosphorylation of PGP and TOPO II (R2 greater than R1), the comparable DNA strand breaks at lower DOX levels with TFP in the resistant sublines suggest that modulation of TOPO II function related to drug-induced DNA damage by calmodulin-mediated events may be an important mode of action.