Sei sulla pagina 1di 9

Carcinogenesis vol.26 no.11 pp.

1956–1964, 2005
doi:10.1093/carcin/bgi157
Advance Access publication June 15, 2005

Lupeol, a fruit and vegetable based triterpene, induces apoptotic death of human
pancreatic adenocarcinoma cells via inhibition of Ras signaling pathway

Mohammad Saleemy, Satwinderjeet Kaury, States in 2005 and almost the same number of pancreatic
Mee-Hyang Kweony, Vaqar Mustafa Adhami, cancer-related deaths are projected (1). Since the mortality
Farrukh Afaq and Hasan Mukhtar from pancreatic cancer compares strikingly with its incidence,
Department of Dermatology, University of Wisconsin, Madison, WI, USA
it has become a significant public health concern (1). Long-

term survival of patients with organ-confined disease is only
To whom correspondence should be addressed at Department of 15%, and in the majority of cases with invasive metastasis
Dermatology, University of Wisconsin, 1300 University Avenue, Medical
Sciences Center, B-25, Madison, WI, 53706, USA. Tel: þ1 608 263 3927, survival is only 4%. Despite these disappointing statistics and
Fax: þ1 608 263 5223. the increase in incidence of pancreatic cancer over the past

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Email: hmukhtar@wisc.edu several decades, the molecular and biochemical determinants
Pancreatic cancer is an exceptionally aggressive disease, of the disease remain poorly understood and no effective
the treatment of which has largely been unsuccessful due therapeutic regimens to significantly ameliorate the clinical
to higher resistance offered by pancreatic cancer cells to course or prognosis of this disease is apparent (1).
conventional approaches such as surgery, radiation and/or Thus, pancreatic cancer presents a clinical and experimental
chemotherapy. The aberration of Ras oncoprotein has been challenge because of its relative resistance to conventional
linked to the induction of multiple signaling pathways and modes of therapies such as chemotherapy and radiation and
to the resistance offered by pancreatic cancer cells to apop- only 4% of such patients are reported to survive after surgery
tosis. Therefore, there is a need for development of new and (1). It is therefore necessary to intensify our efforts for a better
effective chemotherapeutic agents which can target mul- understanding of this disease and for the development of novel
tiple pathways to induce responsiveness of pancreatic can- approaches for its prevention and treatment. In more than
cer cells to death signals. In this study, human pancreatic 90% of pancreatic cancers, the ras oncogene has been shown
adenocarcinoma cells AsPC-1 were used to investigate the to be mutated [(2,3) and references therein]. This mutation is
effect of Lupeol on cell growth and its effects on the modu- considered an early genetic event in the development of
lation of multiple Ras-induced signaling pathways. Lupeol pancreatic cancer and results in constitutive activation of an
caused a dose-dependent inhibition of cell growth as intracellular pathway leading to cellular proliferation. Several
assessed by MTT assay and induction of apoptosis as ras-induced signaling pathways, such as the phosphatidylino-
assessed by flow cytometry, fluorescence microscopy and sitol 3-kinase (PI3K)/Akt (protein kinase B pathway) mitogen-
western blotting. Lupeol treatment to cells was found to activated protein kinases (MAPKs) and nuclear factor kappa B
significantly reduce the expression of Ras oncoprotein and (NFkB) pathways, have been linked to chemo- resistance of
modulate the protein expression of various signaling the pancreatic carcinoma cells (3–6). These findings suggest
molecules involved in PKCa/ODC, PI3K/Akt and that Ras oncoprotein could be an important target for devel-
MAPKs pathways along with a significant reduction in the oping agents against pancreatic cancer.
activation of NFkB signaling pathway. Our data suggest Epidemiological studies have found a lower risk of pancre-
that Lupeol can adopt a multi-prong strategy to target atic cancer in populations with higher consumption of fresh
multiple signaling pathways leading to induction of apop- fruits, vitamin C and dietary fiber (7,8). In addition, consump-
tosis and inhibition of growth of pancreatic cancer cells. tion of white wine has been reported to have an inverse relation
Lupeol could be a potential agent against pancreatic can- with the risk of pancreatic cancer (9). These data suggest that
cer, however, further in-depth in vivo studies are some dietary substances could be effective in reducing pan-
warranted. creatic cancer incidence. We argued that those agents, which
have the ability to intervene at more than one critical pathway
in pancreatic carcinogenic process, will have greater advant-
Introduction age over other single-target agents. Lupeol, a triterpene
(Figure 1A) is found in various edible plants such as olive,
Pancreatic cancer is the most fatal of all cancers and the fifth fig, mango, strawberry, red grapes and medicinal plants used
most common cause of cancer-related deaths among both men by native people in North America, Japan, China, Latin
and women in the western countries. It is estimated that 32 180 America and Caribbean islands (10–12). Lupeol has been
cases of pancreatic cancer will be diagnosed in the United shown to exhibit strong anti-inflammatory, anti-arthritic,
anti-mutagenic and anti-malarial activity in in vitro and
in vivo systems (10). Lupeol has been shown to act as a potent
Abbreviations: DMSO, dimethyl sulfoxide; MAPKs, mitogen-activated inhibitor of protein kinases and serine proteases and of DNA
protein kinases; NFkB, nuclear factor kappa B; ODC, ornithine topoisomerase II, a target for anticancer chemotherapy
decarboxylase; PARP, poly(ADP-ribose)polymerase; PI, propidium iodide; (13,14). Lupeol has been reported to induce differentiation
PI3K, phosphatidylinositol 3-kinase; PKCa, protein kinase Ca; PMSF,
phenylmethylsulfonyl fluoride; TdT, terminal deoxynucleotidyl transferase.
and inhibit the growth of melanoma and leukemia cells
(15–17). Recently, we have shown that Lupeol inhibits
y
These three authors contributed equally to this work. tumor promotion in two-stage mouse skin carcinogenesis by
Carcinogenesis vol.26 no.11 # Oxford University Press 2005; all rights reserved. 1956
Lupeol targets multiple signaling pathways

(B)
24 h Treatment
100 48 h Treatment
72 h Treatment
(A)
80

Cell viability(%)
60

40

20

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Lupeol
0
C V 10 20 30 40 50 75 100 125

Lupeol (µM)
Fig. 1. (A) Structure of Lupeol [Lup-20(29)-en-3b-ol], a triterpene is found in fruits and vegetables. (B) Effect of Lupeol on viability of AsPC-1 cells: The cells
were treated with specified concentrations of Lupeol for 24, 48 and 72 h, and cell viability was determined by MTT assay. The values are represented as the percent
viable cells where vehicle-treated cells were regarded as 100% viable. Data represent mean value of percent viable cells  SE of three independent experiments.
The details are described under Materials and methods.

modulating various signaling pathways (18). In the present was determined. MTT [5 mg/ml in phosphate-buffered saline (PBS)] was
study, employing human pancreatic adenocarcinoma cell line added to each well and incubated for 2 h after which the plate was centrifuged
at 1800 r.p.m. (500 g) for 5 min at 4 C. The supernatant was removed from
AsPC-1, we show that Lupeol is a potent multi-target antican- the wells by aspiration. After careful removal of the medium, 0.1 ml of
cer agent that induces apoptotic cell death of pancreatic cancer buffered DMSO was added to each well, and plates were shaken. The absorb-
cells via modulation of Ras-induced protein kinase Ca ance was recorded on a microplate reader at the wavelength of 540 nm. The
(PKCa)/ornithine decarboxylase (ODC), PI3K/Akt, MAPKs effect of Lupeol on growth inhibition was assessed as percent cell viability
and NFkB signaling pathways. We suggest that Lupeol may be where vehicle-treated cells were taken as 100% viable.
an effective agent that should be tested for its in vivo efficacy Detection of apoptosis and necrosis by fluorescence microscopy
against pancreatic cancer. The Annexin-V-FLUOS apoptosis detection kit (Roche Applied Science,
Indianapolis, IN) was used for the detection of apoptotic and necrotic cells.
This kit uses a dual-staining protocol in which the cells are stained with
Materials and methods annexin and propidium iodide (PI). Cell populations that potentially may be
detected are as follows: viable cells will be non-fluorescent; cells in the
Cell culture and reagents metabolically active stages of apoptosis will stain with annexin (green fluor-
Human pancreatic adenocarcinoma cells AsPC-1 were a gift from Dr Nihal escence) but not with PI (red fluorescence) and necrotic cells with PI staining.
Ahmad, Department of Dermatology, University of Wisconsin-Madison. The In addition, cells undergoing late stage apoptosis bind both annexin and PI.
cells were cultured in RPMI 1640 (ATCC, Manassas VA) in a humidified Briefly, AsPC-1 cells were grown to 50% confluence on slides and then
atmosphere of 95% air and 5% CO2 at 37 C. Antibodies were procured from treated with Lupeol (30, 40, 50 mM) for 48 h. Apoptosis and necrosis was
BD Transduction laboratories (San Jose, CA), Santacruz Biotechnology (Santa detected by the using a Zeiss Axiovert 100 microscope. Briefly, the samples
Cruz, CA) and Upstate (Charlottesville, VA). Lupeol was purchased from were excited at 330–380 nm, and the image was observed and photographed
Sigma Chemical Company (St Louis, MO). under a combination of a 400 nm dichoric mirror and then 420 nm high-pass
filter.
Treatment of cells
Lupeol stock (30 mM) was prepared by dissolving Lupeol in warm alcohol and Quantification of apoptosis
diluting in dimethyl sulfoxide (DMSO) in a 1:1 ratio. Working solutions were Apoptosis was quantified by terminal deoxynucleotidyl transferase-mediated
prepared by diluting the stock solution with DMSO to get desired final con- dUTP-biotin nick and labeling (TUNEL) method. Briefly, the cells were grown
centrations. For dose-dependent studies, the cells (50% confluent) were treated at a density of 1  106 cells in 100-mm culture dishes and then treated with
with Lupeol (30, 40 and 50 mM) for 48 h in complete cell medium. Cells that Lupeol (30, 40 and 50 mM) for 48 h. The cells were trypsinized, washed with
served as controls were incubated with the vehicle (alcohol þ DMSO) alone. PBS and processed for labeling with fluorescein-tagged deoxyuridine triphos-
The final concentration of DMSO and alcohol was 0.25 and 0.075%, phate nucleotide and PI by use of an APO-DIRECT apoptosis kit obtained
respectively, in all treatment protocols. from Phoenix Flow Systems (San Diego, CA) and following the manufac-
turer’s protocol. The labeled cells were then analyzed by flow cytometry.
Cell viability assay
The effect of Lupeol on the viability of AsPC-1 cells was determined by Total cell lysate and nuclear lysate preparation
MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazoliumbromide) assay. The total cell lysate was prepared in cold buffer [50 mM Tris–HCl, 150 mM
The cells were plated at 1  104 cells per well in 200 ml of complete culture NaCl, 1 mM EDTA, 20 mM NaF, 100 mM Na3VO4, 0.5% NP-40, 1% Triton
medium and treated with 10, 20, 30, 40, 50, 75, 100, 125 mM concentrations of X-100, 1 mM phenylmethylsulfonyl fluoride (PMSF), pH 7.4] with freshly
Lupeol in 96-well microtiter plates. Lupeol stock solutions prepared at 30 mM added protease inhibitor cocktail (Protease inhibitor Cocktail Set III; Calbio-
concentration were mixed with fresh medium to achieve the desired final chem, La Jolla, CA) in ice. For the preparation of nuclear fractions, the cells
concentration. Each concentration of Lupeol was repeated in 10 wells. After were washed with cold PBS and suspended in 0.4 ml of lysis buffer (10 mM
incubation for 24, 48 and 72 h at 37 C in a humidified incubator, cell viability HEPES, pH 7.9, 10 mM KCI, 0.1 mM EDTA, 0.1 mM EGTA, 1 mM DTT,

1957
M.Saleem et al.

0.5 mM PMSF, Protease Inhibitor Cocktail Set III) in a microfuge tube. The from the morphology of cells, as assessed by fluorescence
cells were incubated on ice for 15 min, after which 12.5 ml of 10% Nonidet microscopy after labeling the cells with annexin and PI
P-40 was added and the contents were mixed on a vortex and then centrifuged (Figure 2, upper panel). As shown by representative pictures
for 1 min at 4 C at 14 000 g. The nuclear pellet was resuspended in 25 ml of
ice-cold nuclear extraction buffer (20 mM HEPES, pH 7.9, 0.4 M NaCl, 1 mM (Figure 2, upper panel), Lupeol treatment resulted in induction
EDTA, 1 mM EGTA, 1 mM DTT, 1 mM PMSF, Protease Inhibitor Cocktail of apoptosis in a dose-dependent manner. These data also show
Set III) and incubated on ice for 30 min with intermittent mixing. The tube was that Lupeol treatment also resulted in necrosis of these cells,
centrifuged for 5 min at 14 000 g at 4 C and the supernatant (nuclear extract) which may be a secondary event in the apoptotic process. We
was stored at 80 C. The protein concentration was determined by BCA assay
(Pierce, Rockford, IL) as per the manufacturer’s protocol. next quantified the extent of apoptosis by flow-cytometric
Western blot analysis analysis of Lupeol-treated cells labeled with bromo-
For western blot analysis, 40 mg of the protein was resolved over 8–12% deoxyuridine triphosphate and PI. As shown by the data in
polyacrylamide gels and transferred to a nitrocellulose membrane. The blot Figure 2 (bottom panel), compared with control, Lupeol treat-
containing the transferred protein was kept in blocking buffer (5% nonfat ment resulted in 0.3, 8.6 and 22.0% TUNEL-positive cells
dry milk, 1% Tween-20; in 20 mM TBS, pH 7.6) on a shaker for 1 h at room at 30, 40 and 50 mM at 48 h post-treatment, respectively.
temperature followed by incubation with appropriate primary antibody in
blocking buffer for 1 h to overnight at 4 C. This was followed by incubation
Consistent with the fluorescence microscopy data, TUNEL
with anti-mouse or anti-rabbit secondary antibody conjugated with horse- assay by flow cytometry revealed that treatment of AsPC-1
radish peroxidase (Amersham Life Sciences) for 1 h. After several washings, cells with Lupeol resulted in a dose-dependent induction of
immunoblots were detected by chemiluminescence ECL kit (Amersham Life apoptosis.

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Sciences) and autoradiography using XAR-5 film obtained from Eastman
Kodak (Rochester, NY). Immunoblots were scanned by HP Precisionscan Lupeol induces PARP cleavage and increases Bax protein
Pro 3.13 (Hewlett-Packard, Palo Alto, CA). Densitometry measurements
of the scanned bands were performed using digitalized scientific software expression in AsPC-1 cells
program UN-SCAN-IT (Silk Scientific Corporation, Orem, UT). Data were Since poly(ADP-ribosylation) is a post-translational modifica-
normalized to b-actin and expressed as mean values  SE of three separate sets tion of proteins which plays a crucial role in DNA repair and
of experiments.
cell death, poly(ADP-ribose)polymerase (PARP), a DNA nick
Electrophoretic mobility shift assay
sensor, serves as one of the best-known biomarkers of apop-
To detect NFkB-DNA binding, electrophoretic mobility shift assay (EMSA)
was performed using lightshiftÔ chemiluminiscent kit (Pierce, Rockford, IL)
tosis (19). During apoptosis, PARP protein is cleaved into
by following the manufacturer’s protocol. To start with, DNA was biotin an 85 kDa C-terminal fragment, with a reduced catalytic activ-
labeled using the biotin 30 end labeling kit (Pierce, Rockford, IL). Briefly, in ity, and a 24 kDa N-terminal peptide, which retains the DNA
a 50 ml reaction buffer, 5 pmol of double-stranded NFkB oligonucleotide 50 - binding domains. Next, we assessed the effect of Lupeol
AGT TGA GGG GAC TTT CCC AGG C-30 ; 30 -TCA ACT CCC CTG AAA (30–50 mM) treatment of cells on PARP cleavage. The
GGG TCC G-50 , 10 ml of 5 TdT (terminal deoxynucleotidyl transferase)
buffer, 5 ml of 5 mM biotin-N4-CTP, 10 U of diluted TdT, and 25 ml of PARP cleavage analysis showed that the full-size PARP
ultrapure water were incubated at 37 C for 30 min. For supershift assay, the (116 kDa) protein was cleaved to yield an 85 kDa fragment
reaction mixture was incubated for an additional 20 min with or without after treatment of cells with Lupeol at 30, 40 and 50 mM
antibody for NFkB/p65 (1 ml, Geneka Biotechnology). The reaction was concentrations at 48 h post-treatment (Figure 3). The expres-
stopped with 2.5 ml of 0.2 M EDTA. To extract labeled DNA, 50 ml of
chloroform:isoamyl alcohol (24:1) was added to each tube and centrifuged
sion of Bax (pro-apoptotic protein) and Bcl-2 (anti-apoptotic
briefly at 13 000 g. The top aqueous phase containing the labeled DNA was protein) have been reported to play a crucial role in apoptotic
removed and saved for binding reactions. Each binding reaction contained 1- response mediated by many agents (20,21). We next determ-
binding buffer (100 mM Tris, 500 mM KCl, 10 mM DTT, pH 7.5), 2.5% ined the effect of Lupeol treatment of cells on the protein
glycerol, 5 mM MgCl2, 50 ng/ml poly(dI–dC), 0.05% NP-40, 5 mg of nuclear levels of Bax and Bcl-2. The immunoblot analysis exhibited
extract and 20–50 fmol of biotin-end labeled target DNA. The content was
incubated at room temperature for 20 min. To this reaction mixture, 5 ml of a significant increase in the protein expression of Bax in
5 loading buffer was added, subjected to gel electrophoresis on a native Lupeol-treated cells (Figure 3). However, the protein expres-
polyacrylamide gel and transferred to a nylon membrane. When the transfer sion of Bcl-2 did not exhibit any change by Lupeol treatment at
was complete, DNA was cross-linked to the membrane at 120 mJ/cm2 using a any dose.
UV cross-linker equipped with 254 nm bulbs. The biotin end-labeled DNA was
detected using streptavidin–horseradish peroxidase conjugate and a chemilu-
minescent substrate. The membrane was exposed to X-ray film (XAR-5
Lupeol induces activation of caspases in AsPC-1 cells
Amersham Life Science, Arlington Height, IL) and developed using a Kodak Caspases receive the upstream death signals and are the final
film processor. executioners of apoptosis (22,23). In most cancer cells,
caspases are present in the pro-forms (inactive) and require
Results cleavage of protein to become active to participate in the
process of apoptosis. Through this process, the pro-forms of
Lupeol induces growth inhibition and apoptosis in AsPC-1 caspases are lost and the expression of their active forms
cells (cleaved products) increases during apoptosis (22,23). We
We first evaluated the effect of Lupeol on cell viability by next performed immunoblot analysis of the members of cas-
MTT assay. As shown in Figure 1B, treatment of AsPC-1 cells pases in the cell lysates obtained from Lupeol-treated cells. We
with Lupeol (10–125 mM) resulted in a dose-dependent inhibi- have used antibodies that specifically recognize the pro-forms
tion of cell growth when assessed at 24, 48 or 72 h post- and cleaved products (active forms) of caspases. As shown in
treatment (Figure 1B). The IC50 value of Lupeol on growth Figure 4, Lupeol-treated cells did not exhibit any change in the
inhibition was estimated to be 35 mM at 48 h. Based on expression level of procaspase-3, however, the expression
these observations, we selected a dose of 30, 40 and 50 mM of active caspase-3 was found to be increased in such cells
and a time period of 48 h post-Lupeol treatment for further (Figure 4). Further Lupeol treatment induced the expression of
mechanistic studies. caspases-8 and -9 in a dose-dependent manner which is evident
We next determined whether Lupeol-mediated loss of cell from the decrease in the protein expression of their pro-forms
viability in AsPC-1 cells is a result of induction of apoptosis. (inactive forms) with a concomitant increase in their active
The induction of apoptosis by Lupeol (30–50 mM) was evident forms as compared with vehicle-treated control (Figure 4).
1958
Lupeol targets multiple signaling pathways

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Fig. 2. Lupeol induces apoptosis in AsPC-1 cells as assessed by fluorescence microscopy (upper panel) and by flow cytometry (bottom panel). The upper
panel contains the representative micrographs of AsPC-1 cells undergoing apoptosis induced by treatment with Lupeol as assessed by fluorescence microscopy.
The cells were treated with vehicle alone or specified concentration of Lupeol for 48 h. Apoptosis and necrosis was detected by a Zeiss Axiovert 100 microscope
as described in Materials and methods. The samples were excited at 330–380 nm, and the image was observed and photographed under a combination of a
400 nm dichoric mirror and then 420 nm high pass filter. The bottom panel is the quantitative representation of Lupeol-induced apoptosis in AsPC-1 cells
as assessed by flow cytometry. The cells were treated with Lupeol (30–50 mM for 48 h), labeled with deoxyuridine triphosphate using terminal deoxynucleotide
transferase and PI by using an apoptosis APO-DIRECT kit obtained from Phoenix Flow Systems (San Diego, CA) as per vendor’s protocol followed by
flow cytometry. Cells showing deoxyuridine triphosphate fluorescence above that of control population, as indicated by the line in each histogram, are considered
as apoptotic cells. The images shown here are representative of three independent experiments with similar results. The values shown inside each graph indicate
the percent apoptotic cells. The details are described under Materials and methods.

Lupeol decreases the expression of Ras, PKCa, ODC proteins


and inhibits PI3K/Akt in AsPC-1 cells
Many cancers including pancreatic cancer exhibit aberrant Ras
signaling that leads to chronic up-regulation of the Ras path-
way and accumulation of Ras oncoprotein in transformed
cells (24). PKCa has been implicated in Ras signaling and
transformed cells have been reported to have high levels of
PKCa (25). In various cancer types, Ras-induced PKCa has
been reported to activate ODC, an oncogene which is highly
activated during the cellular proliferation (25–27). Next, we
determined the effect of Lupeol treatment of cells on the
expression of Ras, PKCa and ODC proteins. It is evident
from the data of immunoblot analysis that treatment of Lupeol
significantly reduced the protein expression of Ras in a dose-
dependent manner (Figure 5). Similarly, treatment of Lupeol
to cells also caused a significant reduction in the protein
expression of ODC and PKCa (Figure 5). These findings
suggest that Lupeol exerts a preferential inhibitory effect on
Ras/PKCa/ODC pathway leading to reduction in pancreatic
cancer cell growth.
The frequent aberration in Ras signaling is reported to cause
constitutive activation of PI3K/Akt during the development of
pancreatic cancer in humans [(6,28) and references therein].
We next investigated the effect of Lupeol treatment of cells on
Fig. 3. Effect of Lupeol treatment on PARP cleavage and on protein
PI3K/Akt signaling pathway. Lupeol treatment of cells was
expression of Bax, Bcl-2 in AsPC-1 cells. The cells were treated with vehicle
(DMSO þ alcohol) only or specified concentrations of Lupeol for 48 h, found to cause a significant reduction in the expression of
harvested and total cell lysates were prepared. PARP cleavage and the regulatory subunit of PI3K (p85) protein (Figure 5). Similarly,
expression of Bax and Bcl-2 were determined by western blot analysis. Equal Lupeol caused a significant dose-dependent decrease in the
loading was confirmed by stripping immunoblots and reprobing them for phosphorylation of Akt protein in cells (Figure 5). The treat-
b-actin. Values given above each lane on the immunoblot represents the
relative density of the bands normalized to b-actin. The immunoblots shown ment of Lupeol did not cause any change in the protein levels
here are representative of three independent experiments with similar results. of total Akt (Figure 5). These results suggest the inhibitory
The details are described under Materials and methods. potential of Lupeol against PI3K/Akt pathway, which is highly
1959
M.Saleem et al.

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Fig. 4. Effect of Lupeol treatment on protein expression of caspases-3, -8 and
-9 in AsPC-1 cells. The cells were treated with vehicle (DMSO þ alcohol)
only or specified concentrations of Lupeol for 48 h, harvested and total cell
lysates were prepared. The expression of proforms and cleaved products of
caspase -3, -8 and -9 were determined by western blot analysis. Equal
loading was confirmed by stripping immunoblots and reprobing them for
b-actin. Values given above each lane on the immunoblot represents the
relative density of the bands normalized to b-actin. The immunoblots shown
here are representative of three independent experiments with similar results.
The details are described under Materials and methods. Fig. 5. Effect of Lupeol treatment on the expression of Ras, PKCa, ODC,
PI3K (P85), pAkt and total Akt, phospho-p38 and Erk1/2 proteins in AsPC-1
cells: the cells were treated with vehicle (DMSO þ alcohol) only or specified
concentrations of Lupeol for 48 h, harvested and total cell lysates were
activated during the development and progression of pancre- prepared. The expression of Ras, PKCa, ODC, PI3K(P85), pAkt and total
atic cancer. Akt, p38 and Erk1/2 proteins total cell lysate was determined by western blot
analysis. Equal loading was confirmed by stripping immunoblots and
reprobing them for b-actin. Values given above each lane on the immunoblot
Lupeol inhibits the expression of MAPK proteins p38 and represents the relative density of the bands normalized to b-actin. The
Erk1/2 in AsPC-1 cells immunoblots shown here are representative of three independent
Constitutive levels of Erk1/2 protein have been reported to be experiments with similar results. The details are described under Materials
very low in pancreatic cancer cells and studies have linked the and methods.
dysregulation of MAPKs with tumor cell survival in various
cancer types including pancreatic adenocarcinoma (3,29,30).
JNK1 protein (data not shown). Taken together, these obser-
Therefore, we determined whether MAPKs contributed to
Lupeol-induced apoptosis in our model. We addressed this vations indicate that activation of Erk1/2 protein and decrease
in expression levels of phospho-p38 protein might be contrib-
question by determining the effect of Lupeol treatment on
uting towards apoptosis of cells induced by Lupeol.
expression levels of Erk1/2, JNK and p38 MAPK proteins.
As shown in Figure 5, exposure of cells to Lupeol resulted in a
dose-dependent activation (phosphorylation) of Erk1/2. Lupeol inhibits phosphorylation of IkBa and NF-kB/p65 in
Lupeol treatment caused a dose-dependent decrease in AsPC-1 cells
the level of phospho-p38 MAPK (Figure 5). Unlike ERK1/2 A number of studies clarified the role of NFkB in tumor cell
and p38, however, Lupeol treatment did not alter the level of survival by inhibiting apoptosis (31). The activation of NFkB
1960
Lupeol targets multiple signaling pathways

pathway in pancreatic cancer cells has been linked to NFkB–DNA binding is reduced in Lupeol-treated AsPC-1
their resistance to conventional chemotherapy (32). The cells
phosphorylation of inhibitory protein IkBa is a critical The translocation of NFkB to nucleus is marked by its binding
step in the pathway of NFkB activation that results in the with DNA to activate the expression of various genes leading
translocation of NFkB molecule to nucleus from cytosol, to the tumor cell survival (31). We next performed EMSA to
which ultimately leads to the survival of tumor cells. The investigate the effect of Lupeol treatment on NFkB–DNA
translocation of NFkB to nucleus is preceded by the phos- binding activity. As shown in Figure 5B, untreated and vehicle
phorylation of its p65 subunit and therefore is an important control cells exhibited a constitutively increased NFkB–DNA
event. Next, we investigated the effect of Lupeol treatment on binding activity; however, in cells treated with Lupeol, a
the phosphorylation of IkBa and NFkB/p65 in cells. As shown significant decrease in NFkB–DNA binding activity was
by immunoblot analysis (Figure 6A), Lupeol treatment observed (Figure 6B). The effect of Lupeol on NFkB–DNA
resulted in a dose-dependent inhibition of phosphoryla- binding in cells was concomitant with the effect of Lupeol on
tion of NFkB/p65 protein in these cells. The inhibition of phosphorylation of NF-kB/p65 subunit (Figure 6A). As is
phosphorylation of NF-kB/p65 molecule was concomitant evident from results of supershift assay using anti-p65 anti-
with a decrease in the phosphorylation of IkBa protein bodies, Lupeol was observed to specifically decrease the levels
(Figure 6A). of p65 subunit of NF-kB in the nucleus of cells (Figure 6C).

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011

Fig. 6. (A) Effect of Lupeol treatment on the phosphorylation of IkBa protein and p65-subunit of NFkB transcriptional factor in AsPC-1 cells. The cells were
treated with vehicle (DMSO þ alcohol) only or specified concentrations of Lupeol for 48 h, harvested and total cell lysates were prepared. Equal loading was
confirmed by stripping immunoblots and reprobing it for b-actin. Values given above each blot represents the relative density of the bands normalized to b-actin.
The immunoblots shown here are representative of three independent experiments with similar results. The details are described under Materials and methods.
(B) Effect of Lupeol treatment on the binding of NFkB transcriptional factor with DNA in AsPC-1 cells as assessed by EMSA. After 48 h of treatment with
Lupeol, the cells were harvested, nuclear lysates were prepared and DNA binding was determined by EMSA as described under Materials and methods.
I, II, and III refer to internal experimental controls, where I represents biotin–EBNA (Epstein–Barr virus nuclear antigen) control DNA, II represents biotin–EBNA
control DNA and EBNA extract, and III represents biotin–EBNA control DNA and EBNA extract plus 200-fold molar excess of EBNA DNA. In control
number I, no protein extract for DNA to bind resulted in an unshifted band. In control number II, sufficient target protein leads to DNA–protein binding resulting in
shift detected by comparison to band at position I. Control number II demonstrated that the signal shift observed could be prevented by competition from
excess unlabelled DNA. The data shown here are representative of three independent experiments with similar results. (C) Effect of Lupeol treatment on the
binding of p65-subunit of NFkB transcriptional factor with DNA in AsPC-1 cells. The cells were treated with vehicle only or specified concentrations of
Lupeol for 48 h, and then processed for nuclear lysate. NFkB/DNA binding activity was determined by Supershift assay using anti-p65 antibody and EMSA kit
from Pierce (Rockford, IL). The details are described under Materials and methods. The data shown here are representative of three independent experiments
with similar results, ns represents non-specific binding.

1961
M.Saleem et al.

Discussion cellular proliferation (2–6). Targeting the ras gene has been
shown to inhibit the growth of metastatic pancreatic cells (4).
The aggressive nature of pancreatic adenocarcinoma is related In the present study, we observed that treatment of human
to several abnormalities in growth factors and their receptors pancreatic cancer cells with Lupeol decreases the protein
that affect the downstream signal transduction pathways expression of Ras in a dose-dependent manner.
involved in the control of growth and differentiation (4). Ras protein is reported to regulate various downstream
These perturbations confer a tremendous survival and growth signaling pathways such as the PI3K/Akt pathway. The
advantage to pancreatic cancer cells. The acquisition of an growth-promoting potential of the PI3K/Akt pathway and its
apoptosis-resistant phenotype by pancreatic cancer cells may anti-apoptotic properties are closely linked to the resistance of
also upset the success of conventional radio- and chemother- cancer cells to a broad spectrum of apoptotic stimuli and
apies (32). In addition to perturbations in cell cycle control, several studies have demonstrated that treatment with the
resistance to apoptosis may promote tumor growth and meta- PI3K inhibitors substantially enhances apoptosis (4–6,36,37).
stasis. Apoptosis resistance mechanisms seem to vary among Studies have shown that inhibitors of PI3K/Akt induce dose-
different tumor types and have only begun to be characterized dependent apoptosis of pancreatic cancer cells and inhibit
at the molecular level. tumor growth of pancreatic cancer xenografts (37,38). In the
Elucidation of molecular events during pancreatic cancer current study, we show that treatment of pancreatic cancer
development has led to several distinct therapeutic advances cells with Lupeol reduced the expression of PI3K and Akt

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


and many novel agents and inhibitors including monoclonal proteins in a dose-dependent manner. This is consistent with
antibodies have been developed and are undergoing clinical our earlier report where we demonstrated that Lupeol inhibited
trials (4,32–34). However, many of these agents are not effect- the PI3K/Akt signaling pathway in vivo during two-stage
ive alone and have to be combined for maximum efficacy mouse skin tumorigenesis (18). These results suggest that
leading to undesirable and sometimes fatal side effects. One Lupeol has an efficiency to modulate the signaling pathways
promising approach could be identification of non-nutrient which are known to promote pancreatic cancer cell growth
dietary constituents that can target multiple pathways without and survival.
causing any undesirable toxicity. Here, we describe the use- The effects mediated by aberrantly activated Ras in human
fulness of Lupeol, a novel non-nutrient dietary agent against cancers are likely to be very complex and are currently not
human pancreatic cancer cells. In the current study, we dem- completely understood. Both Ras and PI3K/Akt signaling
onstrate that Lupeol treatment induces a dose-dependent death pathways have been reported to regulate the expression of
of human pancreatic cancer cells. From our data, it is evident MAPKs during the progression of various cancer types includ-
that the induction of cell death is mediated through apoptosis. ing pancreatic cancer (3,4,29,30). Chronic stimulation of the
We also found that Lupeol-induced apoptosis of pancreatic Ras/Raf/MEK/ERK pathway by oncogenic Ras is thought to
cells is mediated through the activation of caspases-3, -8 and promote cellular transformation and this pathway has been
-9. The observation that expression levels of procaspase-3 did reported to be activated in various cancer types, however,
not exhibit any significant change upon Lupeol treatment Erk1/2 activation does not always correlate with the expres-
could be explained through a possible involvement of a feed- sion of mutant ras or other oncogenes (3). Constitutive levels
back mechanism, which restores depleted procaspase-3 levels of Erk1/2 proteins have been reported to be very low in
at intracellular level. The only other pro-apoptotic effect of pancreatic cancer cells despite Ras activation and Erk1/2
lupeol reported so far is on mouse melanoma and human inhibition in pancreatic cancer cells have been linked with
leukemia cells (15–17). the resistance to apoptosis [(3, 30) and references therein].
Transformed cells have been shown to possess a dysregu- As evident from our data, the correlation of significant cell
lated apoptotic machinery (35). Several pro-apoptotic proteins death with increased expression of Erk1/2 proteins in Lupeol-
are either lost or diminished and many anti-apoptotic proteins treated cells is evidence that low levels of Erk1/2 protein
are activated during the development of cancer (35). As is have an association with tumor cell survival in pancreatic
evident from our data, induction of pro-apoptotic Bax protein cancer. Our data are supported by recent studies showing that
by Lupeol indicates that Lupeol may rectify the errors in chemotherapeutic agent-induced apoptosis in pancreatic can-
apoptotic machinery of pancreatic cancer cells. Although cer cells is through Erk1/2 activation. It has been demonstrated
many studies have shown that natural agents decrease the that p38 MAPK activation triggers an anti-apoptotic response
expression of anti-apoptotic protein Bcl-2 with a concomitant to tumor cells, although there have also been some contrasting
increase in the expression of pro-apoptotic Bax protein and reports that p38 MAPK activation (phosphorylation) is central
shift the balance towards apoptosis, we did not observe to the pro-apoptotic effects in some cancer cell lines (39,40).
any change in the Bcl-2 protein expression in Lupeol-treated Recent reports have demonstrated a link between the Ras
pancreatic cancer cells. However, with an increase in Bax oncoprotein and p38 MAPK activation in transformed cells
expression the shift is indeed towards apoptosis. Since our (41). It is evident from our data that Lupeol treatment signi-
data suggest that Lupeol is a multi-target agent, other mech- ficantly decreases the expression of phospho-p38 MAPK in
anisms of apoptosis could not be ruled out. cells leading to their apoptotic death.
Other contributing molecular changes in pancreatic adeno- Ras oncoprotein has been shown to contribute to constitutive
carcinoma include activation of oncogenes and inactivation of activation of NFkB signaling in pancreatic cancer through the
tumor suppressor genes (2–5). The ras oncogene is thought to activation of PI3K/Akt and MAPK pathways (32). Wang et al.
play an important role in the growth of pancreatic cancer, reported that p65 subunit of NFkB was constitutively activated
because an activated (mutated) ras gene is found in 90% of in 67% of pancreatic adenocarcinomas and a positive cor-
human pancreatic cancers (2–5). The ras mutation results in relation exists between the activation of NFkB and Ras
the accumulation of Ras oncoprotein, and constitutive activa- oncoprotein during the proliferation of pancreatic cancer
tion of various intracellular signaling pathways, leading to cells (42). NFkB plays an important role in tumor resistance
1962
Lupeol targets multiple signaling pathways

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Fig. 7. A schematic representation of the action of Lupeol with respect to various signaling pathways in AsPC-1 cell. " Represents increased expression;
# represents decreased expression; ‘ represents inhibition; and ! represents induction. See online Supplementary material for a color version of this figure.

to apoptosis and studies have shown that pancreatic cancer growth of pancreatic cancer cells. The schematic representa-
cells resistant to apoptosis exhibit a high basal NFkB activity tion of the action of Lupeol is depicted in Figure 7. This may
(31,43). Interestingly, we found that treatment of Lupeol inhib- be explained by modulation of NFkB mediated by Ras-
ited expression of NFkB and phosphorylation of IkBa protein induced pathways such as PKCa/ODC, PI3K/Akt and MAPK
in pancreatic cancer cells. Because Lupeol inhibits IkBa phos- signaling. Since pancreatic cancer is resistant to conventional
phorylation, our study suggests that the effect of Lupeol on chemotherapeutic regimens, Lupeol could be a potential agent
NFkB/p65 is through inhibition of phosphorylation of IkBa to overcome this resistance. Further in-depth in vivo studies are
which in turn is regulated by Ras and PI3K/Akt pathways. warranted to verify this suggestion and are presently under
The Ras oncoprotein is also reported to regulate PKCa investigation in our laboratory.
(23,24). PKCa has been reported to be involved in the regula-
tion of apoptosis and in animal models, tumorigenicity of Supplementary material
pancreatic cancer cells has been directly related to increased
expression of PKCa protein (25,26). Numerous studies have Supplementary material can be found at: http://carcin.
shown that PKC levels are high in pancreatic cancer patients oxfordjournals.org/
and increased PKC levels offer resistance to apoptosis of
pancreatic cancer cells (32). In this scenario, Lupeol with an Acknowledgements
efficacy of targeting PKCa protein seems to be a promising Author (S.K.) was recipient of BOYSCAST fellowship from Department of
agent against the growth of pancreatic cancer cells as it exhib- Science and Technology, Government of India. This work was supported by
its an inhibitory potential against PKCa protein and induces United States Public Health Service grants RO1 CA 78809 and RO1 CA
apoptosis of pancreatic cancer cells in a dose-dependent 101039.
manner. Conflict of Interest Statement: None declared.
Ras-induced PKCa is known to induce the ODC protein,
the first and the rate-limiting enzyme in the biosynthesis of References
polyamines expression during the development of various
cancer types (26,27,44). Elevated levels of ODC gene products 1. Jemal,A., Murray,T., Ward,E., Samuels,A., Tiwari,R.C., Ghafoor,A.,
Feuer,E.J. and Thun,M.J. (2005) Cancer statistics. CA Cancer J. Clin.,
are consistently detected in transformed cell lines, virtually 55, 10–30.
all-animal tumors and in certain tissues predisposed to tumori- 2. Almoguera,C., Shibata,D., Forrester,K., Martin,J., Arnheim,N. and
genesis (24,25). Because tumor formation can be prevented by Perucho,M. (1988) Most human carcinomas of the exocrine pancreas
the agents that block induction of ODC, several ODC inhibit- contain mutant c-K-ras genes. Cell, 53, 549–554.
ors have been tested against various types of cancers [(18) and 3. Yip-Schneider,M.T., Lin,A. and Marshall,M.S. (2001) Pancreatic tumor
cells with mutant K-ras suppress ERK activity by MEK-dependent
references therein]. These data demonstrate that Lupeol is induction of MAP kinase phosphatase-2. Biochem. Biophys. Res.
a potent inhibitor of ODC expression and is consistent with Commun., 280, 992–997.
our earlier report where we showed that Lupeol inhibits ODC 4. Xiong,H.Q. (2004) Molecular targeting therapy for pancreatic cancer.
activity and expression in vivo (18). Cancer Chemother. Pharmacol., 54, S69–S77.
5. Asano,T., Yao,Y., Zhu,J., Li,D., Abbruzzese,J.L. and Reddy,S.A. (2004)
Based on the outcome of this study, we suggest Lupeol could The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten
provide a multi-prong beneficial strategy for targeting multiple expression and targets transcription factors NF-kappaB and c-Myc in
signaling pathways leading to apoptosis and inhibition of pancreatic cancer cells. Oncogene, 23, 8571–8580.

1963
M.Saleem et al.

6. Stoll,V., Calleja,V., Vassaux,G., Downward,J. and Lemoine,N.R. (2005) 28. Yamamoto,S., Tomita,Y., Hoshida,Y. et al. (2004) Prognostic significance
Dominant negative inhibitors of signalling through the phosphoinositol 3- of activated Akt expression in pancreatic ductal adenocarcinoma. Clin.
kinase pathway for gene therapy of pancreatic cancer. Gut, 54, 109–116. Cancer Res., 10, 2846–2850.
7. Nkondjock,A., Ghadirian,P., Johnson,K.C. and Krewski,D. (2005) Dietary 29. Sebolt-Leopold,J.S. and Herrera,R. (2004) Targeting the mitogen-
intake of lycopene is associated with reduced pancreatic cancer risk. activated protein kinase cascade to treat cancer. Nat. Rev. Cancer, 4,
J. Nutr., 135, 592–597. 937–947.
8. Norell,S.E., Ahlbom,A., Erwald,R., Jacobson,G., Lindberg-Navier,I., 30. Yip-Schneider,M.T., Lin,A., Barnard,D., Sweeney,C.J. and Marshall,M.S.
Olin,R., Tornberg,B. and Wiechel,K.L. (1986) Diet and pancreatic cancer: (1999) Lack of elevated MAP kinase (Erk) activity in pancreatic
a case–control study. Am. J. Epidemiol., 124, 894–902. carcinomas despiteoncogenic K-ras expression. Int. J. Oncol., 15,
9. Bueno de Mesquita,H.B., Maisonneuve,P., Moerman,C.J., Runia,S. and 271–279.
Boyle,P. (1992) Lifetime consumption of alcoholic beverages, tea and 31. Kumar,A., Takada,Y., Boriek,A.M. and Aggarwal,B.B. (2004) Nuclear
coffee and exocrine carcinoma of the pancreas: a population-based case– factor-kappaB: its role in health and disease. J. Mol. Med., 82, 434–448.
control study in The Netherlands. Int. J. Cancer, 50, 514–522. 32. Lampe,E., Heinrich,M., Walczak,H. and Kalthoff,H. (2001) CD95 and
10. Saleem,M., Alam,A., Arifin,S., Shah,M.S., Ahmed,B. and Sultana,S. TRAIL receptor-mediated activation of protein kinase C and NF-kappaB
(2001) Lupeol, a triterpene, inhibits early responses of tumor promotion contributes to apoptosis resistance in ductal pancreatic adenocarcinoma
induced by benzoyl peroxide in murine skin. Pharmacol. Res., 43, cells. Oncogene, 20, 4258–4269.
127–134. 33. Li,Y., Ellis,K.L., Ali,S., El-Rayes,B.F., Nedeljkovic-Kurepa,A., Kucuk,O.,
11. Beveridge,T.H., Li,T.S. and Drover,J.C. (2002) Phytosterol content in Philip,P.A. and Sarkar,F.H. (2004) Apoptosis-inducing effect of chemo-
American ginseng seed oil. J. Agric. Food. Chem., 50, 744–750. therapeutic agents is potentiated by soy isoflavone genistein, a natural
12. Kakuda,R., Iijima,T., Yaoita,Y., Machida,K. and Kikuchi,M. (2002) inhibitor of NF-kappaB in BxPC-3 pancreatic cancer cell line. Pancreas,

Downloaded from carcin.oxfordjournals.org at William Jewell College on February 26, 2011


Triterpenoids from Gentiana scabra. Phytochemistry, 59, 791–794. 28, 90–95.
13. Hodges,L.D., Kweifio-Okai,G. and Macrides,T.A. (2003) Antiprotease 34. Mohammad,R.M., Adsay,N.V., Philip,P.A., Pettit,G.R., Vaitkevicius,V.K.
effect of anti-inflammatory lupeol esters. Mol. Cell. Biochem., 252, and Sarkar,F.H. (2001) Bryostatin 1 induces differentiation and potentiates
97–101. the antitumor effect of Auristatin PE in a human pancreatic tumor (PANC-
14. Wada,S., Iida,A. and Tanaka,R. (2001) Screening of triterpenoids isolated 1) xenograft model. Anticancer Drugs, 12, 735–740.
from Phyllanthus flexuosus for DNA topoisomerase inhibitory activity. 35. Igney,F.H. and Krammer,P.H. (2002) Death and anti-death: tumour
J. Nat. Prod., 64, 1545–1547. resistance to apoptosis. Nat. Rev. Cancer, 2, 277–288.
15. Aratanechemuge,Y., Hibasami,H., Sanpin,K., Katsuzaki,H., Imai,K. and 36. Perugini,R.A., McDade,T.P., Vittimberga,F.J.Jr and Callery,M.P. (2000)
Komiya,T. (2004) Induction of apoptosis by lupeol isolated from Pancreatic cancer cell proliferation is phosphatidylinositol 3-kinase
mokumen (Gossampinus malabarica L. Merr) in human promyelotic dependent. J. Surg. Res., 90, 39–44.
leukemia HL-60 cells. Oncol. Rep., 11, 289–292. 37. Ng,S.S.W., Tsao,M.S., Chow,S. and Hedley,D.W. (2000) Inhibition of
16. Hata,K., Hori,K. and Takahashi,S. (2003) Role of p38 MAPK in lupeol- phosphatidylinositide 3-kinase enhances gemcitabine-induced apoptosis in
induced B16 2F2 mouse melanoma cell differentiation. J. Biochem., 134, human pancreatic cancer cells. Cancer Res., 60, 5451–5455.
441–445. 38. Yau,C.Y., Wheeler,J.J., Sutton,K.L. and Hedley,D.W. (2005) Inhibition
17. Hata,K., Hori,K., Ogasawara,H. and Takahashi,S. (2003) Anti-leukemia of integrin-linked kinase by a selective small molecule inhibitor,
activities of Lup-28-al-20(29)-en-3-one, a lupane triterpene. Toxicol. Lett., QLT0254, inhibits the PI3K/PKB/mTOR, Stat3, and FKHR pathways
143, 1–7. and tumor growth, and enhances gemcitabine-induced apoptosis in human
18. Saleem,M., Afaq,F., Adhami,V.M. and Mukhtar,H. (2004) Lupeol orthotopic primary pancreatic cancer xenografts. Cancer Res., 65,
modulates NF-kappaB and PI3K/Akt pathways and inhibits skin cancer 1497–1504.
in CD-1 mice. Oncogene, 23, 5203–5214. 39. Habiro,A., Tanno,S., Koizumi,K., Izawa,T., Nakano,Y., Osanai,M.,
19. Ivana Scovassi,A. and Diederich,M. (2004) Modulation of poly(ADP- Mizukami,Y., Okumura,T. and Kohgo,Y. (2004) Involvement of p38
ribosylation) in apoptotic cells. Biochem. Pharmacol., 68, 1041–1047. mitogen-activated protein kinase in gemcitabine-induced apoptosis in
20. Hastak,K., Agarwal,M.K., Mukhtar,H. and Agarwal,M.L. (2005) Ablation human pancreatic cancer cells. Biochem. Biophys. Res. Commun., 316,
of either p21 or Bax prevents p53-dependent apoptosis induced by green 71–77.
tea polyphenol epigallocatechin-3-gallate. FASEB J., 19, 789–791. 40. Ding,X.Z. and Adrian,T.E. (2001) MEK/ERK-mediated proliferation is
21. Gupta,S., Afaq,F. and Mukhtar,H. (2002) Involvement of nuclear factor- negatively regulated by P38 map kinase in the human pancreatic cancer
kappa B, Bax and Bcl-2 in induction of cell cycle arrest and apoptosis by cell line, PANC-1. Biochem. Biophys. Res. Commun., 282, 447–453.
apigenin in human prostate carcinoma cells. Oncogene, 21, 3727–3738. 41. Shin,I., Kim,S., Song,H., Kim,H.R. and Moon,A. (2005) H-ras-specific
22. Philchenkov,A. (2004) Caspases: potential targets for regulating cell activation of Rac-MKK3/6-p38 pathway: its critical role in invasion and
death. J. Cell. Mol. Med., 8, 432–444. migration of breast epithelial cells. J. Biol. Chem., 280, 14675–14683.
23. Riedl,S.J. and Shi,Y. (2004) Molecular mechanisms of caspase regulation 42. Wang,W., Abbruzzese,J.L., Evans,D.B., Larry,L., Cleary,K.R. and
during apoptosis. Nat. Rev. Mol. Cell Biol., 5, 897–907. Chiao,P.J. (1999) The nuclear factor-kappa B RelA transcription factor
24. Beaupre,D.M. and Kurzrock,R. (1999) RAS inhibitors in hematologic is constitutively activated in human pancreatic adenocarcinoma cells.
cancers: biologic considerations and clinical applications. Invest. New Clin. Cancer Res., 5, 119–127.
Drugs, 17, 137–143. 43. Li,L., Aggarwal,B.B., Shishodia,S., Abbruzzese,J. and Kurzrock,R. (2004)
25. da Rocha,A.B., Mans,D.R., Regner,A. and Schwartsmann,G. (2002) Nuclear factor-kappaB and IkappaB kinase are constitutively active
Targeting protein kinase C: new therapeutic opportunities against in human pancreatic cells, and their down-regulation by curcumin
high-grade malignant gliomas? Oncologist, 7, 17–33. (diferuloylmethane) is associated with the suppression of proliferation
26. Voskas,D., Mader,R., Lee,J. and Hurta,R.A. (2001) Tumour promoter and the induction of apoptosis. Cancer, 101, 2351–2362.
mediated altered expression and regulation of ornithine decarboxylase and 44. Pignatti,C., Tantini,B., Stefanelli,C. and Flamigni,F. (2004) Signal
S-adenosylmethionine decarboxylase in H-ras-transformed fibrosarcoma transduction pathways linking polyamines to apoptosis. Amino Acids,
cell lines. Biochem. Cell Biol., 79, 69–81. 27, 359–365.
27. Weinstein,I.B. (1991) Nonmutagenic mechanisms in carcinogenesis: role
of protein kinase C in signal transduction and growth control. Environ.
Health Perspect., 93, 175–179. Received April 12, 2005; revised May 26, 2005; accepted June 7, 2005

1964

Potrebbero piacerti anche