ArticlePDF Available

Abstract

The incidence of Barrett esophageal adenocarcinoma (BEAC) has been increasing at an alarming rate in western countries. In this study, we have evaluated the therapeutic potential of sulforaphane (SFN), an antioxidant derived from broccoli, in BEAC. BEAC cells were treated with SFN, alone or in combination with chemotherapeutic, paclitaxel, or telomerase-inhibiting agents (MST-312, GRN163L), and live cell number determined at various time points. The effect on drug resistance/chemosensitivity was evaluated by rhodamine efflux assay. Apoptosis was detected by annexin V labeling and Western blot analysis of poly(ADP-ribose) polymerase cleavage. Effects on genes implicated in cell cycle and apoptosis were determined by Western blot analyses. To evaluate the efficacy in vivo, BEAC cells were injected subcutaneously in severe combined immunodeficient mice, and after the appearance of palpable tumors, mice were treated with SFN. SFN induced both time- and dose-dependent decline in cell survival, cell cycle arrest, and apoptosis. The treatment with SFN also suppressed the expression of multidrug resistance protein, reduced drug efflux, and increased anticancer activity of other antiproliferative agents including paclitaxel. A significant reduction in tumor volume was also observed by SFN in a subcutaneous tumor model of BEAC. Anticancer activity could be attributed to the induction of caspase 8 and p21 and down-regulation of hsp90, a molecular chaperon required for activity of several proliferation-associated proteins. These data indicate that a natural product with antioxidant properties from broccoli has great potential to be used in chemoprevention and treatment of BEAC.
Anticancer Activity of a Broccoli
Derivative, Sulforaphane, in
Barrett Adenocarcinoma:
Potential Use in
Chemoprevention and as
Adjuvant in Chemotherapy
1
Aamer Qazi*
,2
, Jagannath Pal
,,2
,Main Maitah*
,2
,
Mariateresa Fulciniti
,
,DheerajPelluru
,
,
Puru Nanjappa
,
,SaemLee
, Ramesh B. Batchu*,
Madhu Prasad*, Christopher S. Bryant*,
Samiyah Rajput
, Sergei Gryaznov
§
, David G. Beer
,
Donald W. Weaver*, Nikhil C. Munshi
,
, Raj K. Goyal
and Masood A. Shammas
,
*Departments of Surgery/Obstetrics and Gynecology,
Wayne State University and Karmanos Cancer Institute,
Detroit, MI, USA;
Adult Oncology, Harvard (Dana Farber)
Cancer Institute, Boston, MA, USA;
Harvard Medical School
and VA Boston Healthcare System, Boston, MA, USA;
§
Geron
Corporation, Menlo Park, CA, USA;
University of Michigan,
Ann Arbor, MI, USA
Abstract
INTRODUCTION: The incidence of Barrett esophageal adenocarcinoma (BEAC) has been increasing at an alarming
rate in western countries. In this study, we have evaluated the therapeutic potential of sulforaphane (SFN), an
antioxidant derived from broccoli, in BEAC. METHODS: BEAC cells were treated with SFN, alone or in combination
with chemotherapeutic, paclitaxel, or telomerase-inhibiting agents (MST-312, GRN163L), and live cell number
determined at various time points. The effect on drug resistance /chemosensitivity was evaluated by rhodamine
efflux assay. Apoptosis was detected by annexin V labeling and Western blot analysis of poly(ADP-ribose) poly-
merase cleavage. Effects on genes implicated in cell cycle and apoptosis were determined by Western blot
analyses. To evaluate the efficacy in vivo , BEAC cells were injected subcutaneously in severe combined immuno-
deficient mice, and after the appearance of palpable tumors, mice were treated with SFN. RESULTS: SFN induced
both time- and dose-dependent decline in cell survival, cell cycle arrest, and apoptosis. The treatment with SFN
also suppress ed the expression of mult idrug resistance protein, reduced drug efflux, and increased anticancer
activity of other antipro liferative agents including paclitaxel. A significant reduction in tumor volume was also
observed by SFN in a subcutaneous tumor model of BEAC. Anticancer activity could be attributed to the induc-
tion of caspase 8 and p21 and down-regulation of hsp90, a molecular chaperon required for activity of several
proliferation-associated proteins. CONCLUSIONS: These data indicate that a natural product with antioxidant
properties from broccoli has great potential to be used in chemoprevention and treatment of BEAC.
Translational Oncology (2010) 3, 389399
Address all correspondence to: Masood A. Shammas, PhD, Harvard (Dana Farber) Cancer Institute, Boston, MA, Harvard Medical School at VAMC, 1400 VFW Pkwy, Bldg 3,
Room 2A111, West Roxbury, MA 02132. E-mail: masood_shammas@dfci.harvard.edu
1
This work was supported by grants from R01CA125711 to MAS, from the Department of Veterans Affairs Merit Review Awards and from the National Institutes of Health grant
RO1-124929 to NCM and grants P50-100007 and PO1-78378 to NCM and Dr. K. C. Anderson and by funds from Karmanos Cancer Institute and Department of Surgery, Wayne
State University, Detroit, MI.
2
AQ, JP, and MM contributed equally to this work.
Received 22 September 2010; Revised 6 October 2010; Accepted 9 October 2010
Copyright © 2010 Neoplasia Press, Inc. All rights reserved 1944-7124/10/$25.00
DOI 10.1593/tlo.10235
www.transonc.com
Translational Oncology
Volume 3 Number 6 December 2010 pp. 389399 389
Introduction
Va rious epidemiologic studies have indicated that consumption of
broccoli is associated with a lower risk of cancer [1], including breast [2],
prostate [3], lung, stomach [4], and colon [5] cancers. The anticancer
effect of broccoli has been attributed to sulforaphane (SFN), an isothio-
cyanate formed by hydrolysis of a precursor glucosinolate called glu-
coraphanin [1]. Although glucoraphanin is found in varying amounts
in all cruci ferous vegetables, the highest concentration of this co m-
pound is found in broccoli and its sprouts [6]. Among various parts
of mature broccoli, the florets have the maximum amount of SFN.
The amount of SFN in 1 g of dry broccoli florets ranges from 507
to 684 μg [7]. The sprouts of broccoli seem to have 20 to 30 times
higher concentration of glucoraphanin, an SFN precursor [8,9], indi-
cating that 1 oz of sprouts may have the amount of antioxidant present
in 20 oz of mature broccoli. Glucoraphanin is converted to SFN by
myrosinase, an enzyme released from broccoli during its consumption
and also found in our stomach [1]. The reduced risk of cancer after
consumption of broccoli is associated with the ability of SFN to inhibit
phase 1 enzymes (implicated in the conversion of procarcinogens to
carcinogens) and induce phase 2 enzymes (implicated in detoxification
and excretion of carcinogens from body) [1,6].
However, the anticancer activity of SFN is not limited to its ability
to promote detoxification and removal of carcinogens. SFN has been
shown to inhibit cell cycle progression, induce apoptotic cell death,
and inhibit angiogenesis in a variety of cancer cell types [1,6]. Expo-
sure to SFN (20 μM) has been shown to induce a chk2 kinase
dependent arrest at the G
2
/M phase of cell cycle in prostate cancer
(PC3) cells [10]. A G
2
/M arrest after treatment with SFN has also
been demonstrated in human colon cancer (HCT116) cells [10].
Although cell cycle arrest at G
2
M has been frequently observed after
treatment of cancer cells with SFN, arrest at other phases of cell cycle
has also been reported for many cell lines. For example, the treatment
of colon cancer (HT-29) cells with this agent led to the induction of
p21, down-regulation of cyclin D1 and cyclin A, and a G
1
cell cycle
arrest [11], whereas prostate cancer (LnCap) cells arrested at G
1
/S
after treatment with 10 μM drug [12,13].
SFN has also been shown to induce apoptosis, or programmed cell
death, in cancer cells. Treatment with 15 μM SFN induced apoptosis
in both the p53-positive and p53-negative human colon cancer cell
lines [14]. Similarly, exposure to 10 μM SFN caused apoptosis in
prostate cancer DU145 cells [15]. The mechanisms of SFN-induced
apoptosis in these cell lines include activation of caspase 7 and 9 [14]
and/or release of cytochrome C from mitochondria [16].
Anticancer activity of SFN has also been demonstrated in vivo .SFN
has been shown to inhibit growth of human pancreatic cancer [17],
human prostate cancer [18], murine osteosarcoma xenografts [19], and
prevent intestinal polyposis [11], UV lightinduced skin tumors [20],
carcinogen-induced skin tumors [21], and carcinogen-induced stomach
tumors [22] in vivo. SFN has also been reported to increase natural killer
cell activity and antibody-dependent cellular cytotoxicity in both the
control and tumor-bearing mice [23]. These and other modulations
of immune system by SFN have been shown to play important role
in the inhibition of metastatic spread of melanoma in mice [24].
Barrett esophageal adenocarcinoma (BEAC) develops in Barrett
esophagus, a precancerous condition associated with chronic esophageal
reflux. Because the cancer starts to spread before onset of clinical symp-
toms, BEAC patients usually have a dreary outcome with a poor sur-
vival rate [25]. Moreover, the incidence of this cancer has been
increasing at a disturbing rate in Europe and United States [26]. Be-
cause the effect of SFN on BEAC cells has not been demonstrated,
we studied the effect of this natural food ingredient, either alone or
in combination with other anticancer agents including paclitaxel, on
cell cycle and cell viability in BEAC cell lines. We have shown that
SFN induces cell cycle arrest and apoptosis in BEAC cells at concentra-
tions (3-7 μM) lower than (10-20 μM) those of SFN required to kill
other cancer cell lines. Moreover, the SFN also suppressed multidrug
resistance protein (MRP), reduced drug efflux, and significantly
enhanced the anticancer activity of telomerase inhibitors (MST-312,
GRN163L) and paclitaxel, a chemotherapeutic currently used to treat
BEAC, in BEAC cell lines tested. The treatment with SFN was also
associated with induction of caspase 8 and p21 and suppression of
hsp90, a molecular chaperone required for activity of several prolifera-
tion related proteins. A significant anticancer activity of SFN was also
observed in a subcutaneous tumor model in vivo. These data indicate a
marked anticancer activity of SFN in BEAC and provide a rationale for
clinical evaluation.
Materials and Methods
Chemicals
L-Sulforaphane (SFN) and paclitaxel were purchased from Sigma-
Aldrich (St Louis, MO) and dissolved in phosphate-buffered saline.
MST-312 [N ,N -bis(2,3-dihydroxybenzoyl)-1,3-phenylenediamine]
was purchased from Calbiochem/EMD Biosciences (Madison, WI) and
dissolved in DMSO. GRN163L is a palmitoyl (C16) lipidattached
N3-P5 phosphoramidate oligonucleotide, targeting the template
region of RNA subunit of telomerase (hTR) and was obtained from
Geron Corporation (Menlo Park, CA). GRN140833 mismatch oligo-
nucleotide was also obtained from Geron Corporation and used as a
negative control.
Barrett Adenocarcinoma Cell Lines and Normal Cells
BEAC cell line FLO-1 and a lung adenocarcinoma cell line H460
have been described previously [27]. BEAC cell line OE33, from
European Collection Of Cell Cultures, was purchased through Sigma-
Aldrich and has been described previously [28,29]. Normal primary
human esophageal epithelial cells (HEEC) were purchased from Scien-
Cell Research Laboratories (Carlsbad, CA) and have been described
previously [30]. FLO-1 cells were cultured in Dulbecco modified Eagle
medium (Sigma) supplemented with 10% fetal bovine serum (H yClone,
South Logan, UT). OE33 cells were cultured in RPMI-1640 sup-
plemented with 2 mM
L-glutamine and 10% fetal bovine serum.
Normal HEE C (human esophageal epithe lia l cells) were cultured
in epithelial cell medium-2 (ScienCell Research Laboratories). Cells
were maintained in a state of logarithmic growth at 37°C in humidified
air with 5% CO
2
, as described previously [30]. For RNA and protein
analyses, cultures were harvested at the same final cell density (5 ×
10
5
/ml) and immediately processed.
Treatment and Growth of Cells
Cells (5 × 10
5
) were plated in 100-mm dishes and treated with
SFN alone or in combination with paclitaxel (PAC) and MST-312,
at concentrations described in the figure legends. Substrate-attached
viable cell number was counted, and cell viability was confirmed by
trypan blue exclusion or cell proliferation assays at various time points.
Cell proliferation assays were performed using Cell Counting Kit-8
(Dojindo Molecular Technologies, Inc, Gaithersburg, MD) according
to the manufacturers protocol. The method provides a highly sensitive
390 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. Translational Oncology Vol. 3, No. 6, 2010
colorimetric assay for the determination of viable cell number and is
based on the production of a yellow product (formazan) after reduction
of a highly water-soluble tetrazolium salt by dehydrogenases in viable
cells. The amount of the formazan dye generated is measured by a plate
reader and is directly proportional to the number of viable cells.
Rhodamine Efflux Assay
FLO-1 cells, plated in a six-well plate, were pretreated with SFN
(3 or 5 μM) for 5 hours or with cyclosporin A (10 μM; a known
inhibitor of MDR1 gene product) for 1 hour . Rhodamine 123 (3 μM)
was then added into the same medium, and the cells were incubated for
an additional 1.5 hours. Rhodamine-containing medium was then re-
moved and cells were washed with PBS and incubated overnight at
37°C, 5% CO
2
with serum-free medium containing the same concen-
trations of drugs without rhodamine. Cells were harvested, washed with
PBS, and evaluated for rhodamine 123 florescence by flow cytometry.
Cell Cycle Analysis
The effect of SFN and PAC on progression of the cell cycle was
determined using the Cell Cycle Phase Determination Kit (Cayman
Chemical Company, Ann Arbor, MI). Briefly, the control and treated
cells were washed and resuspended in assay buffer at 10
6
/ml. Fixative
(1 ml) was then added, and cells were allowed to permeabilize for at
least 2 hours. The cells were centrifuged, the fixative was removed,
and cells were resuspended in 0.5 ml of staining solution containing
propidium iodide. After 30 minutes of incubation at room temper-
ature, the cells were analyzed in the FL2 channel of a flow cytometer
with a 488-nm excitation filter.
Figure 1. Effect of SFN on BEAC cell survival. BEAC cells were cultured in the medium containing no SFN or various concentrations of
SFN. Cells were harvested at different time points as indicated and proliferative potential was assessed by trypan blue exclusion and/or
proliferation assay, based on the production of a yellow product (formazan) after reduction of a highly water-soluble tetrazolium salt by
dehydrogenases in viable cells. The growth curves show the mean of three independent experiments, with SEM. (A) Barrett adenocar-
cinoma (FLO-1) cells treated with various concentrations of SFN. (B) BEAC (OE33) cells treated with various concentrations of SFN. (C)
Photomicrograph of BEAC (FLO-1 and OE33) cells treated with 3 μM SFN for 72 hours. (D) Photomicrograph of normal diploid fibroblasts
and primary normal esophageal epithelial cells (ScienCell Research Laboratories) treated with 3 μM SFN for 72 hours. (E) FLO-1 cells
were treated with SFN for 48 hours, detached floating cells from the medium and the attached cells (by trypsinization) were collected
separately and evaluated for number and viability using trypan blue exclusion. The number of cells detached after treatment with various
concentrations of SFN is expressed as percent of untreated FLO-1 cells. Total represents the total number of detached cells whereas
Dead reflects the fraction of dead cells in detached cell population. (F) Panel (I): FLO-1 cells were incubated with various concentra-
tions of SFN for 48 hours, and the expression of caspase 8 was detected by Western blot analysis, using anticaspase 8 mouse mono-
clonal antibody (Cell Signaling, Danvers, MA). Panel (II): Bar graph showing caspase 8 expression relative to β-actin.
Translational Oncology Vol. 3, No. 6, 2010 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. 391
Apoptosis Assay
Apoptotic cells were detected using the Annexin VBiotin Apoptosis
Detection Kit (Oncogene Research Products, San Diego, CA). Un-
treated or treated BEAC cells (1 × 10
6
cells/ml) were mixed with annexin
Vbiotin and medium-binding reagent and incubated in the dark for
15 minutes at room temperature. Cells were then centrifuged, and the
medi um was replaced with binding buffer (Oncogene Research
Products) containing fluorescein isothiocyanate (FITC)streptavidin
(Amersham Life Sciences, Inc, Arlington Heights, IL). A portion of
the cell suspension (50 μl) was placed onto a glass slide, covered with
a coverslip, and viewed immediately using a fluorescence microscope
equipped with FITC (green) filter. Two hundred cells, representing
at least five distinct microscopic fields, were analyzed to assess the frac-
tion of FITC-labeled cells for each sample.
Western Blot Analysis
Approximately 50 mg o f protein was suspended in Laemmli
sample buffer (0.1 M Tris-HCl buffer pH 6.8, 1% SDS, 0.05% β-
mercaptoethanol, 10% glycerol, and 0.001% bromphenol blue), boiled
for 2 minutes, and electrophoresed on 4% to 20% glycerol gradient
SDSpolyacrylamide gel for 4 hours at 120 V. Gels were electroblotted
onto Trans-Blot nitrocellulose membrane (Bio-Rad Laboratories,
Hercules, CA) at 40 V for 3 hours in a Tris-glycine buffer system. In-
cubation with indicated antibodies was performed for 2 hours in PBS
Tween 20 (PBST) containing 1% BSA with constant rocking. Blots
were washed with PBST and incubated in either antirabbit or anti-
mouse horseradish peroxidase (Santa Cruz Biotechnology, Inc, Santa
Cruz, C A) conjugates for 2 hours in PBST containing 3% nonfat
dry milk. After washing, specific proteins were detected using an en-
hanced chemiluminescence, according to the instructions provided in
the manual (Amersham Life Sciences, Inc).
In Vivo Study
The in vivo efficacy of SFN was tested in a murine xenograft model
of BEAC in which FLO-1 cells were injected subcutaneously in severe
combined immunodeficient (SCID) mice. After detection of tumors,
mice were treated with either 0.75 mg of SFN or 10% DMSO sub-
cutaneously daily for 2 weeks. Tumor growth was measured in two
perpendicular dimensions once every 3 days using a caliper and the
following formula: V =(a
2
× b) / 2, where a is the width of the tumor
(smaller diameter) and b is the length (larger diameter).
Results
SFN Induces Time- and Dose-Dependent Decline in Survival
of BEAC Cells
FLO-1 and OE33 cells were cultured in the presence or absence of
SFN at various concentrations and for variable length of time, substrate-
Figure 1. (continued).
392 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. Translational Oncology Vol. 3, No. 6, 2010
attached viable cell number was counted, and cell viability was con-
firmed by trypan blue exclusion or CCK 8 assays (Figure 1, A and B).
SFN induced both ti me- and dose-dependent decline in survival of
BEAC cells (Figure 1). Exposure to 7 μM SFN led to a complete cell
death in both BEAC cell lines tested, in a period of 3 to 5 days (Figure 1,
A and B). A substantial antiproliferative activity was also seen by SFN
at 5 μM, which killed 100% of OE33 cells in 3 days and 83% ± 4% of
FLO-1 cells in 5 days. The treatment of FLO-1 cells with 3 μMSFN
ledto74%±4%celldeathin5days.At1μM, SFN was less affective
and killed 54% of OE33 cells in 3 days and 39% of FLO-1 cells in 5 days
(Figure 1). These data indicate that SFN inhibits the proliferation of
BEAC cells. To evaluate the effect of SFN on the morphology of treated
cells, both the cancer (FLO-1 , OE33) and normal (fibroblasts, primary
esophageal epithelial) cells were treated with 3 μMSFNfor72hoursand
photographed under a phase-contrast microscope. As shown in Fig ure 1
(C and D), whereas the cancer (FLO-1 and OE33) cells started to detach
and/or reduce in size and number , both types of normal cells (fibroblasts
and esophageal epithelial cells) remained unaffected by the treatment.
To evaluate the nature of detached cells, FLO-1 cells were treated
with SFN for 48 hours. Detached floating cells from the medium
and the attached cells (by trypsinization) were collected separately
and evaluated for the number and viability using trypan blue exclu-
sion. Figure 1E shows the relative proportion of cells detached after
treatment with various concentrations of SFN, expressed as percent
of untreated FLO-1 cells. The total number of detached cells in-
creased with increasing concentrations of SFN. Although majority
(60%) of detached cells were dead, approxi mately 40% appeared
alive, indicating detachment before complete death (Figure 1E ).
Consistent with decline in cell viability and attachment, Western
blot analysis indicated that treatment of BEAC cells with SFN leads
to a marked induction of caspase 8, an initiator caspase implicated in
death receptorinduced apoptosis (Figure 1F ).
SFN Increases Intracellular Accumulation of Drug in
BEAC Cells
To evaluate if SFN can increase intracellular drug accumulation and
chemosensitivity by inhibiting drug efflux, we used rhodamine efflux
assay. Rhodamine 123 is a substrate of MRP and P-glycoprotein, a prod-
uct of multidrug resistance gene (MDR), implicated in the extrusion of
drugs outside the cell. We therefore exposed BEAC cells, pretreated with
SFN or cyclosporin A (broad-spectrum inhibitor of multidrug resistance
gene products), to rhodamine 123. Rhodamine-containing medium
Figure 2. SFN increases intracellular accumulation of drug and enhances antiproliferative activity of other anticancer agents in BEAC cells.
(A and B) Effect of SFN on intracellular drug accumulation. BEAC (FLO-1) cells, pretreated with SFN or cyclosporin A, were exposed to
rhodamine 123. Rhodamine was then removed, and cells were incubated overnight in the presence of corresponding drugs as described.
Cells were harvested, washed, and evaluated for rhodamine 123 florescence by flow cytometry. (A) The amount of rhodamine 123 fluo-
rescence retained was measured using a fluorescence-activated cell flow analyzer. (B) Bar graph showing dose-dependent increases in
the accumulation of intracellular rhodamine 123 in FLO-1 cells treated with different concentrations of SFN. (C) FLO-1 cells were treated
with various concentrations of SFN for 24 hours, and the expression of MRP was monitored by Western blot analysis using MRP (E-19)
antibody (Santa Cruz Biotechnology, Inc).
Translational Oncology Vol. 3, No. 6, 2010 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. 393
was then removed, and cells were washed with PBS and incubated
overnight in the presence of corresponding drugs without rhodamine.
Cells were harvested, washed with PBS, and evaluated for rhodamine
123 florescence by flow cytometry. Figure 2 (A and B)showsadose-
dependent i ncrease in the accumulation of intracellular rhodamine
123 in FLO-1 cells treated with different concentrations of SFN. These
data indicate th at SFN inhibits the drug efflux, thus increasing the
intracellular concentration and chemosensitivity. Consistent with these
data, Western blot analysis showed down-regulation of MRP in SFN-
treated, relative to untreated FLO-1 cells (Figure 2C ).
SFN Significantly Enhances the Antiproliferative Effect of
Chemotherapeutic and Telomerase-Inhibiting Agents in
BEAC Cells
We next evaluated if SFN can enhance the antiproliferative affect
of other anticancer agents. For this purpose, we chose paclitaxel (a
chemotherapeutic agent used for treatment of BEAC), MST-312 (a
chemical inhibitor of telomerase), and GRN163L (a lipidated oligo-
nucleotide targeting RNA component of telomerase). For SFN-PAC
combination study, we first cultured the FLO-1 and OE33 cells in the
presence of paclitaxel at various concentrations and for variable length
of time and evaluated for cell viability as described (not shown). Con-
centrations of SFN and paclitaxel inducing similar cell death at a given
time point were used for combination study. As shown in Figure 3A,
the treatment of FLO-1 cells with 1 μM paclitaxel and 3 μM SFN for
3 days led to 57% ± 6% and 61% ± 6% cell death, respectively. How-
ever, a combination of both drugs led to 92% ± 3% cell death, indi-
cating a significant (P > .0003) 35% increase in cell death relative to
paclitaxel alone (Figure 3 A). Similarly, in OE33 cells, paclitaxel alone
induced 42% ± 5% cell death, whereas a combination of paclitaxel and
SFN led to 79% ± 4% cell death, showing a significant (P <.01)37%
increase in cell death relative to paclitaxel alone (Figure 3B). These data
show that SFN can significantly enhance the antiproliferative effect of
paclitaxel in BEAC cells.
To evaluate the effect of SFN on antiproliferative effect of telomerase
inhibiting agents, FLO-1 cells were pretreated with a telomerase inhib-
itor MST-312 (1 μM) for 10 days to initiate telomere shortening and
were then treated with SFN (3 or 5 μM) for 48 hours. As shown in
Figure 3C, the treatment of FLO-1 cells pretreated with MST-312
to SFN led to a significant (P .002) decrease in cell growth, relative
Figure 3. Effect of SFN on the antiproliferative activity of other agents. BEAC cells, FLO-1 (A) or OE33 (B), were cultured in the medium
containing no drug, 1 μM paclitaxel, 3 μM SFN, or a combination of both 1 μM paclitaxel and 3 μM SFN. Cells were harvested at indi-
cated time points, substrate-attached live cell number was determined, and cell viability was confirmed by trypan blue exclusion and/or
proliferation assays described. Bar graphs show the mean of three independent experiments, with SEM. (C and D) Effect of SFN on
antiproliferative activity of FLO-1 and H460 adenocarcinoma cells pretreated with telomerase inhibitors. FLO-1 cells (C), pretreated with
MST-312 (1 μM) for 10 days, were treated with SFN (3 or 5 μM) for 48 hours, and live cell number was determined. H460 lung adeno-
carcinoma cells (D), treated with GRN163L or mismatch oligonucleotide at 2 μM for 10 days, were exposed to SFN (2 μM) and evaluated
for live cell number everyday for the next 3 days. Error bars represent SEMs of triplicate experiments.
394 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. Translational Oncology Vol. 3, No. 6, 2010
to cells treated with MST-312 or SFN alone. SFN at 3 and 5 μMin-
creased the cell death by 36% and 48%, respectively, in FLO-1 cells pre-
treated with MST-312. Similar observations were also made in H460
lung adenocarcinoma cells treated with telomerase inhibitor GRN163L,
a lipid-attached oligonucleotide-targeting RNA component of telome-
rase. H460, pretreat ed with GRN163L or mismatch oligonucleotid e at
2 μM for 10 days, was exposed to SFN (2 μM) and evaluated for live
cell number everyday for next 3 days. Addition of SFN to cells pretreated
with 163L reduced the live cell number to 16.3% ± 3.2% in 3 days,
whereas 40% cells were still alive when drugs were used separately
(Figure 3D). These data indicate that SFN significantly enhances the
anticancer activity of chemotherapeutic and other antiproliferative agents.
SFN Inhibits Cell Cycle Progression and Enhances the Ability
of Paclitaxel to Induce Cell Cycle Arrest
Paclitaxel is a microtubule inhibitor and is known to induce cell
cycle arrest at G
2
-M. In this study, we evaluated the effect of SFN on
cell cycle progression and on paclitaxel-induced cell cycle arrest in
BEAC cells. FLO-1 cells were treated with SFN (3 μM), paclitaxel
(1 μM), or both for 3 days, and the effect on cell cycle progression was
determined using the Cell Cycle Phase Determination Kit (Cayman
Chemical Company). Briefly, the control and treated cells were fixed,
stained with propidium iodide, and analyzed using a flow cytometer.
Although 41% of control cells were in the S-phase, the fraction of cells
in the S-phase after treatment with paclitaxel and SFN was only 18%
and 24%, respectively (Figure 4A). Consistent with cell viability data,
a more powerful inhibition of the S-phase was observed when pacli-
taxel was combined with SFN (Figure 4A, panel IV ). Cells treated
with paclitaxel were arrested at G
2
-M (55%) or G
1
(27%), whereas
those treated with SFN were mostly at G
1
. However, when SFN was
added along with paclitaxel, majority of cells were arrested at the G
1
phase of the cell cycle (Figure 4A).
Consistent with cell cycle arrest, the treatment with paclitaxel,
SFN, and the combination of both drugs led to 5-fold, 9-fold, and
Figure 4. Effect of SFN, paclitaxel, and combination treatments on cell cycle in BEAC cells. (A) Barrett adenocarcinoma (FLO-1) cells,
untreated or treated with SFN (3 μM), paclitaxel (1 μM), or both for 72 hours, were processed for evaluation of cell cycle using the Cell
Cycle Phase Determination Kit (Cayman Chemical Company). Phases of cell cycle were analyzed in the FL2 channel of a flow cytometer
with a 488-nm excitation filter. Panels: (I) untreated BEAC (FLO-1) cells, (II) BEAC (FLO-1) cells treated with SFN, (III) BEAC (FLO-1) cells
treated with paclitaxel, and (IV) BEAC (FLO-1) cells treated with both the paclitaxel and SFN. (B) Induction of p21 by SFN. Panels: (I)
Western blot showing protein levels of p21 after the above treatments and (II) bar graph showing fold induction in p21 protein levels in
those treated relative to control cells, after normalization with β-actin. (C) Suppression of hsp90 by SFN: Transcript levels of hsp90 in
FLO-1 cells after treatments described above. Panels: (I) Protein levels of hsp90 after the above treatments and (II) bar graph showing
fold reduction in hsp90 protein levels in treated relative to control cells after normalization with β-actin.
Translational Oncology Vol. 3, No. 6, 2010 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. 395
57-fold induction of cell cycle inhibiting protein, p21 (Figure 4B,
panels I and II ). These data not only confirm that p21 was induced
after SFN treatment but also demonstrate a marked effect of the com-
bination treatment in BEAC cells. SFN treatment also led to t he
down-regulation of hsp90, a molecular chaperone required for activity
of several proliferation-associated proteins (Figure 4C ). The suppres-
sion of hsp90 was also the strongest when SFN was used in combina-
tion with paclitaxel.
SFN Induces Apoptotic Cell Death and Enhances the
Proapoptotic Activity of Paclitaxel
BEAC cells (FLO-1 and OE33) were treated with SFN (3 μM)
and paclitaxel (1 μ M) and analyzed for apoptotic cell death. Both
untreated or BEAC cells were sequentially treated with annexin V
biotin and FITC-streptavidin and apoptotic cells were evaluated by a
fluorescence microscope. Approximately 200 cells, representin g at
least five distinct microscopic fields, were analyzed to assess the frac-
tion of annexin-positive cells for each sample. After a 3-day exposure,
43% ± 6% of cells treated with paclitaxel, 50% ± 7% of cells treated
with SFN, and 80% ± 8% of cells treated with both paclitaxel and
SFN were annexin Vpositive (Figure 5A). These data indicate that
both paclitaxel and SFN induce apoptosis in BEAC cells, and the
addition of SFN significantly increases the fraction of cells undergoing
apoptosis. To further evaluate the effect of these treatments and their
combination on apoptosis, we analyzed poly(ADP-ribose) polymerase
(PARP), a protein which undergoes caspase 3mediated cleavage
during apoptosis. At an earlier time point of 48 hours, the cells treated
with paclitaxel or SFN alone had only 5% to 6% of cleaved PARP
product, whereas a substantial fraction (35%) of PARP was found
to be cleaved in the cells treated with combination of both drugs
(Figure 5B).
In Vivo Efficacy of SFN
In vivo efficacy of SFN was demonstrated in a murine model in
which SCID mice were subcutaneously inoculated in the interscap-
ular area with human Barrett adenocarcinoma (BEAC; FLO-1) cells.
After detection of tumors, mice were treated with either 0.75 mg of
SFN or 10% DMSO subcutaneously daily, for 2 weeks. Tumor
growth was measured in two perpendicular dimensions, once every
3 days, using a caliper. As shown in Figure 6A, the tumor size was
Figure 5. Effect of SFN, paclitaxel, and combination treatments on apoptosis in BEAC cells. (A) BEAC (FLO-1) cells, untreated or treated
with SFN (3 μM), paclitaxel (1 μM), or both for 72 hours, were analyzed for apoptosis using the Annexin VBiotin Apoptosis Detection Kit.
Cells were sequentially treated with annexin Vbiotin and FITC-streptavidin. FITC-streptavidinlabeled apoptotic cells within the same
microscopic field were viewed and photographed by phase-contrast (PC) or by fluorescence emitted at 518 nm (FITC filter). Using the
FITC filter, apoptotic cells appear bright green. Panels: (I) Annexin labeling of FLO-1 cells, untreated or treated as described and (II)
bar graph showing percent apoptotic cells after each treatment. (B) FLO-1 cells were treated as described for panel A but for a duration of
48 hours and analyzed for cleavage of PARP, a marker for apoptosis. Panels: (I) PARP was identified by Western blot analysis using a rabbit
polyclonal antibody against PARP (Santa Cruz Biotechnology, Inc) and (II) bar graph showing percentage of cleaved 89-kDa PARP fragment.
396 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. Translational Oncology Vol. 3, No. 6, 2010
significantly reduced in the mice treated with SFN, relative to control
mice. In Figure 6B, the tumor sizes in individual mice show that tumors
in three of four treated mice were smaller than any control mice. These
data show that SFN has significant antitumor activity in vivo.
Discussion
In this study, we have evaluated the therapeutic potential of SFN, an
antioxidant derived from broccoli, in BEAC. We have found that: 1)
SFN induc es both the cell cycle arrest and apoptosis in BEAC cell
lines tested and inhibits tumor growth in a subcutaneous model of
BEAC in vivo. 2) The concentration of SFN (3-7 μM) required to
induce cell cycle arrest and apoptosis in BEAC cells is lower than that
(10-20 μM) reported for similar affects in several other cancer cell types. 3)
Treatment of FLO-1 cells with SFN is associated with down-regulation
of MRP and reduction in drug efflux. 4) SFN significantly enhances the
antiproliferative activity of other antiproliferative agents (paclitaxel, a
chemotherapeutic used in the treatment of BEAC) and telomerase in-
hibitors MST-312 and GRN163L. 5) Addition of SFN with paclitaxel
leads to a marked increase in the expression of p21, cell cycle arrest, and
apoptosis in BEAC cells. 6) Anticancer activity of SFN could be attrib-
uted, at least in part, to the induction of caspase 8 and p21 and the
suppression of MRP and hsp90, a molecular chaperone required for
activity of several proliferation associated proteins.
Anticancer effects of SFN have been demonstrated in several malig-
nancies including human colon, bladder, prostate, ovarian, lympho-
blastoid, pancreatic, cervical cancer, and lung cancers [15,17,3136].
However, to our knowledge, this is the first report demonstrating the
ability of SFN to induce apoptotic cell death, increase chemosensitivity,
and significantly enhance the antiproliferative effects of chemothera-
peutic and telomerase-inhibiting agents in BEAC cells. Moreover, the
SFN could induce apoptosis in BEAC cell lines at 3 to 7 μM, a con-
centration lower than 10 to 40 μM, reported to induce apoptotic cell
death in most other cancers including colon, ovarian, nonsmall lung,
cervical, breast, l ung ade nocarcinoma, hepat oma, and prostate
[10,15,18,3139], indicating a greater sensitivity of this agent for BEAC.
SFN also significantly enhanced the antiproliferative activity of other
anticancer agents including a chemotherapeutic agent, paclitaxel, used
for the treatment of BEAC. A combination of SFN and paclitaxel led to
a significant increase in the fraction of apoptotic cells, as indicated by
annexin labeling (Figure 5). To further confirm the apoptosis and the
increased efficacy of combination treatment, we analyzed the treated
cells for PARP cleavage at an earlier time point of 48 hours. In the cells
treated with paclitaxel, SFN, or both drugs, the fraction of PARP
cleaved into the 89-kDa fragment was 6%, 5%, and 35%, respectively.
These data indicate that the combination of both the drugs leads to a
remarkable increase in apoptotic activity and is consistent with the sig-
nificant increase in cell death and annexin labeling after exposure of
BEAC cells to both the paclitaxel and SFN.
SFN also significantly enhanced the antiproliferative activity of a
chemical inhibitor of telomerase (MST-312) in BEAC cells. A similar
effect was also observed in lung adenocarcinoma H460 cells in which
SFN significantly enhanced the antiproliferative activity of GRN163L,
an oligon ucleotide targeting the RNA component of telomerase.
Consistent with these observations, we have found that SFN down-
regulates MRP and reduces drug efflux in BEAC cells. Because MRPs
have been implicated in the export of chemotherapeutic drugs includ-
ing paclitaxel [40], it is possible that SFN enhances the chemosensitivity
Figure 6. In vivo efficacy of SFN in BEAC. SCID mice were inoculated subcutaneously in the interscapular area with FLO-1 (BEAC) cells,
and after the appearance of tumors, mice were treated intraperitoneally with PBS alone or SFN 25 mg/kg (5 times per week). Average
tumor sizes in all treated and control mice (A) and tumor sizes in individual mice (B) are shown.
Translational Oncology Vol. 3, No. 6, 2010 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. 397
of other antiproliferative agents, at least in part, by increasing intracel-
lular drug concentration.
Exposure of BEAC cells to SFN was associated with both the cell
cycle arrest and apoptosis, and it is consistent with the observations
made in other cancer cell types including, osteosarcoma, ovarian carci-
noma, colon, prostate, and lung cancers [1,14,19,32,41,42]. As shown
in Figure 4, the treatment with SFN led to the accumulation of FLO-1
cells at the G
1
phase of the cell cycle. Although a G
2
/M cell cycle arrest is
more commonly seen after exposure of cancer cells to SFN [15,18,43],
arrest at G
1
has been observed for human colon cancer (HT-29) cells
[44]. Consistent with our observations in BEAC cells, the G
1
arrest in
colon HT-29 cells was also associated with induction of p21.
Several mechanisms have been proposed for induction of apoptosis
after treatment of cancer cells with SFN. SFN-induced apoptosis in
colon cancer (HT-29) cells is attributed to up-regulation of Bax and
release of cytochrome C from mitochondria [16]. In both the prostate
and colon cancer cells, SFN treatment was associated with inhibition
of histone deacetylase, leading to increased histone acetylation and
apoptosis [42,45]. SFN has also been reported to induce apoptosis
through activation of AP-1 [46], activation of MAPK pathways (ERK,
JNK, p38) [44,47], and down-regulation of nuclear factor κB [48]. In
our study, the apoptosis in BEAC cells could be attributed, at least in
part, to the induction of caspase 8 and p21 and suppression of hsp90.
Exposure of BEAC cells to SFN led to a marked induction of caspase 8,
an initiator caspase implicated in death receptormediated apoptosis
[45]. Consistent with this, the transcript levels of caspase 8 and several
death receptors were also elevated in SFN-treated BEAC cells (not
shown). SFN, paclitaxel, and the combination of both led to 9-fold,
5-fold, and 57-fold induction of p21 in FLO-1 cells, respectively. These
data indicate that there could be a remarkable effect of adding SFN
with paclitaxel in treating BEAC.
In summary, these studies demonstrate that SFN inhibits the prolif-
eration o f BEAC cells at a nontoxic concentration, reduces tumor
growth in vivo, and significantly enhances the anticancer activity of
other chemotherapeutic and antiproliferative agents in BEAC. These
data therefore indicate that a natural product with antioxidant properties
from broccoli has a specific activity against BEAC, making it an ideal
compound for therapy and possible chemoprevention of this disease.
References
[1] Clarke JD, Dashwood RH, and Ho E (2008). Multi-targeted prevention of cancer
by sulforaphane. Cancer Lett 269,291304.
[2] Ambrosone CB, McCann SE, Freudenheim JL, Marshall JR, Zhang Y, and
Shields PG (2004). Breast cancer risk in premenopausal women is inversely asso-
ciated with consumption of broccoli, a source of isothiocyanates, but is not mod-
ified by GST genotype. J Nutr 134,11341138.
[3] Joseph MA, Moysich KB, Freudenheim JL, Shields PG, Bowman ED, Zhang Y,
Marshall JR, and Ambrosone CB (2004). Cruciferous vegetables, genetic poly-
morphisms in glutathione S-transferases M1 and T1, and prostate cancer risk.
Nutr Cancer 50, 206213.
[4] van Poppel G, Verhoeven DT, Verhagen H, and Goldbohm RA (1999). Brassica
vegetables and cancer prevention. Epidemiology and mechanisms. Adv Exp Med
Biol 472, 159168.
[5] Chung FL, Conaway CC, Rao CV, and Reddy BS (2000). Chemoprevention of
colonic aberrant crypt foci in Fischer rats by sulforaphane and phenethyl isothio-
cyanate. Carcinogenesis 21, 22872291.
[6] Zhang Y, Talalay P, Cho CG, and Posner GH (1992). A major inducer of anti-
carcinogenic protective enzymes from broccoli: isolation and elucidation of
structure. Proc Natl Acad Sci USA 89, 23992403.
[7] Campas-Baypoli ON, Sanchez-Machado DI, Bueno-Solano C, Ramirez-Wong
B, and Lopez-Cervantes J (2010). HPLC method validation for measurement of
sulforaphane level in broccoli by-products. Biomed Chromatogr 24 , 387392.
[8] Fahey JW, Zhang Y, and Talalay P (1997). Broccoli sprouts: an exceptionally
rich source of inducers of enzymes that protect against chemical carcinogens.
Proc Natl Acad Sci USA 94, 1036710372.
[9] Zhang Y and Talalay P (1994). Anticarcinogenic activities of organic isothiocyanates:
chemistry and mechanisms. Cancer Res 54, 1976s1981s.
[10] Singh SV, Herman-Antosiewicz A, Singh AV, Lew KL, Srivastava SK, Kamath R,
Brown KD, Zhang L, and Baskaran R (2004). Sulforaphane-induced G
2
/M phase
cell cycle arrest involves checkpoint kinase 2mediated phosphorylation of cell
division cycle 25C. J Biol Chem 279, 2581325822.
[11] Hu R, Khor TO, Shen G, Jeong WS, Hebbar V, Chen C, Xu C, Reddy B,
Chada K, and Kong AN (2006). Cancer chemoprevention of intestinal polyposis
in ApcMin/+ mice by sulforaphane, a natural product derived from cruciferous
vegetable. Carcinogenesis 27,20382046.
[12] Chiao JW, Chung FL, Kancherla R, Ahmed T, Mittelman A, and Conaway CC
(2002). Sulforaphane and its metabolite mediate growth arrest and apoptosis in
human prostate cancer cells. Int J Oncol 20, 631636.
[13] Wang L, Liu D, Ahmed T, Chung FL, Conawa y C, and Chiao JW (2004).
Targeting cell cycle machinery as a molecular mechanism of sulforaphane in
prostate cancer prevention. Int J Oncol 24, 187192.
[14] Pappa G, Lichtenberg M, Iori R, Barillari J, Bartsch H, and Gerhauser C
(2006). Comparison of growth inhibition profiles and mechanisms of apoptosis
induction in human colon cancer cell lines by isothiocyanates and indoles from
Brassicaceae. Mutat Res 599,7687.
[15] Cho SD, Li G, Hu H, Jiang C, Kang KS, Lee YS, Kim SH, and Lu J (2005).
Involvement of c-Jun N-terminal kinase in G
2
/M arrest and caspase-mediated
apoptosis induced by sulforaphane in DU145 prostate cancer cells. Nutr Cancer
52, 213224.
[16] Gamet-Payrastre L, Li P, Lumeau S, Cassar G, Dupont MA, Chevolleau S, Gasc N,
Tulliez J, and Tercé F (2000). Sulforaphane, a naturally occurring isothiocyanate,
induces cell cycle arrest and apoptosis in HT29 human colon cancer cells. Cancer
Res 60, 14261433.
[17] Pham NA, Jacobberger JW, Schimmer AD, Cao P, Gronda M, and Hedley DW
(2004). The dietary isothiocyanate sulforaphane targets pathways of apoptosis,
cell cycle arrest, and oxidative stress in human pancreatic cancer cells and inhibits tumor
growth in severe combined immunodeficient mice. Mol Cancer Ther 3, 12391248.
[18] Singh AV, Xiao D, Lew KL, Dhir R, and Singh SV (2004). Sulforaphane induces
caspase-mediated apoptosis in cultured PC-3 human prostate cancer cells and
retards growth of PC-3 xenografts in vivo. Carcinogenesis 25,8390.
[19] Matsui TA, Murata H, Sakabe T, Sowa Y, Horie N, Nakanishi R, Sakai T, and Kubo T
(2007). Sulforaphane induces cell cycle arrest and apoptosis in murine osteosar-
coma cells in vitro and inhibits tumor growth in vivo. Oncol Rep 18,12631268.
[20] Din kov a- Kos tov a AT, Jenkin s SN, Fahey JW, Ye L , Wehage SL, Li by KT,
Stephenson KK, Wade KL, and Talalay P (2006). Protection against UV-light
induced skin carcinogenesis in SKH-1 high-risk mice by sulforaphane-containing
broccoli sprout extracts. Cancer Lett 240,243252.
[21] Xu C, Huang MT, Shen G, Yuan X, Lin W, Khor TO, Conney AH, and Kong
AN (2006). Inhibition of 7,12-dimethylbenz(a)anthraceneinduced skin
tumorigenesis in C57BL/6 mice by sulforaphane is mediated by nuclear factor
E2related factor 2. Cancer Res 66, 82938296.
[22] Fahey JW, Haristoy X, Dolan PM, Kensler TW, Scholtus I, Stephenson KK,
Talalay P, and Lozniewski A (2002). Sulforaphane inhibits extracellular, intra-
cellular, and antibiotic-resistant strains of Helicobacter pylori and prevents
benzo[a]pyrene-induced stomach tumors. Proc Natl Acad Sci USA 99,76107615.
[23] Thejass P and Kuttan G (2006). Augmentation of natural killer cell and antibody-
dependent cellular cytotoxicity in BALB/c mice by sulforaphane, a naturally occur-
ring isothiocyanate from broccoli through enhanced production of cytokines IL-2
and IFN-gamma. Immunopharmacol Immunotoxicol 28,443457.
[24] Thejass P and Kuttan G (2007). Modulation of cell-mediated immune response
in B16F-10 melanoma-induced metastatic tumor-bearing C57BL/6 mice by
sulforaphane. Immunopharmacol Immunotoxicol 29, 173186.
[25] Blot WJ and McLaughlin JK (1999). The changing epidemiology of esophageal
cancer. Semin Oncol 26,28.
[26] Devesa SS, Blot WJ, and Fraumeni JF Jr (1998). Changing patterns in the incidence
of esophageal and gastric carcinoma in the United States. Cancer 83, 20492053.
[27] Aggar wal S, Taneja N, Lin L, Orringer MB, Rehemtulla A, and Beer DG
(2000). Indomethacin-induced apoptosis in esophageal adenocarcinoma cells
involves upregulation of Bax and translocation of mitochondrial cytochrome
C independent of COX-2 expression. Neoplasia 2, 346356.
[28] Ogunwobi OO and Beales IL (2008). Statins inhibit proliferation and induce apop-
tosis in Barretts esophageal adenocarcinoma cells. Am J Gastroenterol 103,825837.
398 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. Translational Oncology Vol. 3, No. 6, 2010
[29] Tselepis C, Morris CD, Wakelin D, Hardy R, Perry I, Luong QT, Harper E,
Harrison R, Attwood SE, and Jankowski JA (2003). Upregulation of the onco-
gene c-myc in Barretts adenocarcinoma: induction of c-myc by acidified bile acid
in vitro. Gut 52, 174180.
[30] Yos hida N, Katada K, Handa O, Takagi T, Kokura S, Naito Y, Mukaida N,
Soma T, Shimada Y, Yoshikawa T, et al. (2007). Interleukin-8 production via
protease -activated receptor 2 in human esophageal epithelial cells. Int J Mol
Med 19,335340.
[31] Andelova H, Rudolf E, and Cervinka M (2007). In vitro antiproliferative effects
of sulforaphane on human colon cancer cell line SW620. Acta Medica (Hradec
Kralove) 50, 171176.
[32] Chuang LT, Moqattash ST, Gretz HF, Nezhat F, Rahaman J, and Chiao JW
(2007). Sulforaphane induces growth arrest a nd apoptosis in h uman ovarian
cancer cells. Acta Obstet Gynecol Scand,16.
[33] Jin CY, Moon DO, Lee JD, Heo MS, Choi YH, Lee CM, Park YM, and Kim
GY (2007). Sulforaphane sensitizes tumor necr osis factorrelated apoptosis-
inducing ligand-mediated apoptosis through downregulation of ERK and Akt
in lung adenocarcinoma A549 cells. Carcinogenesis 28, 10581066.
[34] Mi L, Wang X, Govind S, Hood BL, Veenstra TD, Conrads TP, Saha DT,
Goldman R, and Chung FL (2007). The role of protein binding in induction
of apoptosis by phenethyl isothiocyanate and sulforaphane in human nonsmall
lung cancer cells. Cancer Res 67, 64096416.
[35] Park SY, Kim GY, Bae SJ, Yoo YH, and Choi YH (2007). Induction of apoptosis
by isothiocyanate sulforaphane in human cervical carcinoma HeLa and hepato-
carcinoma HepG
2
cells through activation of caspase-3. Oncol Rep 18, 181187.
[36] Shan Y, Sun C, Zhao X, Wu K, Cassidy A, and Bao Y (2006). Effect of sulfora-
phane on cell growth, G(0)/G(1) phase cell progression and apoptosis in human
bladder cancer T24 cells. Int J Oncol 29,883888.
[37] Chaudhuri D, Orsulic S, and Ashok BT (2007). Antiproliferative activity of sulfo-
raphane in Akt-overexpressing ovarian cancer cells. Mol Cancer Ther 6,334345.
[38] Pledgie-Tracy A, Sobolewski MD, and Davidson NE (2007). Sulforaphane induces
cell typespecific apoptosis in human breast cancer cell lines. Mol Cancer Ther 6,
10131021.
[39] Yeh CTand Yen GC (2005). Effect of sulforaphane on metallothionein expression
and induction of apoptosis in human hepatoma HepG2 cells. Carcinogenesis 26,
21382148.
[40] Vanhoefer U, Cao S, Minderman H, Tóth K, Scheper RJ, Slovak ML, and Rustum
YM (1996). PAK-104P, a pyridine analogue, reverses paclitaxel and doxorubicin
resistance in cell lin es and nude mice bearing xenografts that overexpress the
multidrug resistance protein. Clin Cancer Res 2,369377.
[41] Mi L, Xiao Z, Hood BL, Dakshanamurthy S, Wang X, Govind S, Conrads TP,
Veenstra TD, and Chung FL (2008). Covalent binding to tubulin by isothio-
cyanates: a mechanism of c ell growth arrest and apoptosis. J Biol Chem 283,
2213622146.
[42 ] Myzak MC, Hardin K, Wang R, Dashwood RH, and Ho E (2006). Sulfo-
raphane inhibits histone deacetylase activity in BPH-1, LnCaP and PC-3 prostate
epithelial cells. Carcinogenesis 27,811819.
[43] Herman-Antosiewicz A, Xiao H, Lew KL, and Singh SV (2007). Induction of p21
protein protects against sulforaphane-induced mitotic arrest in LNCaP human
prostate cancer cell line. Mol Cancer Ther 6,16731681.
[44] Shen G, Xu C, Chen C, Hebbar V, and Kong AN (2006). p53-independent G
1
cell cycle arrest of human colon carcinoma cells HT-29 by sulforaphane is associ-
ated with induction of p21
CIP1
and inhibition of expression of cyclin D1. Cancer
Chemother Pharmacol 57,317327.
[45] Myzak MC, Karplus PA, Chung FL, and Dashwood RH (2004). A novel mech-
anism of chemoprotection by sulforaphane: inhibit ion of histone deacetylase.
Cancer Res 64, 57675774.
[46] Jeong WS, Kim IW, Hu R, and Kong AN (2004). Modulatio n of AP-1 by
natural chemopreventive compounds in human colon HT-29 cancer cell line.
Pharm Res 21, 649660.
[47] Jakubikova J, Sedlak J, Mithen R, and Bao Y (2005). Role of PI3K/Akt and
MEK/ERK signaling pathways in sulforaphane- and erucin-induced phase II
enzymes and MRP2 transcription, G
2
/M arrest and cell death in Caco-2 cells.
Biochem Pharmacol 69, 15431552.
[48] Xu C, Shen G, Chen C, Gelina s C, and Kong AN (2005). Suppression of NF-
κBandNF-κB re gulated gene expression by sulforaphane and PEITC
through IκBα, IKK pathway in human prostate cancer PC-3 cells. Oncogene
24,44864495.
Translational Oncology Vol. 3, No. 6, 2010 SFN Inhibition of Barrett Adenocarcinoma Qazi et al. 399
... A significant decrease in tumor weights was observed in the experimental group compared to the control, and an increase in Egr1 expression was noticed along with a decrease in cyclinB1 and CDC25c expression. [121] exposed esophageal cancer cell lines OE33 and FLO-1 to SFN and witnessed inhibited cell growth through apoptosis induction, G1 phase arrest, upregulation of p21, and downregulation of HSP90. Additionally, Lu et al. [122] treated EC9706 and ECa109 esophageal squamous cancer cells with SFN and observed inhibited cell proliferation. ...
... Qazi et al. [121] also extended their in vitro work to evaluate in vivo efficacy of SFN in mice xenografted with BEAC and FLO-1 tumors. After 2 weeks of daily subcutaneous (s.c.) injections of SFN, tumor growth was significantly reduced compared to the control group; however, anticancer mechanisms were not identified. ...
Article
Full-text available
There is substantial and promising evidence on the health benefits of consuming broccoli and other cruciferous vegetables. The most important compound in broccoli, glucoraphanin, is metabolized to SFN by the thioglucosidase enzyme myrosinase. SFN is the major mediator of the health benefits that have been recognized for broccoli consumption. SFN represents a phytochemical of high interest as it may be useful in preventing the occurrence and/or mitigating the progression of cancer. Although several prior publications provide an excellent overview of the effect of SFN in cancer, these reports represent narrative reviews that focused mainly on SFN’s source, biosynthesis, and mechanisms of action in modulating specific pathways involved in cancer without a comprehensive review of SFN’s role or value for prevention of various human malignancies. This review evaluates the most recent state of knowledge concerning SFN’s efficacy in preventing or reversing a variety of neoplasms. In this work, we have analyzed published reports based on in vitro, in vivo, and clinical studies to determine SFN’s potential as a chemopreventive agent. Furthermore, we have discussed the current limitations and challenges associated with SFN research and suggested future research directions before broccoli-derived products, especially SFN, can be used for human cancer prevention and intervention.
... As a result of glucosinolate degradation products [35,36], studies have demonstrated that considerable inhibition of cancer cells of lung by phenethyl isothiocyanate, benzyl isothiocyanate, allyl isothiocyanate, and sulforaphane. Sulforaphane effectively suppressed the growth of esophageal adenocarcinoma cells [37], cancer cells of colon [38], and lung cancer cells [35]. As reported by Tanaka et al. [33], indole-3-methanol inhibits cancer cell of colon, breast, and tongue s in male ACI/N rats [38]. ...
Chapter
Full-text available
There are numerous secondary plant metabolites found in the crop B. juncea, especially glucosinolates. Isothiocyanates, the by-products of glycosinolate breakdown, are beneficial to human health. A number of studies have also called attention to phenolic compounds and carotenoids, both well known for their anti-oxidant properties. A notable feature is that the profiles and concentrations of secondary plant metabolites vary greatly between varieties and that genetic factors are thought to be the most significant factors. In addition, environmental and agronomic factors have also been noted to change the concentrations of secondary plant metabolites. Secondary plant metabolites are primarily produced for defense purposes. Consequently, the intrinsic quality of Indian mustard, including color, aroma, taste, and medicinal properties, is profoundly influenced by its secondary metabolite profile. The health benefits of glycosinolates and the cancer prevention properties of their breakdown products make them of specific interest. Plant cells that have been injured undergo enzymatic decomposition of glucosinolate by endogenous enzymes such as myrosinase, which releases degradation products such as nitriles, epithionitriles, or isothiocyanates. The main phenolic compounds found in B. juncea are flavonoids and hydroxycinnamic acid derivatives. A diverse secondary metabolite pool is also essential for plant-environment interactions.
... ITCs and indoles commonly found in cabbages such as sulforaphane (SFP), erucin (ER), allyl ITC (AITC), 2-phenyethyl ITC (PEITC), iberin (IB) and indole-3-carbinol (I3C) are reported to be responsible for some of the health-promoting properties of Brassicas [2]. SFP, the most studied of all the ITCs, is reported to possess chemoprotective, antioxidative, antimicrobial and neuroprotective properties [4][5][6][7][8][9]. AITC has been found to be potent against bladder [10,11], breast [12] and lung [13] cancer cells. ...
Article
Full-text available
Glucosinolate hydrolysis products are responsible for the health-promoting properties of Brassica vegetables. The impact of domestic cooking on the myrosinase stability, glucosinolates and hydrolysis products in 18 cabbage accession was investigated. Cabbages were steamed, microwaved, and stir-fried before analysis. Cooking significantly affected myrosinase stability and glucosinolate concentrations within and between cabbage morphotypes. Myrosinase was most stable after stir-frying, with up to 65% residual activity. Steaming and microwaving resulted in over 90% loss of myrosinase activity in some accessions. Stir-frying resulted in the greatest decrease in glucosinolate concentration, resulting in up to 70% loss. Steamed cabbages retained the highest glucosinolates after cooking (up to 97%). The profile and abundance of glucosinolate hydrolysis products detected varied across all cooking methods studied. Cooking reduced the amounts of nitriles and epithionitriles formed compared to raw samples. Steaming led to a significant increase in the concentration of beneficial isothiocyanates present in the cabbage and a significantly lower level of nitriles compared to other samples. Microwaving led to a reduction in the concentrations of both nitriles and isothiocyanates when compared to other cooking methods and raw cabbage. The results obtained help provide information on the optimal cooking methods for cabbage, suggesting that steaming may be the best approach to maximising beneficial isothiocyanate production.
... EGCG has the potential to inhibit vascular endothelial growth factor (VEGF) receptor-2, a key receptor associated with tumor angiogenesis (Choudhari et al., 2020). EGCG inhibited cell proliferation via modulating hepatocyte growth factor in human colon cancer cells (Qazi et al., 2010). Genistein exerted anti-angiogenic effects by inhibiting key enzymes responsible for cell division and survival (Li et al., 2012). ...
Article
Background: Epidemiological studies has revealed that a diet rich in fruits and vegetables could lower the risk of certain cancers. In this setting, natural polyphenols are potent anticancer bioactive compounds to overcome the non-target specificity, undesirable cytotoxicity and high cost of treatment cancer chemotherapy. Purpose: The review focuses on diverse classifications of the chemical diversity of dietary polyphenol and their molecular targets, modes of action, as well as preclinical and clinical applications in cancer prevention. Results: The dietary polyphenols exhibit chemo-preventive activity through modulation of apoptosis, autophagy, cell cycle progression, inflammation, invasion and metastasis. Polyphenols possess strong antioxidant activity and control multiple molecular events through activation of tumor suppressor genes and inhibition of oncogenes involved in carcinogenesis. Numerous in vitro and in vivo studies have evidenced that these dietary phytochemicals regulate critical molecular targets and pathways to limit cancer initiation and progression. Moreover, natural polyphenols act synergistically with existing clinically approved drugs. The improved anticancer activity of combinations of polyphenols and anticancer drugs represents a promising perspective for clinical applications against many human cancers. Conclusion: The anticancer properties exhibited by dietary polyphenols are mainly attributed to their anti-metastatic, anti-proliferative, anti-angiogenic, anti-inflammatory, cell cycle arrest, apoptotic and autophagic effects. Hence, regular consumption of dietary polyphenols as food or food additives or adjuvants can be a promising tactic to preclude adjournment or cancer therapy.
... Gallic acid in combination with gamma irradiation inhibits lipophagy to promote lipophagy associated cell death [73]. A synergistic combination of sulforaphane and paclitaxel in Barrett esophageal adenocarcinoma, an enhanced antiproliferation is evident through the onset of apoptosis [140]. Moreover, it inhibits bronchial dysplasia and cellular proliferation which is evident through reduced Ki-67 expression (Fig. 3, Table 2). ...
Article
Full-text available
Despite the advancement in prognosis, diagnosis and treatment, cancer has emerged as the second leading cause of disease associated with death in the globe. With the remarkable application of synthetic drugs in cancer therapy and the onset of therapy-associated adverse effects, dietary phytochemicals have materialized as potent anticancer drugs owing to their antioxidant, apoptosis and autophagy modulating activities. With dynamic regulation of apoptosis and autophagy in association with cell cycle regulation, inhibition in cellular proliferation, invasion and migration, dietary phytochemicals have emerged as potent anticancer pharmacophores for cancer therapeutics. Dietary phytochemicals or their synthetic analogous as individual drug candidates or in combination with FDA approved chemotherapeutic drugs exhibit potent anticancer efficacy. With the advancement in medical sciences, dietary phytochemicals hold high prevalence for their use as precision and personalized medicine to replace conventional chemotherapeutic drugs. Hence, keeping these perspectives in mind, this review focuses on the diversity of dietary phytochemicals and their molecular mechanism of action in several cancer cells and tumor entities. Understanding the possible molecular key players involved, the use of dietary phytochemicals will thrive a new horizon in cancer therapy.
Article
Full-text available
For centuries, plants have been serving as sources of potential therapeutic agents. In recent years, there has been a growing interest in investigating the effects of plant-derived compounds on epigenetic processes, a novel and captivating Frontier in the field of epigenetics research. Epigenetic changes encompass modifications to DNA, histones, and microRNAs that can influence gene expression. Aberrant epigenetic changes can perturb key cellular processes, including cell cycle control, intercellular communication, DNA repair, inflammation, stress response, and apoptosis. Such disruptions can contribute to cancer development by altering the expression of genes involved in tumorigenesis. However, these modifications are reversible, offering a unique avenue for therapeutic intervention. Plant secondary compounds, including terpenes, phenolics, terpenoids, and sulfur-containing compounds are widely found in grains, vegetables, spices, fruits, and medicinal plants. Numerous plant-derived compounds have demonstrated the potential to target these abnormal epigenetic modifications, including apigenin (histone acetylation), berberine (DNA methylation), curcumin (histone acetylation and epi-miRs), genistein (histone acetylation and DNA methylation), lycopene (epi-miRs), quercetin (DNA methylation and epi-miRs), etc. This comprehensive review highlights these abnormal epigenetic alterations and discusses the promising efficacy of plant-derived compounds in mitigating these deleterious epigenetic signatures in human cancer. Furthermore, it addresses ongoing clinical investigations to evaluate the therapeutic potential of these phytocompounds in cancer treatment, along with their limitations and challenges.
Article
Sulforaphane (SFN) is an isothiocyanate commonly found in cruciferous vegetables. It is formed via the enzymatic hydrolysis of glucoraphanin by myrosinase. SFN exerts various biological effects, including anti-cancer, anti-oxidation, anti-obesity, and anti-inflammatory effects, and is widely used in functional foods and clinical medicine. However, the structure of SFN is unstable and easily degradable, and its production is easily affected by temperature, pH, and enzyme activity, which limit its application. Hence, several studies are investigating its physicochemical properties, stability, and biological activity to identify methods to increase its content. This article provides a comprehensive review of the plant sources, extraction and analysis techniques, in vitro and in vivo biological activities, and bioavailability of SFN. This article highlights the importance and provides a reference for the research and application of SFN in the future.
Article
Full-text available
CGG expansions between 55 and 200 in the 5′-untranslated region of the fragile-X mental retardation gene (FMR1) increase the risk of developing the late-onset debilitating neuromuscular disease Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). While the science behind this mutation, as a paradigm for RNA-mediated nucleotide triplet repeat expansion diseases, has progressed rapidly, no treatment has proven effective at delaying the onset or decreasing morbidity, especially at later stages of the disease. Here, we demonstrated the beneficial effect of the phytochemical sulforaphane (SFN), exerted through NRF2-dependent and independent manner, on pathways relevant to brain function, bioenergetics, unfolded protein response, proteosome, antioxidant defenses, and iron metabolism in fibroblasts from FXTAS-affected subjects at all disease stages. This study paves the way for future clinical studies with SFN in the treatment of FXTAS, substantiated by the established use of this agent in clinical trials of diseases with NRF2 dysregulation and in which age is the leading risk factor.
Article
Full-text available
Naturally occurring isothiocyanates (ITCs) from edible vegetables have shown potential as chemopreventive agents against several types of cancer. The aims of the present study were to study the potential of ITCs in chemoprevention and in potentiating the efficacy of cytotoxic drugs in gastric cancer treatment. The chemoprevention was studied in chemically induced mouse model of gastric cancer, namely N-methyl-N-nitrosourea (MNU) in drinking water, and in a genetically engineered mouse model of gastric cancer (the so-called INS-GAS mice). The pharmacological effects of ITCs with or without cisplatin were studied in human gastric cell lines MKN45, AGS, MKN74 and KATO-III, which were derived from either intestinal or diffused types of gastric carcinoma. The results showed that dietary phenethyl isothiocyanate (PEITC) reduced the tumor size when PEITC was given simultaneously with MNU, but neither when administrated after MNU nor in INS-GAS mice. Treatments of gastric cancer cells with ITCs resulted in a time- and concentration-dependent inhibition on cell proliferation. Pretreatment of gastric cancer cells with ITCs enhanced the inhibitory effects of cisplatin (but not 5-fluorouracil) in time- and concentration-dependent manners. Treatments of gastric cancer cells with PEITC plus cisplatin simultaneously at different concentrations of either PEITC or cisplatin exhibited neither additive nor synergetic inhibitory effect. Furthermore, PEITC depleted glutathione and induced G2/M cell cycle arrest in gastric cancer cells. In conclusion, the results of the present study showed that PEITC displayed anti-cancer effects, particularly when given before the tumor initiation, suggesting a chemopreventive effect in gastric cancer, and that pretreatment of PEITC potentiated the anti-cancer effects of cisplatin, possibly by reducing the intracellular pool of glutathione, suggesting a possible combination strategy of chemotherapy with pretreatment with PEITC.
Article
Full-text available
Vertically aligned ZnO nanorod arrays with a diameter of 40–150 nm were fabricated on Al2O3 substrates with and without GaN interlayers, and consequently covered with a ZnO film in situ by a catalyst-free metal–organic vapour phase epitaxy method. X-ray diffraction and transmission electron microscopy measurements demonstrated that the ZnO film/nanorods hybrid structures had a well-ordered wurtzite structure with no lattice mismatch between the film and nanorods, and that the film was homoepitaxially grown horizontally as well as vertically on the pre-grown nanorods. From n-ZnO film/nanorods/p-GaN heterojunctions, we observed a blue light emission with a wavelength of about 440 nm.
Article
Full-text available
Isothiocyanates (ITCs) found in cruciferous vegetables, including benzyl-ITC (BITC), phenethyl-ITC (PEITC), and sulforaphane (SFN), inhibit carcinogenesis in animal models and induce apoptosis and cell cycle arrest in various cell types. The biochemical mechanisms of cell growth inhibition by ITCs are not fully understood. Our recent study showed that ITC binding to intracellular proteins may be an important initiating event for the induction of apoptosis. However, the specific protein target(s) and molecular mechanisms were not identified. In this study, two-dimensional gel electrophoresis of human lung cancer A549 cells treated with radiolabeled PEITC and SFN revealed that tubulin may be a major in vivo binding target for ITC. We examined whether binding to tubulin by ITCs could lead to cell growth arrest. The proliferation of A549 cells was significantly reduced by ITCs, with relative activities of BITC > PEITC > SFN. All three ITCs also induced mitotic arrest and apoptosis with the same order of activity. We found that ITCs disrupted microtubule polymerization in vitro and in vivo with the same order of potency. Mass spectrometry demonstrated that cysteines in tubulin were covalently modified by ITCs. Ellman assay results indicated that the modification levels follow the same order, BITC > PEITC > SFN. Together, these results support the notion that tubulin is a target of ITCs and that ITC-tubulin interaction can lead to downstream growth inhibition. This is the first study directly linking tubulin-ITC adduct formation to cell growth inhibition.
Conference Paper
This paper first gives an overview of the epidemiological data concerning the cancer-preventive effect of brassica vegetables, including cabbages, kale, broccoli, Brussels sprouts, and cauliflower. A protective effect of brassicas against cancer may be plausible due to their relatively high content of glucosinolates. Certain hydrolysis products of glucosinolates have shown anticarcinogenic properties. The results of six cohort studies and 74 case-control studies on the association between brassica consumption and cancer risk are summarized. The cohort studies showed inverse associations between the consumption of brassica's and risk of lung cancer, stomach cancer, all cancers taken together. Of the case-control studies 64% showed an inverse association between consumption of one or more brassica vegetables and risk of cancer at various sites. Although the measured effects might have been distorted by various types of bias, it is concluded that a high consumption of brassica vegetables is associated with a decreased risk of cancer. This association appears to be most consistent for lung, stomach, colon and rectal cancer, and least consistent for prostatic, endometrial and ovarian cancer. It is not yet possible to resolve whether associations are to be attributed to brassica vegetables per se or to vegetables in general. Further epidemiological research should separate the anticarcinogenic effect of brassica vegetables from the effect of vegetables in general. The mechanisms by which brassica vegetables might decrease the risk of cancer are reviewed in the second part of this paper. Brassicas, including all types of cabbages, broccoli, cauliflower, and Brussels sprouts, may be protective against cancer due to their glucosinolate content. Glucosinolates are usually broken down through hydrolysis catalysed by myrosinase, an enzyme that is released from damaged plant cells. Some of the hydrolysis products, viz. indoles, and isothiocyanates, are able to influence phase 1 and phase 2 biotransformation enzyme activities, thereby possibly influencing several processes related to chemical carcinogenesis, e.g. the metabolism, DNA-binding, and mutagenic activity of promutagens. Most evidence concerning anticarcinogenic effects of glucosinolate hydrolysis products and brassica vegetables has come from studies in animals. In addition, studies carried out in humans using high but still realistic human consumption levels of indoles and brassica vegetables have shown putative positive effects on health. The combination of epidemiological and experimental data provide suggestive evidence for a cancer preventive effect of a high intake of brassica vegetables.
Article
Background and aims C-myc over expression is implicated in malignancy although to date this has not been studied in Barrett’s metaplasia. We sought to determine c-myc expression in the malignant progression of Barrett’s metaplasia and whether it may be induced by bile acids seen in gastro-oesophageal refluxate. Methods C-myc protein and mRNA levels were assessed in 20 Barrett’s metaplasia and 20 oesophageal adenocarcinoma samples by western blotting and real time polymerase chain reaction. Levels of c-myc and proliferation were also assessed in cell lines OE21, OE33, SW-480, and TE-7 stimulated with pulses or continuous exposure to the bile acids deoxycholic acid and chenodeoxycholic acid. Results C-myc protein was upregulated in 50% of Barrett’s metaplasia and 90% of oesophageal adenocarcinoma samples compared with squamous, gastric, and duodenal controls. C-myc immunolocalisation in Barrett’s metaplasia revealed discrete nuclear localisation, becoming more diffuse with progression from low to high grade dysplasia to adenocarcinoma. Both continual and pulsed bile acid induced c-myc at pH 4, with no effect at pH 7 or with acidified media alone. Pulsed bile acid treatment induced proliferation (p<0.05); in contrast, continuous exposure led to suppression of proliferation (p<0.05). Conclusions We have shown upregulation of c-myc with malignant progression of Barrett’s metaplasia and suggest that acidified bile may be a novel agent responsible for induction of this oncogene.
Article
Epidemiological studies have linked consumption of broccoli to a reduced risk of colon cancer in individuals with the glutathione S-transferase M1 (GSTM1) null genotype. GSTs are involved in excretion and elimination of isothiocyanates (ITCs), which are major constituents of broccoli and other cruciferous vegetables and have cancer chemopreventive potential, so it is speculated that ITCs may play a role in protection against human colon cancer. However, there is a lack of data from animal studies to support this. We carried out a bioassay to examine whether sulforaphane (SFN) and phenethyl isothiocyanate (PEITC), major ITCs in broccoli and watercress, respectively, and their corresponding N-acetylcysteine (NAC) conjugates, show any chemopreventive activity towards azoxymethane (AOM)-induced colonic aberrant crypt foci (ACF) in F344 rats. Groups of six male F344 rats were treated with AOM subcutaneously (15 mg/kg body wt) once weekly for 2 weeks. SFN and PEITC and their NAC conjugates were administered by gavage either three times weekly for 8 weeks (5 and 20 μmol, respectively) after AOM dosing (post-initiation stage) or once daily for 3 days (20 and 50 μmol, respectively) before AOM treatment (initiation stage). The bioassay was terminated on week 10 after the second AOM dosing and ACF were quantified. SFN, SFN-NAC, PEITC and PEITC-NAC all significantly reduced the formation of total ACF from 153 to 100-116 (P < 0.01) and multicrypt foci from 52 to 27-38 (more than four crypts/focus; P < 0.05) during the post-initiation treatment. However, only SFN and PEITC were effective during the initiation phase, reducing the total ACF from 153 to 109-115 (P < 0.01) and multicrypt foci from 52 to 35 (more than four crypts/focus; P < 0.05). The NAC conjugates were inactive as anti-initiators against AOM-induced ACF. These findings provide important laboratory evidence for a potential role of SFN and PEITC in the protection against colon cancer.
Article
Sulforaphane, a dietary isothiocyanate, possesses potent chemopreventive effects through the induction of cellular detoxifying/antioxidant enzymes via the transcription factor nuclear factor E2–related factor 2 (Nrf2). To investigate carcinogenesis mechanisms related to the regulation of Nrf2, we examined the tumor incidence and tumor numbers per mouse in Nrf2 wild-type (+/+) and Nrf2 knockout (−/−) mice. 7,12-Dimethylbenz( a )anthracene/12- O -tetradecanoylphorbol-13-acetate treatments resulted in an increase in the incidence of skin tumors and tumor numbers per mouse in both genotypes; however, both indices were markedly higher in Nrf2(−/−) mice as compared with Nrf2(+/+) mice. Western blot analysis revealed that Nrf2 as well as heme oxygenase-1, a protein regulated by Nrf2 were not expressed in skin tumors from mice of either genotype, whereas expression of heme oxygenase-1 in Nrf2(+/+) mice was much higher than that in Nrf2(−/−) mice in nontumor skin samples. Next, we examined the chemopreventive efficacy of sulforaphane in mice with both genotypes. Topical application of 100 nmol of sulforaphane once a day for 14 days prior to 7,12-dimethylbenz( a )anthracene/12- O -tetradecanoylphorbol-13-acetate applications decreased the incidence of skin tumor in the Nrf2(+/+) mice when compared with the vehicle-treated group. Importantly, there was no chemoprotective effect elicited by sulforaphane pretreatment in the Nrf2(−/−) mice group. Taken together, our results show for the first time that Nrf2(−/−) mice are more susceptible to skin tumorigenesis and that the chemopreventive effects of sulforaphane are mediated, at least in part, through Nrf2. (Cancer Res 2006; 66(16): 8293-6)
Article
A simple and specific analytical method was developed and tested for the determination of sulforaphane in broccoli by-products. The method includes the optimization of the conversion of glucoraphanin to sulforaphane, followed by purification of extracts using solid-phase extraction and high-performance liquid chromatography (HPLC) analysis. The response surface methodology was used to find optimum conditions for the preparation and purification procedure. Chromatographic conditions for reversed-phase HPLC with UV photodiode array detection were as follows: column, Exil ODS C(18), 25 x 0.46 cm, 5 microm; column temperature, 36 degrees C; mobile phase, a 30 : 70 (v/v) mixture of acetonitrile:water; flow rate, 0.6 mL/min. The detection wavelength was UV 202 nm. Under these conditions, excellent linearity was obtained (r(2) = 1), and the overall recovery was 97.5 and 98.1% for fresh florets and lyophilized florets, respectively. The precision results showed that the relative standard deviation of the repeatability for florets fresh and lyophilized was 3.0 and 4.0%, respectively. Sulforaphane contents were determined in the edible portion of fresh broccoli, and broccoli crop remains.
Article
Consumption of vegetables, especially crucifers, reduces the risk of developing cancer. Although the mechanisms of this protection are unclear, feeding of vegetables induces enzymes of xenobiotic metabolism and thereby accelerates the metabolic disposal of xenobiotics. Induction of phase II detoxication enzymes, such as quinone reductase [NAD(P)H:(quinone-acceptor) oxidoreductase, EC 1.6.99.2] and glutathione S-transferases (EC 2.5.1.18) in rodent tissues affords protection against carcinogens and other toxic electrophiles. To determine whether enzyme induction is responsible for the protective properties of vegetables in humans requires isolation of enzyme inducers from these sources. By monitoring quinone reductase induction in cultured murine hepatoma cells as the biological assay, we have isolated and identified (-)-1-isothiocyanato-(4R)-(methylsulfinyl)butane [CH3-SO-(CH2)4-NCS, sulforaphane] as a major and very potent phase II enzyme inducer in SAGA broccoli (Brassica oleracea italica). Sulforaphane is a monofunctional inducer, like other anticarcinogenic isothiocyanates, and induces phase II enzymes selectively without the induction of aryl hydrocarbon receptor-dependent cytochromes P-450 (phase I enzymes). To elucidate the structural features responsible for the high inducer potency of sulforaphane, we synthesized racemic sulforaphane and analogues differing in the oxidation state of sulfur and the number of methylene groups: CH3-SOm-(CH2)n-NCS, where m = 0, 1, or 2 and n = 3, 4, or 5, and measured their inducer potencies in murine hepatoma cells. Sulforaphane is the most potent inducer, and the presence of oxygen on sulfur enhances potency. Sulforaphane and its sulfide and sulfone analogues induced both quinone reductase and glutathione transferase activities in several mouse tissues. The induction of detoxication enzymes by sulforaphane may be a significant component of the anticarcinogenic action of broccoli.
Article
Organic isothiocyanates block the production of tumors induced in rodents by diverse carcinogens (polycyclic aromatic hydrocarbons, azo dyes, ethionine, N-2-fluorenylacetamide, and nitrosamines). Protection is afforded by alpha-naphthyl-, beta-naphthyl-, phenyl-, benzyl-, phenethyl-, and other arylalkyl isothiocyanates against tumor development in liver, lung, mammary gland, forestomach, and esophagus. Many isothiocyanates and their glucosinolate precursors (beta-thioglucoside, N-hydroxysulfate) occur naturally and sometimes abundantly in plants consumed by humans, e.g., cruciferous vegetables. Nevertheless, the possible contributions of isothiocyanates and glucosinolates to the well recognized protective effects against cancer of high consumptions of vegetables are unclear. The anticarcinogenic effects of isothiocyanates appear to be mediated by tandem and cooperating mechanisms: (a) suppression of carcinogen activation by cytochromes P-450, probably by a combination of down-regulation of enzyme levels and direct inhibition of their catalytic activities, which thereby lower the levels of ultimate carcinogens formed; and (b) induction of Phase 2 enzymes such as glutathione transferases and NAD(P)H: quinone reductase, which detoxify any residual electrophilic metabolites generated by Phase 1 enzymes and thereby destroy their ability to damage DNA. Since isothiocyanates block carcinogenesis by dual mechanisms and are already present in substantial quantities in human diets, these agents are ideal candidates for the development of effective chemoprotection of humans against cancer.
Article
Induction of phase 2 detoxication enzymes [e.g., glutathione transferases, epoxide hydrolase, NAD(P)H: quinone reductase, and glucuronosyltransferases] is a powerful strategy for achieving protection against carcinogenesis, mutagenesis, and other forms of toxicity of electrophiles and reactive forms of oxygen. Since consumption of large quantities of fruit and vegetables is associated with a striking reduction in the risk of developing a variety of malignancies, it is of interest that a number of edible plants contain substantial quantities of compounds that regulate mammalian enzymes of xenobiotic metabolism. Thus, edible plants belonging to the family Cruciferae and genus Brassica (e.g., broccoli and cauliflower) contain substantial quantities of isothiocyanates (mostly in the form of their glucosinolate precursors) some of which (e.g., sulforaphane or 4-methylsulfinylbutyl isothiocyanate) are very potent inducers of phase 2 enzymes. Unexpectedly, 3-day-old sprouts of cultivars of certain crucifers including broccoli and cauliflower contain 10-100 times higher levels of glucoraphanin (the glucosinolate of sulforaphane) than do the corresponding mature plants. Glucosinolates and isothiocyanates can be efficiently extracted from plants, without hydrolysis of glucosinolates by myrosinase, by homogenization in a mixture of equal volumes of dimethyl sulfoxide, dimethylformamide, and acetonitrile at -50 degrees C. Extracts of 3-day-old broccoli sprouts (containing either glucoraphanin or sulforaphane as the principal enzyme inducer) were highly effective in reducing the incidence, multiplicity, and rate of development of mammary tumors in dimethylbenz(a)anthracene-treated rats. Notably, sprouts of many broccoli cultivars contain negligible quantities of indole glucosinolates, which predominate in the mature vegetable and may give rise to degradation products (e.g., indole-3-carbinol) that can enhance tumorigenesis. Hence, small quantities of crucifer sprouts may protect against the risk of cancer as effectively as much larger quantities of mature vegetables of the same variety.