Cyanidin Rutinoside J. Biol. Chem.-2007-Feng-jbc.M610616200

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

JBC Papers in Press.

Published on March 14, 2007 as Manuscript M610616200


The latest version is at http://www.jbc.org/cgi/doi/10.1074/jbc.M610616200

CYANIDIN-3-RUTINOSIDE, A NATURAL POLYPHENOL ANTIOXIDANT,


SELECTIVELY KILLS LEUKEMIC CELLS BY INDUCTION OF OXIDATIVE
STRESS
Rentian Feng1, Hong-Min Ni1, Shiow Y. Wang2, Irina L. Tourkova 1, Michael R. Shulin1, Hisashi
Harada3 and Xiao-Ming Yin1

From the 1Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA,
2
The Fruit Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture,
Beltsville, MD,
3
Departmetn of Internal Medicine, Virginia Commonwealth University, Richmond, VA

Running title: Polyphenol-induced oxidative stress in leukemic cells

Address corresponding to: Dr. Xiao-Ming Yin, Department of Pathology, University of Pittsburgh
School of Medicine, 3550 Terrace Street, Pittsburgh, PA 15261, Phone: 412-648-8436; Fax: 412-684-
9564; E-mail: xmyin@pitt.edu

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Anthocyanins are a group of naturally of being wildly available and being selective
occurring phenolic compounds widely available against tumors.
in fruits and vegetables of human diets. They
have broad biological activities including anti- Natural products derived from plants have
mutagenesis and anti-carcinogenesis, which are recently received much attention as potential
generally attributed to their antioxidant chemopreventive and chemotherapeutic agents.
activities. We studied the effects and the Among them great attention has been given to
mechanisms of the most common type of natural products with established antioxidant
anthocyanins, cyanidin-3-rutinoside, in several activities and less toxicity in normal cells, such as
leukemia and lymphoma cell lines. We found tea polyphenols and resveratrol (1-3). These
that cyanidin-3-rutinoside extracted and substances appear very promising for cancer
purified from black raspberry cultivar Jewel prevention and treatment in pre-clinical models
induced apoptosis in HL-60 cells in a dose- and and clinical trials (1-4).
time-dependent manner. Paradoxically, this Perhaps the most common type of plant
compound induced the accumulation of polyphenols is flavonoids, which provide much of
peroxides, which are involved in the induction the flavor and color to fruits and vegetables. More
of apoptosis in HL-60 cells. In addition, than 5,000 different flavonoids have been
cyanidin-3-rutinoside treatment resulted in described (5). There are six major subclasses of
reactive oxygen species (ROS)-dependent flavonoids (5): anthocyanins, flavones (e.g.,
activation of p38 MAPK and JNK, which apigenin), flavonols (e.g, quercetin), flavanones
contributed to cell death by activating the (e.g., naringenin), catechins of flavanols (e.g.,
mitochondria pathway mediated by Bim. epicatechin) and isoflavones (e.g., genistein).
Down-regulation of Bim or over-expression of Anthocyanins probably deserve the most attention,
Bcl-2 or Bcl-xL considerably blocked apoptosis. as the daily uptake of anthocyanins in human diet
Notably, cyanidin-3-rutinoside treatment did is remarkable, estimated to be 180 to 215 mg/day
not lead to increased ROS accumulation in in the United States (6), which is much higher than
normal human peripheral blood mononuclear the total intake estimated for other flavonoids (23
cells and had no cytotoxic effects on these cells. mg/day).
These results indicate that cyanidin-3- Anthocyanins are the glycosylated form of
rutinoside have the promising potential to be anthocyanidins, which are polyhydroxyl and
used in leukemia therapy with the advantages polymethoxy derivatives of 2-

Copyright 2007 by The American Society for Biochemistry and Molecular Biology, Inc.
phenylbenzopyrylium or flavylium salts (7). mononuclear cells (PBMC) and had very low
Structurally, there are more than a dozen different toxicity toward these cells. Our study thus
anthocyanidins in the plants but cyanidin is the indicates that anti-oxidative natural products, such
most frequently found naturally occurring as cyanidin 3-rutinoside, could exhibit pro-oxidant
anthocyanidin. Cyanidin is widely available in activities selectively in the leukemic cells, which
human diet through crops, beans, fruits, vegetables could be exploited for the development of anti-
and red wine (7), and cyanidin-3-rutinoside (C-3- tumor agents with a low toxicity toward normal
R)1 is a common glycosylated form (8). cells.
Anthocyanins demonstrate strong anti-oxidant
activities in a variety of in vitro assays (7-10). Experimental Procedures
These natural compounds can react with reactive
oxygen species (ROS) and thus interrupt the Reagents. Cyanidin-3-rutinoside (C-3-R)
propagation of new free radical species. The (>99% purity) was purified from black raspberry
double bonds present in the phenolic ring, the cultivar Jewel extract as previously described (10).
hydroxyl side chains and even the glycosylation Purified C-3-R was stored at -80°C and the stock
contribute to the scavenging activity. solution (150 mM) was freshly prepared before
Anthocyanins, particularly cyanidin glycosides, use by dissolving the powder in DMSO.

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


have been found to possess a broad spectrum of The following primary or secondary
biological activities, including the scavenging antibodies were used: anti-Bak (NT) (Upstate
effects on activated carcinogens and mutagens and Biotechnology, Lake Placid, NY); anti-Bak (Ab-1)
the effects on cell cycle regulation (7,8,11-13) (Oncogene, Cambridge, MA); anti-Bax (N-20)
Previous work indicated that cyanidin-rich berry (Santa Cruz Biotechnology, Santa Cruz, CA); anti-
or pomegranate extracts, or purified cyanidins Bax (clone 6A7), anti-Bcl-2, anti-JNK (clone 666),
possess remarkable chemopreventive activities in anti-cytochrome c and anti-Smac (all from BD
cell culture and animal models (11,14-17) Biosciences, San Jose, CA); anti-Bcl-xL, anti-Bim,
Furthermore, cyanidin glycosides purified from anti-phosphorylated JNK and p38MAP kinase (all
berry extracts possess pro-apoptotic properties in from Cell Signaling, Beverly, MA); anti-β-actin
human cancer cells including leukemia cells (Sigma, St. Louis, MO); anti-COX IV subunit II
(13,18-20). These studies suggest that the anti- (Invitrogen Molecular Probes, Carlsbad, CA); and
tumor activities of cyanidin glycosides could be HRP-conjugated secondary antibodies (Jackson
related to their ability to induce apoptosis and that ImmunoResearch Laboratories, West Grove, PA).
these products may have potential as agents for Cell line and cell culture. The human
cancer treatment as well. All these activities have leukemia and lymphoma cell lines HL-60
been thought to be related to the antioxidant (myeloblastic), MOLT-4 (lymphoblastic) and
properties of the cyanidins (8,21-23). However, Daudi (lymphoblastic) were obtained from
this notion has not been carefully examined, American Type Culture Collection (Manassas,
particularly for the apoptotic activity. VA). Human leukemia cells HL-60/neo, HL-
Furthermore, the molecular mechanisms and 60/Bcl-2, HL-60/Bcl-xL were derived from the
signaling pathways initiated by cyanidin laboratory of Dr. Kapil N. Bhalla (Medical
glycosides in cell death are also poorly College of Georgia) and cultured as previously
understood. described (24). The T cell acute lymphoblastic
In the present study, we have found that leukemia cell line, CCRF-CEM and its subline
cyanidin-3-rutinoside paradoxically increases the stably expressing shRNA against Bim were
level of peroxides in the human leukemia cells, cultured as previously described (25). Human
which is responsible for its apoptotic effects. PBMC were prepared from healthy donors
Cyanidin-3-rutinoside subsequently activates the (Pittsburgh Blood Bank) using Ficoll-Hypaque
p38MAP kinase and the pro-death Bcl-2 family centrifugation. The leukemia/lymphoma cell lines
proteins, leading to mitochondrial release of and PBMC were cultured in RPMI 1640 medium
apoptogenic factors and cell death. In contrast, supplemented with 10% fetal bovine serum (FBS).
cyanidin-3-rutinoside caused little ROS generation Analysis of cell death. Apoptotic and
in the normal human peripheral blood necrotic cell death were determined as previously

2
described with modifications (26). Briefly, cells (DCF) in the presence of superoxide radicals or
were double-stained with 10 µmol/L bis- H2O2, respectively, which can be detected by flow
benzimide Hoechst 33258 and propidium iodide cytometry (26). To measure the intracellular
(PI, 1µg/ml) (Invitrogen, Molecular Probes, accumulation of C-3-R-induced H2O2 production,
Carlsbad, CA) for 30 minutes and analyzed by cells were simultaneously treated with C-3-R and
fluorescence microscopy. Approximately 300-500 H2DCFDA for 1.5-3 hours and then harvested for
cells per condition were randomly selected and flow cytometry analysis. In another set of
assessed. Hoechst 33258 positive cells with experiments, intracellular H2O2 levels were
apoptotic (condensed and/or fragmented) nuclei determined by a method based on horseradish
were considered as apoptotic cells regardless peroxidase-dependent oxidation of phenol red,
whether they are PI positive or not, whereas PI- which is assessed by the increased absorbance at
stained cells without apoptotic nuclear changes 610 nm (30).
were considered as necrotic cells. Immunofluorescence and immunoblot
Caspase activities were measured using 15 µg analysis. Immunofluorescence staining was
(caspase-3) or 20µg (caspase-8 and -9) of proteins carried out as previously described (26). Treated
and 20 µM of fluorescent substrates (Ac-DEVD- cells were harvested and pelleted to glass slides by
AFC, AC-IETD-AFC, and Ac-LEHD-AFC for Cytospin® (Thermo Shandon Inc., Pittsburgh,

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


caspase-3, -8, and -9, respectively)(Biomol, PA). Cells were fixed in 4%
Plymouth Meeting, PA)(26). The fluorescence paraformaldehyde/PBS (pH 7.4) for 1 hour at 4°C
signals were detected by a fluorometer (Tecan and permeabilized in 0.5% Triton X-100 for
GENios, Durham, NC) at excitation and emission another 1 hour. Following the incubation of the
wavelengths of 400 nm and 510 nm, respectively. primary antibodies, the molecules of interest were
Cytochrome c release and Bax translocation to detected using Alexa Fluor-conjugated secondary
the mitochondria were analyzed by immunoblot antibodies and fluorescence microscopy.
assay using the cytosolic and membrane fractions For the immunoblot assay, treated cells were
prepared from treated cells. Subcellular lysed with RIPA buffer supplemented with 1 µM
fractionation was conducted using limited plasma EDTA and a cocktail of protease inhibitors.
membrane permeabilization with 0.05% digitonin Proteins in the lysates were quantified and
as previously described (27). separated by SDS- PAGE, transferred to a PVDF
Measurement of ROS. For the in vitro study, membrane and probed with relevant antibodies.
superoxide radicals were quantified
spectrophotometrically in a xanthine/xanthine- Results
oxidase system as previously described (28).. The
final results were expressed as percent inhibition C-3-R selectively induces apoptosis in
of superoxide radical production in the presence of human leukemic cells. Treatment of the human
an antioxidant (C-3-R, ascorbic acid or Trolox). leukemia cell line HL-60 with C-3-R induced a
The assay for hydrogen peroxide (H2O2) levels significant amount of apoptosis in a time and dose-
was carried out following procedures previously dependent manner (Figure 1A) based on the
described (29). The final results were expressed morphology of the nucleus. About 50% of cells
as percent inhibition of H2O2 levels in Ti(IV)- became apoptotic in 18 hours when 50 µM of C-3-
H2O2 complex system in the presence of R was applied and virtually all cells were
antioxidants. apoptotic in the presence of 120 µM or more of C-
For determination of the cellular ROS level, 3-R. There was very little necrosis as measured by
two different methods were used. First, at various propidium iodide (PI) staining. Consistently, a
times (15 minutes to 4 hours) after C-3-R significant amount of caspase-3 and caspase-9
treatment, HL-60 cells were incubated for another activities could be detected in a dose-dependent
15 minutes with either 2 µM of dihydroethidium manner, which peaked around 16 hours (Figure
bromide or 2 µM of 2’,7’- 1B-C). The detected activity was reduced
dichlorodihydrofluorescein diacetate (H2DCFDA). thereafter, likely due to the increased cell death.
These ROS-dependent fluorescent probes were Consequently, C-3-R induced apoptosis and
converted to ethidium or 2’,7’-dichlorofluorescein caspase activation in HL-60 cells could be

3
suppressed by the effector caspase inhibitor, suggest an overall less oxidative environment in
DEVD-CHO (data not shown), and the pan- these cells (Figure 2B).
caspase inhibitor, z-VAD-FMK (Figure 1D). The In contrast, when the leukemic HL-60 cells
mitochondria pathway would be an important were treated with C-3-R, there was an increased
mechanism contributing to C-3-R induced level of H2O2 accumulated in the cells. In the first
apoptosis, as we found that cytochrome c (Figure assay, we added H2DCFDA only for the last 15
1E) and Smac (see below) was released upon C-3- minutes of the culture at each given time point to
R treatment. detect the intracellular level of H2O2 at that
C-3-R also induced apoptosis in other human particular moment. We found that while similar to
leukemia/lymphoma cell lines including MOLT-4, that in the normal PBMC, C-3-R treatment in HL-
Daudi and CCRF-CEM (Figure 1F). It had little 60 cells led to the reduction of intracellular
toxicity against the normal human PBMC as peroxides in the first 15 minutes of the treatment.
measured by nuclear staining with Hoechst 33258 The peroxide level began to reverse in 30 minutes
and by the MTT assay (Figure 2A-B). This and was significantly above the control level by 1
probably is not surprising, as C-3-R is a common hour of culture (Figure 3C). The accumulated
constituent in human diets (5,6). This property is level of peroxide was gradually reduced thereafter
similar to Resveratrol, a natural product widely to the control level by 4 hours after treatment. To

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


studied for its chemopreventive benefits (4). C-3- examine the overall impact of C-3-R treatment on
R had some toxicity against proliferating PBMC the intracellular accumulation of H2O2, we added
stimulated with PHA, based on Hoechst staining the H2DCFDA at the beginning of the culture
(Figure 2A), although this was not as obvious in through the first 1.5 and 3 hours. This could allow
the MTT assay. This toxicity, however, was much the level of DCF accumulated as the production of
lower than that in the proliferating HL-60 cells H2O2 was increased. The result indicated that
(Figure 1A). We had also observed a similar level despite the initial reduction of H2O2 in the first 15
of toxicity of Resveratrol in the proliferating minutes after treatment, the net level of H2O2 was
PBMC using the Hoechst assay (data not shown). increased during the first 3 hours, which was
These observations suggest that proliferating peaked at the first 1.5 hours (Figure 3D). We
PBMC could be in general more susceptible to confirmed this result using an enzyme-based assay
these chemicals. that measures the concentration of H2O2 based on
C-3-R selectively alters the intracellular the horseradish peroxidase-mediated oxidation of
level of peroxides in HL-60 cells. Structurally, phenol red (30) (Figure 3E). In contrast to the
C-3-R possesses the phenolic rings typical of the observed change in H2O2 levels, there were no
anthocyanidins (Figure 3A), suggesting that it has significant changes in the intracellular superoxide
the similar anti-oxidant activity as the other (O2-) levels using dihydroethidium staining (data
members of the family. Indeed, when using the not shown). These results suggested that C-3-R
standard in vitro assays, we found that C-3-R led to a bi-phasic change in the intracellular redox
demonstrated concentration-dependent inhibition status and the net impact is the increase of the
of H2O2 production in the Ti (IV)-H2O2 complex intracellular H2O2 level.
system (Figure 3B). C-3-R could also effectively C-3-R-induced apoptosis could be mediated
inhibit superoxide and peroxyl radicals (ROO.) by ROS, p38MAP kinase and JNK. Based on
production (data not shown). This activity might the above observations, it was possible that C-3-R-
be related to its chemo-preventive activity in the induced apoptosis could be related to its activity in
normal cells (8,21,22). Consistently, we have enhancing cellular peroxide level. To investigate
found that treatment of normal human PBMC with this possibility, we first determined whether a pre-
C-3-R led to reduced intracellular peroxide levels incubation of a non-toxic low dose of H2O2 would
in the time frame of 15 minutes to 4 hours based inhibit C-3-R-induced apoptosis. Such treatment
on the use of the cell permeable nonfluorescent had been well known to induce an adaptive
H2DCFDA, which is oxidized to generate the response from the cells, which leads to cellular
fluorescent product, DCF, by the peroxides resistance to the subsequent challenge of a larger
(Figure 2C). In addition, the increased reduction dose of H2O2 due to the induction of anti-oxidative
of MTT in the C-3-R treated cells could also gene expression (31). Indeed, this pre-treatment

4
with 0.5 to 4 µM of H2O2 for a period of 3 hours conformation-sensitive anti-Bax antibody (clone
dramatically suppressed C-3-R induced apoptosis 6A7) could also detect Bax in a distinctive
in HL-60 cells (Figure 4A). Consistently, C-3-R- punctated pattern following C-3-R treatment,
induced apoptosis, caspase activity and suggesting that the normally cytosol-located Bax
mitochondrial release of apoptogenic factors could was translocated to the mitochondria and was in an
be also suppressed by the general anti-oxidant, N- activated status (Figure 5A). Fractionation
acetyl-cysteine (NAC) or the H2O2 scavenger, studies confirmed Bax translocation upon C-3-R
catalase (Figure 4B-C). These observations treatment (Figure 5B). Interestingly, catalase,
suggest that C-3-R-induced oxidative stress can SB203580 or SP600125 had much stronger effects
activate the mitochondria apoptosis pathway in on Bax conformational change (Figure 5C) than
HL-60 cells. on Bax translocation (Figure 5B). These data
To explore the signaling events upstream the suggest that ROS and stress kinases mainly
mitochondria, we examined the re-dox sensitive mediate the second step of Bax activation, the
stress kinase pathways, which are involved in oligomerization.
apoptosis (32). In untreated HL-60 cells, neither This activation step of Bax and Bak
p38MAP kinase nor JNK was phosphorylated (oligomerizaiton and conformation change) was
(Figure 4D). Upon C-3-R treatment, there was a most likely caused by the BH3-only Bcl-2 family

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


rapid increase in the phosphorylation of both proteins (33). Among the possible BH3-only
p38MAP kinase and JNK, correlating with the molecules that could be activated under the current
increase of intracellular peroxides levels (Figure experimental system, we focused on Bim as it had
3C-E). Indeed, treatment of cells with the anti- been reported to be activated by stress kinases,
oxidant, NAC, and the H2O2 scavenger, catalase, mostly via direct phosphorylation of protein (34-
suppressed the activation of p38MAP kinase and 37). We found that while the general level of
JNK at the early time point (Figure 4E). BimEL did not change significantly following C-3-
While JNK activation seemed to be relatively R treatment, the migration patterns of BimEL was
transient, p38MAP kinase phosphorylation was altered notably (Figure 5D). There were about
sustained through the time course. However, both three species of BimEL that could be detected in
p38MAP kinase inhibitor, SB203580, and JNK the non-treated HL-60 cells, which represent
inhibitor, SP600125, could significantly inhibit different phosphorylation status of BimEL as
mitochondrial release of cytochrome c and Smac, previously shown (34-36,38). Treatment of C-3-R
caspase activation and apoptosis induced by C-3-R rapidly induced the increase of the slowest
(Figure 4B-C), indicating that both stress kinases migrating species, which remained for at least
were involved in C-3-R-activated mitochondria several hours (Figure 5D). When the inhibitors of
apoptosis pathway following C-3-R-induced the stress kinases were added to the culture, this
oxidative stress. up-shift was significantly suppressed (Figure 5E).
Activation of the mitochondria pathway by These results thus indicate that increased BimEL
C-3-R. To understand how C-3-R-mediated phosphorylation following C-3-R treatment was
oxidative stress and stress kinase activated the correlated with the activation of p38MAP kinase
mitochondria apoptosis pathway, we examined and JNK, which could contribute to its pro-
activation of the pro-death Bcl-2 family proteins, apoptotic activity as shown in earlier studies (34-
which are perhaps the most important molecules 37).
promoting this pathway (33). The multi-domain Since suppression of Bim expression using the
pro-death molecules, Bax and Bak, are responsible RNAi-based approach in HL60 cells was not
for the mitochondrial release of apoptogenic successful, we used another leukemic cell line,
factors. When activated, they become CCRF-CEM, for which a subline stably expressing
oligomerized, which can be detected by a shRNA against Bim had been established and
conformation-sensitive antibodies. Indeed, the characterized (25). This cell line was susceptible
mitochondria-localized Bak could be activated by to C-3-R-mediated apoptosis in a dose-dependent
C-3-R treatment as shown by the immunostaining manner (Figure 1F), which could be further
with a conformation-sensitive antibody (Ab-1) suppressed by p38MAP kinase inhibitor,
(Figure 5A). Immunostaining with a SB203580 and the anti-oxidant, NAC and catalase,

5
to the same extent as observed in HL-60 cells where the intracellular H2O2 levels actually
(Figure 5F), suggesting that a conserved pathway remained lower following the treatment,
is activated. Bim expression was considerably suggesting that C-3-R could have a radical-
reduced in the CCRF-CEM subline stably scavenging activity in these cells. It is not quite
expressing a shRNA against Bim and indeed this clear as to why C-3-R has different impact on the
subline was significantly resistant to C-3-R- redox status in the transformed leukemic cells
indcued apoptosis, compared to the subline versus the normal PBMC. But this could be
expressing the shRNA vector only (Figure 5F). related to the notion that transformed cells and
Furthermore, no further reduction of apoptosis was normal cells may have different resting levels of
seen in the Bim knocked-down cells upon ROS and anti-oxidant capacity (39,40).
treatment of SB203580, NAC or catalase, It may be interesting to note that several other
supporting the notion that Bim is the downstream natural products, such as β-phenylethyl
target of ROS and p38 MAP kinase. isothiocyanate (PEITC), a natural compound
Conversely, HL-60 cells that over-expressed found in consumable cruiferous vegetables (41),
the anti-death Bcl-2 or Bcl-xL (Figure 6A) were which are known to be anti-oxidants in the in vitro
significantly resistant to C-3-R induced apoptosis assays, demonstrate pro-oxidant activities in the in
and caspase activation at all tested doses (Figure vivo studies which are responsible for their

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


6B-C). These results indicate that C-3-R-induced apoptotic effects in tumor cells. Notably, PEITC
HL-60 apoptosis is mainly mediated by the also shows a relatively higher selection toward the
mitochondria pathway, which is regulated by the tumor cells for its killing effects and ROS
Bcl-2 family proteins. generation (41). These observations indicate that
the dual effects on the redox status could be a
Discussion shared feature in some biologically active natural
products that are being evaluated for their
Anthocyanins, such as C-3-R, have broad chemoprevention and/or chemotherapeutic
biological effects in anti-mutagenesis and anti- activities.
carcinogenesis and have been hailed as useful The intracellular redox environment could be
natural agents for chemoprevention (8,18,21,22). determined by the relative rate of production and
These activities are generally attributed to their removal of ROS. Changes in ROS level or the
anti-oxidant properties, as determined in a variety intracellular redox environment can significantly
of in vitro assays. While we have confirmed that modulate physiologic and pathologic processes. It
C-3-R, purified from black raspberries, does has been generally postulated that tumor cells
possess the anti-oxidant activity in these in vitro produce more ROS than normal cells, which is
assays, we find that it actually causes ROS connected with increased proliferation and
accumulation in the leukemic cells and that the tumorigenesis (39,40). Oxygen radicals such as
increased oxidative stress is responsible for its superoxide and hydrogen peroxide can act as
pro-apoptotic activity. signaling molecules directly. Alternatively, a
It is not yet clear as to how C-3-R promotes change in redox environment can affect signaling
oxidative stress in the leukemic cells. We have cascades and redox sensitive transcription factors
observed the increase of peroxides, but not involved in proliferation, survival, and hormonal
superoxides, in the treated cells. The surge of response.
ROS can be cumulative and is sufficient to However, excessive ROS production can lead
activate the downstream apoptotic events. It is to cellular damage and induce cell death (40).
likely that treatment with C-3-R could inactivate Because of the pro-oxidant status of the cancer
the glutathione anti-oxidant system, which is cells, they seem to be more susceptible than
involved in scavenging of the peroxides. This normal cells to treatment with agents that cause
assumption is supported by the protective effects oxidative stress, as epitomized by the study using
of NAC observed in this study, which could regain PEITC (41). Our findings that C-3-R, also a
the glutathione levels in C-3-R-treated cells. natural product commonly found in the fruits and
C-3-R, on the other hand, did not induce vegetables, selectively induces the accumulation
increased levels of peroxide in the normal PBMC, of peroxides in the leukemic cell line, HL-60, but

6
not in the normal PBMC, point to the same Bcl-xL, inhibit mitochondria activation, the pro-
direction and suggest that this approach is valid in death molecules promote it (33). Our findings
seeking new generation of therapeutic agents with indicate that C-3-R induced p38MAP kinase and
lower side effects, considering that many JNK activation promotes the activation of BH3-
anticancer drugs currently used in the clinic have only molecule, BimEL, and Bax and Bak
strong cytotoxicity toward normal cells. conformation change. Earlier studies showed that
Not all tumor cells may be subjected to such a Bim could be sequestered in the cytosol by its
selective killing. While we have observed that binding with the dynein light chain-1, and is
several leukemia/lymphoma cell lines (HL-60, released from the microtubules following
MOLT4, Daudi and CCRF-CEM) are sensitive to phosphorylation in response to apoptotic stimuli
C-3-R, we have also found that a lung (35,38). It binds and antagonizes the anti-death
adenocarcinoma cell line A549 and a Bcl-2 or Bcl-xL, thus activating Bax or Bak (43).
hepatocellular carcinoma cell line SMMC-7721 BimEL could be phosphorylated at Ser65 by JNK
are resistant to the treatment of C-3-R (data not (36) or p38MAP kinase (37) in response to growth
shown). It is possible that the redox status of these factor deprivation, UV stimulation or sodium
cells is different from that of leukemic cells like arsenate, agents that all can readily induce
HL-60. Alternatively C-3-R may not be oxidative stress. p38MAP kinase could also

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


sufficiently potent in changing the re-dox balance transcriptionally up-regulate Bim in leukemic cells
to allow the development of oxidative stress in following glucocorticoids treatment (25). Our
some cancer cells. These hypotheses could be finding that C-3-R could promote p38MAP kinase
tested in future experiments. and JNK-mediated BimEL phosphorylation is
ROS can cause cell death via a variety of consistent with these studies. Furthermore,
mechanisms, among which the activation of stress inhibiting Bim expression led to the suppression of
kinases plays an important role (32). Our studies C-3-R-induced apoptosis to the same extent as the
indicate the activation of p38MAP kinase and JNK p38MAP kinase inhibitor and the anti-oxidant,
by C-3-R in a ROS-dependent manner in the HL- supporting the role of Bim in C-3-R-induced
60 and CCRF-CEM cells. The exact mechanisms apoptosis. It has to be pointed, however, that this
by which ROS activate the MAPK pathways are pathway is likely not the only pro-death
not fully clear in many cases and could depend on mechanism activated by C-3-R as suppressing
the types of cells and the stimulus. ROS could ROS, stress kinases or Bim could block C-3-R-
activate the upstream ASK1 (32), or inactivate the induced apoptosis significantly, but not completely.
MAP kinase phosphatase (42), leading to In conclusion, the selective toxicity of C-3-R
sustained phosphorylation and activation of in the leukemic cells is contributed significantly
p38MAP kinase or JNK. Future work should be by its paradoxical pro-oxidant effects in the cancer
directed to explore these possibilities. cells, which activate the stress kinases and
Both ROS and JNK/p38MAP kinase-initiated subsequently the pro-death Bcl-2 family proteins
apoptosis have been closely linked to the intrinsic and the mitochondria apoptotic pathway. This
pathway characterized by the mitochondrial property of C-3-R could be further explored for
release of apoptogenic factors, such as cytochrome the development of anti-tumor agents with a
c and Smac (32). Mitochondrial activation is most higher therapeutic index.
critically regulated by the Bcl-2 family proteins.
While the anti-death molecules, such as Bcl-2 and

References

1. Surh, Y. J. (2003) Nat Rev Cancer 3(10), 768-780


2. Feng, R., Lu, Y., Bowman, L. L., Qian, Y., Castranova, V., and Ding, M. (2005) J Biol Chem
280(30), 27888-27895
3. Lopez-Lazaro, M. (2002) Curr Med Chem Anticancer Agents 2(6), 691-714

7
4. Jang, M., Cai, L., Udeani, G. O., Slowing, K. V., Thomas, C. F., Beecher, C. W. W., Fong,
H. H. S., Farnsworth, N. R., Kinghorn, A. D., Mehta, R. G., Moon, R. C., and Pezzuto, J. M.
(1997) Science 275(5297), 218-220
5. Ross, J. A., and Kasum, C. M. (2002) Annu Rev Nutr 22, 19-34
6. Hertog, M. G., Hollman, P. C., Katan, M. B., and Kromhout, D. (1993) Nutr Cancer 20(1),
21-29
7. Mazza, G., and Miniati, E. (1993) Anthocyanins in fruits, vegetables, and grains, CRC Press,
Inc, Boca Raton, Florida
8. Galvano, F., La Fauci, L., Lazzarino, G., Fogliano, V., Ritieni, A., Ciappellano, S., Battistini,
N. C., Tavazzi, B., and Galvano, G. (2004) J Nutr Biochem 15(1), 2-11
9. Wang, H., Nair, M. G., Strasburg, G. M., Chang, Y. C., Booren, A. M., Gray, J. I., and
DeWitt, D. L. (1999) J Nat Prod 62(2), 294-296
10. Wang, S. Y., and Zheng, W. (2005) Intl. J. Food & Technology 40, 187-195
11. Hagiwara, A., Miyashita, K., Nakanishi, T., Sano, M., Tamano, S., Kadota, T., Koda, T.,
Nakamura, M., Imaida, K., Ito, N., and Shirai, T. (2001) Cancer Lett 171(1), 17-25
12. Serafino, A., Sinibaldi Vallebona, P., Lazzarino, G., Tavazzi, B., Rasi, G., Pierimarchi, P.,

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Andreola, F., Moroni, G., Galvano, G., Galvano, F., and Garaci, E. (2004) FASEB J. 18(15),
1940-1942
13. Lazze, M. C., Savio, M., Pizzala, R., Cazzalini, O., Perucca, P., Scovassi, A. I., Stivala, L.
A., and Bianchi, L. (2004) Carcinogenesis 25(8), 1427-1433
14. Feng, R., Bowman, L. L., Lu, Y., Leonard, S. S., Shi, X., Jiang, B. H., Castranova, V.,
Vallyathan, V., and Ding, M. (2004) Nutr Cancer 50(1), 80-89
15. Ding, M., Feng, R., Wang, S. Y., Bowman, L., Lu, Y., Qian, Y., Castranova, V., Jiang, B. H.,
and Shi, X. (2006) J Biol Chem 281(25), 17359-17368
16. Malik, A., Afaq, F., Sarfaraz, S., Adhami, V. M., Syed, D. N., and Mukhtar, H. (2005) Proc
Natl Acad Sci U S A 102(41), 14813-14818
17. Fimognari, C., Berti, F., Nusse, M., Cantelli Forti, G., and Hrelia, P. (2005) Chemotherapy
51(6), 332-335
18. Wenzel, U., Kuntz, S., Brendel, M. D., and Daniel, H. (2000) Cancer Res 60(14), 3823-3831
19. Katsuzaki, H., Hibasami, H., Ohwaki, S., Ishikawa, K., Imai, K., Date, K., Kimura, Y., and
Komiya, T. (2003) Oncol Rep 10(2), 297-300
20. Fimognari, C., Berti, F., Nusse, M., Cantelli-Forti, G., and Hrelia, P. (2004) Biochem
Pharmacol 67(11), 2047-2056
21. Kahkonen, M. P., Hopia, A. I., Vuorela, H. J., Rauha, J. P., Pihlaja, K., Kujala, T. S., and
Heinonen, M. (1999) J Agric Food Chem 47(10), 3954-3962
22. Zheng, Y., Wang, C. Y., Wang, S. Y., and Zheng, W. (2003) J Agric Food Chem 51(24),
7162-7169
23. Wang, S. Y., Feng, R., Bowman, L., Penhallegon, R., Ding, M., and Lu, Y. (2005) J Agric
Food Chem 53(8), 3156-3166
24. Amarante-Mendes, G. P., Naekyung Kim, C., Liu, L., Huang, Y., Perkins, C. L., Green, D.
R., and Bhalla, K. (1998) Blood 91(5), 1700-1705
25. Lu, Quearry, and Harada. (2006) FEBS Letters 580(14), 3539-3544
26. Ding, W. X., Ni, H. M., DiFrancesca, D., Stolz, D. B., and Yin, X. M. (2004) Hepatology
40(2), 403-413
27. Mikhailov, V., Mikhailova, M., Pulkrabek, D. J., Dong, Z., Venkatachalam, M. A., and
Saikumar, P. (2001) J. Biol. Chem. 276(21), 18361-18374

8
28. Richmond, R., Halliwell, B., Chauhan, J., and Darbre, A. (1981) Anal Biochem 118(2), 328-
335
29. Patterson, B. D., MacRae, E. A., and Ferguson, I. B. (1984) Anal Biochem 139(2), 487-492
30. Pick, E., and Keisari, Y. (1980) J Immunol Methods 38(1-2), 161-170
31. Davies, K. J. (1999) IUBMB Life 48(1), 41-47
32. Matsuzawa, A., and Ichijo, H. (2005) Antioxid Redox Signal 7(3-4), 472-481
33. Green, D. R., and Kroemer, G. (2004) Science 305(5684), 626-629
34. Ley, R., Ewings, K. E., Hadfield, K., and Cook, S. J. (2005) Cell Death Differ 12(8), 1008-
1014
35. Lei, K., and Davis, R. J. (2003) Proc Natl Acad Sci U S A 100(5), 2432-2437
36. Putcha, G. V., Le, S., Frank, S., Besirli, C. G., Clark, K., Chu, B., Alix, S., Youle, R. J.,
LaMarche, A., Maroney, A. C., and Johnson, E. M. (2003) Neuron 38(6), 899-914
37. Cai, B., Chang, S. H., Becker, E. B. E., Bonni, A., and Xia, Z. (2006) J. Biol. Chem. 281(35),
25215-25222
38. Chen, D., and Zhou, Q. (2004) Proc Natl Acad Sci U S A 101(5), 1235-1240
39. Cerutti, P. A. (1985) Science 227(4685), 375-381

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


40. Schumacker, P. T. (2006) Cancer Cell 10(3), 175-176
41. Trachootham, D., Zhou, Y., Zhang, H., Demizu, Y., Chen, Z., Pelicano, H., Chiao, P. J.,
Achanta, G., Arlinghaus, R. B., Liu, J., and Huang, P. (2006) Cancer Cell 10(3), 241-252
42. Kamata, H., Honda, S., Maeda, S., Chang, L., Hirata, H., and Karin, M. (2005) Cell 120(5),
649-661
43. Strasser, A., Puthalakath, H., Bouillet, P., Huang, D. C., O'Connor, L., O'Reilly, L. A.,
Cullen, L., Cory, S., and Adams, J. M. (2000) Ann N Y Acad Sci 917, 541-548

Footnotes

* We would like to thank Dr. Kapil N. Bhalla (Medical College of Georgia) for the HL-60/neo, HL-
60/Bcl-2 and HL-60/Bcl-xL cell lines, and Drs. Wen-Xing Ding and Wentao Gao for advice in certain
experimental procedures. Ms. Lu Feng (Mt. Lebanon Senior High School) partially took part in the
experiment as a summer student. X.-M. Yin is in part supported by NIH grants (CA83817, CA111456
and NS045252).

1
The abbreviation used are: C-3-R: Cyanidin-3-rutinoside; CAT: catalase; DCF: 2’,7’-
dichlorofluorescein; H2DCFDA: 2’,7’-dichlorodihydrofluorescein diacetate; NAC: N-acetyl-cysteine;
PBMC: peripheral blood mononuclear cells; PEITC: β-phenylethyl isothiocyanate; PI: propidium iodide.
STS: staurosporine.

Figures Legends

Figure 1. C-3-R induces apoptosis in HL-60 cells in a dose- and time-dependent way. (A-C). HL-60
cells were incubated with C-3-R at difference dose for 18 hours (A, B) or at 60 µM for indicated hours
(A, C). Cell death was analyzed by Hoechst 33258 and PI staining. Apoptotic and necrotic cell death
were defined in the Experimental Procedure Section. Caspase-3 and caspase-9 activities were measured
using 20 µM of Ac-DEVD-AFC or Ac-LEHD-AFC, respectively (B-C). (D) HL-60 cells were exposed
to C-3-R (60 µM) for 18h in the absence or presence of the pan-caspase inhibitor z-VAD (50 µM).
Percentage of apoptotic cells (upper panel) or caspase-3 activity (lower panel) was measured as above.
Data (mean±SD) represent at least three independent experiments. (E) HL-60 cells were treated with 80

9
µM of C-3-R for the indicated times. Both total cell lysates and cytosolic fractions were prepared and
subjected to immunoblot analysis using antibodies against cytochrome c and β-actin. (F). MOLT-4,
Daudi and CCRF-CEM cells were treated with C-3-R for 18h at the indicated concentrations and then
stained with Hoechst 33258. Cells with the typical apoptotic nuclear changes were quantified (mean ±
SD).

Figure 2. Normal human peripheral blood mononuclear cells are resistant to C-3-R-induced
apoptosis and redox stress. (A-B). Normal resting human PBMC or PMBC stimulated with PHA (10
µg/ml) for 48 hours were incubated with the designated chemicals for 18 hours. C-3-R was used at 80
µM and STS (staurosporine) was used at 1.0 µM or as indicated. Percentage of apoptotic cells was
determined using Hoechst 33258 staining (A). Alternatively, cells were subjected to MTT assay (B).
(C). Normal resting PBMC were incubated with C-3-R (80 µM) or vehicle control for the designated time
point. H2DCFDA (2 µM) was added 15 minutes before the harvest and cells were then analyzed by flow
cytometry. Light line: control untreated samples; Dark line: C-3-R-treated samples.

Figure 3. C-3-R treatment perturbs ROS production in HL-60 cells. (A) The polyphenol structure of
C-3-R. (B) C-3-R suppressed hydrogen peroxide formation in the in vitro H2O2-Ti (IV) system.

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Titanium reagent was added to 200 µl of C-3-R of various concentrations in the presence of NH4OH and
H2O2. The absorbance of the supernatant from the Ti- H2O2 complex was measured at 410 nm and the
inhibition of H2O2 production by C-3-R was determined against blank control. (C). HL-60 cells were
treated with or without C-3-R for the indicated time. H2DCFDA (2 µM) was added in the last 15 minutes
of the culture before cells were harvested for flow cytometry analysis. Light line: control untreated
samples; Dark line: C-3-R treated samples. (D) HL-60 cells were treated with C-3-R (80 µM) for 0, 1.5
or 3 hours and then harvested for flow cytometry analysis. H2DCFDA was added at the beginning of the
culture. (E) HL-60 cells were incubated with C-3-R at the designated dose for 1 hour. The intracellular
H2O2 levels were determined by the red-phenol method as described in Materials and Methods.

Figure 4. C-3-R-induced apoptosis and mitochondria activation can be mediated by ROS via p38
MAPK and JNK. (A). HL-60 cells were pretreated with H2O2 for 3 hours and then cultured in fresh
medium without H2O2, but treated with C-3-R or vehicle control for 18 hours. Apoptotic cells were
determined by Hoechst 33258 staining. (B) HL-60 cells were pretreated with N-acetyl-cysteine (NAC, 5
mM), catalase (CAT, 500 U/ml), SB203580 (SB, 10 µM) or SP600125 (SP, 1 µM) for 1 hour, followed
by incubation with C-3-R (80 µM) for another 18 hours. Apoptosis was determined by Hoechst 332598
staining (upper panel) and capase-3 activities in the total cell lysates were measured using Ac-DEVD-
AFC as the substrate (lower panel). (C) HL-60 cells were pretreated with various reagents as in B,
followed by incubation with C-3-R (80 µM) for another 8 hours. Subcellular fractionation was performed
and the cytosolic fraction was subjected to immunoblot analysis with the indicated antibodies. (D). HL-
60 cells were treated with 80 µM of C-3-R for various times and subjected to immunoblot analysis using
antibodies against total and phosphorylated p38MAP kinase or JNK. (E). HL-60 cells were pretreated
with vehicle control, NAC (5 mM) or CAT (500 U/ml) for 1 hour, followed by incubation with C-3-R (80
µM) for another 2 hours. Immunoblot analysis was carried out as in E.

Figure 5. C-3-R activates the pro-death Bcl-2 family proteins via ROS, p38MAP kinase and JNK.
(A) HL-60 cells were treated with C-3-R (80 µM) for 8 hours and then immunostained with an anti-Bax
(6A7) or anti-Bak (Ab-1) to detect conformationally changed Bax or Bak. (B-C) HL-60 cells were pre-
treated with catalase (CAT, 500 U/ml), SB203580 (SB, 10 µM), or SP600125 (SP, 2 µM) for 1 hour,
followed by incubation with C-3-R (80 µM) for another 8 hours. The mitochondria-enriched heavy
membrane fraction was analyzed by immunoblot assay with the anti-Bax (NT) and anti-COX IV subunit
II antibodies (B). The latter serves as the loading control for the mitochondria fraction. Alternatively,
percentage of HL-60 cells showing positive 6A7 staining in the punctated pattern was quantified (C). (D)

10
HL-60 cells were treated with C-3-R (80 µM) for the designated time point and subjected to immunoblot
analysis using an anti-Bim antibody. (E) HL-60 cells were treated as in B. Total cell lysates were
prepared and analyzed by immunoblot using the anti-Bim and anti-β-actin antibodies. (F). CCRF-CEM
cells expressing the shRNA-vector or expressing the specific shRNA against Bim were treated with
vehicle control (white column), C-3-R (80 µM) in the absence (black column) or presence of SB203580
(10 µM, gray column), NAC (5 mM, horizontal line column) or CAT (500 U/ml, vertical line column) for
24 hours. Percentages of apoptotic cells were determined following Hoechst staining. The insert shows
the expression of BimEL in the two cell lines used.

Figure 6. Bcl-2 or Bcl-xL can protect HL-60 cells from apoptosis induced by C-3-R. (A) HL-60 cell
lines stably expressing the mammalian expression vector (neo) or the human Bcl-xL or Bcl-2 were
analyzed by immunoblot assay for Bcl-xL (left panel) or Bcl-2 (right panel). (B-C) HL-60 stable cell lines
expressing the vector, Bcl-xL or Bcl-2 were treated with C-3-R (0-80 µM) for 18 hours. The percentage of
cells with apoptotic nuclei as determined by Hoechst 33258 staining were quantified (B). Alternatively,
cell lysates were prepared and analyzed for the activities of caspase-3 (C, upper panel) and -9 activities
(C, lower panel).

Downloaded from http://www.jbc.org/ by guest on August 29, 2018

11
Figure 1

A B C
100 Apoptosis
40,000 40,000
Necrosis
Cell Death (%)

80

Caspase-3 Activity
Caspase-3 Activity
60 30,000 30,000

40
20,000 20,000
20
10,000 10,000
0
0 10 20 40 60 80 100 120 160
0 0
C-3-R (µM)
15,000 15000
100
Caspase-9 Activity
Apoptosis

Caspase-9 Activity
Cell Death (%)

80
Necrosis 10,000 10000
60

40 5,000 5000

20

0 0 0

0 8 16 24 48 0 20 40 60 80 0 8 16 24 32
Time (Hr) C-3-R (µM) Time (Hrs)

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Figure 1

D E
80
0 6 8 16 Time (Hrs)

60
Apoptotic Cells %)

Cytosol Cytochrome c

β-Actin
40
Total Cytochrome c

20
F
0 70
30,000
60

Apoptotic Cells (%)


Caspase-3 Activity

50
20,000
40
30
10,000 20
10
0 0
Control C-3-R C-3-R C-3-R (µM): 0 20 40 80 0 20 40 80 0 20 40 80
+ z-VAD
MOLT-4 Daudi CCRF-CEM

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


C Figure 2
A 15 min
100 C-3-R Control
Apoptotic Cells (%)
80

60
30 min
40
C-3-R Control

20

0
0 40 80 40 80 1.0 0 80 1.0 (µM)
C-3-R Resveratrol STS C-3-R STS 1 hr
- + PHA C-3-R Control
B
0.6
0.5
2 hr
0.4 C-3-R Control
OD 570 (nm)

0.3
0.2
0.1
4 hr
0 C-3-R Control
Control C-3R STS Control C-3-R STS
- + PHA

DCF Fluorescence Intensity

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


C 15 min Figure 3
C-3-R Control

A D

Control
30 min
Control

Events
C-3-R
3 Hrs 1.5 Hrs

Rutinoside 1 hr
Control
C-3-R
DCF Fluorescence Intensity
B E
Inhibition of H2O2 Formation (%)

30
25 3
2 hr
20

H2O2 (µM)
Control
C-3-R 2
15
10
1
5
0 4 hr 0
Control
10 20 40 60 80 100 0 20 40 80 160
C-3-R (µM) C-3-R C-3-R (µM)

DCF Fluorescence Intensity

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


A 100
Figure 4

Apoptotic Cells (%)


80
C
- + C-3-R
60
- - SB SP CAT NAC
40 Cytochrome c

20 Smac

0 β-Actin
H2O2 (µM): 0 0 0.5 1 2 4
C-3-R: - +
B 100
D
0 0.5 1 2 4 8 16 Time (Hr)
Apoptotic Cells (%)

80 Phospho-p38

Total p38
60
Phospho-JNKs
40
Total JNKs
20

0
50,000
E
- + C-3-R
Caspase-3 Activity

40,000
- - NAC CAT
30,000 Phospho-p38

Total p38
20,000
Phospho-JNKs
10,000
Total JNKs
0
C-3-R: - +
- - SP SB NAC CAT

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Figure 5
A Control C-3-R
B
- + C-3-R
- - CAT SB SP
Bax (6A7)

Bax

COX-IV

D
Bak (ab-1)

0 1 2 4 8 (Hrs.)

BimEL

β-actin

C
30 E
- + C-3-R
6A7+ Cells (%)

20 - - SB SP

BimEL
10

β-actin
0
- - SP SB CAT
C-3-R: - +

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


F Figure 5

80
Control
70 shRNA
C-3-R
Vector Bim
C-3-R+SB
60 BimEL

Apoptotic Cells (%)


C-3-R+NAC
C-3-R+CAT
50

40

30

20

10

0
shRNA-Vector shRNA-Bim

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Figure 6

A C 30,000

Cell Line: Neo Bcl-xL Neo Bcl-2 25,000

Relative Caspase-3 Activity


20,000

Bcl-xL Bcl-2 15,000

B 10,000

5,000

80
0
Apoptotic Cells (%)

8,000
60
7,000

Relative Caspase-9 Activity


6,000
40
5,000

20 4,000
3,000
0 2,000
C-3-R (µM): 0 20 40 80 0 20 40 80 0 20 40 80
Cell lines: HL-60-Neo HL-60-Bcl-XL HL-60-Bcl-2 1,000
0
C-3-R (µM): 0 40 80 0 40 80 0 40 80
Cell lines: HL-60-Neo HL-60-Bcl-xL HL-60-Bcl-2

Downloaded from http://www.jbc.org/ by guest on August 29, 2018


Cyanidin-3-rutinoside, a natural polyphenol antioxidant, selectively kills leukemic
cells by induction of oxidative stress
Rentian Feng, Hong-Min Ni, Shiow Y. Wang, Irina L. Tourkova, Michael R. Shulin,
Hisashi Harada and Xiao-Ming Yin
J. Biol. Chem. published online March 14, 2007

Access the most updated version of this article at doi: 10.1074/jbc.M610616200

Alerts:
• When this article is cited
• When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

Downloaded from http://www.jbc.org/ by guest on August 29, 2018

You might also like