ArticlePDF Available

Capsaicin-induced genotoxic stress does not promote apoptosis in A549 human lung and DU145 prostate cancer cells

Authors:

Abstract and Figures

Capsaicin is the major pungent component of the hot chili peppers of the genus Capsicum, which are consumed worldwide as a food additive. More recently, the selective action of capsaicin against cancer cells has been reported. Capsaicin was found to induce apoptosis and inhibit proliferation of a wide range of cancer cells in vitro, whereas being inactive against normal cells. As data on capsaicin-induced genotoxicity are limited and the effects of capsaicin against human lung A549 and DU145 prostate cancer cells were not explored in detail, we were interested in determining whether capsaicin-associated genotoxicity may also provoke A549 and DU145 cell death. Capsaicin-induced decrease in metabolic activity and cell proliferation, and changes in the cell cycle were limited to high concentrations used (≥100μM), whereas, at lower concentrations, capsaicin stimulated both DNA double strand breaks and micronuclei production. Capsaicin was unable to provoke apoptotic cell death when used up to 250μM concentrations. Capsaicin induced oxidative stress, but was ineffective in provoking the dissipation of the mitochondrial inner transmembrane potential. A different magnitude of p53 binding protein 1 (53BP1) recruitment contributed to diverse capsaicin-induced genotoxic effects in DU145 and A549 cells. Capsaicin was also found to be a DNA hypermethylating agent in A549 cells. In summary, we have shown that genotoxic effects of capsaicin may contribute to limited susceptibility of DU145 and A549 cancer cells to apoptosis in vitro, which may question the usefulness of capsaicin-based anticancer therapy, at least in a case of lung and prostate cancer. Copyright © 2015 Elsevier B.V. All rights reserved.
Content may be subject to copyright.
Mutation Research 779 (2015) 23–34
Contents lists available at ScienceDirect
Mutation Research/Genetic Toxicology and
Environmental Mutagenesis
journal homepage: www.elsevier.com/locate/gentox
Community address: www.elsevier.com/locate/mutres
Capsaicin-induced genotoxic stress does not promote apoptosis in
A549 human lung and DU145 prostate cancer cells
Anna Lewinskaa,, Paulina Jaroszb, Joanna Czechb, Iwona Rzeszutekb,
Anna Bielak-Zmijewskac, Wioleta Grabowskac, Maciej Wnukb
aDepartment of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza 4, 35-601 Rzeszow, Poland
bDepartment of Genetics, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
cLaboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Pasteura 3, 02-093 Warsaw, Poland
article info
Article history:
Received 29 November 2014
Received in revised form 8 February 2015
Accepted 10 February 2015
Available online 16 February 2015
Keywords:
Capsaicin
Cancer cells
DNA damage
Micronuclei
Oxidative stress
DNA methylation
abstract
Capsaicin is the major pungent component of the hot chili peppers of the genus Capsicum, which are
consumed worldwide as a food additive. More recently, the selective action of capsaicin against can-
cer cells has been reported. Capsaicin was found to induce apoptosis and inhibit proliferation of a wide
range of cancer cells in vitro, whereas being inactive against normal cells. As data on capsaicin-induced
genotoxicity are limited and the effects of capsaicin against human lung A549 and DU145 prostate can-
cer cells were not explored in detail, we were interested in determining whether capsaicin-associated
genotoxicity may also provoke A549 and DU145 cell death. Capsaicin-induced decrease in metabolic
activity and cell proliferation, and changes in the cell cycle were limited to high concentrations used
(100 M), whereas, at lower concentrations, capsaicin stimulated both DNA double strand breaks and
micronuclei production. Capsaicin was unable to provoke apoptotic cell death when used up to 250 M
concentrations. Capsaicin induced oxidative stress, but was ineffective in provoking the dissipation of the
mitochondrial inner transmembrane potential. A different magnitude of p53 binding protein 1 (53BP1)
recruitment contributed to diverse capsaicin-induced genotoxic effects in DU145 and A549 cells. Cap-
saicin was also found to be a DNA hypermethylating agent in A549 cells. In summary, we have shown
that genotoxic effects of capsaicin may contribute to limited susceptibility of DU145 and A549 cancer
cells to apoptosis in vitro, which may question the usefulness of capsaicin-based anticancer therapy, at
least in a case of lung and prostate cancer.
© 2015 Elsevier B.V. All rights reserved.
1. Introduction
Capsaicin, a homovanillic acid derivate (8-methyl-N-vanillyl-6-
nonenamide), is the most abundant pungent ingredient found in
pepper plants (genus Capsicum, family Solanaceae) that is widely
used as a spice [1]. Capsaicin has been previously used to treat pain
and inflammation associated with neuropathic pain conditions,
such as rheumatoid arthritis, cluster headaches, herpes zoster and
vasomotor rhinitis [2–5]. More recently, anticancer activity of cap-
saicin has been demonstrated [6]. Capsaicin-mediated apoptosis
and/or antiproliferative potential has been reported for numer-
ous cancer cell lines, e.g., in leukemia cells [7], multiple myeloma
cells [8], cutaneous cell carcinoma [9], glioma cells [10], tongue
cancer cells [11], nasopharyngeal carcinoma cells [12], esophageal
Corresponding author. Tel.: +48 17 785 54 03; fax: +48 17 872 12 65.
E-mail address: alewinska@o2.pl (A. Lewinska).
carcinoma cells [13], gastric cancer cells [14], pancreatic cancer
cells [15], hepatocarcinoma cells [16], colon carcinoma cells [17],
small cell lung cancer [18], breast cancer cells [19] and prostate
cancer cells [20]. It is widely accepted that capsaicin-associated
anticancer effects may be executed by reactive oxygen species
(ROS) production, cell cycle arrest, regulation of transcription fac-
tor expression and changes in growth/survival signal transduction
pathways, such as EGFR/HER-2 pathway or NF-B inactivation
[9,15,19,21–24]. In contrast, capsaicin-induced autophagy may
result in the suppression of endoplasmic reticulum (ER) stress-
mediated apoptosis, which, in turn, may cause an attenuation of
cancer cell death response, e.g., in malignant breast cells [25].
Because capsaicin carcinogenic and tumorigenic properties have
also been suggested [26], the mechanisms underlying capsaicin
cytotoxicity and chemotherapeutic activity require clarification.
It has been postulated and rebutted that capsaicin may stim-
ulate genotoxic effects [27–30]. These discrepancies may rely on
different protocols and diverse biological materials used to address
http://dx.doi.org/10.1016/j.mrgentox.2015.02.003
1383-5718/© 2015 Elsevier B.V. All rights reserved.
24 A. Lewinska et al. / Mutation Research 779 (2015) 23–34
capsaicin-induced genotoxicity [27–30]. Of course, in a case of anti-
cancer therapy it would be beneficial if capsaicin-induced excessive
DNA damage would provoke apoptotic cell death and cancer cell
elimination.
In the present study, a link between capsaicin-induced geno-
toxicity and cytotoxicity has been evaluated. To address capsaicin
genotoxic potential, we used two distinct cancer cell lines, namely,
A549 lung adenocarcinoma and DU145 prostate adenocarci-
noma cells, because data on capsaicin-induced cytotoxicity and
genotoxicity against these cancer cells are limited and lacking,
respectively. Surprisingly, capsaicin-associated genotoxic effects
were not accompanied by apoptosis, which may have implications
for capsaicin-based anticancer therapy.
2. Materials and methods
2.1. Reagents
Capsaicin (12084, analytical standard grade, 99%) was pur-
chased from Sigma (Poznan, Poland), phosphate-buffered saline
(PBS) was obtained from Gibco, Invitrogen Corporation (Grand
Island, NY, USA), rhodamine G6 and dihydroethidium were pur-
chased from Molecular Probes (Leiden, Netherlands) and dimethyl
sulfoxide (DMSO) was purchased from BioShop (LabEmpire, Rzes-
zow, Poland). Capsaicin was dissolved in DMSO and added to the
medium to a given final concentration. The DMSO concentration
in the cell culture did not exceed 0.1%, which did not influence
the cell survival. All other reagents, if not mentioned otherwise,
were purchased from Sigma (Poznan, Poland) and were of analytical
grade.
2.2. Cell culture
Human prostate carcinoma cells (DU145) and human lung car-
cinoma cells (A549) were obtained from ATCC (LGC Standards,
Lomianki, Poland). Cells (3000 cells/cm2) were cultured at 37 Cin
Dulbecco’s Modified Eagle’s medium (DMEM) supplemented with
10% fetal calf serum (FCS) and antibiotic and antimycotic mix solu-
tion (100 U/ml penicillin, 0.1 mg/ml streptomycin and 0.25 g/ml
amphotericin B) in a humidified atmosphere in the presence of 5%
CO2until they reached confluence. Typically, cells were passaged
by trypsinization and maintained in DMEM.
2.3. MTT assay
Capsaicin-mediated metabolic activity was estimated as a
function of redox potential as an ability of live cells to
metabolize 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium
bromide (MTT) to formazan [31]. The ability of cells to reduce
MTT provides an indication of the mitochondrial integrity and
activity, which, in turn, may be interpreted as a measure of cell
number/proliferation/viability/survival/toxicity. Cells were seeded
onto 96-well plates at a density of 2 ×103cells per well, grown
for 24 h and then the medium was discarded and replaced with
fresh medium with a range of concentrations of capsaicin (from 5 to
250 M) and incubated for another 24 h or 48h. Then, the medium
was removed and cells were supplemented with MTT-containing
medium (500 g/ml working concentration). After 4-h incubation
at 37 C, the medium was removed and formazan crystals were
dissolved in DMSO (30 min, room temperature). Absorbance was
read at 570 nm (measurement wavelength) and at 630 nm (refer-
ence wavelength) using a Tecan Infinite®M200 absorbance mode
microplate reader. Metabolic activity (activity of mitochondrial
dehydrogenases) was calculated as a A(A570 A630). Metabolic
activity at standard growth conditions is considered as 100%.
2.4. BrdU incorporation test
Capsaicin-mediated changes in cell proliferation were esti-
mated as the ability of cells to synthesize DNA, namely,
5-bromo-2-deoxyuridine (BrdU) incorporation using Cell Prolif-
eration ELISA, BrdU (colorimetric) test (Roche) according to the
manufacturer’s instructions. Briefly, cancer cells were seeded onto
96-well plates at a density of 2 ×103cells per well, grown for
24 h and then the medium was discarded and replaced with fresh
medium with a range of concentrations of capsaicin (from 5 to
150 M) and incubated for another 24 h. Then, the medium was
removed and cells were incubated with 10 M BrdU for 6 h. After
cell fixation, BrdU-labeled DNA was denatured and detected with
anti-BrdU antibody and substrate solution. Photometric detection
was performed with a Tecan Infinite®M200 absorbance mode
microplate reader (measurement wavelength: 370 nm, reference
wavelength: 492 nm). The amount of BrdU-labeled DNA reflecting
cell proliferation was calculated as a A(A370 A492). Cell prolifer-
ation at standard growth conditions is considered as 100%.
2.5. Annexin V staining and multi-caspase assay
After 24-h treatment with capsaicin (0–250 M), apoptotic
markers, namely, phosphatidylserine externalization and pan
caspase activity were evaluated. The live, early apoptotic, late apop-
totic and dead cells were assessed using the MuseTM Cell Analyzer
and both the MuseTM Annexin V and Dead Cell Assay Kit and the
MuseTM Multi-caspase Kit according to manufacturer’s instructions
(Merck Millipore, Warsaw, Poland). Briefly, Annexin V was used
to detect phosphatidylserine on the external membrane of apop-
totic cells and a derivatized VAD-peptide (Val-Ala-Asp tripeptide)
was used to detect the activity of multiple caspases (caspase-1,
3, 4, 5, 6, 7, 8, and 9) [32]. The VAD peptide (fluorescent-labeled
inhibitor of caspases, FLICA), derivatized with a fluorescent group
and a fluoromethyl ketone (FMK) moiety, is membrane permeable
and non-cytotoxic and binds to the activated caspases, resulting
in a fluorescent signal proportional to the number of active cas-
pases in the cell [32]. A dead cell marker, 7-aminoactinomycin D
(7-AAD) was also used as an indicator of cell membrane integrity.
Four populations of cells were detected:
non-apoptotic cells (live cells): Annexin V () and 7-AAD (); and
caspase () and 7-AAD (),
early apoptotic cells: Annexin V (+) and 7-AAD (); and caspase
(+) cells exhibiting pan caspase activity: caspase (+) and 7-AAD
(),
late stage apoptotic and dead cells: Annexin V (+) and 7-AAD (+);
and late stage of caspase activity cells: caspase (+) and 7-AAD (+),
mostly nuclear debris (necrotic cells): Annexin V () and 7-AAD
(+); and caspase () and 7-AAD (+).
The calculations were performed automatically, and the
Annexin V and multi-caspase profiles (dot plots) were displayed
using MuseTM Annexin V and dead cell software module and the
MuseTM multi-caspase software module, respectively. As positive
controls, 0.5-h and 3-h treatments with 10 mM hydrogen peroxide
and 10 mM tert-butyl hydroperoxide (tBOOH) were used [33].
2.6. Cell cycle analysis (DNA content)
After 24-h treatment with capsaicin, DNA content analysis was
performed using flow cytometry method as described by Korwek at
al. [34]. The analysis of 104cells was performed using FACSCalibur
and CellQuestPro software.
A. Lewinska et al. / Mutation Research 779 (2015) 23–34 25
2.7. Oxidative stress parameters
After 24-h treatment with capsaicin, steady-state level of
reactive oxygen species (ROS) in the cell culture medium, total
intracellular ROS production and superoxide production were
measured with 2,7-dichlorodihydrofluorescein diacetate (H2DCF-
DA) and dihydroethidium, respectively. Briefly, 5 MH
2DCF-DA
was added to the medium (supernatant obtained after cell
centrifugation) and fluorescence of the 2,7-dichlorofluorescein
(DCF) formed was monitored in a Tecan Infinite®M200 flu-
orescence mode microplate reader. Measurement conditions
were: ex = 495 nm and em =525 nm. Steady-state level of ROS
is presented as RFU (relative fluorescence unit). For ROS and
superoxide kinetics, cells (2.5 ×105cells/ml) were washed and
suspended in PBS containing 0.1% glucose, 0.5 mM EDTA and
5MH
2DCF-DA or 5 M dihydroethidium, respectively. After
15-min incubation in the dark at 37 C, fluorescence intensity
due to oxidation of 2,7-dichlorodihydrofluorescein diacetate
(H2DCF-DA) to 2,7-dichlorofluorescein (DCF) and dihydroethid-
ium to ethidium was monitored in a Tecan Infinite®M200
fluorescence mode microplate reader. Measurement conditions
were: ex = 495 nm and em = 525 nm; temperature 37 C and
ex = 518 nm and em = 605 nm; temperature 37 C, respectively.
Data are presented as RFU (relative fluorescence unit) per minute.
2.8. Mitochondrial membrane potential (m)
After 24-h treatment with capsaicin, cancer cells (2.5 ×105
cells/ml) were incubated with 5 M rhodamine G6 in PBS con-
taining 0.1% glucose, 0.5 mM EDTA at 37 C for 15 min, washed
and fluorescence intensity reflecting mitochondrial membrane
potential (MMP) was monitored in a Tecan Infinite®M200 fluo-
rescence mode microplate reader. Measurement conditions were:
ex = 528 nm and em = 551 nm; temperature 37 C. Mitochondrial
membrane potential is presented as RFU (relative fluorescence
unit).
2.9. Cytokinesis-block micronucleus (CBMN) assay
To evaluate capsaicin-mediated micronuclei generation, after
24-h treatment with capsaicin, a CBMN assay was performed using
BDTM Gentest Micronucleus Assay Kit using the standard proto-
col according to the manufacturer’s instructions. Briefly, the cells
were cultured for 24 h, then capsaicin (5–250 M) was added for
another 24 h and after that the cells were subjected to cytokinesis
blocking solution for another 24 h. A total of 500 binucleated cells
per well [35,36] were scored using an In Cell Analyzer 2000 (GE
Healthcare, UK) equipped with a high performance CCD camera.
Three replicates per concentration were used (a total of 1500 cells
per concentration). For a positive control, 24-h treatment with
100 ng/ml mitomycin C was used [37].
2.10. Comet assay
DNA double strand breaks (DSBs) were assessed using neutral
single-cell microgel electrophoresis (comet assay). After 24-h treat-
ment with capsaicin, cells were suspended in PBS, mixed with low
melting (LM) agarose (0.7%), added to agarose (LM) slides, lyzed
with proteinase K (0.5 mg/ml) and reduced glutathione (2 mg/ml)
in lysis solution (1.25 M NaCl, 50 mM EDTA, 100 mM Tris–HCl, 0.01%
N-lauroylsarcosine sodium salt, pH 10) at 37 C for 2 h and pro-
ceeded with electrophoresis (neutral comet assay buffer: 100 mM
Tris–HCl, 0.5 M NaCl, 1 mM EDTA, 0.2% DMSO, pH 10). Subse-
quently, slides were stained with 0.25 M YOYO-1 (Invitrogen
Corporation, Grand Island, NY, USA) in 2.5% DMSO and 0.5% sucrose,
mounted with a coverslip and digital comet images were imme-
diately captured with an Olympus BX61 fluorescence microscope
equipped with a DP72 CCD camera and Olympus CellF software. The
CCD capture conditions were: exposure time 81 ms, magnification
400×. Images were saved as TIFF files. At least 150 comets were
measured per each sample triplicate using AutoComet Software
http://autocomet.com/index.php (TriTek Corp.). The % tail DNA was
used as a parameter of DNA damage. For a positive control, 4-h
treatment with 10 mM hydrogen peroxide was used.
2.11. 53BP1 immunostaining
For 53BP1 immunostaining, interphase nuclei were used. After
24-h treatment with capsaicin in the 96-well plate, cancer cells
were fixed with 3.7% formaldehyde containing 0.1% Triton X-100
in PBS for 20 min. Subsequently, the cells were incubated with 1%
bovine serum albumin (BSA) in PBST (phosphate buffered saline
containing 0.25% Triton X-100) at room temperature for 30 min.
After washing with PBST, the cells were incubated with a rab-
bit polyclonal antibody against 53BP1 (Novus Biologicals, Poland)
(diluted 1:200 in PBST–BSA (PBST containing 1% BSA)) overnight
at 4 C, and with, a FITC-conjugated, secondary polyclonal anti-
body against rabbit IgG (BD Biosciences, Germany) (diluted 1:200
in PBST–BSA) at room temperature for 1 h. Nuclei were visualized
with Hoechst 33,342. Digital cell images were captured with an In
Cell Analyzer 2000 (GE Healthcare, UK) equipped with a high per-
formance CCD camera. Cells with 0, 1, 2–5 and more than 5 53BP1
foci were scored [%].
2.12. Global DNA methylation
DNA methylation was estimated as a 5-methyl-2-
deoxycytidine (5mdC) level using both high performance liquid
chromatography (HPLC) and enzyme-linked immunosorbent assay
(ELISA).
For HPLC analysis, DNA from control and capsaicin-treated cells
was purified by phenol chloroform extraction and then digested
by a nuclease P1/alkaline phosphatase DNA degradation method
[38]. Typically, 50 g of good quality DNA was used for diges-
tion. Using HPLC, in DNA hydrolysates from cancer cells treated
for 24 h with capsaicin (5–100 M), 5-methyl-2-deoxycytidine
(5mdC) and 2-deoxycytidine (dC) were determined using UV
detector. Separation of the 2-deoxyribonucleosides was performed
using 250 ×4.6 mm 5 m Hypersil GOLD Column equipped with
10 ×4mm 5m Hypersil GOLD drop in guards precolumn. The
nucleosides were eluted with 5% methanol using a HPLC grade
mixture: methanol:water (20%:80%) (v/v) with sodium phosphate
monobasic, pH 3.2–3.4 (34,899 Sigma, Poland) and a flow rate of
1 ml/min. The injection volume of sample was 20 l. Typically,
three replicate injections were performed for each biological sam-
ple. The eluate was monitored with UV detector at 279 nm (5mdC
detection) and 271 nm (dC detection). Linear calibration curves
were obtained in the concentrations ranging from 2.5 nmol/20 lto
20 nmol/20 l for dC and from 0.25 nmol/20 l to 2nmol/20 l for
5mdC. Calibration curves were constructed from triplicate injec-
tions for each concentration. The 5mdC content in genomic DNA
was expressed as a ratio of 5mdC/(dC + 5mdC) [%]. Acquisition
and quantitative analysis of the chromatograms were carried out
using Chromeleon 4.3 software (Dionex Corporation). For a neg-
ative control, 24-h treatment with a DNA methylation inhibitor
5-aza-2-deoxycytidine (5-aza-dC) (5 M) was used [37]. For a pos-
itive control, 24-h treatment with a DNA hypermethylating agent
hydroxyurea (HU) (100 M) was used [37].
For ELISA, MethylFlashTM Methylated DNA Quantification Kit
(Epigentek, Farmingdale, NY, USA) was used according to the man-
ufacturer’s instructions. Briefly, after 24-h treatment with 10 or
26 A. Lewinska et al. / Mutation Research 779 (2015) 23–34
Fig. 1. Capsaicin-mediated changes in metabolic activity (A) and proliferation (B). (A) MTT assay. Metabolic activity at standard growth conditions is considered as 100%.
The bars indicate the SD, n=5, ***p< 0.001 compared with 24-h control, ###p< 0.001 compared with 48-h control (ANOVA and Dunnett’s a posteriori test). DU145 cells (left),
A549 cells (right). (B) Cell proliferation was estimated as the ability of cells to synthesize DNA: BrdU incorporation assay using Cell Proliferation ELISA. Cell proliferation at
standard growth conditions is considered as 100%. The bars indicate the SD, n=5, *** p< 0.001 compared with 24-h control (ANOVA and Dunnett’s a posteriori test). DU145
cells (left), A549 cells (right).
100 M capsaicin, DNA from cancer cells (100 ng) was subjected
to 5mdC content analysis. The positive and negative controls were
always performed. The positive control is a methylated polynu-
cleotide containing 50% of 5mdC, whereas the negative control
is an unmethylated polynucleotide containing 50% of cytosine.
Absorbance was read at 450 nm using a Tecan Infinite®M200
absorbance mode microplate reader. The calculation was made on
the basis of a standard curve obtained for positive control solutions
and 5mdC content is presented as ng 5mdC per 100 ng DNA.
2.13. Statistical analysis
The results represent the mean ±SD from at least three inde-
pendent experiments. The obtained data conform the ANOVA
assumptions as evaluated using Shapiro–Wilk normality test and
Levene test for the equality of variances. Differences between
capsaicin-treated cells versus control cells were assessed using the
one-way analysis of variance (ANOVA) with post hoc testing using
Dunnett’s multiple comparison test. A p-value <0.05 was consid-
ered significant.
The statistical analyses were performed using StatSoft, Inc.
(2005), STATISTICA, version 7.0 (www.statsoft.com).
3. Results
3.1. Capsaicin-mediated cytotoxicity, apoptosis and changes in
the cell cycle
Firstly, cytotoxic/cytostatic potential of capsaicin was inves-
tigated. Because capsaicin was dissolved initially in DMSO, the
possibility of solvent interference was evaluated and we were
unable to observe any DMSO-mediated effects (data not shown).
We used a wide range of capsaicin concentrations from 5 Mto
250 M and two different cancer cell lines: DU145 prostate cancer
cells and A549 lung cancer cells. Both cell lines were found to be
insensitive to 24-h treatment with nutraceutical, when capsaicin
was used at concentrations up to 50 M(Fig. 1A).
For both cell lines, IC50 value was established to be approx-
imately 200 M(Fig. 1A). At the highest concentration used,
250 M, and 24-h incubation time, capsaicin caused a reduction
of metabolic activity of about 80% and 65% in DU145 and A549
cells compared with untreated control, respectively (p< 0.001)
(Fig. 1A). After 48-h treatment, capsaicin was slightly more effec-
tive, especially when higher concentrations were used (Fig. 1A).
Capsaicin also caused a moderate inhibition of cancer cell prolif-
eration (Fig. 1B). At concentration of 150 M, capsaicin lowered
BrdU incorporation of about 30% and 50% in DU145 and A549
cells compared with untreated control, respectively (p< 0.001)
(Fig. 1B).
The level of dead cells was unaffected after capsaicin treatment
using trypan blue exclusion assay (data not shown). Capsaicin,
when used up to 250 M, was also unable to provoke apoptotic
cell death in cancer cells as estimated using Annexin V stain-
ing and multi-caspase assay (Fig. 2). Capsaicin treatment did not
result in either phosphatidylserine externalization or pan caspase
activity, which are widely accepted apoptotic markers (Fig. 2). In
contrast, treatment with the well-known oxidants: hydrogen per-
oxide and tert-butyl hydroperoxide induced apoptosis (Fig. 2). It
is possible that, at the highest concentrations used, the capsaicin-
mediated decrease in metabolic activity estimated using the MTT
assay reflects the antiproliferative and cytostatic action of capsaicin
rather than its ability to stimulate apoptotic cell death.
Capsaicin-associated cell cycle analysis revealed some minor
changes in the particular phases of the cell cycle of DU145 cells,
when capsaicin was used up to 200 M(Fig. 3). The changes in
the G1, S and G2/M phases were statistically insignificant (Fig. 3).
However, at the highest concentration examined (250 M), the
percentage of cells in the S phase dropped from 26% (control con-
ditions) to 17% (250 M capsaicin) and the percentage of cells
in the G1 phase increased from 46% (control conditions) to 58%
(250 M capsaicin) (Fig. 3). These effects were statistically sig-
nificant (p< 0.05) (Fig. 3). The level of polyploidy events was
comparable between control conditions and capsaicin treatment
(Fig. 3). Similar observations were noticed for A549 cells (data not
shown).
A. Lewinska et al. / Mutation Research 779 (2015) 23–34 27
Fig. 2. Capsaicin-mediated apoptosis. After 24-h treatment with capsaicin (0–250 M), apoptosis was assessed using the MuseTM Cell Analyzer and the MuseTM Annexin V
and Dead Cell Assay Kit (panel I) and the MuseTM Multi-caspase Kit (panel II). Panel I: representative Annexin V profiles (dot plots) of DU145 cells are presented. (A) control,
(B) 50 M capsaicin, (C) 100 M capsaicin, (D) 150 M capsaicin, (E) 200M capsaicin, (F) 250 M capsaicin. Panel II: representative multi-caspase profiles (dot plots) of
A549 cells (top) and DU145 cells (bottom) are presented. (A and F) control, (B and G) 150 M capsaicin, (C and H) 250M capsaicin, (D) 10 mM H2O2, 3 h, (E) 10 mM tBOOH,
3 h, (I) 10 mM H2O2, 0.5 h, (J) 10 mM tBOOH, 0.5 h.
28 A. Lewinska et al. / Mutation Research 779 (2015) 23–34
Fig. 3. Cell cycle analysis after capsaicin treatment. After 24-h treatment with capsaicin (0–250 M), cell cycle analysis was performed using flow cytometry. (A) Repre-
sentative histograms of DU145 cells are shown. M1: G1, M2: sub-G1, M3: G2/M, M4: S, M5: polyploidy. (B) Graph showing % of cells in a particular phase of cell cycle and
polyploidy events. The bars indicate the SD, n=3, *p< 0.05 compared with 24-h control (ANOVA and Dunnett’s a posteriori test).
3.2. Capsaicin-induced oxidative stress
Capsaicin acted as a prooxidant, because it stimulated both
extracellular and intracellular redox imbalance (Fig. 4). The effects
were concentration-dependent and similar for two cancer cell lines
examined (Fig. 4).
After capsaicin treatment, culture medium was found more
oxidative compared with untreated control (Fig. 4A). The level
of reactive oxygen species (ROS) was increased about 15% after
treatment with 150 M capsaicin compared with the control con-
ditions (p< 0.001) (Fig 4A). After treatment with 150 M capsaicin,
intracellular ROS and superoxide production was augmented
approximately 2-fold and 4-fold, respectively (p< 0.001) (Fig. 4B
and C). As cells were suffered from oxidative stress, we were inter-
ested in determining whether the disequilibrium in the redox
homeostasis may affect mitochondria, namely, the mitochondrial
membrane potential (MMP). Surprisingly, no changes in rhodamine
G6 fluorescence reflecting MMP were observed (Fig. 4D). The effect
was comparable for two cancer cell lines used (Fig. 4D).
3.3. Capsaicin-mediated genotoxicity
Capsaicin, at a very mild inhibitory concentration of 100 M
(15% reduction of metabolic activity, Fig. 1A), was found an inducer
of DNA double strand breaks (DSBs) (Fig. 5A).
Capsaicin, in a concentration-dependent manner, caused
increase in DNA double strand breaks (Fig. 5A). After treatment with
100 M capsaicin, the level of DNA DSBs was elevated 2.5- and 3-
fold compared with untreated control in A549 cells and DU145 cells,
respectively (p< 0.05) (Fig. 5A). Capsaicin also caused micronuclei
(MN) formation (p< 0.05) (Fig. 5B). However, the effect was not
concentration-dependent, especially for DU145 cells (Fig. 5B). Cap-
saicin, at concentrations up to 100 M, increased MN production up
to 4-fold (p< 0.05), whereas at the highest concentration (250 M),
a decrease in MN production to control level was observed in DU145
cells (Fig. 5B). In contrast, treatment with capsaicin, at concentra-
tions up to 250 M, stimulated MN production from 2- to 3-fold
compared with untreated control in A549 cells (p< 0.05). We also
investigated capsaicin-mediated formation of p53 binding protein
(53BP1) foci, which are considered to be accumulated at site of DSBs
being a part of DNA repair process. Surprisingly, an inverse correla-
tion between 53BP1 foci and MN generation was shown in DU145
cells (Fig. 5C). In contrast, in A549 cells, in which the level of MN pro-
duction was relatively high after treatment with capsaicin at a wide
range of concentrations, and did not decrease at higher concentra-
tions, the formation of 53BP1 foci was also high and comparable
between concentrations used (Fig. 5C).
3.4. Capsaicin-mediated epigenetic changes
Capsaicin-mediated changes in the global DNA methylation
were also evaluated. After capsaicin treatment, the level of
5-methyl-2-deoxycytidine (5mdC) was unchanged in DU145,
whereas capsaicin was found to be a potent DNA hypermethylating
agent in A549 cells (Fig. 6).
A. Lewinska et al. / Mutation Research 779 (2015) 23–34 29
Fig. 4. Capsaicin-mediated oxidative stress (A–C) and changes in mitochondrial membrane potential (D). (A) Steady-state level of reactive oxygen species (ROS) in the cell
culture medium. (B) Intracellular ROS production. (C) Intracellular superoxide production. Fluorescence intensity was monitored in a Tecan Infinite®M200 fluorescence
mode microplate reader. The bars indicate the SD, n=5, ***p< 0.001 compared with 24-h control (ANOVA and Dunnett’s a posteriori test). DU145 cells (left), A549 cells (right).
(D) Fluorescence intensity of rhodamine G6 reflecting mitochondrial membrane potential (MMP) was monitored in a Tecan Infinite®M200 fluorescence mode microplate
reader. The bars indicate the SD, n= 5. DU145 cells (left), A549 cells (right).
30 A. Lewinska et al. / Mutation Research 779 (2015) 23–34
Fig. 5. Capsaicin-mediated genotoxicity. After 24-h treatment with capsaicin, DNA double strand breaks (DSBs) (A), micronuclei production (B) and 53BP1 foci (C) were
analyzed. (A) DSBs were assessed using neutral comet assay. As a DNA damage marker, the % tail DNA was used. A positive control (4-h treatment with 10 mM hydrogen
peroxide) is also shown. The bars indicate the SD, n= 150, ** p< 0.01, *p< 0.05 compared with 24-h control (ANOVA and Dunnett’s a posteriori test). DU145 cells (left), A549
cells (right). (B) CBMN assay. A positive control (24-h treatment with 100 ng/ml mitomycin C) is also shown. The bars indicate the SD, n=1500, *p< 0.05 compared with 24-h
control (ANOVA and Dunnett’s a posteriori test). DU145 cells (left), A549 cells (right). (C) 53BP1 foci were revealed using 53BP1 immunostaining. Cells with 0, 1, 2–5 and
more than 5 53BP1 foci were scored [%]. DU145 cells (left), A549 cells (right).
HPLC analysis revealed that a concentration of 10 M of cap-
saicin caused an increase in 5mdC level of approximately 20%
compared with untreated control in A549 cells (p< 0.001), whereas
ELISA-based 5mdC quantification showed a 40% increase in 5mdC
level compared with control conditions (p< 0.05) (Fig. 6).
4. Discussion
In the present study, capsaicin genotoxic potential in two can-
cer cell lines DU145 and A549 has been demonstrated. Capsaicin
induced DNA double strand breaks (DSBs) and chromosomal dam-
age, and capsaicin-associated genomic instability affected capsaicin
pro-apoptotic activity and cytotoxicity, which, in turn, may lead to
limited effectiveness of capsaicin-based anticancer therapy. DNA
damaging agents may affect DNA yielding modified bases, intra-
and inter-strand cross-links, cyclobutane pyrimidine dimers, 6-
4 photoproducts, single- and double-stranded DNA breaks. Upon
sensing DNA damage, a well co-ordinated network of signaling cas-
cade, termed the DNA damage response (DDR), is induced [39].
Depending on the magnitude of DNA damage, different cellular
processes may be activated, such as cell cycle checkpoint control,
transcription, DNA repair machinery, senescence and/or apoptotic
cell death [39]. Thus, there can be two outcomes of genotoxic injury:
survival or apoptosis.
The concentration- and time-dependent anticancer potential
of capsaicin in vitro has been well-documented [1]. Capsaicin is
believed to be toxic to cancer cells when added at the micromo-
lar range [1]. The maximal antiproliferative activity of capsaicin
A. Lewinska et al. / Mutation Research 779 (2015) 23–34 31
Fig. 6. Capsaicin-mediated changes in the global DNA methylation. After 24-h treatment with capsaicin, DNA methylation was estimated as a 5-methyl-2-deoxycytidine
(5mdC) level using HPLC (A) and ELISA (B). The 5mdC content in genomic DNA was expressed as a ratio of 5mdC/(dC + 5mdC) [%] (A) and as ng 5mdC/100ng DNA (B). (A)
The cells treated with a DNA methylation inhibitor 5-aza-2-deoxycytidine (5-aza-dC) (5 M, 24-h treatment) served as a negative control, whereas cells treated with a DNA
hypermethylating agent hydroxyurea (HU) (100 M, 24-h treatment) served as a positive control. The bars indicate the SD, n=5, *p< 0.05, *** p< 0.001 compared with 24-h
control (ANOVA and Dunnett’s a posteriori test). DU145 cells (left), A549 cells (right).
has been observed at about 200–300 M[1]. The response to cap-
saicin may also be cancer cell type-dependent [1]. The sensitivity
of A549 and DU145 cells to capsaicin was comparable as estimated
using MTT test. We have established IC50 value to be approximately
200 M for both cell lines examined. Capsaicin was not able to
inhibit completely the proliferation of cancer cells. The maximum
antiproliferative effect of approximately 30% and 50% for DU145
and A549 cells compared with untreated control was observed,
respectively. Moreover, capsaicin did not stimulate apoptosis or
massive changes in the cell cycle. We were unable to observe any
significant changes in sub-G1 population or changes in Annexin V
staining or pan caspase activity after capsaicin treatment. Data on
capsaicin effects on DU145 and A549 cells are scarce and contradic-
tory. Cytotoxicity of dihydrocapsaicin (DHC), a saturated structural
analog of capsaicin, against A549 cells was found moderate [40].
A549 cells did not respond to DHC treatment when DHC was used
up to 200 M, whereas 400 M DHC treatment resulted in decrease
in metabolic activity of about 30% compared with untreated con-
trol [40]. After 200 M DHC treatment, the level of apoptotic cells
increased to about 10% [40]. In contrast, 100 M capsaicin was
established to be IC50 value in A549 cells, whilst 200 M capsaicin
accounted for 90% inhibition of cell metabolic activity [41]. After
100 M capsaicin treatment, clonal proliferation of DU145 cells
was reduced to 20% of control level [20].10M capsaicin was
established to be IC50 value in DU145 cells [20]. Beside cancer cell
type-specific response to capsaicin [33], capsaicin-based toxicolog-
ical data may be also affected by some other factors, e.g., capsaicin
stability during particular experimental conditions, which, in turn,
may yield contradictory results, e.g., discrepancies between IC50
values across studies may occur [1].
It is widely accepted that cancer cells are characterized by
higher levels of reactive oxygen species (ROS) compared with
non-cancerous cells due to oncogenic stimulation and increased
metabolic activities [42], which promotes their proliferation and
cell growth [43]. The role of ROS in cancer biology is rather complex,
because both cancer-suppressing and cancer-promoting actions of
ROS have been documented, which depends on ROS levels [43].
In general, cancer cells upregulate glycolysis even when oxygen
is abundant, a phenomenon called the Warburg effect (or aero-
bic glycolysis) [44,45]. The upregulation of glycolysis may serve
for the synthesis of ATP, ribonucleotides and amino acids, but
also for reduced nicotinamide adenine dinucleotide phosphate
(NADPH) production [46], which can remove ROS [46]. Pyruvate
kinase M2 (PKM2), an essential regulator of aerobic glycolysis
in cancer cells, was also found a participant in a negative feed-
back loop controlled by cellular oxidative stress [46]. Oxidation of
PKM2 on Cys358 caused its inactivation, thereby inducing NAPDH
production by the pentose phosphate pathway, and increasing
cellular redox buffering capacity [46]. This strategy allows can-
cer cells to control a proper redox status and may be considered
a survival mechanism for cancer cells, especially under hypoxic
conditions [45,46]. Thus, a combination of targeting the Warburg
effect and the manipulations in cancer cell redox homeostasis may
have anticancer effects [45]. Indeed, redox-based cancer therapy
involving nutraceutical-mediated modulation in ROS levels and sig-
naling molecules/pathways associated with drug resistance seems
promising [43].
Because anticancer effects of capsaicin are accompanied by
oxidative stress [15,17,47], we were interested in determin-
ing whether cellular redox homeostasis is affected in A549 and
DU145 cells after capsaicin treatment. Indeed, after capsaicin
treatment, ROS level in the culture medium was increased,
and ROS and superoxide production was elevated in A549 and
DU145 cells. Nevertheless, capsaicin-mediated disequilibrium in
the intracellular redox state did not provoke the dissipation of
the mitochondrial inner transmembrane potential (m)-based
32 A. Lewinska et al. / Mutation Research 779 (2015) 23–34
apoptotic cell death in cancer cells, which is inconsistent with avail-
able data on capsaicin-induced ROS-based apoptosis. Capsaicin
was reported to inhibit cancer cell-specific isoform of the outside
cell surface-localized NADH oxidase (ENOX2), which may result in
ROS elevation triggering mitochondrial-dependent apoptosis [6].
Capsaicin-induced ROS may also inhibit antioxidant protein thiore-
doxin, a negative regulator of apoptosis signal-regulating kinase
1 (ASK1), leading to activation of the ASK1 cascade and apopto-
sis in pancreatic cancer cells [48]. Moreover, capsaicin may affect
the activities of mitochondrial complexes: I and III, which may
cause ROS accumulation, ATP level depletion and downregulation
of antioxidant enzymes, which, in turn, may contribute to apoptosis
in pancreatic cancer cells [15].
In contrast to limited cytotoxicity, capsaicin was found an
inducer of genotoxic events in A549 and DU145 cells. Capsaicin
promoted DNA double strand breaks (DSBs) and micronuclei for-
mation. It has been previously reported that capsaicin may induce
DNA damage both in nonmalignant cells and malignant cells
[27,30]. Capsaicin caused sister chromatid exchanges and micronu-
clei production in human lymphocytes [30] and stimulated DNA
strand breaks in human neuroblastoma cells SHSY-5Y and MCF-7
breast cancer cells [27,49]. However, genotoxic potential of cap-
saicin has been also questioned [29]. Using four genotoxicity assays,
namely, the Ames test, mouse lymphoma cell mutation test, mouse
in vivo bone marrow micronucleus test and chromosomal aberra-
tion in human peripheral blood lymphocytes (HPBL) assay, weak
genotoxic activity of capsaicin has been observed [29]. The authors
suggested that inadequate purity of capsaicin and the use of pepper
plant extracts instead of pure, analytical standard grade capsaicin
may yield false positive results on capsaicin genotoxic potential
[29]. Nevertheless, we used high purity capsaicin, so genotoxic
activity of capsaicin against A549 and DU145 cells was not due to
impurities.
When DSBs occur, a complex cellular response is induced to
promote DNA repair and maintain genome integrity [50]. During
cellular response to DNA damage, p53 binding protein (53BP1)-
dependent pathway is activated: 53BP1 is recruited to sites of
DNA damage due to methylation state-specific recognition of his-
tone H4-K20 by 53BP1 [51]. The biological role of 53BP1 is far
from being understood. Nevertheless, 53BP1 has been shown to be
involved in the regulation of activation of the G2/M phase check-
point [52,53] and the intra-S phase checkpoint [54], and repair
of DNA DSBs via non-homologous end-joining (NHEJ) [55].We
have monitored 53BP1 foci during capsaicin-mediated genotoxicity
and found a negative correlation between micronuclei production
and 53BP1 foci, especially in DU145 cells. A link between recruit-
ment of 53BP1 and resolution of DNA damage has been already
reported [56]. In 53BP1-depleted WI38 human fibroblasts sub-
jected to DNA damage-promoting conditions, the percentage of
micronuclei-positive cells was increased [56]. Moreover, a more
robust activation or upregulation of 53BP1 in response to DNA
damage resulted in lower level of chromosomal damage [56]. Thus,
53BP1 was suggested to contribute to genomic stability. Perhaps,
a more potent recruitment of 53BP1 may account for diminished
MN generation after DU145 cell treatment with capsaicin at higher
concentrations.
The introduction of DNA damage in a cancer cell by geno-
toxic agents/chemicals would be a beneficial approach if DNA
damage-associated cell death would occur. However, unresolved
DNA damage may also promote gene mutation and/or genomic
instability, which, in turn, may affect cell susceptibility to cell
cycle checkpoint control and apoptosis. Thus, nutraceutical-based
genotoxic agents with the ability to eliminate rapidly cancer
cells without causing potentially detrimental genomic instabili-
ties and/or mutagenic side effects should be screened. Perhaps,
capsaicin is not a promising candidate for anticancer therapy,
because capsaicin-induced genotoxicity is accompanied by lim-
ited cytotoxicity in A549 and DU145 cells. Capsaicin-mediated
genomic instability may be a part of an adaptive response of can-
cer cells that allow them to survive after capsaicin treatment. More
recently, capsaicin-induced autophagy was found to protect human
breast cancer cells against apoptosis [25]. Autophagy is a lysosome-
dependent and energy supplying degradation process that is a part
of cellular stress response promoting cell survival [57]. Capsaicin-
induced DNA damage provoked autophagy through AMPK–mTOR
signaling pathway, which, in turn, regulated activation of ATM,
DNA–PKcs and PARP-1, and prolonged breast cancer cell survival
[49]. Capsaicin-mediated induction of DNA repair signaling and
autophagy may interrupt the treatment of human breast cancer
[49]. However, autophagy genes may be considered another target
of anticancer therapy.
Epigenetic changes in cancer cells may involve global DNA
hypomethylation resulting in the expression of quiescent proto-
oncogenes and prometastatic genes and promoting tumor
progression, and promoter-localized hypermethylation causing
transcriptional silencing of tumor suppressor genes and affecting
the control of tumorigenesis [58]. Nutraceuticals may modulate
cancer cell epigenome, e.g., dietary phytochemicals have been
shown to affect DNA methylation, histone modifications and
miRNA expression, which may promote cancer cell chemosensiti-
zation, being a particularly attractive anticancer strategy [59,60].
Because data on capsaicin-mediated epigenetic changes in can-
cer cells are lacking, we decided to evaluate capsaicin-associated
global DNA methylation. The level of 5-methyl-2-deoxycytidine
(5mdC) was unaffected in DU145 cells after capsaicin treatment,
whereas capsaicin was found to be a potent DNA hypermethy-
lating agent in A549 cells. Thus, capsaicin epigenetic action may
be considered cancer cell-specific and at least in A549 cells cap-
saicin may promote DNA hypermethylation-mediated changes in
gene expression. More recently, capsaicin has been documented a
potent modulator of microRNAs in cancer cells, which may affect
gene expression profiles [61,62]. Capsaicin restored the p53/miR-
34a regulatory axis and decreased survival of non-small cell lung
carcinoma cells [61]. Capsaicin also downregulated miR-520a-5p in
human chronic myeloid leukemia cells (K562 cells), which resulted
in cell growth retardation and apoptosis via targeting STAT3 [62].
In conclusion, we showed that capsaicin may induce DNA and
chromosomal damage in A549 and DU145 cancer cells, which may
contribute to limited susceptibility of these cells to apoptotic cell
death and may challenge the use of capsaicin in anticancer thera-
pies, at least in lung and prostate cancer treatment.
Conflict of interest
The authors declare that there are no conflicts of interest.
Acknowledgments
This study was supported by Grant from the National Science
Center, 2013/11/D/NZ7/00939. Experiments using flow cytometry
were performed in the Laboratory of Cytometry at the Nencki Insti-
tute of Experimental Biology.
References
[1] K. Bley, G. Boorman, B. Mohammad, D. McKenzie, S. Babbar, A comprehensive
review of the carcinogenic and anticarcinogenic potential of capsaicin,
Toxicol. Pathol. 40 (2012) 847–873.
[2] S. Marabini, P.G. Ciabatti, G. Polli, B.M. Fusco, P. Geppetti, Beneficial effects of
intranasal applications of capsaicin in patients with vasomotor rhinitis, Eur.
Arch. Otorhinolaryngol. 248 (1991) 191–194.
A. Lewinska et al. / Mutation Research 779 (2015) 23–34 33
[3] M. Matucci Cerinic, G. McCarthy, A. Lombardi, A. Pignone, G. Partsch,
Neurogenic influences in arthritis: potential modification by capsaicin, J.
Rheumatol. 22 (1995) 1447–1449.
[4] F. Sicuteri, B.M. Fusco, S. Marabini, V. Campagnolo, C.A. Maggi, P. Geppetti, M.
Fanciullacci, Beneficial effect of capsaicin application to the nasal mucosa in
cluster headache, Clin. J. Pain 5 (1989) 49–53.
[5] C.P. Watson, R.J. Evans, V.R. Watt, Post-herpetic neuralgia and topical
capsaicin, Pain 33 (1988) 333–340.
[6] D.J. Morre, P.J. Chueh, D.M. Morre, Capsaicin inhibits preferentially the NADH
oxidase and growth of transformed cells in culture, Proc. Nat. Acad. Sci. U. S.
A. 92 (1995) 1831–1835.
[7] K. Ito, T. Nakazato, K. Yamato, Y. Miyakawa, T. Yamada, N. Hozumi, K. Segawa,
Y. Ikeda, M. Kizaki, Induction of apoptosis in leukemic cells by homovanillic
acid derivative, capsaicin, through oxidative stress: implication of
phosphorylation of p53 at Ser-15 residue by reactive oxygen species, Cancer
Res. 64 (2004) 1071–1078.
[8] M. Bhutani, A.K. Pathak, A.S. Nair, A.B. Kunnumakkara, S. Guha, G. Sethi, B.B.
Aggarwal, Capsaicin is a novel blocker of constitutive and
interleukin-6-inducible STAT3 activation, Clin. Cancer Res. 13 (2007)
3024–3032.
[9] N. Hail Jr., R. Lotan, Examining the role of mitochondrial respiration in
vanilloid-induced apoptosis, J. Natl. Cancer Inst. 94 (2002) 1281–1292.
[10] C. Amantini, M. Mosca, M. Nabissi, R. Lucciarini, S. Caprodossi, A. Arcella, F.
Giangaspero, G. Santoni, Capsaicin-induced apoptosis of glioma cells is
mediated by TRPV1 vanilloid receptor and requires p38 MAPK activation, J.
Neurochem. 102 (2007) 977–990.
[11] S.W. Ip, S.H. Lan, A.C. Huang, J.S. Yang, Y.Y. Chen, H.Y. Huang, Z.P. Lin, Y.M.
Hsu, M.D. Yang, C.F. Chiu, J.G. Chung, Capsaicin induces apoptosis in SCC-4
human tongue cancer cells through mitochondria-dependent and
-independent pathways, Environ. Toxicol. 27 (2012) 332–341.
[12] S.W. Ip, S.H. Lan, H.F. Lu, A.C. Huang, J.S. Yang, J.P. Lin, H.Y. Huang, J.C. Lien, C.C.
Ho, C.F. Chiu, W. Wood, J.G. Chung, Capsaicin mediates apoptosis in human
nasopharyngeal carcinoma NPC-TW 039 cells through mitochondrial
depolarization and endoplasmic reticulum stress, Hum. Exp. Toxicol. 31
(2012) 539–549.
[13] C.C. Wu, J.P. Lin, J.S. Yang, S.T. Chou, S.C. Chen, Y.T. Lin, H.L. Lin, J.G. Chung,
Capsaicin induced cell cycle arrest and apoptosis in human esophagus
epidermoid carcinoma CE 81T/VGH cells through the elevation of intracellular
reactive oxygen species and Ca2+ productions and caspase-3 activation,
Mutat. Res. 601 (2006) 71–82.
[14] J.D. Kim, J.M. Kim, J.O. Pyo, S.Y. Kim, B.S. Kim, R. Yu, I.S. Han, Capsaicin can
alter the expression of tumor forming-related genes which might be followed
by induction of apoptosis of a Korean stomach cancer cell line, SNU-1, Cancer
Lett. 120 (1997) 235–241.
[15] K.C. Pramanik, S.R. Boreddy, S.K. Srivastava, Role of mitochondrial electron
transport chain complexes in capsaicin mediated oxidative stress leading to
apoptosis in pancreatic cancer cells, PLoS One 6 (2011) e20151.
[16] S.P. Huang, J.C. Chen, C.C. Wu, C.T. Chen, N.Y. Tang, Y.T. Ho, C. Lo, J.P. Lin, J.G.
Chung, J.G. Lin, Capsaicin-induced apoptosis in human hepatoma HepG2 cells,
Anticancer Res. 29 (2009) 165–174.
[17] H.F. Lu, Y.L. Chen, J.S. Yang, Y.Y. Yang, J.Y. Liu, S.C. Hsu, K.C. Lai, J.G. Chung,
Antitumor activity of capsaicin on human colon cancer cells in vitro and colo
205 tumor xenografts in vivo, J. Agric. Food Chem. 58 (2010) 12999–13005.
[18] K.C. Brown, T.R. Witte, W.E. Hardman, H. Luo, Y.C. Chen, A.B. Carpenter, J.K.
Lau, P. Dasgupta, Capsaicin displays anti-proliferative activity against human
small cell lung cancer in cell culture and nude mice models via the E2F
pathway, PLoS One 5 (2010) e10243.
[19] N.H. Thoennissen, J. O’Kelly, D. Lu, G.B. Iwanski, D.T. La, S. Abbassi, A. Leiter, B.
Karlan, R. Mehta, H.P. Koeffler, Capsaicin causes cell-cycle arrest and
apoptosis in ER-positive and -negative breast cancer cells by modulating the
EGFR/HER-2 pathway, Oncogene 29 (2010) 285–296.
[20] A. Mori, S. Lehmann, J. O’Kelly, T. Kumagai, J.C. Desmond, M. Pervan, W.H.
McBride, M. Kizaki, H.P. Koeffler, Capsaicin a component of red peppers,
inhibits the growth of androgen-independent, p53 mutant prostate cancer
cells, Cancer Res. 66 (2006) 3222–3229.
[21] H.J. Kang, Y. Soh, M.S. Kim, E.J. Lee, Y.J. Surh, H.R. Kim, S.H. Kim, A. Moon, Roles
of JNK-1 and p38 in selective induction of apoptosis by capsaicin in
ras-transformed human breast epithelial cells, Int. J. Cancer 103 (2003)
475–482.
[22] Y.S. Lee, Y.S. Kang, J.S. Lee, S. Nicolova, J.A. Kim, Involvement of NADPH
oxidase-mediated generation of reactive oxygen species in the apototic cell
death by capsaicin in HepG2 human hepatoma cells, Free Radical Res. 38
(2004) 405–412.
[23] J.K. Min, K.Y. Han, E.C. Kim, Y.M. Kim, S.W. Lee, O.H. Kim, K.W. Kim, Y.S. Gho,
Y.G. Kwon, Capsaicin inhibits in vitro and in vivo angiogenesis, Cancer Res. 64
(2004) 644–651.
[24] Y.J. Surh, More than spice: capsaicin in hot chili peppers makes tumor cells
commit suicide, J. Natl. Cancer Inst. 94 (2002) 1263–1265.
[25] C.H. Choi, Y.K. Jung, S.H. Oh, Autophagy induction by capsaicin in malignant
human breast cells is modulated by p38 and extracellular signal-regulated
mitogen-activated protein kinases and retards cell death by suppressing
endoplasmic reticulum stress-mediated apoptosis, Mol. Pharmacol. 78 (2010)
114–125.
[26] Y.J. Surh, S.S. Lee, Capsaicin in hot chili pepper: carcinogen co-carcinogen or
anticarcinogen? Food Chem. Toxicol. 34 (1996) 313–316.
[27] F. Richeux, M. Cascante, R. Ennamany, D. Saboureau, E.E. Creppy, Cytotoxicity
and genotoxicity of capsaicin in human neuroblastoma cells SHSY-5Y, Arch.
Toxicol. 73 (1999) 403–409.
[28] F. Richeux, M. Cascante, R. Ennamany, D. Sanchez, A. Sanni, D. Saboureau, E.E.
Creppy, Implications of oxidative stress and inflammatory process in the
cytotoxicity of capsaicin in human endothelial cells: lack of DNA strand
breakage, Toxicology 147 (2000) 41–49.
[29] S. Chanda, G. Erexson, C. Riach, D. Innes, F. Stevenson, H. Murli, K. Bley,
Genotoxicity studies with pure trans-capsaicin, Mutat. Res. 557 (2004) 85–97.
[30] S. Marques, N.G. Oliveira, T. Chaveca, J. Rueff, Micronuclei and sister
chromatid exchanges induced by capsaicin in human lymphocytes, Mutat.
Res. 517 (2002) 39–46.
[31] T. Mosmann, Rapid colorimetric assay for cellular growth and survival:
application to proliferation and cytotoxicity assays, J. Immunol. Methods 65
(1983) 55–63.
[32] E. Bedner, P. Smolewski, P. Amstad, Z. Darzynkiewicz, Activation of caspases
measured in situ by binding of fluorochrome-labeled inhibitors of caspases
(FLICA): correlation with DNA fragmentation, Exp. Cell Res. 259 (2000)
308–313.
[33] A. Lewinska, P. Chochrek, K. Smolag, E. Rawska, M. Wnuk, Oxidant-based
anticancer activity of a novel synthetic analogue of capsaicin, capsaicin
epoxide, Redox Rep. (2014),
http://dx.doi.org/10.1179/1351000214Y.0000000113.
[34] Korwek, T. Sewastianik, A. Bielak-Zmijewska, G. Mosieniak, O. Alster, M.
Moreno-Villanueva, A. Burkle, E. Sikora, Inhibition of ATM blocks the
etoposide-induced DNA damage response and apoptosis of resting human T
cells, DNA Repair (Amst) 11 (2012) 864–873.
[35] M. Fenech, The in vitro micronucleus technique, Mutat. Res. 455 (2000) 81–95.
[36] M. Fenech, W.P. Chang, M. Kirsch-Volders, N. Holland, S. Bonassi, E. Zeiger,
HUMN project: detailed description of the scoring criteria for the
cytokinesis-block micronucleus assay using isolated human lymphocyte
cultures, Mutat. Res. 534 (2003) 65–75.
[37] J. Mytych, A. Lewinska, A. Bielak-Zmijewska, W. Grabowska, J. Zebrowski, M.
Wnuk, Nanodiamond-mediated impairment of nucleolar activity is
accompanied by oxidative stress and DNMT2 upregulation in human cervical
carcinoma cells, Chem. Biol. Interact. 220 C (2014) 51–63.
[38] M. Berdasco, M.F. Fraga, M. Esteller, Quantification of global DNA methylation
by capillary electrophoresis and mass spectrometry, Methods Mol. Biol. 507
(2009) 23–34.
[39] A. Ciccia, S.J. Elledge, The DNA damage response: making it safe to play with
knives, Mol. Cell 40 (2010) 179–204.
[40] C.H. Choi, Y.K. Jung, S.H. Oh, Selective induction of catalase-mediated
autophagy by dihydrocapsaicin in lung cell lines, Free Radical Biol. Med. 49
(2010) 245–257.
[41] C.A. Reilly, J.L. Taylor, D.L. Lanza, B.A. Carr, D.J. Crouch, G.S. Yost, Capsaicinoids
cause inflammation and epithelial cell death through activation of vanilloid
receptors, Toxicol. Sci. 73 (2003) 170–181.
[42] T. Fiaschi, P. Chiarugi, Oxidative stress, tumor microenvironment, and
metabolic reprogramming: a diabolic liaison, Int. J. Cell Biol. 2012 (2012)
762825.
[43] S.C. Gupta, D. Hevia, S. Patchva, B. Park, W. Koh, B.B. Aggarwal, Upsides and
downsides of reactive oxygen species for cancer: the roles of reactive oxygen
species in tumorigenesis, prevention, and therapy, Antioxid. Redox Signaling
16 (2012) 1295–1322.
[44] O. Warburg, The chemical constitution of respiration ferment, Science 68
(1928) 437–443.
[45] X. Chen, Y. Qian, S. Wu, The Warburg effect: evolving interpretations of an
established concept, Radical Biol. Med. (2014),
http://dx.doi.org/10.1016/j.freeradbiomed.2014.08.027.
[46] D. Anastasiou, G. Poulogiannis, J.M. Asara, M.B. Boxer, J.K. Jiang, M. Shen, G.
Bellinger, A.T. Sasaki, J.W. Locasale, D.S. Auld, C.J. Thomas, M.G. Vander
Heiden, L.C. Cantley, Inhibition of pyruvate kinase M2 by reactive oxygen
species contributes to cellular antioxidant responses, Science 334 (2011)
1278–1283.
[47] A.M. Sanchez, M.G. Sanchez, S. Malagarie-Cazenave, N. Olea, I. Diaz-Laviada,
Induction of apoptosis in prostate tumor PC-3 cells and inhibition of xenograft
prostate tumor growth by the vanilloid capsaicin, Apoptosis 11 (2006) 89–99.
[48] K.C. Pramanik, S.K. Srivastava, Apoptosis signal-regulating kinase
1-thioredoxin complex dissociation by capsaicin causes pancreatic tumor
growth suppression by inducing apoptosis, Antioxid. Redox Signaling 17
(2012) 1417–1432.
[49] J.H. Yoon, S.G. Ahn, B.H. Lee, S.H. Jung, S.H. Oh, Role of autophagy in
chemoresistance: regulation of the ATM-mediated DNA-damage signaling
pathway through activation of DNA-PKcs and PARP-1, Biochem. Pharmacol.
83 (2012) 747–757.
[50] G.S. Stewart, Solving the RIDDLE of 53BP1 recruitment to sites of damage, Cell
Cycle 8 (2009) 1532–1538.
[51] M.V. Botuyan, J. Lee, I.M. Ward, J.E. Kim, J.R. Thompson, J. Chen, G. Mer,
Structural basis for the methylation state-specific recognition of histone
H4-K20 by 53BP1 and Crb2 in DNA repair, Cell 127 (2006) 1361–1373.
[52] R.A. DiTullio Jr., T.A. Mochan, M. Venere, J. Bartkova, M. Sehested, J. Bartek, T.D.
Halazonetis, 53BP1 functions in an ATM-dependent checkpoint pathway that
is constitutively activated in human cancer, Nat. Cell Biol. 4 (2002) 998–1002.
[53] B. Wang, S. Matsuoka, P.B. Carpenter, S.J. Elledge, 53BP1, a mediator of the
DNA damage checkpoint, Science 298 (2002) 1435–1438.
34 A. Lewinska et al. / Mutation Research 779 (2015) 23–34
[54] J. Silverman, H. Takai, S.B. Buonomo, F. Eisenhaber, T. de Lange, Human Rif1
ortholog of a yeast telomeric protein, is regulated by ATM and 53BP1 and
functions in the S-phase checkpoint, Genes Dev. 18 (2004) 2108–2119.
[55] S. Difilippantonio, E. Gapud, N. Wong, C.Y. Huang, G. Mahowald, H.T. Chen,
M.J. Kruhlak, E. Callen, F. Livak, M.C. Nussenzweig, B.P. Sleckman, A.
Nussenzweig, 53BP1 facilitates long-range DNA end-joining during V(D)J
recombination, Nature 456 (2008) 529–533.
[56] L.S. Fink, M. Roell, E. Caiazza, C. Lerner, T. Stamato, S. Hrelia, A. Lorenzini, C.
Sell, 53BP1 contributes to a robust genomic stability in human fibroblasts,
Aging (Albany NY) 3 (2011) 836–845.
[57] T. Yorimitsu, D.J. Klionsky, Autophagy: molecular machinery for self-eating,
Cell Death. Differ. 12 (Suppl 2) (2005) 1542–1552.
[58] P.M. Das, R. Singal, DNA methylation and cancer, J. Clin. Oncol. 22 (2004)
4632–4642.
[59] S. Reuter, S.C. Gupta, B. Park, A. Goel, B.B. Aggarwal, Epigenetic changes
induced by curcumin and other natural compounds, Genes Nutr. 6 (2011)
93–108.
[60] S. Fu, R. Kurzrock, Development of curcumin as an epigenetic agent, Cancer
116 (2010) 4670–4676.
[61] S. Chakraborty, M. Mazumdar, S. Mukherjee, P. Bhattacharjee, A. Adhikary, A.
Manna, P. Khan, A. Sen, T. Das, Restoration of p53/miR-34a regulatory axis
decreases survival advantage and ensures Bax-dependent apoptosis of
non-small cell lung carcinoma cells, FEBS Lett. 588 (2014)
549–559.
[62] B.T. Kaymaz, V.B. Cetintas, C. Aktan, B. Kosova, MicroRNA-520a-5p displays a
therapeutic effect upon chronic myelogenous leukemia cells by targeting
STAT3 and enhances the anticarcinogenic role of capsaicin, Tumour Biol. 35
(2014) 8733–8742.
... The pungency of chili peppers (genus Capsicum) is formed from a mixture of substances called capsaicinoids, of which the most abundant is capsaicin (8-methyl-N-vanylyl-6-nonenamide), followed by dihydrocapsaicin, nordihydrocapsaicin, homocapsaicin, and homodihydrocapsaicin (Zhigila et al., 2014;Lewinska et al., 2015). Capsaicin (CAP), besides giving a spicy flavor to chili peppers, also has a high therapeutic potential (Freitas et al., 2018), with anti-inflammatory, antioxidant, anti-obesity, analgesic, antiparasitic, immunomodulatory, and anti-tumor properties, in addition to being efficient in the treatment of arthritis-related pain, cystitis, and diabetic neuropathy (Chapa-Oliver and Mejía-Teniente, 2016;Bogusz Junior et al., 2018;Xiang et al., 2021). ...
... These cytotoxic effects related to capsaicin, as demonstrated by other authors, may have occurred through the production of reactive oxygen species (ROS). This would stop the cell cycle, regulating the expression of the transcription factor and changes in the transduction signal pathways, cell growth, and survival, resulting in decreased viability and consequent cell death (Lewinska et al., 2015;Gómez-Sierra et al., 2013). ...
Article
Full-text available
Capsaicin (CAP) is the main compound responsible for the spicy flavor of Capsicum plants. However, its application can be inhibited due to its pungency and toxicity. This study aimed to evaluate and compare the cytotoxic effect of CAP and its analogs N-benzylbutanamide (AN1), N-(3-methoxybenzyl) butanamide (AN2), N-(4-hydroxy-3-methoxybenzyl) butanamide (AN3), N-(4-hydroxy-3-methoxybenzyl) hexanamide (AN4) and N-(4-hydroxy-3-methoxybenzyl) tetradecanamide (AN5) on the hepatoma cells of Rattus norvegicus using the MTT test. The results showed cytotoxicity of CAP at concentrations of 100, 150, 175, and 200 μM (24 hours), AN1 at 150 and 175 μM (48 hours), AN2 at 50 μM (24 hours) and 10, 25, 50, and 75 μM (48 hours), AN4 at 175 μM (24 hours), and AN5 at 50 μM (48 hours). Removing the hydroxyl radical from the vanillyl group of capsaicin, together with reducing the acyl chain to 3 carbons, which is the case of AN2, resulted in the best biological activity. Increasing the carbon chain in the acyl group of the capsaicin molecule, which is the case of AN5, also showed evident cytotoxic effects. The present study proves that the chemical modifications of capsaicin changed its biological activity.
... Alterations in this protein disrupt the function of the NHEJ pathway and promote the utilization of error-prone alt-NHEJ, which can lead to genomic instability and tumorigenesis [125,126]. Studies have shown that mutations in TP53BP1 are present in PC and its expression decreases with cancer progression [127][128][129][130]. In addition, the study by Jaworski et al. indicates no correlation between TP53BP1 expression and GS or GP, while Gzil et al. observed decreased TP53BP1 expression in lymph node metastases compared to primary PC [54,55]. ...
Article
Full-text available
Prostate cancer remains a leading cause of cancer-related death in men worldwide. Recent research advances have emphasized the critical roles of mismatch repair (MMR) and double-strand break (DSB) in prostate cancer development and progression. Here, we provide a comprehensive review of the molecular mechanisms underlying DSB and MMR defects in prostate cancer, as well as their clinical implications. Furthermore, we discuss the promising therapeutic potential of immune checkpoint inhibitors and PARP inhibitors in targeting these defects, particularly in the context of personalized medicine and further perspectives. Recent clinical trials have demonstrated the efficacy of these novel treatments, including Food and Drugs Association (FDA) drug approvals, offering hope for improved patient outcomes. Overall, this review emphasizes the importance of understanding the interplay between MMR and DSB defects in prostate cancer to develop innovative and effective therapeutic strategies for patients.
... Cluster headaches, rheumatoid arthritis, post-mastectomy pain syndrome, diabetic neuropathy, and herpes zoster are just a few of the conditions that capsaicin has been used to treat [4]. Particularly lung cancer, have been the subject of in-depth research on the action of capsaicin, which may have anticancer and antiproliferative properties [3][4][5]. Most often, capsaicin-mediated cytotoxicity is linked to cancer cell apoptosis through the activation of a number of processes, such as the production of reactive oxygen species (ROS), the beginning of endoplasmic reticulum (ER) stress, and changes in protein kinases [6,7]. ...
Article
Capsaicin is the main component of many hot peppers, exerts anticancer effects on various cancer cells by inducing apoptotic pathways. In addition, the ineffectiveness of this apoptotic effect of capsaicin on healthy cells provides a wide-ranging use of capsaicin. In recent years, many studies have been carried out to determine the safety of capsaicin use in lung cancer, which is a common cancer type worldwide with high mortality rates. In the present study anticancer effects of capsaicin were studied on A549 human lung adenocarcinoma cell line. Also, citotoxicity of this compound was studied on L929 mouse embryonic fibroblast cell line. Various concentration of capsaicin (3.90625-500 μM) effectively decreased cell viability in A549 cell line in a dose-dependent manner. Also, this antiproliferative effect of capsaicin was selective compared to L929 cell line. On the other hand, apoptosis inducing effects of capsaicin were studied by Annexin V-FITC and Caspase 3 assays by using IC50 and IC50/2 concentrations on A549 cell by flow cytometric methods. Our results revealed that the anticancer effects induced by capsaicin on A549 cell line involved apoptosis by inducing Caspase 3 pathway.
... The general perception is that capsaicin's ability to either prevent cancer or encourage its growth depends on its concentration, how long it has been used, the sort of tumors it effects, and when it is consumed with food-the presence of other toxic substances [53,133]. The observation that capsaicin may affect cancer calls without damaging normal cells is of great value in cancer chemoprevention [134]. Based on the present survey and the findings in animal models it is essential to increase the efforts to investigate the anticancer potential of capsaicin in humans and to detect the appropriate administration dosage in order to add it in the list of the anti-cancer drugs. ...
Article
The pungent component of the chili pepper known as capsaicin is popular both as a spice and as a carcinopreventer. In vitro experiments and studies with animal models suggest that capsaicin may inhibit gastrointestinal, pancreatic, hepatic and other sorts of cancer development through a number of different pathways. Additionally, capsaicin may improve the therapeutic potential of traditional anti-cancer medications when taken with them. However, several reports have raised concerns that capsaicin may have a dual impact on cancer cells that promotes both cell proliferation and metastatic abilities. The purpose of this review is to examine the contradicting information regarding capsaicin’s ability to prevent cancer.
... The effects of autophagy induced by capsaicin in cancer cells are still controversial. For instance, Lewinska et al. reported that up to 250 μM capsaicin induces genotoxic stress but does not promotes apoptosis in A549 and DU145 cancer cells, and suggested that the higher concentrations of capsaicin might decrease metabolic activity to exert antiproliferative and cytostatic actions (Lewinska et al., 2015). In contrast, capsaicin-induced autophagy was reported to play important roles in the DNA damage responses of breast cancer cells, activating the ATM-DNA-PKcs-PARP-1 axis to protect cells against apoptosis (Yoon et al., 2012). ...
Article
Full-text available
The ability of capsaicin co-treatment to sensitize cancer cells to anticancer drugs has been widely documented, but the detailed underlying mechanisms remain unknown. In addition, the role of ribophorin II turnover on chemosensitization is still uncertain. Here, we investigated capsaicin-induced sensitization to chemotherapeutic agents in the human oral squamous carcinoma cell lines, HSC-3 and SAS. We found that capsaicin (200 μM) did not induce remarkable apoptotic cell death in these cell lines; instead, it significantly enhanced autophagy with a concomitant decrease of ribophorin II protein. This capsaicin-induced decrease in ribophorin II was intensified by the autophagy inducer, rapamycin, but attenuated by the autophagy inhibitors, ULK1 inhibitor and chloroquine, indicating that the autophagic process was responsible for the capsaicin-induced down-regulation of ribophorin II. Co-administration of capsaicin with conventional anticancer agents did, indeed, sensitize the cancer cells to these agents. In co-treated cells, the induction of apoptosis was significantly reduced and the levels of the necroptosis markers, phospho-MLKL and phospho-RIP3, were increased relative to the levels seen in capsaicin treatment alone. The levels of DNA damage response markers were also diminished by co-treatment. Collectively, our results reveal a novel mechanism by which capsaicin sensitizes oral cancer cells to anticancer drugs through the up-regulation of autophagy and down-regulation of ribophorin II, and further indicate that the induction of necroptosis is a critical factor in the capsaicin-mediated chemosensitization of oral squamous carcinoma cells to conventional anticancer drugs.
... In healthy cells, phosphatidylserine (PS) was only distributed in the inner leaflet of the lipid bilayer; however, in the early stage of apoptosis, PS on the cell membrane traversed from the inner leaflet to the outer leaflet. Annexin V can bind to PS and is used as an indicator of early apoptosis [48]. The 7-aminoactinomycin D (7-AAD) can bind to DNA and is a fluorescent probe used to detect late apoptotic and dead cells [49]. ...
Article
Full-text available
Methods: Hypoxia in hBMSCs was induced for 0, 4, and 12 hours, and cellular senescence was evaluated by senescence-associated β-galactosidase (SA-β-gal) staining. Tandem mass tag (TMT) labeling was combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) for differential proteomic analysis of hypoxia in hBMSCs. Parallel reaction monitoring (PRM) analysis was used to validate the candidate proteins. Verifications of signaling pathways were evaluated by western blotting. Cell apoptosis was evaluated using Annexin V/7-AAD staining by flow cytometry. The production of reactive oxygen species (ROS) was detected by the fluorescent probe 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA). Results: Cell senescence detected by SA-β-gal activity was higher in the 12-hour hypoxia-induced group. TMT analysis of 12-hour hypoxia-induced cells identified over 6000 proteins, including 686 differentially expressed proteins. Based on biological pathway analysis, we found that the senescence-associated proteins were predominantly enriched in the cancer pathways, PI3K-Akt pathway, and cellular senescence signaling pathways. CDK1, CDK2, and CCND1 were important nodes in PPI analyses. Moreover, the CCND1, UQCRH, and COX7C expressions were verified by PRM. Hypoxia induction for 12 hours in hBMSCs reduced CCND1 expression but promoted ROS production and cell apoptosis. Such effects were markedly reduced by the PI3K agonist, 740 Y-P, and attenuated by LY294002. Conclusions: Hypoxia of hBMSCs inhibited CCND1 expression but promoted ROS production and cell apoptosis through activating the PI3K-dependent signaling pathway. These findings provided a detailed characterization of the proteomic profiles related to hypoxia-induced senescence of hBMSCs and facilitated our understanding of the molecular mechanisms leading to stem cell senescence.
... 163 Bunun yanı sıra kapsaisin tarafından indüklenen otofajinin endoplazmik retikulum stresi aracılı apoptozun baskılanması sonucu ortaya çıktığı ve bazı kanser türlerinde kanserli hücrelerin ölümünü baskıladığı da belirtilmiştir. 164 Kapsaisinin kemokoruyucu potansiyelinin yanı sıra antioksidan ve antiinflamatuar özelliklere sahip olması, hipokolesteromik etkisi sayesinde kardiyo koruyucu etki göstermesi, insan sağlığına olan olumlu etkileri açısından beslenmede ayrıcalıklı bir yere koymaktadır. 163 ...
Article
Hücrelerin kontrolsüz ve anormal bir şekilde çoğalması ile karakterize olan kanser, gelişen ve gelişmekte olan ülkelerde mortalitesi ve morbiditesi en yüksek hastalıktır. Kanserin farklı evrelerinde tedavi amacıyla kemoterapi, radyoterapi ve cerrahi yöntemler kullanılmaktadır. İlaveten prevantif olarak yani hastalıktan korunma amacıyla alternatif tamamlayıcı yöntemlere ve yaşam şartlarında değişikliklere de başvurulmaktadır. Bu bağlamda beslenme tarzı ve kanser oluşum riski arasındaki olası ilişki göz önünde bulundurulunca, kanserden korunmada beslenme tarzının önemi üzerine odaklanan araştırmalar da hız kazanmıştır. Bu derlemenin öncelikli amacı dünya üzerinde sıklıkla kullanılan besinlerin aktif bileşenlerini temel alarak etkiledikleri moleküler yolakları açıklamaktır. Yapılan multidisipliner bilimsel araştırmalar göstermiştir ki bu etkileşimler ancak genomik, proteomik ve metabolomik çalışmaların entegre edilmesi ile sağlıklı bir şekilde açıklanmaktadır. “Foodomik” olarak adlandırılan yaklaşım ile çoklu omik çalışmalar kullanılarak moleküllerin insan sağlığı üzerindeki etkisi ileri analitik yöntemlerle aydınlatılmaktadır. Bu derleme kapsamında dünya nüfusunun büyük bir kısmının beslenme alışkanlıklarını teşkil eden dört büyük mutfak ve vazgeçilmez unsurları sunulmuştur. Akdeniz mutfağı ve önemli unsurlarından zeytinyağı, domates, sarımsak, biberiye, adaçayı; Hint mutfağı ve önemli unsurlarından zerdeçal, sumak, kimyon, zencefil, safran; Çin mutfağı ve önemli unsurlarından soya fasulyesi, pirinç, brokoli; Meksika mutfağı ve önemli unsuru olan Şili biberi incelenmiştir. İlgili aktif bileşenlerin kanserden korunmada veya kanseri engellemede etkin olduğu metabolik yolaklar üzerine literatür taraması yapılmıştır. Ayrıca, bölgelere göre gözlemlenen kanser türlerinin de genel bir değerlendirmesi gerçekleştirilerek literatür taraması sonucu elde edilen bulgularla ilişkilendirilmiştir. Bu derlemenin; klinisyenler, diyetisyenler ve eczacılar için yararlı bir özet kaynak olması amaçlanmıştır.
Chapter
Humans are subjected to an array of external and internal stimuli during their lives, which injure genetic material and generate somatic mutations that lead to accumulating genomic stress. Several human pathologies like chronic degenerative diseases, including hepatic, neurodegenerative, and cardiovascular disorders; diabetes; arthritis; cancer; chronic inflammation; and aging, are associated with genotoxic stress. In the past few decades, a great deal of research has been done to find substances that can shield humans from DNA damage and its impact. Natural substances continue to show a beneficial impact on genome integrity. Scientists have identified and reviewed unique bioactive plant-derived compounds, such as flavonoids, phenolic compounds, and carotenoids, that can mitigate the impact of genotoxic stress; however, the genoprotective potential of alkaloids is not being reviewed so far. Herbs high in alkaloidal content have a plethora of pharmacological traits, which are utilized as drugs, and are significant to the pharmaceutical sector. This chapter summarizes the literature on the genoprotective/antigenotoxic action of alkaloids acting against endogenously and exogenously produced genetic damage. The current knowledge regarding the function of alkaloids to guard the genome can be exploited to create dietary plans and natural health products that are beneficial in the sequel of chemotherapeutic-associated toxicities.
Article
TRDMT1 methyltransferase is postulated to be a novel target in anticancer therapy as TRDMT1-mediated RNA methylation is involved in DNA damage response (DDR) and TRDMT1 deficient cells are sensitive to PARP1 inhibitors. However, the effects of TRDMT1 gene knockout (KO) during cancer cell selection upon drug stimulation and the involvement of exogenous RNA were not addressed. In the present study, osteosarcoma (OS) cells lacking active TRDMT1 gene were subjected to short-term treatment of etoposide in the presence of exogenous RNA and long-term effects were analyzed after drug removal. Changes in cell proliferation and cell viability, genetic stability and DDR, telomere length and shelterin complex, retrotransposon activity and the levels of selected pro-inflammatory cytokines were considered. Long-term selection of TRDMT1 KO OS cells resulted in modified DDR, changes in telomere length and increased retrotransposon activity that was modulated by the addition of exogenous RNA. Thus, TRDMT1 gene KO may promote cellular and genetic heterogeneity that may modulate cancer cell responses to chemotherapeutic drugs.
Article
Full-text available
Background: The vanilloids capsaicin and resiniferatoxin are natural products that contain a vanillyl moiety (4-hydroxy-3-methoxybenzyl). Both vanilloids can induce apoptosis in certain cell types by a mechanism that has not been fully elucidated but may involve plasma membrane or mitochondrial targets. We investigated the role of mitochondrial respiration in vanilloid-induced apoptosis. Methods: Cytofluorometric analysis was used to evaluate the effects of vanilloids on apoptosis, Ca 2+ mobilization, hydroperoxide generation, and DNA content in cells from two human cutaneous squamous cell carcinoma (SCC) cell lines (parental cells) and in their respiration-deficient clones. Oxygen consumption by the cells was determined polarographically. Results: The majority of the parental SCC cells underwent apoptosis after a 12-hour exposure to 100 μM capsaicin or 10 μM resiniferatoxin. The induction of apoptosis was associated with the mitochondrial permeability transition (i.e., an increase in the permeability of the inner mitochondrial membrane associated with the opening of a nonspecific pore). Exposure of parental cells to either vanilloid was not associated with an increase in intracellular free Ca 2+ levels but was associated with a rapid increase in hydroperoxide generation and a decrease in oxygen consumption. After vanilloid treatment, the respiration-deficient clones generated less hydroperoxide and were resistant to the mitochondrial permeability transition and the induction of apoptosis. Moreover, vanilloid treatment inhibited cell proliferation in the respiration-deficient clones by promoting G 1 arrest. Conclusions: Vanilloid-induced apoptosis in the parental SCC cells appears to involve the inhibition of mitochondrial respiration. The apoptogenic effects promoted by vanilloid treatment in parental SCC cells, as well as the antiproliferative effects observed in their respiration-deficient clones, suggest that vanilloids may be useful for preventing or treating skin cancers or other hyperproliferative skin disorders.
Article
Full-text available
Objectives: Plant-derived natural substances, such as capsaicin, with potent antiproliferative activity against cancer cells in vitro are considered to be promising nutraceuticals in anticancer therapy. Nevertheless, the limited systemic bioavailability of phytochemicals may raise questions regarding the physiological relevance of their phytochemical effects in vivo. Thus, the search for novel phytochemical-based substances with more efficient anticancer action is needed. Methods: In the present study, a capsaicin analogue, namely, capsaicin epoxide, was synthesized, and its cytotoxic potential against cancer cells was evaluated and compared to that of capsaicin through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and multi-caspase assays. The abilities of capsaicin and capsaicin epoxide to induce oxidative stress were estimated using redox-sensitive fluorogenic probes: 2',7'-dichlorodihydrofluorescein diacetate (H2DCF-DA) and dihydroethidium. Results: The structure and purity of the synthesized product were confirmed by nuclear magnetic resonance spectroscopy, electrospray ionization mass spectrometry, and gas chromatography. Normal human dermal fibroblasts were not susceptible to treatment with the agent, whereas a cancer cell type-specific response was observed. Human breast carcinoma cells were found to be the most sensitive to capsaicin epoxide treatment compared with capsaicin treatment, and the action of capsaicin epoxide was oxidant based. Discussion: Our data indicate that the antiproliferative activity of capsaicin epoxide is potentiated in vitro, when used at much lower concentrations compared with capsaicin at similar concentrations. Thus, the findings of this study may have implications for phytochemical-based anticancer drug development.
Article
Background: The vanilloids capsaicin and resiniferatoxin are natural products that contain a vanillyl moiety (4-hydroxy3-methoxybenzyl). Both vanilloids can induce apoptosis in certain cell types by a mechanism that has not been fully elucidated but may involve plasma membrane or mitochondrial targets. We investigated the role of mitochondrial respiration in vanilloid-induced apoptosis. Methods: Cytofluorometric analysis was used to evaluate the effects of vanilloids on apoptosis, Ca2+ mobilization, hydroperoxide generation, and DNA content in cells from two human cutaneous squamous cell carcinoma (SCC) cell lines (parental cells) and in their respiration-deficient clones. Oxygen consumption by the cells was determined polarographically. Results: The majority of the parental SCC cells underwent apoptosis after a 12-hour exposure to 100 muM capsaicin or 10 muM resiniferatoxin. The induction of apoptosis was associated with the mitochondrial permeability transition (i.e., an increase in the permeability of the inner mitochondrial membrane associated with the opening of a nonspecific pore). Exposure of parental cells to either vanilloid was not associated with an increase in intracellular free Ca2+ levels but was associated with a rapid increase in hydroperoxide generation and a decrease in oxygen consumption. After vanilloid treatment, the respiration-deficient clones generated less hydroperoxide and were resistant to the mitochondrial permeability transition and the induction of apoptosis. Moreover, vanilloid treatment inhibited cell proliferation in the respiration-deficient clones by promoting G(1) arrest. Conclusions: Vanilloid-induced apoptosis in the parental SCC cells appears to involve the inhibition of mitochondrial respiration. The apoptogenic effects promoted by vanilloid treatment in parental SCC cells, as well as the antiproliferative effects observed in their respiration-deficient clones, suggest that vanilloids may be useful for preventing or treating skin cancers or other hyperproliferative skin disorders.
Article
The study of DNA damage at the chromosome level is an essential part of genetic toxicology because chromosomal mutation is an important event in carcinogenesis. The micronucleus assays have emerged as one of the preferred methods for assessing chromosome damage because they enable both chromosome loss and chromosome breakage to be measured reliably. Because micronuclei can only be expressed in cells that complete nuclear division a special method was developed that identifies such cells by their binucleate appearance when blocked from performing cytokinesis by cytochalasin-B (Cyt-B), a microfilament-assembly inhibitor. The cytokinesis-block micronucleus (CBMN) assay allows better precision because the data obtained are not confounded by altered cell division kinetics caused by cytotoxicity of agents tested or sub-optimal cell culture conditions. The method is now applied to various cell types for population monitoring of genetic damage, screening of chemicals for genotoxic potential and for specific purposes such as the prediction of the radiosensitivity of tumours and the inter-individual variation in radiosensitivity. In its current basic form the CBMN assay can provide, using simple morphological criteria, the following measures of genotoxicity and cytotoxicity: chromosome breakage, chromosome loss, chromosome rearrangement (nucleoplasmic bridges), cell division inhibition, necrosis and apoptosis. The cytosine-arabinoside modification of the CBMN assay allows for measurement of excision repairable lesions. The use of molecular probes enables chromosome loss to be distinguished from chromosome breakage and importantly non-disjunction in non-micronucleated binucleated cells can be efficiently measured. The in vitro CBMN technique, therefore, provides multiple and complementary measures of genotoxicity and cytotoxicity which can be achieved with relative ease within one system. The basic principles and methods (including detailed scoring criteria for all the genotoxicity and cytotoxicity end-points) of the CBMN assay are described and areas for future development identified.
Article
Capsaicin ( trans -8-methyl- N -vanillyl-6-nonenamide), a natural product of Capsicum species, is known to induce excitation of nociceptive terminals involved in pain perception. Recent studies have also shown that capsaicin not only has chemopreventive properties against certain carcinogens and mutagens but also exerts anticancer activity. Here, we demonstrated the antiangiogenic activity of capsaicin using in vitro and in vivo assay systems. In vitro , capsaicin inhibited vascular endothelial growth factor (VEGF) -induced proliferation, DNA synthesis, chemotactic motility, and capillary-like tube formation of primary cultured human endothelial cells. Capsaicin inhibited both VEGF-induced vessel sprouting in rat aortic ring assay and VEGF-induced vessel formation in the mouse Matrigel plug assay. Moreover, capsaicin was able to suppress tumor-induced angiogenesis in chick chorioallantoic membrane assay. Capsaicin caused G1 arrest in endothelial cells. This effect correlated with the down-regulation of the expression of cyclin D1 that led to inhibition of cyclin-dependent kinase 4-mediated phosphorylation of retinoblastoma protein. Signaling experiments show that capsaicin inhibits VEGF-induced p38 mitogen-activated protein kinase, p125FAK, and AKT activation, but its molecular target is distinct from the VEGF receptor KDR/Flk-1. Taken together, these results demonstrate that capsaicin is a novel inhibitor of angiogenesis and suggest that it may be valuable to develop pharmaceutical drugs for treatment of angiogenesis-dependent human diseases such as tumors.
Article
Metabolic reprogramming and altered bioenergetics have become emerged as a hallmark of cancer and an area of active basic and translational cancer research. Drastically upregulated glucose transport and metabolism in most cancers regardless the oxygen supply, a phenomenon called the Warburg effect, is one of major focuses of the research. Warburg speculated that cancer cells, due to defective mitochondrial oxidative phosphorylation (OXPHOS), switch to glycolysis for ATP synthesis, even in the presence of oxygen. Studies in the recent decade indicated that while glycolysis is indeed drastically upregulated in almost all cancer cells, mitochondrial respiration continues to operate normally at rates proportional to oxygen supply. There is no OXPHOS-to-glycolysis switch but rather upregulation of glycolysis. Furthermore, upregulated glycolysis appears to be for synthesis of biomass and reducing equivalents in addition to ATP production. The new finding that a significant amount of glycolytic intermediates are diverted to the pentose phosphate pathway (PPP) for production of NADPH has profound implications in how cancer cells use the Warburg effect to cope with reactive oxygen species (ROS) generation and oxidative stress, opening the door for anti-cancer interventions taking advantage of this. Recent findings in the Warburg effect and its relationship with ROS and oxidative stress controls will be reviewed. Cancer treatment strategies based on these new findings will be presented and discussed.
Article
Nanoscale diamond has recently received considerable attention due to the various possible applications such as luminescence imaging, drug delivery, quantum engineering, surface coatings, seeding etc. For most of these fields a suitable surface termination and functionalization of the diamond materials are required. In this feature article we discuss recent achievements in the field of surface modification of nanoscale diamond including the establishment of a homogeneous initial surface termination, the covalent and non-covalent immobilization of different functional moieties as well as the subsequent grafting of larger (bio)molecules onto previously functionalized nanodiamond.