ArticlePDF Available

Cellular effects of a turmeric root and rosemary leaf extract on canine neoplastic cell lines

Authors:
  • Royal Canin SAS

Abstract and Figures

Background: The use of nutraceuticals is gaining in popularity in human and canine oncology with a relatively limited understanding of the effects in the vastly different tumor types seen in canine oncology. We have previously shown that turmeric root (TE) and rosemary leaf (RE) extracts can work synergistically to reduce neoplastic cell growth, but the mechanisms are poorly understood and require further elucidation. Results: Three different canine cell lines (C2 mastocytoma, and CMT-12 mammary carcinoma, D17 osteosarcoma) were treated with 6.3 μg mL-1 extract individually, or 3.1 μg mL-1 of each extract in combination based on studies showing synergy of these two extracts. Apoptosis, antioxidant effects, cellular accumulation of curcumin, and perturbation of signaling pathways were assessed. The TE + RE combination treatment resulted in Caspase 3/7 activation and apoptosis in all cell lines, beyond the effects of TE alone with the CMT-12 cell line being most susceptible. Both extracts had antioxidant effects with RE reducing reactive oxygen species (ROS) by 40-50% and TE reducing ROS by 80-90%. In addition RE treatment enhanced the c-jun N-terminal kinase (JNK) activity in the C2 cell line and TE + RE exposure increased activated JNK by 4-5 times in the CMT-12 cell line. Upon further examination, it was found that RE treatment caused a significant increase in the cellular accumulation of curcumin by approximately 30% in the C2 and D17 cell lines, and by 4.8-fold in the CMT-12 cell line. This increase in intracellular curcumin levels may play a role in the synergy exhibited when using TE and RE in combination. Conclusions: The use of RE in combination with TE induces a synergistic response to induce apoptosis which is better than either extract alone. This appears to be related to a variable increased TE uptake in cells and activation of pathways involved in the apoptotic response.
This content is subject to copyright. Terms and conditions apply.
R E S E A R C H A R T I C L E Open Access
Cellular effects of a turmeric root and
rosemary leaf extract on canine neoplastic
cell lines
Corri B. Levine
1
, Julie Bayle
2
, Vincent Biourge
2
and Joseph J. Wakshlag
1*
Abstract
Background: The use of nutraceuticals is gaining in popularity in human and canine oncology with a relatively
limited understanding of the effects in the vastly different tumor types seen in canine oncology. We have previously
shown that turmeric root (TE) and rosemary leaf (RE) extracts can work synergistically to reduce neoplastic cell growth,
but the mechanisms are poorly understood and require further elucidation.
Results: Three different canine cell lines (C2 mastocytoma, and CMT-12 mammary carcinoma, D17 osteosarcoma) were
treated with 6.3 μgmL
1
extract individually, or 3.1 μgmL
1
of each extract in combination based on studies showing
synergy of these two extracts. Apoptosis, antioxidant effects, cellular accumulation of curcumin, and perturbation of
signaling pathways were assessed. The TE + RE combination treatment resulted in Caspase 3/7 activation and apoptosis
in all cell lines, beyond the effects of TE alone with the CMT-12 cell line being most susceptible. Both extracts had
antioxidant effects with RE reducing reactive oxygen species (ROS) by 4050% and TE reducing ROS by 8090%. In
addition RE treatment enhanced the c-jun N-terminal kinase (JNK) activity in the C2 cell line and TE + RE exposure
increased activated JNK by 45 times in the CMT-12 cell line. Upon further examination, it was found that RE treatment
caused a significant increase in the cellular accumulation of curcumin by approximately 30% in the C2 and D17 cell
lines, and by 4.8-fold in the CMT-12 cell line. This increase in intracellular curcumin levels may play a role in the synergy
exhibited when using TE and RE in combination.
Conclusions: The use of RE in combination with TE induces a synergistic response to induce apoptosis which is better
than either extract alone. This appears to be related to a variable increased TE uptake in cells and activation of
pathways involved in the apoptotic response.
Keywords: Apoptosis, Canine cancer, Mammary carcinoma, Osteosarcoma, Mastocytoma, Curcumin, Rosemary
Background
The use of natural remedies, or nutraceuticals, in the
treatment of cancer and a variety of other diseases ap-
pears prevalent in human and veterinary medicine. The
use of plant extracts has been around for centuries, but
investigations into the mechanisms of action across vari-
ous cancer cell lines are more recent, and appear to be
highly variable in cell culture systems [13]. The effect-
ive compounds of interest have been purified from a var-
iety of plants and are used in treating various diseases,
including cancer [4]. The benefit of using these plant
extracts to treat cancer is the potential synergy of mul-
tiple compounds found within a single extract whereby
the major compound may have one or more targets,
while other molecules in the extract may be affecting
other targets or influencing absorption kinetics [5].
The effects of these purified compounds have been ex-
amined in vitro in a variety of human cell lines derived
from tumors of the colon, skin, and breast tissue [6], but
only a few studies have looked at the effects in canine
cancer cells lines [79]. The major types of cancer found
in the dog differ from humans with lymphoma, mast cell
disease, osteosarcoma, and mammary neoplasia being
most often diagnosed in canine oncology [10]. We previ-
ously identified two extracts, turmeric extract rich in
curcuminoids (TE) and rosemary leaf extract rich in
* Correspondence: jw37@cornell.edu
1
Department of Clinical Sciences,Veterinary Medical Center C2-009, Cornell
University College of Veterinary Medicine, Ithaca, NY 14853, USA
Full list of author information is available at the end of the article
© The Author(s). 2017 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Levine et al. BMC Veterinary Research (2017) 13:388
DOI 10.1186/s12917-017-1302-2
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
carnosic acid (RE), which were shown to be cytotoxic
and reduce proliferation in a synergistic manner in ca-
nine mastocytoma, mammary carcinoma, and osteosar-
coma cell lines [11]. The use of TE and its major
compound of interest, curcumin, has been extensively
studied to treat a variety of diseases and ailments, per-
haps due to its ability to bind and interact with a variety
of cellular proteins [12]. Unfortunately, the use of TE in
vivo has been limited by its poor bioavailability and ef-
forts are still underway to increase the absorption and
bioavailability of the curcuminoids found in this extract
[13]. This obstacle may be overcome through the use of
combination treatments with other extracts that improve
bioavailability or hinder additional pathways [1416]. In
our previous study, RE worked in a synergistic manner
with TE to decrease cellular proliferation when used in
combination. Carnosic acid, the compound of interest in
RE, can target a variety of signaling pathways, many of
which overlap with those targeted by curcumin. The
effects of these two compounds in combination have
been examined in acute myeloid leukemia cells and
breast cancer cells [17, 18], showing synergy in anti-
proliferative effects and increased pro-apoptotic signal-
ing. The safety of these commonly used feed ingredients
and continual synergy between the extracts make them
candidates for inclusion in the diet as a potential adju-
vant treatment for dogs diagnosed with neoplasia, if ap-
propriate serum concentrations can be achieved.
The objective of this in vitro study was to determine
the effects on canine cancer cell death and possible
mechanisms by which TE and RE exert anti-proliferative
and cytotoxic effects individually and in combination on
canine mastocytoma, mammary carcinoma, and osteo-
sarcoma cell lines. Concentrations were chosen based on
our prior publication surrounding the effective concen-
trations for synergy between the two extracts of interest
[11]. Markers of apoptosis, antioxidant capabilities, and
changes in the activation of common cell signaling path-
ways were analyzed after treatment.
Methods
Natural extracts
Turmeric extract (TE; 88% total curcuminoids, #DA251471
Naturex, Avignon, France) and rosemary extract (RE; 67%
carnosic acid, #302036 Vitiva, Markovcih, Slovenia) were
solubilized in 100% dimethyl sulfoxide (DMSO; Sigma-
Aldrich, St. Louis, MO, USA) at 20 mg mL
1
.Freshextract
stock solutions were prepared and used for every
experiment.
Cell culture
Three canine neoplastic established cell lines, represent-
ing hematopoietic, epithelial, and mesenchymal tumor
types were used for all experiments; mastocytoma C2
(Dr. Warren Gold, University of California, San Fran-
cisco, USA), mammary gland carcinoma CMT-12 (Dr. R.
Curtis Bird, Auburn University, Alabama, USA), and
osteosarcoma D17 (#CCL-183; ATCC, Manassas, VA,
USA). These cell lines were chosen for initial screening
as representative cell lines of the three major cell line-
ages of cancer in dogs in hopes of finding a similar glo-
bal effect across different cell lineages. Cell lines were
grown on tissue culture-treated plates (Laboratory Prod-
uct Sales, Rochester, NY, USA) at 37 °C and 5% CO
2
for
all experiments and passage of cells, unless otherwise
noted. Cell lines were cultured in appropriate complete
medium as previously described.
11
All culture reagents
were purchased from Invitrogen, Carlsbad, CA, USA,
unless otherwise indicated.
Apoptosisassociated caspase 3/7 activation assay
Cells were plated at a density of 4 × 10
3
cells per well on
white walled 96-well tissue culture-treated plates (Ther-
moFisher Scientific, Waltham, MA, USA) and incubated
overnight in complete medium. Cells were treated the
following day with DMSO vehicle control, 6.3 μgmL
1
extract alone, or 3.1 μgmL
1
each extract in combin-
ation for 36 h. Chemotherapeutic drugs at a 50% inhibi-
tory concentration (IC
50
) were used as a positive
control; 12.5 nM toceranib phosphate (Palladia, Zoetis
Animal Health, Florham Park, NJ) was used for the C2
cell line, and 0.3 or 0.5 μM doxorubicin hydrochloride
(Sigma Aldrich, St Louis, MO) was used for the CMT-12
and D17 cell lines, respectively. Background fluorescence
and luminescence was measured in wells containing
treatments but no cells. Caspase 3/7 activation was
quantified using the ApoLive-GloMultiplex Assay
(Promega, Madison, WI, USA) following manufacturers
instructions. Briefly, after 36 h of treatment, viability re-
agent was added to the wells and incubated at 37 °C for
30 m and fluorescence was measured at 400
Ex
/505
Em
.
Next, Caspase-Glo 3/7 Reagent was added to all wells,
incubated for 30 m at room temperature, and lumines-
cence was measured. Fluorescence and luminescence
was measured using SpectraMax M3 Microplate Reader
(Molecular Devices, Sunnyvale, CA, USA).
Flow Cytometry
Cells were plated on 60 mm tissue culture-treated plates
(LPS, Rochester, NY) and incubated in complete medium
until 60% confluent. Cells were then treated with medium,
DMSO vehicle control, extract alone, or extracts in com-
bination. Cells were treated for 12 h (reactive oxygen spe-
cies generation), 24 h (curcumin accumulation), or 48 h
(Apoptosis/Necrosis, Cell Cycle). All flow cytometric ana-
lysis was performed on BD FACSCalibur (BD Biosciences,
San Jose, CA, USA).
Levine et al. BMC Veterinary Research (2017) 13:388 Page 2 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Cell cycle analysis
Cell cycle effects were analyzed after 24 h (data not
shown) and 48 h treatment using propidium iodide
staining to label DNA content. Briefly, cells were de-
tached with Accumax cell dissociation solution (Innova-
tive Cell Technologies, San Diego, CA USA), collected
in tubes with 1% fetal bovine serum (FBS) in Phosphate
Buffered Saline (PBS) and centrifuged for 5 m at 300 rcf
at 4 °C. The cell pellet was washed twice with 1% FBS in
PBS, filtered, and resuspended in 70% cold ethanol for
overnight fixation. The following day, samples were cen-
trifuged for 10 m at 500 rcf at 4 °C, resuspended in cold
PBS. Samples were centrifuged again for 5 m at 300 rcf
at 4 °C and resuspended in DNA staining solution [2%
propidium iodide (Sigma Aldrich), 0.1% Triton X-100
(Sigma Aldrich), in PBS]. Samples were then incubated for
30 m at room temperature and analyzed with an excita-
tion of wavelength of 488 nm and emission of 617 nm.
Only C2 and D17 cell lines were analyzed due to the pres-
ence of frequent doublets with CMT-12 cells resulting in
an artificial accumulation in the G2/M phase.
Apoptosis and necrosis assay
Apoptosis and necrosis was measured after 48 h treat-
ment using Annexin-V and 7-AAD staining. Briefly, cells
were detached with Accumax dissociation solutions (In-
novative Cell Technologies, San Diego, CA, USA), col-
lected and centrifuged for 10 m at 500 rcf at 4 °C. The
cell pellet was washed once with PBS before resuspen-
sion in Annexin Binding Buffer (ABB; 10 mM HEPES,
140 mM NaCl, 2.5 mM CaCl
2
, pH 7.4) at a density of
1×10
6
cell mL
1
. Annexin-V 488 conjugate and 7-
Aminoactinomycin D (7-AAD) were added to the cell
suspensions and incubated for 15 m at room
temperature. After the incubation, ABB was added to
the cell suspension and kept on ice until fluorescence
analysis. Events labeled only Annexin-V positive were
considered to represent apoptotic cells; events labeled
Annexin-V positive and 7-AAD positive were considered
to represent necrotic cells.
Intracellular reactive oxygen species (ROS) analysis
Since the main constituents of TE and RE (curcumin
and carnosic acid, respectively) have been implicated as
antioxidants, Dihydrorhodamine123 (DHR123; Invitro-
gen, Carlsbad, CA, USA) assay was used to determine
the amount of reactive oxygen species (ROS) present
after 12 h treatment with each extract according to lit-
erature [19]. Briefly, cells were detached using Accumax
dissociation solution (Innovative Cell Technologies), col-
lected and centrifuged for 10 m at 500 rcf at 4 °C. The
pellet was washed once with PBS before resuspension in
1 mL of stain (30 μM DHR123 in DMEM). The cell sus-
pension was then incubated at 37 °C for 30 m, pelleted,
and resuspended in 1 mL DMEM and filtered before
fluorescence analysis of cells.
Cellular accumulation of curcumin
The cellular accumulation of curcumin was measured by
exploiting the auto-fluorescent properties of this com-
pound [20]. After 24 h treatment, cells were detached
with Accumax dissociation solution (Innovative Cell
Technologies), collected and centrifuged for 10 m at 500
rcf at 4 °C. The cell pellet was washed once with PBS be-
fore resuspension in DMEM, and filtered before fluores-
cence analysis when excited at a wavelength of 488 nm
and then measuring emission using a 530/30 filter.
Western blotting assessment of affected signaling pathways
Cells were plated on 100 mm tissue culture-treated
plates (LPS) and incubated overnight in complete
medium until 60% confluency was reached. Cells were
treated the following day with DMSO vehicle control,
6.3 μgmL
1
extract alone, or 3.1 μgmL
1
each extract
in combination. Cells were harvested and lysed at 12 h
and 24 h after treatment using Mammalian Lysis Buffer
(MLB; 25 mM Tris, 100 mM NaCL, 1 mM EDTA, 1%
Triton X-100, 0.004% NaF, 1 mM NaVO4, 25 mM -gly-
cerophosphoric acid, 100 μg/ml phenylmethanesulfonyl
fluoride, and 1 μg/ml each aprotinin and leupeptin,
pH 7.4) and sonication, and then centrifuged for 5 m at
14,000 rcf at 4 °C. The supernatant was collected and
the protein concentration was determined using the
Bradford assay (Coomassie-dye; ThermoFisher Scientific
Pierce, Waltham, MA, USA). Samples were equilibrated
to a common volume (μgμL
1
) in MLB and 5× laemmili
loading buffer (300 mM Tris-HCl pH 6.8, 10% Sodium
dodecyl sulfate, 50% glycerol, 12.5% β-Mercaptoethanol,
0.025% Bromophenol blue). For each protein of interest,
30 μg total proteins were subjected to sodium dodecyl
sulfate polyacrylamide gel electrophoresis (SDS-PAGE)
on gels ranging from 6 to 15% based on the molecular
weight of the protein of interest. The proteins were then
transferred to 0.45 μm pore size polyvinylidene fluoride
membrane (Immobilon-P Membrane; EMD Millipore,
Billerica, MA, USA) for 1 h at 333 mA and then blocked
in 5% milk in Tris-buffered saline/0.05% Tween-20 solu-
tion (TBST). Membranes were incubated overnight in
primary antibody solutions at a dilution of 1:1000 in
TBST on a rocking platform at 4 °C. Primary antibodies
included mouse anti- phosphorylated-gamma H2A.X
and extracellular regulated kinase (ERK) (R&D Biosciences,
Boston, MA, USA); mouse anti- Thr202/Tyr204 phosphor-
ylated p44/42 MAPK (ERK1/2) and STAT3 (Cell Signaling
Technology,Danvers,MA,USA);rabbitanti-proteinkin-
ase B (AKT), Ser473 phosphorylated-AKT, stress-activated
protein kinase/jun-N-terminal kinase (SAPK/JNK),
Thr183/Tyr185 phosphorylated-SAPK/JNK, focal adhesion
Levine et al. BMC Veterinary Research (2017) 13:388 Page 3 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
kinase (FAK), Tyr397 phosphorylated-FAK, Tyr576/Tyr577
phosphorylated-FAK, Tyr925 phosphorylated-FAK, Src,
Tyr416 phosphorylated-Src, Tyr527 phosphorylated-Src,
mammalian target of rapamycin (mTOR), Ser2448
phosphorylated-mTOR, Janus kinase 2 (JAK2), Tyr1007/
Tyr1008 phosphorylated-JAK2, Ser727 phosphorylated-
signal transducer and activator of transcription 3 (STAT3),
Tyr705 phosphorylated-STAT3, B-Cell CLL/Lymphoma 2
(BCL2), and BCL2-Associated X Protein (BAX) (Cell Sig-
naling Technology). Membranes were washed three times
with TBST and incubated at room temperature for 1 h in
the corresponding secondary anti-mouse IgG or anti-
rabbit IgG horseradish peroxidase-conjugated antibody at
a dilution of 1:2000 (Cell Signaling Technology). Mem-
branes were washed three times with TBST and visualized
with a chemi-luminescent reagent (Clarity Western ECL
Substrate; Bio-Rad, Hercules, CA, USA). Digital images
were captured using an imaging system (Biospectrum 410;
UVP, Upland, CA, USA). After images were collected,
membranes were washed three times in TBST and incu-
bated with a 1:10,000 dilution in TBST of the house-
keeping antibody β-Actin (Sigma-Aldrich) for 1 h at room
temperature. Membranes were washed, incubated with
mouse secondary antibody at a dilution of 1:2000, and im-
aged as described.
Data management and statistical analysis
For all flow cytometry experiments, 10,000 events were
collected per sample and then gated based on a forward-
scatter/side-scatter plot. Minimally three independent ex-
periments were examined for each treatment through the
different assays (percent of gated cells in each phase cycle,
percent of apoptotic cells, intracellular ROS and curcumin
level) and analyzed with Cell Quest software (BD Biosci-
ences). DMSO was compared against cells in media alone
showing no significant differences, therefore DMSO
treated cells were used as the control sample for all com-
parisons. The geometric mean fluorescence (GMF) from
each treatment was compared to the DMSO treated sam-
ples and represented as fold change for all experiments
using GMF due to the differences in fluorescence intensity
across cell lines. In addition, for measurements of ROS, an
unstained control was used to determine the baseline
GMF of each extract. This value was subtracted from the
GMF of stained samples to correct for any shift due to
auto-fluorescence of the extract with cells alone. Caspase
3/7 activation was determined as caspase activation per
total viable cells for each treatment. Raw data from the
viability portion of the assay (individual fluorescence
values of each well) were normalized to the vehicle alone
treatment for each cell line, considered to represent 100%
proliferating cells. The ratio of caspase activation to viable
cells is represented as fold increase over DMSO treatment
alone. Each of the treatment conditions were completed
in duplicate and averaged in four independent experi-
ments. Western blots were run in three independent time
course experiments and densitometry was completed
using ImageJ [21]. Values are represented as a ratio of
phosphorylated protein to total protein and standardized
to DMSO vehicle control at every time point examined.
All statistical analyses were performed using JMP Pro
(v. 11.2.1; SAS Institute Inc., Cary, NC, USA). The resid-
uals of all statistical models were found to be normally
distributed therefore parametric statistics were utilized.
The fold-change data from caspase 3/7 activation, per-
cent of apoptotic cells, intracellular ROS level and cur-
cumin accumulation assays and the ratio data from
western blot assay were processed using analysis of vari-
ance with Tukeys method for multiple comparisons be-
tween all treatment conditions (single, combination and
DMSO control). In the case of cell cycle dynamics, Dun-
netts method was used to control for multiple compari-
sons when studying the percent of gated events
difference between single treatment or dual combination
and DMSO control only at each time point. Differences
were considered statistically significant at p< 0.05.
Results
Turmeric and rosemary extracts effects on cell cycle
The effects of TE and/or RE on cell cycle progression were
measured on C2 and D17 tumor cell lines using propi-
dium iodide staining. Cell cycle dynamics were analyzed
after 24 h and 48 h of incubation with the different treat-
ments; no significant difference was seen between these
two time-points therefore only data from the 48 h time
point is shown (Fig. 1A-B). Representative histograms are
shown for C2 (Fig. 1C) and D17 (Fig. 1D) cell lines. Single
treatment with 6.3 μgmL
1
TE resulted in a significant
decrease in S phase (DNA replication) in the D17 cell line
compared to DMSO control. Treatment with 6.3 μgmL
1
RE induced a significant decrease in G
1
/G
0
phase in the
D17 cell line, a reduction in S phase in both cell lines, and
an increase in G
2
/M phase (cell division) in the D17 cell
line. The combination treatment using 3.1 μgmL
1
both
extracts induced a small decrease in S phase in only the
C2 cell line, and a modest increase in G
2
/M phase in only
the D17 cell line. While these differences were significant,
the mild alterations in cell cycle in the D17 and C2 of 5
10% decrease in the G
1
/G
0
and increase in G
2
/M phases
with RE and less consistent yet similar changes with both
TE and RE combined were not considered large enough
to continue examining pathways related to cell cycle arrest
as observed in prior studies using these cell lines [22].
Cellular apoptosis is induced by turmeric and rosemary
extract treatments.
After treating cells for 36 h, TE alone (6.3 μgmL
1
) re-
sulted in a significant increase in apoptotic cells in the
Levine et al. BMC Veterinary Research (2017) 13:388 Page 4 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
C2 and CMT-12 cell lines as determined by Caspase 3/7
activation, 2- and 2.5-fold, respectively (Fig. 2), and
Annexin-V staining which increased from 6% to 11%
apoptotic cells in the C2 (Fig. 3E) and from 4% to 13%
in the CMT-12 cell line (represented in Fig. 3A and C
lower right quadrant; full analysis Fig. 3E). A treatment
with 6.3 μgmL
1
RE alone resulted in a statistically
significant increase of 1.4-fold in Caspase 3/7 activation
in all three cell lines when compared to vehicle control.
When the dual combination treatment (3.1 μgmL
1
TE
+ 3.1 μgmL
1
RE) was used, a significant increase in
Annexin-V positive cells compared to vehicle control
was seen in the 3 cell lines, but this was not significant
compared to 6.3 μgmL
1
TE alone in C2 cell line.
Fig. 1 Effects of turmeric and rosemary extract on cell cycle in C2 and D17 cell lines. Cells were treated with indicated concentrations of extracts
or DMSO for 48 h and the DNA contents were analyzed using propidium iodide staining by flow cytometry. Percentages of cells within each cell
cycle phase (G1, S, and G2/M) were expressed as mean ±standard deviation in (a) C2 and (b) D17 cell lines. Bar graphs represent the average of
three individual experiments performed in duplicate and representative cell cycle histograms of DMSO control treated (c) C2 and (d) D17 cell
lines are shown at 48 h of treatment. All treatments were compared to DMSO vehicle control. Treatments which induced a statistically significant
change from DMSO within each cell cycle phase are indicated
Fig. 2 Caspase 3/7 activation induced by turmeric and rosemary extracts in C2, CMT-12, and D17 cell lines. Cells were treated with indicated
concentrations of extracts or DMSO for 36 h. Activated caspase 3/7 per viable cells was expressed as mean fold change from DMSO control values
± standard deviation from three independent replicates. Within each cell line, values with different letters are significantly different from each
other (C2 p< 0.001; CMT-12 p< 0.005; D17 p< 0.05)
Levine et al. BMC Veterinary Research (2017) 13:388 Page 5 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
However, in the CMT-12 and D17 cell lines, the com-
bination treatment induced a significantly greater per-
centage of apoptotic cells, 40% and 13%, respectively,
compared to 6.3 μgmL
1
TE alone (13% and 7%) and
6.3 μgmL
1
RE alone (5%) (Fig. 3). This was further val-
idated with the caspase activation assay in which all
Fig. 3 Apoptosis induction by turmeric and rosemary extracts in C2, CMT-12, and D17 cell lines. Cells were incubated with the indicated treatments
for 48 h and the induction of apoptosis was detected by Annexin V-FITC and 7-AAD staining followed by flow cytometric analysis. Representative
quadrant plots of the CMT-12 cell line treated with (a)DMSO,(b)6.3μgmL
1
TE, (c)6.3μgmL
1
RE, or (d)3.1μgmL
1
TE + 3.1 μgmL
1
RE are shown.
Each quadrant represents the number of events considered live (lower left), early apoptotic (lower right), or late apoptotic/necrotic (upper right). e
Percent early apoptotic cells (lower right quadrant of Annexin V positive and 7-AAD negative cells) are represented as mean ± standard deviation
(three independent replicates). Within each cell line, means with different letters are significantly different from each other (p<0.05)
Levine et al. BMC Veterinary Research (2017) 13:388 Page 6 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
three cell lines (C2, CMT-12 and D17) showed a signifi-
cant increase in cleaved Caspase 3/7 when the combin-
ation treatment was used (2.8, 5- and 2.2-fold,
respectively), compared to TE alone (2-, 2.5- and 1.2-
fold) or RE alone (1.4-fold across all three cell lines).
Antioxidant activity of TE and RE in cancer cell lines
TE alone was a significantly stronger antioxidant than
RE alone using same extract concentration (6.3 μgmL
1
) in all the three cell lines (C2, CMT-12 and D17) with
TE reducing ROS by about 7590-80%, respectively and
RE reducing ROS by about 5040-40%, respectively. The
dual combination treatment using half the concentration
(3.1 μgmL
1
each extract) was as effective as 6.3 μgmL
1
TE alone in all three cancer cell lines (Fig. 4).
Cellular accumulation of curcumin induced by RE
treatment
Observation from previous flow cytometry experiments
showed an unexpected increase in the GMF when cells
were treated with TE alone when excited at a wavelength
of 488 nm, whereas no change was observed when RE
was used alone (Fig. 5D). A similar increase in GMF was
also seen when half the concentration of each extract
was used in combination (data not shown). Therefore,
the possibility that RE could increase the cellular accu-
mulation of the fluorescent compound curcumin was in-
vestigated when these compounds were used in
combination. TE alone (3.1 μgmL
1
) significantly in-
creased the GMF in the C2 and D17 cell lines, 1.7- and
1.8-fold, respectively; while when using RE with TE the
increase was 2.2 and 2.3 fold, respectively (Fig. 5A, C; p
< 0.0001). The addition of RE at the same concentration
to TE resulted in a significant increase in GMF of 4.8-
fold in the CMT-12 cell line beyond that of TE alone
(Fig. 5B; p< 0.0001).
TE and RE SAPK/JNK activation
After examination of several cell signaling pathways, no
consistent trend was seen in the phosphorylation status
of the variety of signaling proteins, alterations in the
mitochondrial proteins involved in apoptosis or markers
of DNA damage (data not shown). However, changes in
Thr183/Tyr185 phosphorylated-SAPK/JNK (p-SAPK/
JNK; Fig. 6) were detected in the three cancer cell lines.
Treatment with 6.3 μgmL
1
TE resulted in an increase
from a densitometry value of 1.1 at 12 h to 1.5 at 24 h in
p-SAPK/JNK in the C2 cell line, stable activation from
12 h to 24 h in the CMT-12 cell line (1.5 and 1.8, re-
spectively). In the D17 cell line only a minor non-
significant increase was observed (1.2 at 12 h, 1.1 at
24 h). Activated SAPK/JNK increased from 12 h to 24 h
in the C2 cell line (1.8 and 2.1, respectively) and the
CMT-12 cell line (1.2 at 12 h to 1.5 at 24 h) which was
not significant over time. Minimal change was seen in
the D17 cell lines after treatment with 6.3 μgmL
1
RE,
1.1 at both time points. The greatest increase in p-
SAPK/JNK was seen with the combination of 3.1 μg
mL
1
each of TE and RE in the CMT-12 cell line where
densitometry values increased from 1.0 with DMSO
treatment to 4.3 after 12 h incubation and 4.8 after
24 h incubation (p< 0.05 from DMSO treatment). Al-
though there were similar increases in SAPK/JNK acti-
vation with TE and dual treatment of TE/RE (half
doses) in C2 and D17 cell lines, these were not signifi-
cantly different from DMSO control at 12 h or 24 h in-
cubation time points. These results demonstrate
possible mechanisms behind the observed susceptibility
differences across the three cell lines, particularly in
light of the heightened response of lesser doses of RE
and TE in combination when compared to higher con-
centrations each extract independently in the CMT12
cell line.
Fig. 4 Antioxidant effects of turmeric and rosemary extracts in C2, CMT-12 and D17 cell lines. Cells were treated with the indicated concentrations of
extracts for 12 h followed by determination of intracellular levels of reactive oxygen species using Dihydrorhodamine123 staining. Values are expressed
as mean ± standard deviation of four independent replicates. Reported values are represented as fold change compared to DMSO vehicle control.
Within each cell line, means with different letters are significantly different from each other (C2 p <0.05; CMT-12 and D17 p< 0.0001)
Levine et al. BMC Veterinary Research (2017) 13:388 Page 7 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Discussion
Bioactive molecules derived directly from plants, or
modeled after plant compounds, continue to be an ac-
tive area of cancer research. The majority of these stud-
ies have been focused on human and rodent cancer
models and the effects of these plant extracts and select
compounds vary depending on species and cell origin
[23, 24]. Few studies have been completed in dogs or re-
lated cell lines, therefore it is necessary to examine the
effects of these compounds in vitro before using them in
clinical veterinary trials. The aim of the current in vitro
study was to examine the molecular effects of two nat-
ural extracts, turmeric root extract (rich in curcumi-
noids) and rosemary leaf extract (rich in carnosic acid),
previously shown to inhibit proliferation synergistically
in three established canine cancer cell lines [11]. These
experiments were designed to focus on concentrations
that may have utility in vivo and concentrations that
showed synergistic effects of the compounds in our prior
experiments (focusing on synergistic concentrations of
3μg/mL of TE and RE versus 6 μg/mL
1
of each extract
independently) [11]. In agreement with our previous
proliferation and cytotoxicity results, cell treatment
using TE alone was more potent than RE single treat-
ment using the same extract concentrations and experi-
mental conditions. TE had a greater effect on inducing
cell apoptosis as measured by Caspase 3/7 activation
and Annexin-V staining, and the combination treatment
Fig. 5 Effect of rosemary extract on intracellular accumulation of curcumin in canine tumor cell lines. The C2 (a), CMT-12 (b), and D17 (c) cell lines
were treated with the indicated concentration of extracts for 24 h and then cellular accumulation of curcumin was quantified by flow cytometry. Y-axis
values represent the fold change in geometric mean fluorescence (GMF) of all cells compared to DMSO control. Reported data are expressed as mean
± standard deviation of 4 independent replicates. Within each cell line, means with different letters are significantly different from each other (p
< 0.0001). dRepresentative histogram of emission intensity in CMT-12 cell line after 24 h treatment with DMSO, 3.1 μgmL
1
TE alone,
3.1 μgmL
1
RE alone, or 3.1 μgmL
1
TE + RE combination is shown
Levine et al. BMC Veterinary Research (2017) 13:388 Page 8 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
using only half the concentration of each extract induced
a similar, if not greater, cell response. Varying degrees of
susceptibility were detected across the three cancer cell
lines used, with the CMT-12 cell line being the most
susceptible to these treatments, perhaps due to a greater
increase in intracellular curcumin accumulation as
shown by flow cytometry. These differences across cell
lines suggest the complexity in cellular response and po-
tential susceptibility of various cell lines to treatment,
further clarifying the need to understand what types of
cancer may be more responsive to these interventions.
Prior studies have shown the autofluorescence of cur-
cumin can be examined with flow cytometry [20]. An in-
crease in curcumin fluorescence was seen across the
three cell lines with the greatest signal measured in the
CMT-12 cell line, especially when the two extracts were
used in combination. A previous study in the human
breast cancer cell line, MCF-7, showed an increase in
intracellular accumulation of various chemotherapeutic
drugs which was attributed to competitive inhibition of
transmembrane transport pump P-glycoprotein by rose-
mary extract [25]. As this was not within the scope of
our investigation, further experiments examining P-
glycoprotein inhibition or curcumin utilization of other
channels to enter cells are warranted to better under-
stand the mechanisms by which rosemary enhances the
accumulation of curcumin within cells.
The global effects of both TE, RE, and the two in com-
bination showed no appreciable alteration in cell cycle
kinetics. Of the three cell lines used in our experiments,
only the C2 and D17 cells could be examined as a single
cell suspension for cell cycle dynamics, while the CMT-
Fig. 6 Changes in the protein expression levels of SAPK/JNK pathway in turmeric and rosemary-treated cells. C2, CMT-12 and D17 cell lines were
harvested and lysed after 12 h or 24 h treatment with DMSO vehicle control, or 6.3 μgmL
1
Turmeric extract (TE) alone, or 6.3 μgmL
1
Rosemary
extract (RE) alone, or combination of 3.1 μgmL
1
each of TE + RE. Expression level of Thr183/Tyr185 phosphorylated-SAPK/JNK (p46/p54) and total
SAPK/JNK were determined by Western blot analysis. Each blot is a representative of three independent experiments. Densitometry values
represent a ratio of phosphorylated protein to total protein and normalized to DMSO vehicle control of the same time point (mean of
three separate experiments). Changes in densitometry compared to DMSO control with significance of p< 0.05 represented by *. β-Actin
was used as a loading control for every blot to ensure even loading of samples
Levine et al. BMC Veterinary Research (2017) 13:388 Page 9 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
12 cells displayed an artificial accumulation of cells in
the G
2
/M phase. This was attributed to cell clumping in
this cell line due to the fixation method used and use of
propidium iodide causing cell-doublets to be inappropri-
ately represented as G
2
M phase. There were some mild
alterations in cell cycle in the D17 and C2 cell lines
showing decreases in the G
1
/G
0
and increases in G
2
/M
phases with RE and the combination treatment. There
were minimal to no cell cycle changes with TE extract
alone, therefore further examination of cell cycle path-
way analysis was not pursued. Prior literature has shown
that curcumin can have significant effects on cell cycle
dynamics through the upregulation of cyclins or cyclin
dependent kinase activity [2628]. In this previous lit-
erature, there was complete loss or greater than 50% re-
duction or increase in various portions of the cell cycle
warranting further examination of cellular pathways in-
volved. Generally, these differences may be due to the
lower concentrations utilized in our experiments com-
pared to the prior studies.
In line with pathway disruption and cell cycle dynam-
ics, TE and RE are often thought to be antioxidants,
however curcumin has been shown to cause oxidative
damage to DNA and induction of the DNA damage re-
sponse pathways that are intimately involved in cell cycle
alteration or apoptosis [29, 30]. Our assessment of anti-
oxidant status after treatment unanimously indicated
that the cells are under less oxidative stress after treat-
ment with either TE or RE alone or in dual combination
within 12 h of incubation. To further assess the oxida-
tive status, western blot analysis for gamma-histone
H2A.X phosphorylation status was assessed in the three
cell lines with TE, RE or dual treatment showing no
phosphorylation in DMSO control or treated cells.
Gamma-histone H2A.X is a marker of DNA oxidation
and initiation of repair and was not detected when com-
pared to UV irradiation as a positive control for DNA
damage (data not shown). Under our cell culture condi-
tions and extract concentrations used, we could not
elicit a pro-oxidative response from TE or RE in any cell
line used. This anti-oxidant property has been thought
to be involved in cell survival and possible resistance to
chemotherapeutic intervention [31]. Our data at these
concentrations only suggest pro-apoptotic responses,
suggesting that the mechanisms are unlikely to rely on
oxidative damage.
Further examination of the cellular effects into cell sig-
naling pathways previously implicated in TE and RE
treatment were performed [2535]. Concentrations that
appeared to be most synergistic at inhibiting prolifera-
tion from our prior publication [11] were used to ob-
serve enhanced or diminished signaling events over
extended periods of time from 12 to 24 h that might
provide insights into the modest apoptotic response.
Apoptosis could be, in part, due to overlapping effects
on various signaling pathways including SAPK/JNK,
ERK 1/2, STAT3, FAK, Src, mTOR, and membrane per-
meability proteins Bcl-2 and Bax [36, 37]. Previous lit-
erature has shown a synergistic effect between these two
extracts, specifically the cleavage of poly ADP-ribose
polymerase and Caspase-8, 9, and 3 on human cell
lines [17]. Though there is relatively little primary litera-
ture on canine cell lines, one study has shown that a
curcumin analog effectively alters STAT phosphorylation
and activation in canine osteosarcoma cells [38]. After
screening several signaling pathways, a consistent in-
crease in the phosphorylated, or active, form of SAPK/
JNK was detected with no consistent alterations in any
other pathways examined via western blotting. This
pathway has been implicated in driving cells to apoptosis
when faced with environmental stressors such as oxida-
tive stress, inhibition of protein synthesis, changes in the
cell-matrix interaction, or signaling from inflammatory
cytokines [3941]. Consistent with our results, studies
have shown that the downstream effects of SAPK/JNK
activation are both cell and context dependent: pathway
activation can be either pro-apoptotic or pro-
proliferative. [42, 43] Changes in activation of MAPK/
ERK were not observed after treatment in any of the cell
lines examined. In general, early, transient activation of
JNK may lead to cell survival, while sustained activation
can induce apoptosis and curcumin or rosemary extracts
appear to be involved in this constitutive activation of
SAPK/JNK [4446].
Differences in treatment responses were observed in
the three cell lines. Our results showed an increase in
phosphorylated SAPK/JNK after 12 h and 24 h of treat-
ment with TE alone. RE induced a significant increase in
phosphorylation after 12 h and 24 h of treatment in the
CMT-12 cell line, while in the C2 cell line this increase
was only seen at 12 h and returned to baseline by 24 h.
The dual combination treatment had the greatest effect
in the CMT-12 cell line, resulting in phosphorylated
SAPK/JNK at levels greater than either extract alone
(even using twice the concentration). Only the CMT-12
cell line showed sustained activation of SAPK/JNK with
the combination treatment which may be the underlying
reason behind the increased susceptibility of this cell
line. SAPK/JNK has been implicated as a therapeutic tar-
get in certain contexts and patterns of activation
whereby constitutive activation appears to be beneficial
towards a pro-apoptotic response in a variety of cell
lines and animal models [4749]. The transient nature
of activated SAPK/JNK in the C2 and D17 cell lines lead
us to believe this may be involved in the diminished pro-
liferation, however other pathways may be involved in
the induction of apoptosis in these cancer cell lines. This
data further demonstrates that the cell line and context
Levine et al. BMC Veterinary Research (2017) 13:388 Page 10 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
specific effects of these extracts are vastly different and
other approaches are needed to completely understand
the complex interaction of the pathways involved during
apoptosis induction.
Although the results of these experiments are promis-
ing, the clinical utility is complex due to the absorption,
transformation and elimination kinetics of these com-
pounds in general. The use of highly bioavailable curcu-
min is currently being examined and has 1015%
bioavailability [50]. Carnosic acid bioavailability from
rosemary extract, though good, has not been studied ex-
tensively; however there is rapid glucuronidation, methy-
lation, and oxidation of carnosic acid and related
molecules from rosemary extract, and the bioactivity of
these modified derivatives are unknown at this time [51].
Conclusions
The results of this study provides some insights into
possible mechanisms by which TE and RE induce apop-
tosis across three canine neoplastic cell lines. Our results
indicate that different tumor types are likely to have a
differential response to such interventions. The en-
hanced susceptibility found in the CMT-12 mammary
cancer cell line may be due to the increased accumula-
tion of curcumin when the combination treatment was
used. In addition, sustained activation and signaling
through the SAPK/JNK pathway may play a role in this
cell lines increased sensitivity to apoptosis. The results
of this study warrant further investigations into the
pharmacodynamics and pharmacokinetics of these ex-
tracts in dogs when incorporated into feed to determine
if clinical trials are feasible.
Abbreviations
AKT: Protein kinase B; BAX: BCL2-Associated X Protein; BCL2: B-Cell CLL/
Lymphoma 2; DMSO: Dimethyl sulfoxide; ERK: Extracellular related kinase;
FAK: Focal adhesion kinase; FBS: Fetal bovine serum; GMF: Geometric mean
fluorescence; JAK: Janus kinase; mTOR: Mammalian target of rapamycin;
PBS: Phosphate buffered saline; RE: Rosemary extract; ROS: Reactive oxygen
species; SAPK/JNK: Stress activated kinase/jun-N-terminal kinase; STAT: Signal
transducer and activator of transcription; TBST: Tris buffered saline tween;
TE: Turmeric extract
Acknowledgements
We are grateful to the University of California San Francisco, especially Dr.
Warren Gold and Dr. George Caughey, for supplying the C2 canine
mastocytoma cell lines and to Auburn University, especially Dr. R. Curtis Bird,
for supplying CMT-12 canine mammary gland carcinoma cell line.
Funding
The aforementioned study was funded by Royal Canin (JB and VB)
collaborated on the study design and interpretation of data collected.
Availability of data and materials
Datasets used and analyzed during this study are available from the
corresponding author upon reasonable request.
Author contributions
CBL carried out the technical experimentation, performed statistical analysis,
and was primary author in the manuscript. JB and VB conceived the study
and participated in its design and participated in manuscript editing. JW
helped conceive the study, supervised the study and helped in manuscript
drafting and editing. All authors have read and accepted the final manuscript.
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The research leading to these results was supported by Royal Canin SAS.
Royal Canin participated in writing the protocol, analyzing the data,
contributing compounds, revising the manuscript for publication. JB and VB
are employed by Royal Canin. JJW has received compensation from Nestle
Purina, Mars, Annamaet Pet Food Company, and Veterinary Recommend
Solutions for consultation.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Author details
1
Department of Clinical Sciences,Veterinary Medical Center C2-009, Cornell
University College of Veterinary Medicine, Ithaca, NY 14853, USA.
2
Royal
Canin Research Center, Airmargues, France.
Received: 9 June 2017 Accepted: 27 November 2017
References
1. Vidak M, Rozman D, Komel R. Effects of flavonoids from food and dietary
supplements on glial and Glioblastoma Multiforme cells. Mol Ther. 2015 Oct
23;20(10):1940632.
2. Li Y, Go VL, Sarkar FH. The role of Nutraceuticals in pancreatic cancer
prevention and therapy: targeting cellular signaling, MicroRNAs, and
Epigenome. Pancreas. 2015 Jan;44(1):110.
3. Niedzwiecki A, Roomi MW, Kalinovsky T, Rath M. Anticancer Efficacy of
Polyphenols and Their Combinations. Nutrients. 2016 Sep 9;8(9). pii: E552.
4. Newman DJ, Cragg GM, Snader KM. The influence of natural products upon
drug discovery. Nat Prod Rep. 2000 Jun;17(3):21534.
5. Gilbert B, Alves LF. Synergy in plant medicines. Curr Med Chem. 2003 Jan;
10(1):1320.
6. González-Vallinas M, González-Castejón M, Rodríguez-Casado A, Ramírez de
Molina A. Dietary phytochemicals in cancer prevention and therapy: a
complementary approach with promising perspectives. Nutr Rev. 2013;71:
58599.
7. Helmerick EC, Loftus JP, Wakshlag JJ. The effects of baicalein on canine
osteosarcoma cell proliferation and death. Vet Comp Oncol. 2014;12:299
309.
8. Wakshlag JJ, Balkman CE. Effects of lycopene on proliferation and death of
canine osteosarcoma cells. Am J Vet Res. 2010;71:136270.
9. Wakshlag JJ, Balkman CA, Morgan SK, McEntee MC. Evaluation of the
protective effects of all-trans-astaxanthin on canine osteosarcoma cell lines.
Am J Vet Res. 2010;71:8996.
10. Kelsey JL, Moore AS, Glickman LT. Epidemiologic studies of risk factors for
cancer in pet dogs. Epidemiol Rev. 1998;20:20147.
11. Levine CB, Bayle J, Biourge V, Wakshlag JJ. Effects and synergy of feed
ingredients on canine neoplastic cell proliferation. BMC Vet Res. 2016 Aug 2;
12(1):159.
12. Sharma RA, McLelland HR, Hill KA, Ireson CR, Euden SA, Manson MM, et al.
Pharmacodynamic and pharmacokinetic study of oral curcuma extract in
patients with colorectal cancer. Clin Cancer Res. 2001;7:1894900.
13. Lee WH, Loo CY, Young PM, Traini D, Mason RS, Rohanizadeh R. Recent
advances in curcumin nanoformulation for cancer therapy. Expert Opin
Drug Deliv. 2014 Aug;11(8):1183201.
14. Douglass BJ, Clouatre DL. Beyond yellow curry: assessing commercial
Curcumin absorption technologies. J Am Coll Nutr. 2015;34(4):34758.
15. Berginc K, Trontelj J, Basnet NS, Kristl A. Physiological barriers to the oral
delivery of curcumin. Pharmazie. 2012 Jun;67(6):51824.
16. Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MN. Nanoparticle
encapsulation improves oral bioavailability of curcumin by at least 9-fold
Levine et al. BMC Veterinary Research (2017) 13:388 Page 11 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
when compared to curcumin administered with piperine as absorption
enhancer. Eur J Pharm Sci. 2009 Jun 28;37(34):22330.
17. Pesakhov S, Khanin M, Studzinski GP, Danilenko M. Distinct combinatorial
effects of the plant polyphenols curcumin, carnosic acid, and silibinin on
proliferation and apoptosis in acute myeloid leukemia cells. Nutr Cancer.
2010;62:81124.
18. Einbond LS, HA W, Kashiwazaki R, He K, Roller M, Su T, et al. Carnosic acid
inhibits the growth of ER-negative human breast cancer cells and
synergizes with curcumin. Fitoterapia. 2012;83:11608.
19. Passos JF, Miwa S, von Zglinicki T. Measuring reactive oxygen species in
senescent cells. Methods Mol Biol. 2013;965:25363.
20. Kunwar A, Barik A, Mishra B, Rathinasamy K, Pandey R, Priyadarsini KI.
Quantitative cellular uptake, localization and cytotoxicity of curcumin in
normal and tumor cells. Biochim Biophys Acta. 2008 Apr;1780(4):6739.
21. Rasband, WS. ImageJ. U.S. National Institutes of Health, Bethesda, Maryland,
USA. http://imagej.nih.gov/ij/, 1997-2016.
22. Wakshlag JJ, Kallfelz FA, Wakshlag RR, Davenport GM. The effects of
branched-chain amino acids on canine neoplastic cell proliferation and
death. J Nutr. 2006 July;136(7 Suppl):2007S10S.
23. Ramos S. Effects of dietary flavonoids on apoptotic pathways related to
cancer chemoprevention. J Nutr Biochem. 2007 Jul;18(7):42742.
24. Ravindran J, Prasad S, Aggarwal BB. Curcumin and cancer cells: how many
ways can curry kill tumor cells selectively? AAPS J. 2009 Sep;11(3):495510.
25. Plouzek CA, Ciolino HP, Clarke R, Yeh GC. Inhibition of P-glycoprotein
activity and reversal of multidrug resistance in vitro by rosemary extract. Eur
J Cancer. 1999 Oct;35(10):15415.
26. Chen J, Li L, Su J, Li B, Zhang X, Chen T. Proteomic analysis of G2/M arrest
triggered by natural Borneol/Curcumin in HepG2 cells, the importance of
the reactive oxygen species-p53 pathway. J Agric Food Chem. 2015 Jul 22;
63(28):64409.
27. Lim TG, Lee SY, Huang Z, Lim DY, Chen H, Jung SK, et al. Curcumin
suppresses proliferation of colon cancer cells by targeting CDK2. Cancer
Prev Res (Phila). 2014 Apr;7(4):46674.
28. Baharuddin P, Satar N, Fakiruddin KS, Zakaria N, Lim MN, Yusoff NM, et al.
Curcumin improves the efficacy of cisplatin by targeting cancer stem-like
cells through p21 and cyclin D1-mediated tumour cell inhibition in non-
small cell lung cancer cell lines. Oncol Rep. 2016 Jan;35(1):1325.
29. Kumar D, Basu S, Parija L, Rout D, Manna S, Dandapat J, Debata PR.
Curcumin and Ellagic acid synergistically induce ROS generation, DNA
damage, p53 accumulation and apoptosis in HeLa cervical carcinoma cells.
Biomed Pharmacother. 2016 July;81:317.
30. Mosieniak G, Sliwinska MA, Przybylska D, Grabowska W, Sunderland P,
Bielak-Zmijewska A, Sikora E. Curcumin-treated cancer cells show mitotic
disturbances leading to growth arrest and induction of senescence
phenotype. Int J Biochem Cell Biol. 2016 May;74:3343.
31. Tong L, Chuang CC, Wu S, Zuo L. Reactive oxygen species in redox cancer
therapy. Cancer Lett. 2015 Oct 10;367(1):1825.
32. Deng Y, Verron E, Rohanizadeh R. Molecular mechanisms of anti-metastatic
activity of Curcumin. Anticancer Res. 2016 Nov;36(11):563947.
33. Shehzad A, Lee YS. Molecular mechanisms of curcumin action: signal
transduction. Biofactors. 2013 Jan-Feb;39(1):2736.
34. Khuda-Bukhsh AR, Das S, Saha SK. Molecular approaches toward targeted
cancer prevention with some food plants and their products: inflammatory
and other signal pathways. Nutr Cancer. 2014;66(2):194205.
35. Moore J, Yousef M, Tsiani E. Anticancer Effects of Rosemary (Rosmarinus
officinalis L.) Extract and Rosemary Extract Polyphenols. Nutrients. 2016 Nov
17;8(11). pii: E731.
36. Tai J, Cheung S, Wu M, Hasman D. Antiproliferation effect of rosemary
(Rosmarinus Officinalis) on human ovarian cancer cells in vitro.
Phytomedicine. 2012;19:43643.
37. Tsai CW, Lin CY, Lin HH, Chen JH. Carnosic acid, a rosemary phenolic
compound, induces apoptosis through reactive oxygen species-mediated
p38 activation in human neuroblastoma IMR-32 cells. Neurochem Res. 2011;
36:244251.
38. Fossey SL, Bear MD, Lin J, Li C, Schwartz EB, Li PK, et al. The novel curcumin
analog FLLL32 decreases STAT3 DNA binding activity and expression, and
induces apoptosis in osteosarcoma cell lines. BMC Cancer. 2011 Mar 28;11:112.
39. Verheij M, Ruiter GA, Zerp SF, van Blitterswijk WJ, Fuks Z, Haimovitz-
Friedman A, Bartelink H. Role of the stress-activated protein kinase (SAPK/
JNK) signaling pathway in radiation-induced apoptosis. Radiother Oncol.
1998 Jun;47(3):22532.
40. Cul J, Zhang M, Zhang YQ, JNK XZH. Pathway: diseases and therapeutic
potential. Acta Pharmacol Sin. 2007 May;28(5):6018.
41. Bogoyevitch MA, Ngoei KR, Zhao TT, Yeap YY, Ng DC. CpJun N-terminal
kinase (JNK) signaling: recent advances and challenges. Biochim Biophys
Acta. 2010 Mar;1804(3):46375.
42. Basu S, Kolesnick R. Stress signals for apoptosis: ceramide and c-Jun kinase.
Oncogene. 1998 Dec 24;17(25):327785.
43. Johnson CR, Jiffar T, Fischer UM, Ruvolo PP, Jarvis WD. Requirement for
SAPK-Jnk signaling in the induction of apoptosis by ribosomal stress in REH
lymphoid leukemia cells. Leukemia. 2003 Nov;17(11):21408.
44. Ventura JJ, Hübner A, Zhang C, Flavell RA, Shokat KM, Davis RJ. Chemical
genetic analysis of the time course of signal transduction by JNK. Mol Cell.
2006 Mar 3;21(5):70110.
45. Li J, Xiang S, Zhang Q, Wu J, Tang Q, Zhou J, et al. Combination of
curcumin and bicalutamide enhanced the growth inhibition of androgen-
independent prostate cancer cells through SAPK/JNK and MEK/ERK1/2-
mediated targeting of NF-κB/p65 and MUC1-C. J Exp Clin Cancer Res. 2015
May 15;34:46.
46. Kuwabara M, Takahashi K, Inanami O. Induction of apoptosis through the
activation of SAPK/JNK followed by the expression of death receptor Fas in
X-irradiated cells. J Radiat Res. 2003 Sep;44(3):20139.
47. Davies C, Tournier C. Exploring the function of the JNK (c-Jun N-terminal
kinase) signaling pathway in physiological and pathological processes to
design novel therapeutic strategies. Biochem Soc Trans. 2012 Feb;40(1):859.
48. Kim J, Freeman MRJNK. SAPK mediates doxorubicin-induced differentiation
and apoptosis in MCF-7 breast cancer cells. Breast Cancer Res Treat. 2003
Jun;79(3):3218.
49. Lee LF, Li G, Templeton DJ, Ting JP. Paclitaxel (Taxol)-induced gene
expression and cell death are both mediated by the activation of c-Jun
NH2-terminal kinase (JNK/SAPK). J Biol Chem. 1998 Oct 23;273(43):2825360.
50. Kanai M, Otsuka Y, Otsuka K, Sato M, Nishimura T, Mori Y, Kawaguchi M,
Hatano E, Kodama Y, Matsumoto S, Murakami Y, Imaizumi A, Chiba T,
Nishihira J, Shibata H. A phase I study investigating the safety and
pharmacokinetics of highly bioavailable curcumin (Theracurmin) in cancer
patients. Cancer Chemother Pharmacol. 2013;71 Jun,(6):152130.
51. Song Y, Yan H, Chen J, Wang Y, Jiang Y, Tu P. Characterization of in vitro
and in vivo metabolites of carnosic acid, a natural antioxidant, by high
performance liquid chromatography coupled with tandem mass
spectrometry. J Pharm Biomed Anal. 2014;89:18396.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Levine et al. BMC Veterinary Research (2017) 13:388 Page 12 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Synergistic interactions between the compounds of medicinal plant extracts are a vital part of their therapeutic efficacy [34,35]. Levine et al. [36] and Mohamed et al. [37] have demonstrated that plants such as R. officinalis that have potent antiinflammatory and antioxidant effect can positively support the action of Curcuma longa due to a greater increase in intracellular curcumin accumulation when the combination treatment was used. However, whether this synergic interaction will be effective in neurological disorders in animal model has not been tested to the best of our knowledge. ...
... As a result, inhibiting acetylcholine esterase causes an increase in acetylcholine in the brain, which reduces plaque formation [60]. These results are in accordance with Levine et al. [36] and Mohamed et al. [37] who reported that, combination of R. officinalis and C. longa is more effective than C. longa extract alone in improving the outcome due to a greater increase in intracellular curcumin accumulation when the combination treatment was used. ...
Article
Full-text available
Background: Neurological disorders remain a global health challenge. Chronic alcohol consumption induces damage to the brain that can cause various forms of dementia. An abundance of acetaldehyde is produced by excessive alcohol consumption and accumulates in the body to induce oxidative stress, apoptosis, and inflammation in neuronal cells, which results in brain damage leading to memory loss. This study is aimed at investigating the effect of mixed ethanol extract of Curcuma longa rhizome and Rosmarinus officinalis leaf on oxidative stress and proinflammatory markers in the brain of alcohol- treated male Wistar rats. Methods: Twenty-five male Wistar rats were divided into five groups: control, mixed extract only (600 mg/kg), alcohol only (15 ml/kg of 40% ethanol), and mixed extract + alcohol groups at 300 mg/kg and 600 mg/kg doses. Treatments were administered orally for 21 days. Brain homogenates were analyzed for superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), reduced glutathione (GSH), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and AChE activity. Histopathological evaluations assessed neuroprotective effects. Results: Alcohol administration significantly decreased SOD (16.2 ± 1.3 U/mg protein), catalase (12.8 ± 0.9 U/mg protein), GPx (10.7 ± 0.8 U/mg protein), and GSH (9.2 ± 0.6 mg/g protein) compared to the control group (32.8 ± 2.2 U/mg, 23.5 ± 1.7 U/mg, 18.9 ± 1.2 U/mg, and 18.3 ± 1.1 mg/g, respectively; p<0.05). MDA levels increased to 7.6 ± 0.3 nmol/mg protein compared to 2.8 ± 0.2 nmol/mg in the control group. Treatment with 600 mg/kg of mixed extracts significantly restored SOD (31.5 ± 2.4 U/mg), catalase (22.6 ± 1.4 U/mg), GPx (17.8 ± 1.0 U/mg), GSH (17.4 ± 1.2 mg/g), and reduced MDA (3.2 ± 0.3 nmol/mg; p<0.05). TNF-α and IL-1β levels were elevated in alcohol-only rats (123.5 ± 4.3 pg/mL and 85.6 ± 3.2 pg/mL) compared to controls (68.3 ± 2.8 pg/mL and 43.2 ± 1.7 pg/mL, p<0.05) but were significantly reduced with mixed extract treatment. Histopathological analyses confirmed reduced neurodegeneration and restoration of normal brain architecture in treated groups. Conclusion: The mixed extract demonstrated significant neuroprotective effects by mitigating oxidative stress, inflammation, and neuronal damage in alcohol-treated rats. These findings suggest its potential as a therapeutic candidate for preventing alcohol-induced dementia.
... Caspase 3/7 activation and Annexin V staining demonstrated that TE had a higher effect on triggering cell apoptosis and a similar result was obtained from the combination treatment with just half the concentration of each extract. 48 To ascertain which stage of apoptosis the rosemary extract mostly causes, the Annexin V assay and caspase 3/7 assays were performed after the extract was administered. It was discovered that A549 cells treated with 5 µg/ml rosemary extract induced apoptosis and necrosis also reduced cell viability. ...
Article
Full-text available
Objective: Apoptosis resistance and increased proliferation rates are characteristics of cancer cells. The anticancer properties of rosemary (Rosmarinus officinalis L.) extract (RE) have been demonstrated in a small number of in vivo and in vitro animal studies; however, no research has investigated the role of RE in human non-small cell lung cancer (NSCLC) A549 and H1299 cells, and its underlying mechanism of action remains unknown. In the current study, we examined the effects of RE on human non-small cell lung cancer cell proliferation, survival, and apoptosis. Methods: Human non-small cell lung cancer (NSCLC) A549 and H1299 were incubated with (2.5 µg/ml, 5 µg/ml, 7.5 µg/ml, 10 µg/ml, and 12.5 µg/ml) doses of RE for 12, 24, and 48 hours. MTT, Annexin V-PI, and caspase 3/7 assay kit were performed to detect the cell viability and apoptosis. Results: According to MTT analysis, the viability of A549 and H1299 human lung cancer cells was reduced by approximately 49.74% and 47.76%, respectively, for 24 hours by treatment with a dose of 5 µg/ml RE. The results of Annexin V-PI staining and Caspase 3/7 activation showed that RE had a greater effect on inducing cell death. Conclusion: In conclusion, we can say that rosemary extract has both apoptotic and antiproliferative properties on human lung cancer cells. We might propose that additional investigation is necessary to ascertain the therapeutic impacts of rosemary extract.
... [84] In one study, the synergistic effect of turmeric root and rosemary leaf extract on the cellular effect of a canine neoplastic cell line (CMT-12 mammary carcinoma, Cc2 mastocytoma, and D-17 osteosarcoma) resulted in the activation of caspase 3/7 and apoptosis of cell lines. [85] In addition to turmeric, phytochemicals from ginger and berberis are used as anticancer phytomedicines in CBCs. The active ingredients of wild ginger, that is, zerumbone (ZER), are used for antioxidant, antiproliferative, anticancerous, and antiinflammatory properties. ...
Article
Full-text available
Breast cancer in canines is one of the leading causes of death globally due to client misinterpretation and improper diagnosis and treatment. In past centuries, the diagnosis and treatment of breast carcinoma in dogs followed conventional techniques adopted from human oncology. However, with increasing demand and scientific advancements in the upcoming future, there is an emerging necessity to modernize the diagnostic and treatments in canine breast cancer (CBC) patients. This review explores recent advances in diagnostic techniques and novel therapeutic approaches such as adjuvant‐based targeted therapy, nanomaterial therapy, immune‐based therapy, adoptive cell therapy, tumor vaccine, oncolytic virotherapy, and the use of noncoding RNAs in CBCs. In addition, the review discusses the healthcare policies aimed at improving diagnostic and therapeutic efficacy and future directions for translation from human oncology into veterinary oncology. By adopting these modern strategies, the quality of care can be significantly enhanced by translating them into practical applications with better outcomes and improved survival rates for canine patients.
... Piroxicam, in particular, has been evaluated in combination with chemotherapy for a number of canine cancers, including lymphoma, soft tissue sarcoma, squamous cell carcinoma, hemangiosarcoma, oral malignant melanoma, and osteosarcoma [30][31][32][33][34][35]. Curcumin has been shown to decrease cell proliferation in canine cell lines, including osteosarcoma, melanoma, mammary carcinoma, and mast cell tumors [36,37]. ...
Article
Full-text available
Simple Summary The renin–angiotensin system (RAS) is a well-known hormonal system that controls blood pressure and blood volume. Recent work has suggested that it also plays a role in cancer. Various studies in humans and animals have investigated blocking different parts of the RAS; many, but not all, studies have shown decreased tumor growth and spread. The RAS consists of various bypass pathways, which may explain the lack of a response to treatment in some studies. A treatment has been developed to simultaneously block multiple parts of the RAS (multimodal blockade). This treatment has been used in one clinical trial in humans with glioblastoma (a brain tumor) and another trial in cats with squamous cell carcinoma (skin cancer). The aim of this study was to assess the safety of multimodal blockade of the RAS in dogs with two different types of cancer (osteosarcoma and oral malignant melanoma). Two mild adverse effects were observed: one dog developed intermittent vomiting; another dog had a mildly increased serum SDMA concentration (a kidney function biomarker) at one time point. This sets the stage for conducting a larger-scale trial to assess the efficacy of this treatment for cancer in dogs. Abstract The renin–angiotensin system (RAS) is increasingly being recognized to play a role in the tumor microenvironment, promoting tumor growth. Studies blocking a single part of the RAS have shown mixed results, possibly due to the existence of different bypass pathways and redundancy within the RAS. As such, multimodal blockade of the RAS has been developed to exert more complete inhibition of the RAS. The aim of the present study was to assess the safety of multimodal RAS blockade in dogs. Five dogs (four with appendicular osteosarcoma, one with oral malignant melanoma) were treated with atenolol, benazepril, curcumin, meloxicam, and metformin. The dogs underwent clinical examination, blood pressure measurement, and hematology and serum biochemistry tests performed at 0, 1, 3, 6, 9, and 12 weeks, then every 3 months thereafter. End-of-life decisions were made by the owners. None of the dogs developed hypotension. One dog had intermittent vomiting during the 64 weeks it was on the trial. One dog had a one-off increase in serum SDMA(symmetrical dimethylarginine) concentration. Dogs were euthanized at weeks 3 (osteosarcoma), 10 (osteosarcoma), 17 (osteosarcoma), and 26 (oral malignant melanoma), and one dog was still alive at the end of the trial at 64 weeks (osteosarcoma). This is the first assessment of multimodal blockade of the RAS in dogs, and the results suggest it causes only mild adverse effects in some animals. The efficacy of the treatment was not assessed due to the small number of dogs. This pilot study allows for future larger studies assessing multimodal RAS blockade for the treatment of canine cancer.
... But still some alternative strategies are needed to be used, such as combination chemotherapy to synergize the effect and decrease the chance of tumors, reduce the cancer symptoms and death numbers, and prolong the life span [43]. Furthermore, curcumin and its analogs are being tested in combination of FDA-approved drugs, clinical trial drugs [44,45], curcumin fused or conjugated by spacer to other natural products such as carnosic acid [46], aloe vera [46], docosahexaenoic acid [46], trans-farnesylthiosalicylic acid [47], piperine [48], retinoic acid [49], α-tomatine [49], several carotenoids [50], ursolic acid [50], bicalutamide [51], quercetin [52], epigallocatechin-3-gallate, curcurbitacin B [53], rosemary [54], silymarin [55], vincristine, etoposide, carboplatin [56], etc.) for regulating various signaling pathways such as inhibiting the Bcl-2 gene [57], COX-2 [57], MMP-9/2 [58], metastatic proteins, suppressing the NF-KB [58,59], TNF-α and its regulated gene, modulate AMPK activity [58], increasing phosphorylation of ERK1/2 [60], and SAPK/JNK [60], which reducing the p65 [60], AKT/IKKa MUC1-C pathways [60], suppression of β-3 gene expression [60], suppressing the activation of JAK/STAT3/IL-8 signaling pathway [61], suppression of STAT3 [59,61] pathway, apoptosis induction via caspase-3/8 [59], Bax upregulation [59], downregulation of VEGF, VEGFR-2 expressions, upregulation of PTEN, p53, BRCA1, BRCA2, and ERCC [62]. The synergistic effects of this combination chemotherapy revealed that the antitumor effect is far greater than the effect of the native form of curcumin, showing more stability, better bioavailability, and pharmacokinetic properties [17,18]. ...
Book
Curcumin is always at the forefront of prominent plant secondary metabolite that has been inspected for its ability to show pleiotropic pharmacological activities. It has proved its worth in targeting multiple proteins involved in various signaling and metabolic pathways at preclinical stages. On pinpoint, it gains significant importance for its usage as an anticancer agent in cancer therapies viz, breast, bone, lung, hepatic, gastric, leukemia, colon, ovarian, prostate, melanoma, and pancreatic cancers. Despite the plethora of importance in cancer therapy, snag in therapeutic efficacy, bioavailability, and pharmacokinetics are the checkpoints in the passage of curcumin to be super curcumin medicinal contender. Present chapter is a solemn effort toward the transitory explanation of synthetic approaches used to overcome the checkpoints and revamped potency of curcumin. Fine importance has been given to novel curcumin-based structural analogs endowed with anticancer potential.
... In order to find the difference in the S. mutans colonies before and after bonding, paired t-test was used and one-way analysis of variance (ANOVA) was used to find the difference in the S. mutans colonies sustained activation and signaling of pathways that were critical to promote apoptosis of neoplastic cells. 26 Numerous types of research have been conducted to analyze the effects of curcumin and rosemary extracts in mouthwashes individually but not together. Hence, studying their synergistic effects on the plaque microflora was imperative. ...
... The number of dead cells treated with different extracts via the apoptosis mechanism was determined by flow cytometric assay kit (BD bioscience, NJ, USA) at 488 (excitation wavelength) and 617 nm (emission wavelength) [23,24]. To perform a flow cytometry test, we added 2 × 10 5 cells to each 6-cell well plate. ...
Article
Full-text available
Background The genus Artemisia of the Asteraceae family has different species that are used in the treatment of a wide range of diseases, including cancers due to the presence of valuable compounds and important medicinal properties. Various studies on the anti-tumor effect of different species of Artemisia have proven the cytotoxic properties of these plants in cancer treatment, and several anti-cancer compounds of this genus have been purified. Objective The objective of this study was to investigate the cytotoxicity and related mortality mechanisms of Artemisia marschalliana essential oil and extracts. Methods The essential oil and various extracts of Artemisia marschalliana were elicited using a Soxhlet extractor. Anti-cancer to anti-proliferative activity as MTT assay is measuring cancerous and non-cancerous cell viability. In the next step, the strongest extract fractions were obtained by using the vacuum liquid chromatography method. Flow cytometry was applied to identify the mechanism of cell death, and a Real-time polymerase chain reaction test of apoptosis genes, which encode apoptosis-regulating proteins, was measured to confirm the flow cytometry results. Results The strongest extract belonged to dichloromethane extract 60% fraction of the extract on breast cancer cells and 80% fraction on liposarcoma cancer cells showed the most cytotoxicity within 48 h, while, the fractions did not notable cytotoxicity of non-cancerous cells cell. Flow cytometry analysis illustrated the mentioned extract and its fractions kill cancer cell lines through the apoptosis mechanism. Our findings confirmed the flow cytometry results. In addition, the essential oil of Artemisia marschalliana showed a considerable cytotoxic property. Conclusion Dichloromethane extract of Artemisia marschalliana shoot and its 60 and 80% fraction selectively inhibited the growth of cancer cells by inducing the apoptosis mechanism. Regarding obtained results, 60 and 80% fractions of dichloromethane extract can be a good candidate for future studies in the field of identification and separation of pure cytotoxic compounds.
Article
Full-text available
Background and Objective: Plant crude extracts are analyzed to evaluate their potency as a starting material for preparation of eco-friendly and natural product-based drugs and substances for their potential therapeutic benefits. This study investigated the antileishmanial, anti-cancer, anti-inflammatory, cytotoxic and antibacterial activities of Ricinodendron heudelotii seed extracts. Materials and Methods: The seeds were collected, extracted and fractionated using hexane, DCM and ethyl acetate. The 96 microtitre plate bioassay, MTT HeLa cells, Brine shrimp assay and MABA assays were employed for antileishmanial, anti-cancer, anti-inflammatory and cytotoxicity activities of Ricinodendron heudelotii seed extract respectively. Results: The antileishmanial activity showed that all the extracts inhibited in vitro growth of Leishmania tropica promastigotes at the experimental dose with an IC 50 of 3.16, 3.49 and 3.34 µg/mL for hexane, ethyl acetate and DCM fractions, respectively. Hexane fraction showed the highest inhibition (60%) of ROS with IC 50 of 32.4 The cytotoxic and in vitro anticancer activity of the extracts against human HeLa cervical cancer cell lines at 30 µg/mL showed that hexane, ethyl acetate and DCM fractions had 64.5, 59.9 and 15.6% cell growth inhibition, respectively. The cytotoxic activity using Brine shrimp (Artemia salina) lethality bioassay showed that hexane fraction had cytotoxicity at the highest concentration with LC 50 of 1548.82 antibacterial activity revealed that ethyl acetate fraction had a moderate inhibition against Staphylococcus aureus. Conclusion: This study revealed that Ricinodendron heudelotii seeds had in vitro antileishmanial, anti-cancer, anti-inflammatory, cytotoxic and antibacterial activities. The study thus provides evidence for the biological activities of Ricinodendron heudelotii seeds as the extracts showed varying potency levels for all investigated activities.
Article
Full-text available
Estudos recentes vêm demonstrando a eficácia de produtos naturais como agentes terapêuticos anti-tumorais, avaliando-se o mecanismo de ação frente a diferentes células neoplásicas na medicina humana e veterinária. Dentre eles, a cúrcuma, conhecida como açafrão, destaca-se por suas propriedades anti-inflamatórias, anti-proliferativas e pró-apoptóticas. A curcumina é o principal composto ativo da cúrcuma e possui efeito antiproliferativo e pró apoptótico satisfatório, já descrito em diferentes linhagens neoplásicas. Sua eficácia, quando associada a diversos quimioterápicos, tem sido avaliada na busca de aumentar seus efeitos e biodisponibilidade. No trato urinário, a bexiga é o órgão mais acometido por neoplasias, sendo 80% destas malignas, incluindo o carcinoma urotelial canino (CUB), que corresponde a cerca de 2% de todos os tumores malignos do trato urinário inferior e superior. Comumente apresentada como uma neoplasia invasiva de alto grau, localizada na região do trígono e metastática. Seu tratamento inclui excisão cirúrgica, quimioterapia, emprego de inibidores de cicloxigenase (COX), radioterapia, ou a combinação desses. Entretanto, a intervenção cirúrgica muitas vezes é inviabilizada pela localização da lesão. Além disso, estudos recentes demonstraram que esta neoplasia possui semelhança com CUB humano em diferentes aspectos, servindo como um modelo natural na busca de novas estratégias terapêuticas e diagnósticas. Abordamos a respeito do uso de curcumina na terapêutica do carcinoma de bexiga, buscando estimular o desenvolvimento de mais pesquisas acerca dessa temática.
Article
Full-text available
Cancer cells display enhanced growth rates and a resistance to apoptosis. The ability of cancer cells to evade homeostasis and proliferate uncontrollably while avoiding programmed cell death/apoptosis is acquired through mutations to key signaling molecules,which regulate pathways involved in cell proliferation and survival. Compounds of plant origin,including food components,have attracted scientific attention for use as agents for cancer prevention and treatment. The exploration into natural products offers great opportunity to evaluate new anticancer agents as well as understand novel and potentially relevant mechanisms of action. Rosemary extract has been reported to have antioxidant,anti-inflammatory,antidiabetic and anticancer properties. Rosemary extract contains many polyphenols with carnosic acid and rosmarinic acid found in highest concentrations. The present review summarizes the existing in vitro and in vivo studies focusing on the anticancer effects of rosemary extract and the rosemary extract polyphenols carnosic acid and rosmarinic acid,and their effects on key signaling molecules.
Article
Full-text available
Cancer is the leading cause of death worldwide. Although cancer occurs as a localized disease, its morbidity and mortality rates remain high due to the ability of cancer cells to break-off from the primary tumor and spread to distant organs. Currently, chemotherapy is the main treatment for cancer; however, the increase in proportion of drug-resistant cancer cells and unpleasant side-effects of chemotherapy are still the major challenges in cancer therapy. Curcumin is a natural polyphenol compound and the main bioactive constituent of Indian spice turmeric, widely used in Indian and Chinese medicines. Curcumin has well-known therapeutic actions, including anti-inflammatory, anti-microbial, anti-oxidant and anti-cancer properties. Curcumin induces cancer cell apoptosis through regulating various signaling pathways and arresting tumor cell cycle. Curcumin's therapeutic/preventative actions on metastatic cancers have not been yet fully understood and studied. The present review explores the potential anti-metastatic mechanisms of curcumin, including inhibition of transcription factors and their signaling pathways (e.g., NF-κB, ApP-1 and STAT3), inflammatory cytokines (e.g., CXCL1, CXCL2, IL-6, IL-8), multiple proteases (e.g., uPA, MMPs), multiple protein kinases (e.g., MAPKs, FAK), regulation of miRNAs (e.g., miR21, miR181b) and heat shock proteins (HLJ1). In addition, possible synergistic actions of combination therapy of curcumin with current chemotherapies are discussed in this review.
Article
Full-text available
Polyphenols, found abundantly in plants, display many anticarcinogenic properties including their inhibitory effects on cancer cell proliferation, tumor growth, angiogenesis, metastasis, and inflammation as well as inducing apoptosis. In addition, they can modulate immune system response and protect normal cells against free radicals damage. Most investigations on anticancer mechanisms of polyphenols were conducted with individual compounds. However, several studies, including ours, have indicated that anti-cancer efficacy and scope of action can be further enhanced by combining them synergistically with chemically similar or different compounds. While most studies investigated the anti-cancer effects of combinations of two or three compounds, we used more comprehensive mixtures of specific polyphenols and mixtures of polyphenols with vitamins, amino acids and other micronutrients. The mixture containing quercetin, curcumin, green tea, cruciferex, and resveratrol (PB) demonstrated significant inhibition of the growth of Fanconi anemia head and neck squamous cell carcinoma and dose-dependent inhibition of cell proliferation, matrix metalloproteinase (MMP)-2 and -9 secretion, cell migration and invasion through Matrigel. PB was found effective in inhibition of fibrosarcoma HT-1080 and melanoma A2058 cell proliferation, MMP-2 and -9 expression, invasion through Matrigel and inducing apoptosis, important parameters for cancer prevention. A combination of polyphenols (quercetin and green tea extract) with vitamin C, amino acids and other micronutrients (EPQ) demonstrated significant suppression of ovarian cancer ES-2 xenograft tumor growth and suppression of ovarian tumor growth and lung metastasis from IP injection of ovarian cancer A-2780 cells. The EPQ mixture without quercetin (NM) also has shown potent anticancer activity in vivo and in vitro in a few dozen cancer cell lines by inhibiting tumor growth and metastasis, MMP-2 and -9 secretion, invasion, angiogenesis, and cell growth as well as induction of apoptosis. The presence of vitamin C, amino acids and other micronutrients could enhance inhibitory effect of epigallocatechin gallate (EGCG) on secretion of MMPs. In addition, enrichment of NM with quercetin (EPQ mix) enhanced anticancer activity of NM in vivo. In conclusion, polyphenols, especially in combination with other polyphenols or micronutrients, have been shown to be effective against multiple targets in cancer development and progression, and should be considered as safe and effective approaches in cancer prevention and therapy.
Article
Full-text available
Background Adjunctive use of nutraceuticals in human cancer has shown promise, but little work has been done in canine neoplasia. Previous human research has shown that polyphenols and carotenoids can target multiple pathways in vitro and in vivo. These compounds could synergize or antagonize with currently used chemotherapies, either increasing or decreasing the effectiveness of these drugs. Considering the routine and well controlled feeding practices of most dogs, the use of nutraceuticals incorporated into pet food is attractive, pending proof that the extracts are able to improve remission rates. The aim of this study was to examine five feed ingredients for antiproliferative effects, as well as the interaction with toceranib phosphate and doxorubicin hydrochloride, when treating canine neoplastic cell lines in vitro. Results Screening using MTT proliferation assays showed that green tea, turmeric, and rosemary extracts were the most effective. Turmeric extract (TE) was the most potent and exhibited synergy with a rosemary extract (RE) at concentrations from 1 to 25 μg mL−1. This combination had an additive or synergistic effect with chemotherapeutic agents at selected concentrations within each cell line. No significant effects on cell viability were observed when the combination therapy was used with normal primary cells. Conclusions The use of turmeric and rosemary extracts in combination may be worthwhile to investigate in the pre-clinical and clinical neoplastic considering there are no negative effects on traditional chemotherapy treatment. Further studies into the pharmacokinetics and mechanisms of action of these extracts should be investigated.
Article
Full-text available
Natural compounds such as curcumin have the ability to enhance the therapeutic effectiveness of common chemotherapy agents through cancer stem‑like cell (CSC) sensitisation. In the present study, we showed that curcumin enhanced the sensitivity of the double‑positive (CD166+/EpCAM+) CSC subpopulation in non‑small cell lung cancer (NSCLC) cell lines (A549 and H2170) to cisplatin‑induced apoptosis and inhibition of metastasis. Our results revealed that initial exposure of NSCLC cell lines to curcumin (10‑40 µM) markedly reduced the percentage of viability to an average of ~51 and ~54% compared to treatment with low dose cisplatin (3 µM) with only 94 and 86% in both the A549 and H2170 cells. Moreover, sensitisation of NSCLC cell lines to curcumin through combined treatment enhanced the single effect induced by low dose cisplatin on the apoptosis of the double‑positive CSC subpopulation by 18 and 20% in the A549 and H2170 cells, respectively. Furthermore, we found that curcumin enhanced the inhibitory effects of cisplatin on the highly migratory CD166+/EpCAM+ subpopulation, marked by a reduction in cell migration to 9 and 21% in the A549 and H2170 cells, respectively, indicating that curcumin may increase the sensitivity of CSCs to cisplatin‑induced migratory inhibition. We also observed that the mRNA expression of cyclin D1 was downregulated, while a substantial increased in p21 expression was noted, followed by Apaf1 and caspase‑9 activation in the double‑positive (CD166+/EpCAM+) CSC subpopulation of A549 cells, suggested that the combined treatments induced cell cycle arrest, therefore triggering CSC growth inhibition via the intrinsic apoptotic pathway. In conclusion, we provided novel evidence of the previously unknown therapeutic effects of curcumin, either alone or in combination with cisplatin on the inhibition of the CD166+/EpCAM+ subpopulation of NSCLC cell lines. This finding demonstrated the potential therapeutic approach of using curcumin that may enhance the effects of cisplatin by targeting the CSC subpopulation in NSCLC.
Article
Full-text available
Quercetin, catechins and proanthocyanidins are flavonoids that are prominently featured in foodstuffs and dietary supplements, and may possess anti-carcinogenic activity. Glioblastoma multiforme is the most dangerous form of glioma, a malignancy of the brain connective tissue. This review assesses molecular structures of these flavonoids, their importance as components of diet and dietary supplements, their bioavailability and ability to cross the blood-brain barrier, their reported beneficial health effects, and their effects on non-malignant glial as well as glioblastoma tumor cells. The reviewed flavonoids appear to protect glial cells via reduction of oxidative stress, while some also attenuate glutamate-induced excitotoxicity and reduce neuroinflammation. Most of the reviewed flavonoids inhibit proliferation of glioblastoma cells and induce their death. Moreover, some of them inhibit pro-oncogene signaling pathways and intensify the effect of conventional anti-cancer therapies. However, most of these anti-glioblastoma effects have only been observed in vitro or in animal models. Due to limited ability of the reviewed flavonoids to access the brain, their normal dietary intake is likely insufficient to produce significant anti-cancer effects in this organ, and supplementation is needed.
Article
The role of reactive oxygen species (ROS) in cancer cells has been intensively studied for the past two decades. Cancer cells mostly have higher basal ROS levels than their normal counterparts. The induction of ROS has been shown to be associated with cancer development, metastasis, progression, and survival. Various therapeutic approaches targeting intracellular ROS levels have yielded mixed results. As widely accepted dietary supplements, antioxidants demonstrate both ROS scavenging ability and anti-cancer characteristics. However, antioxidants may not always be safe to use since excessive intake of antioxidants could lead to serious health concerns. In this review, we have evaluated the production and scavenging systems of ROS in cells, as well as the beneficial and harmful roles of ROS in cancer cells. We also examine the effect of antioxidants in cancer treatment, the effect of combined treatment of antioxidants with traditional cancer therapies, and the side effects of excessive antioxidant intake. Copyright © 2015. Published by Elsevier Ireland Ltd.
Article
Curcumin (Cur), an active ingredient from the rhizome of the plant, Curcuma longa, has wide anticancer activities. However, due to its poor solubility and hence poor absorption, Cur has limited clinical applications. It is therefore important to develop an effective method to improve its absorption. Natural borneol (NB), a terpene and bicyclic organic compound, has been extensively used as a food additive and our previous studies show that it can improve the uptake of Cur in cancer cells. However, the anticancer mechanism of NB/Cur remains unclear. In this study, the effects of NB/Cur on HepG2 cells were investigated by proteomic analysis. The results showed that 32 differentially expressed proteins identified by MALDI-TOF-MS were significantly changed after NB/Cur treated HepG2 cells for 24 h. Moreover, 17 proteins increased and 12 proteins decreased significantly. Biological progress categorization demonstrated that the identified proteins were mainly associated with cell cycle and apoptosis (28.1%). Subcellular location categorization exhibited that the identified proteins were mainly located in nucleus (28.1%) and mitochondrion (21.9%). Among of all proteins, we selected three differentially proteins (hnRNPC1/C2, NPM and PSMA5), which were associated with the p53 pathway. Down-regulation of hnRNPC1/C2 and NPM contributed to the enhancement of phosphorylated p53. Activated p53 and down-regulation of PSMA5 resulted in an increase in p21 protein. Further studies showed that NB/Cur induced reactive oxygen species (ROS) generation, indicating that ROS might be upstream of the G2/M arrest signaling pathway. In summary, the results exhibited that the whole proteomic response of HepG2 cells to NB/Cur, which might lead to a better understanding of its underlying anticancer mechanisms.