ArticlePDF Available

Dichloromethane fractions of Calystegia soldanella induce S‑phase arrest and apoptosis in HT‑29 human colorectal cancer cells

Authors:

Abstract and Figures

Calystegia soldanella is a halophyte and a perennial herb that grows on coastal sand dunes worldwide. Extracts from this plant have been previously revealed to have a variety of bioactive properties in humans. However, their effects on colorectal cancer cells remain poorly understood. In the present study, the potential biological activity of C. soldanella extracts in the colorectal cancer cell line HT‑29 was examined. First, five solvent fractions [n‑hexane, dichloromethane (DCM), ethyl acetate, n‑butanol and water] were obtained from the crude extracts of C. soldanella through an organic solvent extraction method. In particular, the DCM fraction was demonstrated to exert marked dose‑ and time‑dependent inhibitory effects according to results from the cell viability assay. Data obtained from the apoptosis assay suggested that the inhibition of HT‑29 cell viability induced by DCM treatment was attributed to increased apoptosis. The apoptotic rate was markedly increased in a dose‑dependent manner, which was associated with the protein expression levels of apoptosis‑related proteins, including increased Fas, Bad and Bax, and decreased pro‑caspase‑8, Bcl‑2, Bcl‑xL, pro‑caspase‑9, pro‑caspase‑7 and pro‑caspase‑3. A mitochondrial membrane potential assay demonstrated that more cells became depolarized and the extent of cytochrome c release was markedly increased in a dose‑dependent manner in HT‑29 cells treated with DCM. In addition, cell cycle analysis confirmed S‑phase arrest following DCM fraction treatment, which was associated with decreased protein expression levels of cell cycle‑related proteins, such as cyclin A, CDK2, cell division cycle 25 A and cyclin dependent kinase inhibitor 1. Based on these results, the present study suggested that the DCM fraction of the C. soldanella extract can inhibit HT‑29 cell viability whilst inducing apoptosis through mitochondrial membrane potential regulation and S‑phase arrest. These results also suggested that the DCM fraction has potential anticancer activity in HT‑29 colorectal cells. Further research on the composition of the DCM fraction is warranted.
Content may be subject to copyright.
MOLECULAR MEDICINE REPORTS 25: 60, 2022
Abstract. Calystegia soldanella is a halophyte and a peren-
nial herb that grows on coastal sand dunes worldwide.
Extracts from this plant have been previously revealed to
have a variety of bioactive properties in humans. However,
their effects on colorectal cancer cells remain poorly under-
stood. In the present study, the potential biological activity of
C. soldanella extracts in the colorectal cancer cell line HT-29
was examined. First, ve solvent fractions [n‑hexane, dichlo-
romethane (DCM), ethyl acetate, n-butanol and water] were
obtained from the crude extracts of C. soldanella through
an organic solvent extraction method. In particular, the
DCM fraction was demonstrated to exert marked dose- and
time-dependent inhibitory effects according to results from
the cell viability assay. Data obtained from the apoptosis assay
suggested that the inhibition of HT-29 cell viability induced
by DCM treatment was attributed to increased apoptosis. The
apoptotic rate was markedly increased in a dose-dependent
manner, which was associated with the protein expression
levels of apoptosis-related proteins, including increased Fas,
Bad and Bax, and decreased pro-caspase-8, Bcl-2, Bcl-xL,
pro-caspase-9, pro-caspase-7 and pro-caspase-3. A mitochon-
drial membrane potential assay demonstrated that more cells
became depolarized and the extent of cytochrome c release
was markedly increased in a dose-dependent manner in
HT-29 cells treated with DCM. In addition, cell cycle analysis
conrmed S‑phase arrest following DCM fraction treatment,
which was associated with decreased protein expression levels
of cell cycle-related proteins, such as cyclin A, CDK2, cell
division cycle 25 A and cyclin dependent kinase inhibitor 1.
Based on these results, the present study suggested that the
DCM fraction of the C. soldanella extract can inhibit HT-29
cell viability whilst inducing apoptosis through mitochondrial
membrane potential regulation and S-phase arrest. These
results also suggested that the DCM fraction has potential
anticancer activity in HT-29 colorectal cells. Further research
on the composition of the DCM fraction is warranted.
Introduction
Colorectal cancer is prevalent and a leading cause of death
worldwide (1). The mortality rate of colorectal cancer has
been declining over the past number of decades due to early
diagnosis using improved screening and treatment strategies.
However, the incidence remains high (2). Over the past several
decades, in the United States, the incidence and mortality rates
of colorectal cancer have been steadily decreasing among
those aged >50 years, but the number of those aged between
20 and 49 years is increasing. It is estimated that the incidence
and mortality rates of colorectal cancer according to these
age groups increase uniformly with economic development
due to environmental changes, such as lifestyle, increased
obesity and overall lifespan extension, and the consumption
of processed foods, alcohol and meat (1,2). To date, colorectal
cancer treatment involves radiotherapy and traditional thera-
pies, including surgery and chemotherapy (3). However, these
treatments are limited by toxicity, adverse events and drug
resistance (3). A number of studies have previously reported
that colorectal and colon cancer is negatively associated with
dietary factors, including plants, seaweeds, vegetables and
fruits, which contain a variety of phytochemicals (4-6). These
phytochemicals have been demonstrated to protect cells from
damage that leads to cancer (7-13).
The halophyte Calystegia soldanella (Linnaeus) Roem.
et Schult (Convolvulaceae) is a perennial herb that grows
on coastal sand dunes worldwide (14). This plant has been
extensively used in traditional medicine for general consump-
tion and as a type of herbal treatment, since it is considered
to confer bioactive effects against rheumatic arthritis, sore
throat, dropsy, scurvy, fever and diarrhea (15-17). In partic-
ular, fractions of C. soldanella have been reported to exhibit
anti‑inammatory (18‑20), antifungal (21), antiviral (22‑25),
anticancer (26,27) and analgesic effects (28). Although the
various bioactivities of C. soldanella have been assessed, its
effects on colon cancer have not been explored.
Dichloromethane fractions of Calystegia soldanella induce S‑phase
arrest and apoptosis in HT‑29 human colorectal cancer cells
IN-HYE KIM1*, TAEKIL EOM1*, JOON-YOUNG PARK2, HYUNG-JOO KIM1 and TAEK-JEONG NAM1
1Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041;
2Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
Received September 8, 2021; Accepted November 11, 2021
DOI: 10.3892/mmr.2021.12576
Correspondence to: Professor Taek-Jeong Nam, Future Fisheries
Food Research Center, Institute of Fisheries Sciences, Pukyong
National University, 474 Ilgwang-ro, Busan 46041, Republic of Korea
E-mail: namtj@pknu.ac.kr
*Contributed equally
Key word s: apoptosis, Calystegia soldanella, dichloromethane
fraction, apoptosis, HT-29 cells
KIM et al: DICHLOROMETHA NE FRACTION INDUCES S-PHASE ARREST AND APOPTOSIS
2
In a previous study, the viability of numerous cancer
cell lines was assessed, including the hepatocarcinoma
HepG2, gastric cancer AGS, colorectal cancer HT-29 and the
breast cancer cell line MCF-7, following treatment with the
C. soldanella crude extract (27). Similar effects, including a
decrease in cell viability, were observed in HT-29 and HepG2
cells (27). Therefore, the aim of the present study was to
evaluate the mechanism underlying any changes in HT-29 cell
physiology after treatment with C. soldanella extract fractions.
Materials and methods
Sample collection and preparation. Whole-plant C. soldanella
samples were collected from Gijang, Busan, Korea. A voucher
specimen was deposited at the Herbarium of the Division of
Marine Environment and Bioscience, Korea Maritime and
Ocean University (Busan, Korea). The entire plant samples
were briey air‑dried at room temperature for 1 month, ground
into a ne powder using a blender and stored at ‑20˚C.
Extraction and fractionation. The crude extract of the plant
samples (500 g) was eluted in 99% ethanol for 3 h at room
temperature before being filtered and concentrated three
times. The concentrated crude extracts were evaporated under
reduced pressure at 40˚C using rotary vacuum evaporator
and partitioned between H2O-methanol (9:1) and n-hexane
(4.3 g). The organic layer was further partitioned into dichloro-
methane (DCM; 15.2 g) and ethyl acetate (2.3 g). The aqueous
layer was also fractionated into n-butanol (14.6 g) and water
(16.4 g). Each fraction used was completely removed using a
reux condenser and was subsequently freeze‑dried for use
in experiments. All solvent reagents used for extraction were
of analytical grade. The DCM fraction was diluted to a nal
concentration of 0.2% DMSO so as not to induce toxicity. For
the control, an equivalent volume of 0.2% DMSO was added
to the culture medium.
Ultra‑performance liquid chromatography coupled with
electrospray ionization quadrupole time‑of‑flight mass
spectrometry (UPLC‑ESI‑Q‑TOF‑MS) analysis. The DCM
fractions were analyzed using UPLC-ESI-Q-TOF-MS.
The UPLC system (Agilent innity 1260 series; Agilent
Technologies Deutschland GmbH), with an incorporated
photodiode array detector (DAD) and Impact II Q-TOF
mass spectrometer (Bruker Corporation), was equipped
with an ESI source that operated on the negative ion
mode. A reverse phase Kintex core-shell C-18 column
(100x2.1 mm, 1.7 µm, Phenomenex) was used at a flow
rate of 0.5 ml/min. The mobile phase consisted of water
containing 0.1% TFA (A) and 0.1% TFA containing aceto-
nitrile (B) using the following gradient conditions: 0-1 min,
10% B; 1-4 min, 10-20% B; 4-6 min, 20-25% B; 6-8 min,
25% B; 8-9 min, 25-30% B; 9-11 min, 30% B; 11-12 min,
30-50% B; 12-14 min, 50-60% B; 14-15 min, 60-80% B;
and 15-17 min, 80% B. The injection volume was 2 µl. Mass
spectra in positive-ion or negative-ion mode were recorded
within 20 min. The UPLC profiles of the extracts were
measured using a DAD. The analyses were conducted in the
negative ion mode in a mass range from m/z 50 to 1,000.
The ESI source parameters were: Capillary voltage, 4.5 KV;
nebulizing gas pressure, 1.5 bar; drying gas temperature,
200˚C, drying gas ow, 9.0 l/min; Funnel 1RF 250.0 Vpp;
transfer time, 50.0 µs; and prepulse storage, 2.0 µs. The
MS data were analyzed using Data Analysis 4.2 software
(Bruker Corporation).
Cell culture. The human colorectal HT-29 cell line
(cat. no. 30038) was purchased from the Korean Cell Line
Bank, Korean Cell Line Research Foundation. The STR
prole of the HT‑29 cell line was as follows: D3S1358, 15/17;
von Willebrand factor type A, 17/19; fibrinogen α chain,
20/22; amelogenin, X; tyrosine hydroxylase 1, 6/9; thyroid
peroxidase, 8/9; CSF1P0, 11/12; D5S818, 11/12; D13S317,
11/12; and D7S820, 10. The cells were cultured at 37˚C with
5% CO2 in RPMI-1640 medium (Welgene, Inc.) supplemented
with 10% FBS (Welgene, Inc.) containing 100 U/ml penicillin
and 100 µg/ml streptomycin (cat no. CA005-10; GenDEPOT,
LLC). The culture medium was refreshed every 2 days and the
cells were subcultured for use in subsequent experiments.
Cell viability assays. Cell viability was analyzed using
a EZ-Cytox Kit (cat. no. EZ-1000; DoGenBio Co., Ltd.)
according to the manufacturer's protocol. Cells were seeded
into 96-well plates at 4x104 cells/well and allowed to attach
for 24 h. First, cell viability was examined for each fraction
(hexane, DCM, ethyl acetate, butanol, water) at concentra-
tions of 0, 25, 50 or 100 µg/ml for 24 h. Next, attached
cells were treated with 0, 10, 20, 40, 60, 80 or 100 µg/ml
of the DCM fraction in serum-free medium for 24 or 48 h.
Subsequently, cells were incubated with the EZ-Cytox
solution (100 µl/well) for 30 min at 37˚C before absor-
bance at 450 nm was quantied using the FilterMAX F5
microplate reader (Molecular Devices LLC.). In addition,
morphological cell changes were subsequently observed
using a light microscope (magnification, x200; Eclipse
TS100-F; Nikon Corporation).
Apoptosis assay. Apoptosis was assessed using the Muse®
Annexin V and Dead Cell Kit (cat. no. MCH100105; Luminex
Corporation) according to the manufacturer's protocol. Cells
were seeded into six-well plates at 1x105 cells/well and
treated with 20, 40 or 80 µg/ml concentrations of the DCM
fraction for 20 h. The cells were then harvested at a density
of 5x104 cells/well and washed twice with PBS and stained
with FITC-Annexin V and dead cell reagent for 20 min at
room temperature in the dark. The percentage of apoptotic
cells was determined using the Guava® Muse® Cell Analyzer
(2012model; Luminex Corporation).
Assessment of mitochondrial membrane potential (MMP).
The MMP was assessed using the Muse® MitoPotential Kit
(cat. no. MCH100110; Luminex Corporation.) according to
the manufacturer's protocol. Cells were seeded into six-well
plates at 1x105 cells/well and treated with 0, 20, 40 or 80 µg/ml
concentrations of the DCM fraction for 20 h. The cells were
harvested at a density of 5x104 cells/well, washed twice with
PBS, stained with MitoPotential working solution containing
MitoPotential dye and incubated for 20 min in a 37˚C CO2
incubator. The MMP was determined using the Guava Muse
Cell Analyzer (2012 model).
MOLECULAR MEDICINE REPORTS 25: 60, 2022 3
Cell cycle assay. The cell cycle was assessed using the
Muse® Cell Cycle Assay Kit (cat. no. MCH100106; Luminex
Corporation) according to the manufacturer's protocol. Cells
were seeded into six-well plates at 1x105 cells/well and treated
with 0, 20, 40 or 80 µg/ml concentrations of the DCM fraction
for 20 h. The cells were harvested, washed twice with PBS,
xed in ice cold 70% ethanol and frozen at ‑20˚C for 3 h. The
xed cells were stained at a density of 5x104 cells/ml with
200 µl Muse cell cycle reagent for 30 min at room temperature.
The cell cycle phase was determined using the Guava Muse
Cell Analyzer (2012 model).
Preparation of total cell lysate. HT-29 cells were treated
with 0, 20, 40 or 80 µg/ml of the DCM fraction in serum-free
medium for 24 h at 37˚C. The cells were washed with PBS
and lysed in M-PER Mammalian Protein Extraction Reagent
(cat. no. 78501; Thermo Fisher Scientific, Inc.) containing
phosphate inhibitor cocktail (cat. no. 1862495; Thermo Fisher
Scientic, Inc.) and ProteaseArrest™ protease inhibitor cock-
tail (cat. no. 786-108; G-Biosciences; Geno Technology, Inc.)
on ice for 30 min. The extracts were centrifuged at 12,000 x g
for 10 min at 4˚C and the supernatants were subsequently
used for western blotting. The mitochondrial and cytosolic
fractions were extracted using a Mitochondria Isolation Kit
(cat. no. 89874; Thermo Fisher Scientic, Inc.), according to
the manufacturer's protocols. Protein concentrations were
measured using a BCA Protein Assay Kit (cat. no. 23225;
Thermo Fisher Scientic, Inc.).
Western blotting. Total protein (20-40 µg protein/la n e) wa s
electrophoresed via SDS-PAGE on a 8-15% acrylamide gel
and transferred onto polyvinylidene fluoride immobilon-P
membranes (cat. no. MLP.IPVH00010; MilliporeSigma). The
membranes were blocked with 1% bovine serum albumin
(BSA; cat. no. A0100; GenDEPOT, LLC) in TBS with 0.1%
Tween-20 (TBST; 5 mM Tris, 20 mM sodium chloride,
pH 7.4) and incubated with primary antibodies (1:1,000) in
1% BSA /TBST with gentle agitation at 4˚C overnight. The
membranes were then washed twice for 15 min in TBST
each and incubated with the corresponding HRP-conjugated
secondary antibodies (1:10,000) for 2 h at room temperature,
before being washed again using TBST. Immunoreactive
bands were detected using the WesternBright® ECL HRP
Substrate (cat. no. K12045; Advansta, Inc.) and visualized
using the GeneGnome 5 (model 75000; Syngene). Differences
in protein levels were determined by semi-quantifying the
western blotting band densities using ImageJ software version
IJ.146r (National Institutes of Health).
The antibodies used were as follows: Anti-Fas
(cat. no. sc-7886; rabbit; Santa Cruz Biotechnology,
Inc.), anti-caspase-8 (cat. no. sc-7890; rabbit; Santa Cruz
Biotechnology, Inc.), anti-Bcl-2 (cat. no. sc-7382; mouse;
Santa Cruz Biotechnology, Inc.), anti-Bcl-extra-large (xL;
cat. no. sc-7195; rabbit; Santa Cruz Biotechnology, Inc.),
anti-Bad (cat. no. sc-8044; mouse; Santa Cruz Biotechnology,
Inc.), anti-Bax (cat. no. sc-7480; mouse; Santa Cruz
Biotechnology, Inc.), anti-caspase-9 (cat. no. sc-7885;
rabbit; Santa Cruz Biotechnology, Inc.), anti-caspase-7
(cat. no. sc-6138; rabbit; Santa Cruz Biotechnology,
Inc.), anti-caspase-3 (cat. no. sc-7148; rabbit; Santa Cruz
Biotechnology, Inc.), anti-X-linked inhibitor of apop-
tosis protein (XIAP; cat. no. 2045; rabbit; Cell Signaling
Technology, Inc.), anti-cellular inhibitor of apoptosis protein
(cIAP)-1 (cat. no. 7065; rabbit; Cell Signaling Technology, Inc.),
anti-cIAP-2 (cat. no. 3130; rabbit; Cell Signaling Technology,
Inc.), anti-cytochrome c (cat. no. 4272; rabbit; Cell Signaling
Technology, Inc.), anti-cytochrome c oxidase subunit IV (COX
IV; cat. no. 4844; rabbit; Cell Signaling Technology, Inc.),
anti-cyclin A (cat. no. BS-0571R; rabbit; BIOSS), anti-CDK2
(cat. no. sc-163; rabbit; Santa Cruz Biotechnology, Inc.),
anti-cell division cycle 25 A (Cdc25A; cat. no. sc-7389; mouse;
Santa Cruz Biotechnology, Inc.) and anti-cyclin dependent
kinase inhibitor 1 (p21; cat. no. sc-271532; rabbit; Santa
Cruz Biotechnology, Inc.). Anti-β-actin (cat. no. sc-47778;
mouse; Santa Cruz Biotechnology, Inc.) antibody was used
as the loading control. The secondary antibodies used were
HRP-conjugated anti-mouse IgG (cat. no. 7076; Cell Signaling
Technology, Inc.) and anti-rabbit IgG (cat. no. 7074; Cell
Signaling Technology, Inc.).
Statistical analysis. All data are presented as the mean ± SD
of three independent experiments. Means between >2 groups
were compared using one-way or two-way ANOVA followed
by Bonferroni's multiple comparison test using GraphPad
Prism version 7 software (GraphPad Software, Inc). P<0.05
was considered to indicate a statistically signicant difference.
Results
Solvent fractions of C. soldanella reduce HT‑29 cell viability.
To determine the effects of the C. soldanella fractions on cell
viability, HT‑29 cells were treated with each of the ve solvent
fractions (n-hexane, DCM, ethyl acetate, n-butanol and water)
at different concentrations (0, 25, 50 and 100 µg/ml) for 24 h,
after which cell viability was examined. Cell viability was
signicantly decreased following treatment with the DCM
fraction compared with that in the 0 µg/ml group (Fig. 1A).
Therefore, the DCM fraction with the highest dose-dependent
effect was selected for further study.
Reductions in the viability of HT‑29 cells was conrmed
following treatment with different concentrations (0-100 µg/ml)
of DCM fraction for 24 and 48 h (Fig. 1B). HT-29 cell viability
appeared to be decreased following DCM fraction treat-
ment in a time- and dose-dependent manner compared with
those in the 0 µg/ml group. After treatment with 0, 10, 20,
40, 60, 80 and 100 µg/ml DCM, cell viability was 100±4.7,
89.0±3.5, 74.6±6.7, 73.2±2.0, 53.5±7.1, 26.7±1.6 and 25.4±1.7%
at 24 h, respectively, whereas it was 100±3.4, 92.6±8.3, 57.9± 6.6,
35.4±3.6, 20.2±1.6, 9.3±0.5 and 8.7±0.2% at 48 h, respectively
(Fig. 1B). In addition, it was observed that the morphological
changes conrmed via microscope were reduced in the same
way as the results of the cell viability assay (Fig. 1C).
DCM fraction from C. soldanella induces apoptosis in
HT‑29 cells. The Annexin V and Dead Cell Kit was used to
determine whether this decrease in cell viability induced by
DCM fraction treatment resulted from apoptosis. The rates
of early and late apoptosis were signicantly increased in a
dose-dependent manner following treatment with the DCM
fraction compared with those in the 0 µg/ml group (Fig. 2).
KIM et al: DICHLOROMETHA NE FRACTION INDUCES S-PHASE ARREST AND APOPTOSIS
4
The proportions of early apoptotic cells were 0.7±0.67,
38.2±1.81, 23.0±3.07 and 1.0±0.38%, whilst those of late apop-
totic cells were 0±0.05, 2.1±0.40, 12.9±7.12 and 35.1±0.58%,
following DCM fraction treatment at concentrations of 0,
20, 40 and 80 µg/ml, respectively.
Regarding the protein expression levels of apoptosis-related
proteins, DCM fraction treatment at 40 and 80 µg/ml signi-
cantly increased the protein expression levels of Fas protein
whilst significantly decreasing those of pro-caspase-8,
an extrinsic signaling pathway-related protein (Fig. 3),
compared with those in the 0 µg/ml group. Other intrinsic
apoptosis signaling pathway-related proteins that also
showed signicantly decreased protein expression levels after
40 and 80 µg/ml DMC treatment were Bcl-2 and Bcl-xL, whilst
those that showed signicantly increased protein expression
levels following 40 and 80 µg/ml DCM fraction treatment
were Bad and Bax (Fig. 3). Consequently, the Bax/Bcl-2 ratio
was significantly increased in the 40 and 80 µg/ml DCM
fraction treatment groups compared with that in the 0 µg/ml
group. In addition, 40 and 80 µg/ml DCM fraction treatment
also signicantly decreased the expression of pro‑caspase‑9,
pro-caspase-7 and pro-caspase-3 levels compared with those
in the 0 µg/ml group. The protein expression levels of XIAP,
cIAP-1 and cIAP-2, caspase inhibitors involved in apoptosis
inhibition, were signicantly decreased by 40 and 80 µg/ml
DCM treatment compared with those in the 0 µg/ml group.
These results suggest that the treatment of HT-29 cells with
DCM from C. soldanella may induce apoptosis by regulating
the expression of pro-apoptotic, pre-apoptotic and caspase
inhibitor proteins.
DCM fraction from C. soldanella induces MMP changes in
HT‑29 cells. Since MMP changes are also associated with
apoptosis (29-31), the effects of DCM fract ion treatment on the
MMP in HT-29 cells were investigated. The proportions of live
and dead cells with depolarized mitochondria were markedly
increased following DCM fraction treatment compared with
those in the 0 µg/ml group. The proportions of depolarized
Figure 1. DCM from Calystegia soldanella reduces the viability of HT‑29 colorectal cancer cells. (A) HT‑29 cells were incubated with the ve different solvent
fractions at 0‑100 µg/ml for 24 h. The letters displayed above the concentrations represent signicant differences (P<0.05) as determined by Bonferroni's
multiple comparisons test; groups with different letters are signicantly different to one another, whereas those with the same letter are not. (B) HT‑29 cells were
incubated with t he DCM fraction at 0-100 µg/ml for 24 and 48 h. Cell viability was examined using the EZ-Cytox proliferation assay. These data represent the
percentage of surviving cells compared with the control (0 µg/ml). (C) Cell mor phology changes were observed using light m icroscopy. Scale bar, 100 µm. Data
are presented as the mea n ± SD of three i ndependent exper iments. *P<0.05 vs. 0 µg/ml DCM; #P<0.05. DCM, dichloromethane; NS, not signicant.
MOLECULAR MEDICINE REPORTS 25: 60, 2022 5
live cells were 5.2±0.75, 43.6±1.49, 2.9±0.21 and 1.2±0.05%,
whereas the proportions of depolarized dead cells were
0.8±0.24, 10.5±1.48, 89.7±0.33 and 95.8±0.35%, at concen-
trations of 0, 20, 40 and 80 µg/ml, respectively (Fig. 4A).
The protein expression levels of MMP-related proteins were
examined using western blotting. DCM fraction treatment
at 40 and 80 µg/ml signicant ly incr eased the rele ase of cy to-
chrome c into the cytosol from the mitochondria compared
with that in the 0 µg/ml group (Fig. 4B). In addition, DCM
fraction treatment also resulted in the signicantly increased
translocation of Bax into the mitochondria from the cytosol
in a dose-dependent manner compared with the 0 µg/ml
group.
DCM from C. solda nella induces S‑phas e arrest in HT‑29 cells.
To determine whether decreased cell viability was associated
with the cell cycle, HT-29 cell cycle progression was analyzed
using a cell cycle kit following DCM fraction treatment. DCM
significantly induced S-phase arrest in a dose-dependent
manner compared with the 0 µg/ml group, with S-phase cell
proportions of 20.7±1.8, 33.8±0.2, 39.9±5.6 and 45.3±0.2% at
concentrations of 0, 20, 40 and 80 µg/ml of the DCM fraction,
respectively (Fig. 5A). Subsequently, the relative protein expres-
sion levels of S-phase-related proteins were analyzed using
western blotting. DCM fraction treatment at 40 and 80 µg/ml
led to the signicant downregulation of cyclin A, CDK2 and
Cdc25A protein expression and the signicant upregulation of
p21 protein expression compared with those in the 0 µg/ml
group (Fig. 5B).
ESI‑Q‑TOF‑MS analysis of DCM f raction from C. soldanella.
UPLC-ESI-Q-TOF-MS analysis was applied to analyze the
polyphenolic compounds in the DCM fraction of C. soldanella
to screen for any anticancer substances. The HPLC-UV chro-
matogram (350 nm) and total ion current chromatogram in the
DCM fraction are presented in the Fig. 6A and B.
Molecular ion mass, MS/MS fragment ion mass and
MS-based compound analysis data are all provided in Table I.
UPLC-ESI-Q-TOF MS analysis demonstrated that the major
compounds in the DCM fraction were hydroxybenzoic acid,
hydrosinapinic acid and coumaric acid, which are phenolic
acid derivatives, and quercetrin, which is a avonoid quercetin
derivative.
Discussion
The ultimate aim of discovering novel chemotherapeutic
strategies is to overcome drug resistance (32). A number of
studies have previously investigated the potential anticancer
activity of natural compounds/products and suggested them
to be promising sources of new anticancer drugs (4-13). For
example, the natural compound S-adenosylmethionine has
Figure 2. DCM f raction treatment induces apoptosis in HT-29 cells. HT-29 cells were incubated with various concent rations of the DCM fraction for 20 h.
FITC‑Annexin V staining followed by ow cytometry was used to determine the percentage of apoptotic cells. Data are presented as the mean ± SD of three
independent experiments. The letters displayed above the concentrations represent signicant differences (P<0.05) as determined by Bonferroni's multiple
comparisons test; groups with different letters are signicantly different to one another, whereas those with the same letter a re not. DCM, dichloromethane;
7-AAD, 7-Aminoactinomycin D; apop, apoptotic.
KIM et al: DICHLOROMETHA NE FRACTION INDUCES S-PHASE ARREST AND APOPTOSIS
6
been found to exert anti-tumor properties, including reduction
of cell proliferation, induction of apoptosis, autophagy and
inhibition of invasion and metastasis, in various cancer cell
types, such as human hepatocellular carcinoma, human breast
cancer and head and neck squamous cancer cells, in previous
in vitro and in vivo stud ies (33 -36).
To date, anticancer-associated studies related to
C. soldanella have revealed cytotoxic effects of methanol
and chloroform extracts on the lung cancer A545 and colon
cancer Col2 cell lines (26) and the anticancer effects of an
85% aqueous methanol fraction on the liver cancer cell line
HepG2 (27). However, only a few studies have investigated the
anticancer effects of C. soldanella.
In the present study, solvent fractions were obtained from
C. soldanella, which have been previously reported to exert
anticancer activity in human liver cancer cell line HepG2 (27),
before an effective fraction that can exhibit anticancer effects
on the colorectal cancer cell li ne HT-29 was selected. Following
the crude extraction of C. soldanella with ethanol, the crude
extracts were fractionated into n-hexane, DCM, ethyl acetate,
n‑butanol and water. The effects of these ve fractions on
HT-29 cell viability were examined before the DCM fraction
was selected due to its signicant time‑ and dose‑dependent
effects.
Apoptosis serves a critical role in the regulation of cell
development and proliferation (37,38). This process has
become a target of cancer treatments due to its association
with a number of different types of cancer (39,40). Two major
pathways of apoptosis have been identied: i) The extrinsic
death receptor pathway; and ii) the endoplasmic reticulum
stress pathway and intrinsic mitochondrial apoptosis (41).
To determine the effect of DCM fraction treatment on cell
Figure 3. Effects of DCM fraction t reatment on the expression of apoptosis-related proteins in HT-29 cells. HT-29 cells were incubated with various concentra-
tions of the DCM fraction for 20 h. The expression levels apoptosis-related proteins Fas, pro-caspase-8, Bcl-2, Bcl-xL, Bad, Bax, pro-caspase-9, pro-caspase-7,
pro-ca spase-3, XIA P, cIA P-1 and cIAP-2 were then det ermined by western blott ing. β-acti n was used as the loading contr ol. The band s were semi- quantitatively
analyzed by ImageJ software and the relative protein expression levels are presented. All results were norma lized to the untreated control (0 µg/ml). Data are
presented as t he mean ± SD of three independent experiments. *P<0.05 vs. 0 µg/ml DCM. DCM, dichloromethane; xL, extra-la rge; XIAP, X-linked inhibitor
of apoptosis protein; cIAP-1, cellular inhibitor of apoptosis protein-1; cIAP-2, cellular inhibitor of apoptosis protein-2.
MOLECULAR MEDICINE REPORTS 25: 60, 2022 7
viability and apoptosis, apoptosis and apoptosis-related
protein expression levels in HT-29 cells that were treated with
the DCM fraction were investigated. The results demonstrated
signicantly increased apoptotic rates and marked changes
in the expression levels of apoptosis-related proteins in both
the extrinsic and intrinsic signaling pathways. Treatment with
DCM fraction increased Fas and decreased pro-caspase-8,
which corresponds to the extrinsic signaling pathway. In addi-
tion, treatment with DCM fraction increased Bad and Bax, and
decreased Bcl-2, Bcl-xL, pro-caspase-9, pro-caspase-7 and
pro-caspase-3, which corresponds to the intrinsic signaling
pathway.
The mitochondrial apoptosis pathway involves a number
of components, including pre-apoptotic proteins Bcl-2 and
Bcl-xL and pro-apoptotic proteins Bax and Bak (42-44).
The ratio of Bax/Bcl-2 determines the direction of apoptosis
regulation. An increased Bax/Bcl-2 ratio can lead to the loss
of mitochondrial membrane potentials, which is an important
process in the initiation of apoptosis (45). Furthermore, it has
been reported that an increased Bax/Bcl-2 ratio can activate
caspase-3 to in turn activate apoptosis (46,47). In the present
study, caspase-3 was also markedly activated following
DCM fraction treatment. Since the Bax/Bcl-2 ratio was also
markedly increased with DCM fraction treatment, it was
hypothesized that DCM may activate apoptosis by altering
the mitochondrial membrane potential.
Apoptosis involves the regulation of a series of proteins
mainly in the mitochondrial signaling pathway (48). The
Figure 4. DCM fraction treatment induces depolarization of mitochondria l membrane potential in HT-29 cells. (A) HT-29 cells were incubated with various
concentrations of the DCM fraction for 20 h. MitoPotential kit staining followed by ow cytometry was used to determine the percentages of depola rized cells.
The letters displayed above the concentrations represent signicant differences (P<0.05) as determined by Bonferroni's multiple comparisons test; groups with
different letters are signicantly different to one another, whereas those with the same letter are not. (B) Protein expression levels of MMP‑related proteins
cytochrome c and Bax were determ ined using western blotting analysis. β-actin and COX IV were used as loading controls for the cytosol and mitochon-
dria, respectively. All results were normalized to the untreated control (0 µg/ml). Data are presented as the mean ± SD of three independent experiments.
*P<0.05 vs. 0 µg/ml DCM. DCM, dichloromethane; 7-AAD, 7-Aminoactinomycin D; COX IV, cytochrome c oxidase subunit IV; mi, mitochondr ial; cy, cytosol.
KIM et al: DICHLOROMETHA NE FRACTION INDUCES S-PHASE ARREST AND APOPTOSIS
8
mitochondria maintain the cellular energy balance and regu-
late cell death processes (49). Cellular energy generated during
mitochondrial respiration is stored as an electrochemical
gradient across the mitochondrial membrane, which allows
the mitochondria to induce ATP synthesis (49). Changes
in the MMP are associated with apoptosis, necrosis and
caspase-independent cell death processes in addition to the
opening of mitochondrial transition pores, to release cyto-
chrome c into the cytosol and initiate apoptotic and caspase
cascades (31,49). In the present study, MMP changes following
DCM fraction treatment led to matrix condensation and expo-
sure of cytochrome c to the intermembrane space, which may
have activated apoptosis (Fig. 4).
Numerous proteins associated with cell cycle regulation
are known to be involved in apoptosis (50). In the present study,
cell cycle assay was performed and the protein expression
levels of several cell cycle-related proteins were investigated
to determine whether apoptosis induced by DCM fraction
treatment was due to cell cycle regulation in HT-29 cells. The
results demonstrated that DCM fraction treatment signicantly
increased the proportion of cells in the S-phase. Cyclins and
cyclin-dependent kinases serve important roles in cell cycle
regulation, where changes in the composition of cyclin/CDK
complexes can either increase or decrease cell proliferation
and/or differentiation through apoptosis (51,52). Since cyclin
A, CDK2 and Cdc25A serve critical roles in S-phase regula-
tion (53,54), their protein expression levels following DCM
fraction treatment were examined. The results showed marked
downregulation of cyclin A, CDK2 and Cdc25A expression
following DCM treatment. CDK1, p21 (Waf1/Cip1) have been
previously shown to induce cell cycle arrest by inhibiting CDK
activity (55,56). The results of the present study demonstrated
a dose-dependent increase in p21 protein expression levels in
response to DCM, which may have inhibited Cyclin A-CDK2
complex formation and contributed to cell cycle arrest between
the S and G2/M phases.
Figure 5. DCM fraction treatment induces S-phase arrest in HT-29 cells. (A) HT-29 cells were incubated with various concentrations of the DCM fraction
for 20 h. Flow cytometry was used to deter mine the percentages of cells in each phase of the cell cycle. The letters displayed above the concentrations
represent signicant differences (P<0.05) as determined by Bonferroni's multiple comparisons test; groups with different letters are signicantly different
to one another, whereas those with the sa me letter a re not. (B) Protein expression levels of S-phase-related proteins cyclin A, CDK2, Cdc25A and p21 were
measured using western blotti ng. All results were nor malized to the untreated control (0 µg/ml). Data are presented as the mean ± SD of three independent
experiments. *P<0.05 vs. 0 µg/ml DCM. DCM, dichloromethane; CDK2, cyclin- dependent kinase 2; Cdc25A, cell division cycle 25 homolog A; p21, cyclin
dependent kinase inhibitor 1.
MOLECULAR MEDICINE REPORTS 25: 60, 2022 9
It has been frequently reported that numerous compounds
can exert anticancer activity independent of p53 (57-60). The
tumor suppressor p53 has been found to be dysfunctional in
≥50% colorectal cancer cases (58,59). Colorectal cancer cells
and tumors with p53 mutation are reported to be more aggres-
sive and resistant to chemotherapy (60). In the present study,
the HT-29 cell line is a p53 mutant cell line (genotype R273H),
to which DCM exerted anticancer effects in the absence of p53.
Although not conducted in the present study, anticancer effects
following DCM fraction treatment are also hypothesized to be
present in p53 wild-type cell lines and warrants further study.
Previous studies have reported that C. soldanella contains
a variety of polyphenolic compounds, including avonoids,
avonoid glycosides and phenolic acid derivatives (56,61).
Molecular ion mass, MS/MS fragment ion mass and
MS-based compound analysis data are all provided in Table I.
UPLC-ESI-Q-TOF MS analysis demonstrated that the major
compounds in the DCM fraction were hydroxybenzoic acid,
hydrosinapinic acid and coumaric acid, which are phenolic
acid derivatives, and quercetrin, which is a avonoid quer-
cetin derivative. Various phenolic acids, such as coumaric
acid, caffeic acid and ferulic acid, function as secondary
plant metabolites (62). In particular, cinnamic acid-based
phenoic acid compounds, such as coumaric acid, caffeic acid,
ferulic acid and sinapinic acid, have been reported to exert
anticancer activity in various colon cancer cell lines (63).
Coumaric acid is reported to induce apoptosis in the colon
cancer HCT-115 cell line and induce G1/S arrest in the colon
cancer cell line Caco-2 (64-66). These aforementioned
phenolic acids also have inhibitory activities on the prolifera-
tion of various colorectal/colon cancer cell lines (67-69).
In conclusion, to the best of our knowledge, the present
study was the rst to report the anticancer effects of DCM
from C. soldanella on HT-29 colorectal cancer cells, which
possibly occurred by MMP alteration, S-phase arrest and
induction of apoptosis through the intrinsic/extrinsic signaling
pathways. The present study also demonstrated that phenolic
acid derivatives are the main components of the DCM frac-
tion, which exerted inhibitory activities on HT-29 colorectal
cancer cell by apoptosis and cell cycle arrest.
Acknowledgements
Not applicable.
Funding
The present study was part of the Future Fisheries Food
Research Center Project, funded by the Ministry of Oceans
and Fisheries (grant no. 201803932).
Figure 6. UPLC-ESI-Q-TOF-MS analysis of DCM fraction. (A) UV chromatogram at 350 nm a nd (B) UPLC-ESI-Q-TOF MS total ion chromatogram of the
dichloromethane fraction. intens, intensity; UPLC-ESI-Q-TOF-MS, ultra-performance liquid chromatography coupled with electrospray ionization quadru-
pole time‑of‑ight mass spectrometry.
Table I. Identied compounds in the DCM fraction.
Peak no. Compound Retention time, min Mass m/z MS fragment
1 Hydroxybenzoic acid 1.6 138.12 137.02 112.98
2 Hydrosinapinic acid 2.0 226.08 225.11 181.12, 121.02
3 Coumaric acid 2.2 164.04 163.03 119.05
4 Quercetrin 3.0 448.38 447.09 301.03, 146.93
Peaks correspond to peaks observed in Fig. 6B. DCM, dichloromethane; MS, mass spectrometry.
KIM et al: DICHLOROMETHA NE FRACTION INDUCES S-PHASE ARREST AND APOPTOSIS
10
Availability of data and materials
The datasets used and/or analyzed in the current study are
available from the corresponding author on reasonable request.
Authors' contribution
IHK, TE and TJN conceived and designed the experiments. IHK,
JYP and HJK performed the experiments. IHK, TE and HJK
analyzed and interpreted the results and wrote and revised the
manuscript. IHK, TE and TJN conrm the authenticity of all the
raw data. All authors have read and approved the na l manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
1. Rawla P, Sunkara T and Barsouk A: Epidemiology of colorectal
cancer: Incidence, mortality, survival, and risk factors. Prz
Gastroenterol 14: 89-103, 2019.
2. Cancer Trends Progress Report; Colorectal Cancer Treatment.
National Cancer Institute, 2020.
3. Xie YH, Chen YX and Fang JY: Comprehensive review of
targeted therapy for colorectal cancer. Signal Transduct Target
Ther 5: 22, 2020.
4. Pandey R, Singh PK and Shrivastava AK: Seaweeds: Potential
candidates in human colon cancer therapy. Colon Cancer
Diagnosis Ther: Jun 5, 2021 (Epub ahead of print). doi: 10.1007/
978-3-030-64668-4_13.
5. Zhao Y and Jiang Q: Roles of the polyphenol-gut microbiota
interaction in alleviating colitis and preventing colitis-associated
colorectal cancer. Adv Nutr 12: 546-565, 2021.
6. García-Lafuente A, Guillamón E, Villares A, Rostagno MA
and Martínez JA: Flavonoids as anti-inflammatory agents:
Implications in cancer and cardiovascular disease. Inflamm
Res 58: 537-552, 2009.
7. Akiyama Y, Kimura Y, Enatsu R, Mikami T, Wanibuchi M and
Mikuni N: Advantages and disadvantages of combined chemo-
therapy with carmustine wafer and bevacizumab in patients with
newly diagnosed glioblastoma: A single-institutional experience.
World Neurosurg 113: e508-e514, 2018.
8. Bar-Shalom R, Bergman M, Graossman S, Azzam N, Sharvit L
and Fares F: Inula viscosa extract inhibits growth of colorectal
cancer cells in vitro and in vivo through induction of apoptosis.
Front Oncol 9: 227, 2019.
9. Bravo L: Polyphenols: Chemistry dietary sources metabolism
nutritional signicance. Nutr Rev 56: 317‑333, 1998.
10. Moreira H, Slezak A, Szyjka A, Oszmianski J and Ga siorowski K:
Antioxidant and cancer chemopreventive activities of cistus and
pomegranate polyphenols. Acta Pol Pharm 74: 688-698, 2017.
11. del Mar Blanquer-Rosselló M, Hernández-López R, Roca P,
Oliver J and Valle A: Resveratrol induces mitochondrial respira-
tion and apoptosis in SW620 colon cancer cells. Biochim Biophys
Acta Gen Sugj 1861: 431-440, 2017.
12. Kim IH and Nam TJ: Fucoidan downregulates insulin-like
growth factor-I receptor levels in HT-29 human colon cancer
cells. Oncol Rep 39: 1516-1522, 2018.
13. Kim IH, Kwon MJ and Nam TJ: Differences in cell death and cell
cycle following fucoidan treatment in high-density HT-29 colon
cancer cells. Mol Med Rep 15: 4116-4122, 2017.
14. Bae CY, Hwang JS, Bae JJ, Choi SC, Lim SH, Choi DG, Kim JG
and Choo YS: Physiological responses of Calystegia soldanella
under drought stress. J Ecol Environ 36: 255-265, 2013.
15. Bae KH: The Medicinal Plants of Korea. Korea, Kyo-Hak
publishing, Seoul, 2000.
16. Lee YS, Kwak CG and Kim NW: Nutritional characteristics
of Calystegia japonica. Korean J Food Preserv 19: 619-625,
2012.
17. Takagi S, Yamaki M, Masuda K and Kubota M: Studies on the
purgative drugs. IV. On the constituents of Calystegia japonica
Choisy (author's transl). Yakugaku Zasshi 97: 1369-1371, 1977
(In Japanese).
18. Kim Y, Min HY, Park HJ, Lee EJ, Park EJ, Hwang HJ, Jin C,
Lee YS and Lee SK: Suppressive effects of nitric oxide produc-
tion and inducible nitric oxide synthase (iNOS) gene expression
by Calystegia soldanella methanol extract on lipopolysaccha-
ride-activated RAW 264.7 cells. Eur J Cancer Prev 13: 419-424,
2004.
19. Huang Z and Feng C: Experimental study on anti‑inammatory
and analgesic effects of water extracts of Calystegia soldanella.
Chin Arch Tradit Chin Med 6: 7, 2010.
20. Lee JI, Kim IH, Choi YH, Kim EY and Nam TJ: PTP1B inhibi-
tory effect of alkyl p-coumarates from Calystegia soldanella.
Nat Prod Commun 9: 1585-1588, 2014.
21. Nidiry ES, Ganeshan G and Lokesha AN: Antifungal activity and
isomerization of octadecyl p-coumarates from Ipomoea carnea
subsp. stulosa. Nat Prod Commun 6: 1889‑1892, 2011.
22. Ono M, Kanemaru Y, Yasuda S, Okawa M, Kinjo J, Miyashita H,
Yokomizo K, Yoshimitsu H and Nohara T: A new resin glycoside
from Calystegia soldanella and its antiviral activity towards
herpes. Nat Prod Res 31: 2660-2664, 2017.
23. Ono M, Takigawa A, Kanemaru Y, Kawakami G, Kabata K,
Okawa M, Kinjo J, Yokomizo K, Yoshimitsu H and Nohara T:
Calysolins V-IX, resin glycosides from Calystegia soldanella
and their antiviral activity toward herpes. Chem Phar m Bull
(Tokyo) 62: 97-105, 2014.
24. Ono M, Kawakami G, Takigawa A, Kabata K, Okawa M,
Kinjo J, Yokomizo K, Yoshimitsu H and Nohara T: Calysolins
X-XIII, resin glycosides from Calystegia soldanella, and their
antiviral activity toward herpes simplex virus. Chem Pharm Bull
(Tokyo) 62: 839-844, 2014.
25. Ono M, Takigawa A, Muto H, Kabata K, Okawa M, Kinjo J,
Yokomizo K, Yoshimitsu H and Nohara T: Antiviral activity
of four new resin glycosides calysolins XIV-XVII from
Calystegia soldanella against Herpes Simplex Virus. Chem
Pharm Bull (Tokyo) 63: 641-648, 2015.
26. Min HY, Kim Y, Lee EJ, Hwang HJ, Park EJ and Lee SK:
Cytotoxic activities of indigenous plant extracts in cultured
human cancer cells. Nat Prod Sci 8: 170-172, 2002.
27. Lee JI, Kim IH and Nam TJ: Crude extract and solvent fractions
of Calystegia soldanella induce G1 and S phase arrest of the cell
cycle in HepG2 cells. Int J Oncol 50: 414-420, 2017.
28. Ly JD, Grubb DR and Lawen A: The mitochondrial membrane
potential (deltapsi(m)) in apoptosis; an update. Apoptosis 8:
115-128, 2003.
29. Gottlieb E, Armour SM, Harris MH and Thompson CB:
Mitochondrial membrane potential regulates matrix congura-
tion and cytochrome c release during apoptosis. Cell Death
Differ 10: 709-717, 2003.
30. Yang J, Cao L, Li Y and Liu H, Zhang M, Ma H, Wang B, Yuan X
and Li Q: Gracillin isolated from Reineckia carnea induces
apoptosis of A549 cells via the mitochondrial pathway. Drug Des
Devel Ther 15: 233-243, 2021.
31. Christensen ME, Jansen ES, Sanchez W and Waterhouse NJ:
Flow cytometry-based assays for the measurement of apop-
tosis-associated mitochondrial membrane depolarisation and
cytochrome c release. Methods 61: 138-145, 2013.
32. Mosca L, Pagano M, Pecoraro A, Borzacchiello L,
Mele L, Cacciapuoti G, Porcelli M, Russo G and Russo A:
S-Adenosyl-L-methionine overcomes uL3-mediated drug resis-
tance in p53 deleted colon cancer cells. Int J Mol Sci 22: 103,
2020.
33. Lu SC and Mato JM: S-Adenosylmethionine in cell growth,
apoptosis and liver cancer. J Gastroenterol Hepatol 23 (Suppl 1):
S73-S77, 2008.
34. Cave DD, Desiderio V, Mosca L, Ilisso CP, Mele L, Caraglia M,
Cacciapuoti G and Porcelli M: S-Adenosylmethionine-mediated
apoptosis is potentiated by autophagy inhibition induced by
chloroquine in human breast cancer cells. J Cell Physiol 233:
1370-1383, 2018.
MOLECULAR MEDICINE REPORTS 25: 60, 2022 11
35. Ilisso CP, Delle Cave D, Mosca L, Pagano M, Coppola A, Mele L,
Caraglia M, Cacciapuot i G and Porcelli M: S-Adenosylmethionine
regulates apoptosis and autophagy in MCF-7 breast cancer cells
through the modulation of specic microRNAs. Cancer Cell
Int 18: 197, 2018.
36. Mosca L, Minopoli M, Pagano M, Vitiello F, Carriero MV,
Cacciapuoti G and Porcell i M: Effects of S-adenosyl-L-methion ine
on the invasion and migration of head and neck squamous cancer
cells and analysis of the underlying mechanisms. Int J Oncol 56:
1212-1224, 2020.
37. D'Arcy MS: Cell death: A review of the major forms of apoptosis,
necrosis and autophagy. Cell Biol Int 43: 582-592, 2019.
38. Elmore S: Apoptosis: A review of programmed cell death.
Toxicol Pathol 35: 495-516, 2007.
39. Pistritto G, Trisciuoglio D, Ceci C, Garu A and D'Orazi G:
Apoptosis as anticancer mechanism: Function and dysfunction
of its modulators and targeted therapeutic strategies. Aging
(Albany NY) 8: 603-619, 2016.
40. Goldar S, Khaniani MS, Derak hshan SM and Baradaran B:
Molecular mechanisms of apoptosis and roles in cancer devel-
opment and treatment. Asian Pac J Cancer Prev 16: 2129-2144,
2015.
41. Pan Y, Ye C, Tian Q, Yan S, Zeng X, Xiao C, Wang L and
Wang H: miR-145 suppresses the proliferation, invasion and
migration of NSCLC cells by regulating the BAX/BCL-2 ratio
and the caspase-3 cascade. Oncol Lett 15: 4337-4343, 2018.
42. Knight T, Luedtke D, Edwards H, Taub JW and Ge Y: A delicate
balance-The Bcl-2 family and its role in apoptosis, oncogenesis,
and cancer therapeutics. Biochem Pharmacol 162: 250-261, 2019.
43. Orrenius S: Mitochondrial regulation of apoptotic cell death.
Toxicol Lett 149: 19-23, 2004.
44. Scor rano L and Korsmeyer SJ: Mechanisms of cytochrome
c release by proapoptotic BCL-2 family members. Biochem
Biophys Res Commun 304: 437-444, 2003.
45. Meeran SM and Katiyar SK: Grape seed proanthocyanidins
promote apoptosis in human epidermoid carcinoma A431 cells
through alterations in Cdki-Cdk-cyclin cascade, and caspase-3
activation via loss of mitochondrial membrane potential. Exp
Dermatol 16: 405-415, 2007.
46. Siu WP, Pun PB, Latchoumycandane C and Boelsterli UA:
Bax-mediated mitochondrial outer membrane permeabiliza-
tion (MOMP), distinct from the mitochondrial permeability
transition, is a key mechanism in diclofenac-induced hepatocyte
injury: Multiple protective roles of cyclosporin A. Toxicol
Appl Pharmacol 227: 451-461, 2008.
47. Ohtsuka T, Buchsbaum D, Oliver P, Makhija S, Kimberly R and
Zhou T: Synergistic induction of tumor cell apoptosis by death
receptor antibody and chemotherapy agent through JNK/p38 and
mitochondrial death pathway. Oncogene 22: 2034-2044, 2003.
48. Wang X, Lu X, Zhu R, Zhang K, Li S, Chen Z and Li L:
Betulinic acid induces apoptosis in differentiated PC12 cells
via ROS-mediated mitochondrial pathway. Neurochem Res 42:
1130 -114 0, 2 017.
49. Estaquier J, Vallette F, Vayssiere JL and Mignotte B: The
mitochondrial pathways of apoptosis. Adv Exp Med Biol 942:
157-18 3, 201 2.
50. Pucci B, Kasten M and Giordano A: Cell cycle and apoptosis.
Neoplasia 2: 291-299, 2000.
51. Canavese M, Santo L and Raje N: Cyclin dependent kinases
in cancer: Potential for therapeutic intervention. Cancer Biol
Ther 13: 451-457, 2012.
52. Sperka T, Wang J and Rudolph KL: DNA damage checkpoints in
stem cells, ageing and cancer. Nat Rev Mol Cell Biol 13: 579-590,
2012.
53. George Rosenker KM, Paquette WD, Johnston PA, Sharlow ER,
Vogt A, Bakan A, Lazo JS and Wipf P: Synthesis and biological
evaluation of 3-aminoisoquinolin-1(2H)-one based inhibitors of
the dual‑specicity phosphatase Cdc25B. Bioorg Med Chem 23:
2810 -2 818, 2015.
54. Tilaoui M, Mouse HA, Jaafari A and Zyad A: Differential effect
of artemisinin against cancer cell lines. Nat Prod Bioprospect 4:
189 -19 6, 2 014.
55. Zhu H, Zhang L, Wu S, Teraishi F, Davis JJ, Jacob D and Fang B:
Induction of S-phase arrest and p21 overexpression by a small
molecule 2‑[[3‑(2,3‑dichlorophenoxy) propyl]amino]ethanol in
correlation with activation of ERK. Oncogene 23: 4984-4992,
2004.
56. Ahn NR, Ko JM and Cha HC: Comparison of flavonoid
proles between leaves and stems of Calystegia soldanella and
Calystegia japonica. Am J Plant Sci 3: 1073-1076, 2012.
57. Al Aaraj L, Hayar B, Jaber Z, Saad W, Saliba NA, Darwiche N
and Ghaddar T: The effect of different ester chain modica-
tions of two guaianolides for inhibition of colorectal cancer cell
growth. Molecules 26: 5481, 2021.
58. Takayama T, Miyanishi K, Hayashi T, Sato Y and Niitsu Y:
Colorectal cancer: Genetics of development and metastasis.
J Gastroenterol 41: 185-192, 2006.
59. Nakayama M and Oshima M: Mutant p53 in colon cancer. J Mol
Cell Biol 11: 267-276, 2019.
60. Li XL, Zhou J, Chen ZR and Chng WJ: p53 mutations in
colorectal cancer-molecular pathogenesis and pharmacological
reactivation. World J Gastroenterol 21: 84-93, 2015.
61. Murai Y, Setoguchi H, Ono E and Iwashina T: Flavonoids and
their qualitative variation in Calystegia soldanella and related
species (Convolvulaceae) Nat Prod Commun 10: 429-432, 2015.
62. Abotaleb M, Liskova A, Kubatka P and Busselberg D:
Therapeutic potential of plant phenolic acids in the treatment of
cancer. Biomolecules 10: 221, 2020.
63. Anantharaju PG, Gowda PC, Vimalambike MG and
Madhunapantula SV: An overview on the role of dietary pheno-
lics for the treatment of cancers. Nutr J 15: 99, 2016.
64. Jaganathan SK, Supriyanto E and Mandal M: Events associated
with apoptotic effect of p-coumaric acid in HCT-15 colon cancer
cells. World J Gastroenterol 19: 7726-7734, 2013.
65. Janicke B, Hegardt C, Korgh M, Onning G, Akesson B,
Cirenajwis HM and Oredsson SM: The antiproliferative effect
of dietary ber phenolic compounds ferulic acid and p‑coumaric
acid on the cell cycle of Caco-2 cells. Nutr Cancer 63: 611-622,
2011.
66. Janicke B, Onning G and Oredsson SM: Differential effects of
ferulic acid and p-coumaric acid on S phase distribution and
length of S phase in the human colonic cell line Caco-2. J Agric
Food Chem 53: 6658-6665, 2005.
67. García-Gutiérrez N, Maldonado-Celis ME, Rojas-López M,
Loarca-Piñaa GF and Campos-Vega R: The fermented
non-digestible fraction of spent coffee grounds induces apoptosis
in human colon cancer cells (SW480). J Funct Foods 30: 237-246,
2017.
68. Ekbatan SS, Li XQ, Ghorbani M, Azadi B and Kubow S:
Chlorogenic acid and its microbial metabolites exert anti-prolif-
erative effects, S-phase cell-cycle arrest and apoptosis in human
colon cancer Caco-2 cells. Int J Mol Sci 19: 723, 2018.
69. Hernández-Arriaga AM, Oomah BD and Campos-Vega R:
Microbiota source impact in vitro metabolite colonic production
and anti-proliferative effect of spent coffee grounds on human
colon cancer cells (HT-29). Food Res Int 97: 191-198, 2017.
This work is licensed under a Creat ive Commons
Attribution-NonCommercial-NoDerivatives 4 .0
International (CC BY-NC-ND 4.0) License.
... The S phase is a critical phase during which DNA synthesis occurs. Previous research has shown that inhibition of cyclin A-cdk2 during S phase leads to S phase arrest and apoptosis (Kim et al. 2022). When the cell activates cyclin-CDK-dependent transcription, it signals the decision to initiate a new round of cell division and enter S phase (Ditano, Sakurikar & Eastman 2021). ...
Article
Full-text available
Ovarian cancer is a deadly disease with a poor prognosis, highlighting the urgent need for novel therapeutic alternatives. Pyranocycloartobiloxanthone A (PA), an exceptional xanthone compound has garnered attention due to its diverse medicinal properties. This study aimed to investigate the anticancer properties of PA on metastatic ovarian cancer SKOV-3 cells. Cytotoxicity was evaluated using an MTT assay, while apoptotic mechanisms were determined using AO/PI double staining, annexin V-fluorescein isothiocyanate, multiple cytotoxicity-3, reactive oxygen species (ROS) production, caspases, real-time PCR, western blot, human apoptosis protein profile array and cell cycle analysis. PA inhibited SKOV-3 cell proliferation in a time-dependent manner, with an IC50 of 7.0 ± 0.5 µg/mL and a selectivity index of 13.2 after 72 h of treatment. PA induced apoptosis through the intrinsic apoptotic pathway and arrested the cell cycle at the S phase. PA stimulated ROS production and disrupted the mitochondrial membrane potential, releasing cytochrome c from mitochondria to the cytosol. Additionally, results from human apoptotic protein profile indicated that 21 proteins were upregulated while 22 proteins were downregulated, including Bcl-2, survivin, and HSP70. These findings suggest that PA has the potential as a lead molecule in the development of a chemotherapy drug for ovarian cancer. However, further research is necessary to evaluate the safety and efficacy of PA in preclinical and clinical settings.
... et Schult is a perennial herb that grows on coastal sand dunes worldwide. This plant has been extensively used in oriental traditional medicine for general consumption and as a type of herbal treatment, since it is considered to confer bioactive effects against rheumatic arthritis, sore throat, dropsy, scurvy, fever and diarrhoea (Kim et al. 2022, Hotta et al. 1989. Extracts from C. soldanella have been also used in Portuguese ethno-medicine to cure hydrops, paralysis, rheumatism, and scurvy (Gaspar 1999). ...
Article
Full-text available
a laboratory of ethnobotany and Natural substances, eNs de Kouba, algiers, Faculty of sciences, university of M'sila, algeria; b department of Biological, chemical and Pharmaceutical sciences and technologies (steBIceF), university of Palermo, Palermo, Italy; c NBFc, National Biodiversity Future center, Palermo, Italy; d centro Interdipartimentale di ricerca "riutilizzo bio-based degli scarti da matrici agroalimentari" (rIVIVe), university of Palermo, Palermo ABSTRACT Convolvulus L. and Calystegia R.Br. are two closely related genera of the Convolvulaceae family distributed in Asia, Mediterranean, Macaronesia, East Africa, and Arabia, including about 210 and 30 accepted species, respectively, of flowering plants, present as trees, shrubs, and herbs. The ethnomedical use of Convolvulus species dates to 1730s as they displayed profuse medicinal properties. In the present study, the not previously investigated chemical compositions of the essential oils from aerial parts of Convolvulus althaeoides subsp. tenuissimus (Sm.) Bat., collected in Sicily, and Calystegia sylvatica (Kit.) Griseb., collected in Algeria, were evaluated by GC-MS. The main components of the essential oil of the first one were β-caryophyllene (28.68%), γ-muurolene (23.75%), and γ-elemene (17.55%), whereas the C. silvatica essential oil was shown to be rich of valeranone (10.77%), viridiflorol (9.45%), and germacrene D (8.61%). Furthermore, a complete literature review on the ethno-pharmacological uses of Convolvulus and Calystegia species was performed.
... CDK2 is activated by binding to Cyclin E, which drives the transition from late G1 phase to S phase 25 . P21, an important protein inhibitor in the cell cycle, can also bind to CDK, resulting in the blockage of cell cycle progression 26 . In this study, HCT-8 cells were treated with the indicated concentrations of Corilagin (33.6 μM), 5-FU (58.9 μM) and combination treatment (Corilagin+5-FU 4.6 + 13.8 μM) for 48 h, and then the WB assay was performed to evaluate protein expression levels. ...
Article
Full-text available
Colorectal cancer is one of the most common malignancies worldwide. Although initially effective, patients who receive chemotherapy ultimately experience various complications and develop chemo-resistance, leading to cancer recurrence. Therefore, we aimed to find a drug with good efficacy and low toxicity that could enhance the treatment with 5-Fluorouracil (a commonly used clinical drug) and reduce its dosing. Corilagin, an anti-tumor natural product, has received widespread attention. Glucose regulated protein 78 (GRP78) is overexpressed in colorectal cancer cells and plays a key role in the proliferation, migration and drug resistance of cancer cells. Importantly, GRP78 can affect the apoptosis induced by 5-fluorouracil in CRC cells. In the present study, we determined the synergistic anti-tumor activity of the combination treatment by cell proliferation assay, apoptosis assay, fluorescent staining, cell cycle analysis, WB and PCR assays. This synergistic effect was associated with S-phase blockade, intracellular reactive oxygen species production and downregulation of GRP78. Taken together, our results indicate that Corilagin acts as a potentiator of 5-fluorouracil and may have therapeutic potential for patients with CRC.
... Furthermore, the extract caused an increase in the apoptosisrelated proteins expression levels of Fas, Bad, and Bax, and a decrease in Bcl 2, Bcl xL, procaspase 9, procaspase 8, Bcl 2, procaspase 3, and procaspase 7. Additionally, more cells were depolarized with increased cytochrome c release, S-phase arrest, and reduced cell cycle-related proteins (cyclin A, CDK2, cell division cycle 25 A, and cyclin-dependent kinase inhibitor-1). These results suggest that the DCM fraction of the C. soldanella extract can inhibit cancer in HT 29 cells [76]. ...
... Furthermore, the extract caused an increase in the apoptosisrelated proteins expression levels of Fas, Bad, and Bax, and a decrease in Bcl 2, Bcl xL, procaspase 9, procaspase 8, Bcl 2, procaspase 3, and procaspase 7. Additionally, more cells were depolarized with increased cytochrome c release, S-phase arrest, and reduced cell cycle-related proteins (cyclin A, CDK2, cell division cycle 25 A, and cyclin-dependent kinase inhibitor-1). These results suggest that the DCM fraction of the C. soldanella extract can inhibit cancer in HT 29 cells [76]. ...
Article
Full-text available
Fruits, vegetables, and other edible plants in our diet have numerous health benefits, due to the bioactive compounds in these food items, including polyphenols. These plants are a rich and promising source of natural products and phytochemicals that can be used to treat and prevent numerous diseases and prevent the progression of cancer. Dietary polyphenols exhibit chemo-preventive and therapeutic effects against various ailments, including several types of cancer. The current study focuses on polyphenol’s traditional and advanced extraction methods, with supercritical extraction as a novel approach. It also deals with their identification, bioavailability, and role in preventing and treating colorectal and prostate cancers. Additionally, the article covers the literature that deals with the anticancer activities of polyphenols, as well as their potential use as anticancer agents.
Article
Autophagy is a self‐eating pathway for maintaining normal cellular physiology, while dysregulation of autophagy is associated with cancer progression. Autophagy‐related 4B gene (ATG4B) is a cysteine protease to regulate autophagosome formation and is positively correlated with poor prognosis of colorectal cancer (CRC) patients. An increasing number of reports have implied that ATG4B might be an attractive drug target for CRC. Natural products are the most important source of drug development for cancer therapy due to their high degree of diversity in chemical structure. However, there are few natural products targeting autophagy regulation, especially targeting ATG4B. We aim to identify effective natural compounds from costal plants against ATG4B as potential CRC therapies. We extracted the whole plants, stem, and leaves from nine coastal plant species of Taiwan using different solvents including acetone, methanol, or chloroform. We then evaluated their effects on ATG4B activity and cancer malignancy in CRC cells (DLD‐1, HCT116, and SW620). Among these 26 extracts, we found that the methanol leaf extract of A. elliptifolia significantly inhibited ATG4B proteolytic activity. Moreover, cell viability and colony formation and mobility were decreased in CRC cells treated with the extract. The extract further reduced the number of living cells and induced subG 1 proportion of CRC cells. The cytotoxicity of A. elliptifolia leaf extract was also enhanced in CRC cells under starvation, whereas it had no additional effects in ATG4B or autophagy deficient cells. Taken together, the methanol leaf extract of A. elliptifolia might contains bioactive compounds for inhibiting ATG4B and autophagy activity to diminish viability and mobility of CRC cells, indicating its potential as an anti‐CRC drug for future development.
Article
Land-use change and tourism development have seriously threatened the ecosystems of coastal protection forests and beaches. Light and nutrients are spatially heterogeneously distributed between the two ecosystems. Clonal plants, such as Calystegia soldanella, which play a crucial role in maintaining the ecological stability of coast habitats, are likely to encounter diverse environments. In this study, we investigated clonal integration and the division of labor in C. soldanella under heterogeneous (high nutrient and low light [HNLL]; low nutrient and high light [LNHL]) and homogeneous habitats. We cultivated pairs of connected and severed ramets of C. soldanella in these environments. Our results showed the total biomass (TB) of connected ramets was higher than that of severed ramets in heterogeneous environments, suggesting clonal integration enhances growth in heterogeneous habitats. The root shoot ratio was significantly lower in HNLL than in LNHL conditions for connected ramets, demonstrating a division of labor in growth under heterogeneous conditions. However, parameters of clonal propagation of C. soldanella did not significantly differ between connected and severed ramets in heterogeneous environments, indicating no division of labor in clonal propagation. In homogeneous environments, the growth of C. soldanella did not benefit from clonal integration. Connected ramets in heterogeneous habitats exhibited higher TB than in homogeneous habitats. The TB of one ramet in HNLL was consistently higher than that in LNHL, irrespective of ramet’s states, which suggests that high soil nutrients may enhance the growth. We conclude that C. soldanella has the capability of clonal integration to achieve high biomass in heterogeneous but not in homogeneous conditions, and the establishment of coastal protection forests (high nutrient and low light) may foster the growth of C. soldanella.
Article
Full-text available
Necroptosis is considered a programmed necrosis that requires receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and pore-forming mixed lineage kinase domain-like protein (MLKL) to trigger a regulated cell membrane lysis. Membrane rupture in necroptosis has been shown to fuel innate immune response due to release of damage-associated molecular patterns (DAMPs). Recently published studies indicate that mature erythrocytes can undergo necroptosis as well. In this review, we provide an outline of multiple cell death modes occurring in erythrocytes, discuss possible immunological aspects of diverse erythrocyte cell deaths, summarize available evidence related to the ability of erythrocytes to undergo necroptosis, outline key involved molecular mechanisms, and discuss the potential implication of erythrocyte necroptosis in the physiology and pathophysiology. Furthermore, we aim to highlight the interplay between necroptosis and eryptosis signaling in erythrocytes, emphasizing specific characteristics of these pathways distinct from their counterparts in nucleated cells. Thus, our review provides a comprehensive summary of the current knowledge of necroptosis in erythrocytes. To reflect critical differences between necroptosis of nucleated cells and necroptosis of erythrocytes, we suggest a term erythronecroptosis for necroptosis of enucleated cells.
Article
Full-text available
Colorectal cancer (CRC) is one of the most prevalent and deadliest illnesses all around the world. Growing proofs demonstrate that tumor-associated macrophages (TAMs) are of critical importance in CRC pathogenesis, but their mechanisms remain yet unknown. The current research was designed to recognize underlying biomarkers associated with TAMs in CRC. We screened macrophage-related gene modules through WGCNA, selected hub genes utilizing the LASSO algorithm and COX regression, and established a model. External validation was performed by expression analysis using datasets GSE14333, GSE74602, and GSE87211. After validating the bioinformatics results using real-time quantitative reverse transcription PCR, we identified SPP1, C5AR1, MMP3, TIMP1, ADAM8 as potential biomarkers associated with macrophages in CRC.
Article
Full-text available
Several sesquiterpene lactones (STLs) have been tested as lead drugs in cancer clinical trials. Salograviolide-A (Sal-A) and salograviolide-B (Sal-B) are two STLs that have been isolated from Centaurea ainetensis, an indigenous medicinal plant of the Middle Eastern region. The parent compounds Sal-A and Sal-B were modified and successfully prepared into eight novel guaianolide-type STLs (compounds 1–8) bearing ester groups of different geometries. Sal-A, Sal-B, and compounds 1–8 were tested against a human colorectal cancer cell line model with differing p53 status; HCT116 with wild-type p53 and HCT116 p53−/− null for p53, and the normal-like human colon mucosa cells with wild-type p53, NCM460. IC50 values indicated that derivatization of Sal-A and Sal-B resulted in potentiation of HCT116 cell growth inhibition by 97% and 66%, respectively. The effects of the different molecules on cancer cell growth were independent of p53 status. Interestingly, the derivatization of Sal-A and Sal-B molecules enhanced their anti-growth properties versus 5-Fluorouracil (5-FU), which is the drug of choice in colorectal cancer. Structure-activity analysis revealed that the enhanced molecule potencies were mainly attributed to the position and number of the hydroxy groups, the lipophilicity, and the superiority of ester groups over hydroxy substituents in terms of their branching and chain lengths. The favorable cytotoxicity and selectivity of the potent molecules, to cancer cells versus their normal counterparts, pointed them out as promising leads for anti-cancer drug design.
Article
Full-text available
Background Reineckia carnea is commonly used to treat cough, pneumonia and other diseases in China. In our previous study, it was found that the ethanol extracts of Reineckia carnea have a strong inhibitory effect on the proliferation of human lung cancer A549 cells. Here, we isolated gracillin from ethanol extracts for the first time. Purpose Clarify the antiproliferation effect of gracillin on A549 cells and further explore its mechanisms via the mitochondrial pathway. Methods Gracillin was isolated and purified by silica gel, D-101 macroporous resin and preparative RP-HPLC, then identified by NMR and HR-MS. The inhibitory effects of gracillin on the proliferation of A549 cells were detected by the MTS method. Its mechanisms were further explored by flow cytometry and Western blot. Results A steroid saponin, gracillin, was isolated and identified from Reineckia carnea for the first time. In a concentration-dependent and time-dependent manner, gracillin significantly inhibited the proliferation of A549 cells with an IC50 value at 2.54 μmol/L and induced morphological changes. The results of flow cytometry analysis showed that the apoptosis rate of A549 cells was significantly increased (p < 0.05), and the cells proportion was obviously arrested in S phase. The concentration of intracellular calcium was raised (p < 0.01), and the mitochondrial membrane potential was greatly decreased (p < 0.01). In addition, the expression levels of Bax, caspase-3, cleaved caspase-3, and cytochrome C were dramatically up-regulated while Bcl-2 was down-regulated (p < 0.05) in A549 cells. Conclusion This study confirmed that gracillin has a significant antiproliferative effect on A549 cells. Gracillin could induce the apoptosis of A549 cells through the mitochondrial pathway, which might be associated with regulation of the concentration of intracellular calcium, the mitochondrial membrane potential and the expression levels of Bax, Bcl-2, caspase-3, cleaved caspase-3, and cytochrome C.
Article
Full-text available
Purpose: In order to study novel therapeutic approaches taking advantage of natural compounds showing anticancer and anti-proliferative effects, we focused our interest on S-adenosyl-l-methionine, a naturally occurring sulfur-containing nucleoside synthesized from adenosine triphosphate and methionine by methionine adenosyltransferase, and its potential in overcoming drug resistance in colon cancer cells devoid of p53. Results: In the present study, we demonstrated that S-adenosyl-l-methionine overcomes uL3-mediated drug resistance in p53 deleted colon cancer cells. In particular, we demonstrated that S-adenosyl-l-methionine causes cell cycle arrest at the S phase; inhibits autophagy; augments reactive oxygen species; and induces apoptosis in these cancer cells. Conclusions: Results reported in this paper led us to propose S-adenosyl-l-methionine as a potential promising agent for cancer therapy by examining p53 and uL3 profiles in tumors to yield a better clinical outcomes.
Article
Full-text available
Accumulating evidence indicates that the gut microbiota can promote or inhibit colonic inflammation and carcinogenesis. Promotion of beneficial gut bacteria is considered a promising strategy to alleviate colonic diseases including colitis and colorectal cancer. Interestingly, dietary polyphenols, which have been shown to attenuate colitis and inhibit colorectal cancer in animal models and some human studies, appear to reach relatively high concentrations in the large intestine and to interact with the gut microbial community. This review summarizes the modulatory effects of polyphenols on the gut microbiota in humans and animals under healthy and diseased conditions including colitis and colitis-associated colorectal cancer (CAC). Existing human and animal studies indicate that polyphenols and polyphenol-rich whole foods are capable of elevating butyrate producers and probiotics that alleviate colitis and inhibit CAC, such as Lactobacillus and Bifidobacterium. Studies in colitis and CAC models indicate that polyphenols decrease opportunistic pathogenic or proinflammatory microbes and counteract disease-induced dysbiosis. Consistently, polyphenols also change microbial functions, including increasing butyrate formation. Moreover, polyphenol metabolites produced by the gut microbiota appear to have anticancer and anti-inflammatory activities, protect gut barrier integrity, and mitigate inflammatory conditions in cells and animal models. Based on these results, we conclude that polyphenol-mediated alteration of microbial composition and functions, together with polyphenol metabolites produced by the gut microbiota, likely contribute to the protective effects of polyphenols on colitis and CAC. Future research is needed to validate the causal role of the polyphenol-gut microbiota interaction in polyphenols' anti-colitis and anti-CAC effects, and to further elucidate mechanisms underlying such interaction.
Article
Full-text available
Colorectal cancer (CRC) is among the most lethal and prevalent malignancies in the world and was responsible for nearly 881,000 cancer-related deaths in 2018. Surgery and chemotherapy have long been the first choices for cancer patients. However, the prognosis of CRC has never been satisfying, especially for patients with metastatic lesions. Targeted therapy is a new optional approach that has successfully prolonged overall survival for CRC patients. Following successes with the anti-EGFR (epidermal growth factor receptor) agent cetuximab and the anti-angiogenesis agent bevacizumab, new agents blocking different critical pathways as well as immune checkpoints are emerging at an unprecedented rate. Guidelines worldwide are currently updating the recommended targeted drugs on the basis of the increasing number of high-quality clinical trials. This review provides an overview of existing CRC-targeted agents and their underlying mechanisms, as well as a discussion of their limitations and future trends.
Article
Full-text available
S‑Adenosyl‑L‑methionine (AdoMet) is the principal methyl donor in transmethylation reactions fundamental to sustaining epigenetic modifications. Over the past decade, AdoMet has been extensively investigated for its anti‑proliferative, pro‑apoptotic and anti‑metastatic roles in several types of human cancer. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common type of cancer worldwide, and is an aggressive type of cancer that is associated with a high recurrence rate, metastasis and poor treatment outcomes. The present study demonstrates, for the first time, to the best of our knowledge, that AdoMet induces cell cycle arrest and inhibits the migratory and invasive ability of two different HNSCC cell lines, oral Cal‑33 and laryngeal JHU‑SCC‑011 cells. In both cell lines, AdoMet attenuated cell cycle progression, decreased the protein level of several cyclins and downregulated the expression of p21 cell cycle inhibitor. Moreover, AdoMet was able to inhibit Cal‑33 and JHU‑SCC‑011 cell migration in a dose‑dependent manner after 24 and 48 h, respectively, and also induced a significant reduction in the cell invasive ability, as demonstrated by Matrigel invasion assay monitored by the xCELLigence RTCA system. Western blot analysis of several migration and invasion markers confirmed the inhibitory effects exerted by AdoMet on these processes and highlighted AKT, β‑catenin and small mothers against decapentaplegic (SMAD) as the main signaling pathways modulated by AdoMet. The present study also demonstrated that the combination of AdoMet and cisplatin synergistically inhibited HNSCC cell migration. Taken together, these findings demonstrate that the physiological compound, AdoMet, affects the motility and extracellular matrix invasive capability in HNSCC. Thus, AdoMet may prove to be a good candidate for future drug development against metastatic cancer.
Article
Full-text available
Globally, cancer is the second leading cause of death. Different conventional approaches to treat cancer include chemotherapy or radiotherapy. However, these are usually associated with various deleterious effects and numerous disadvantages in clinical practice. In addition, there are increasing concerns about drug resistance. In the continuous search for safer and more effective treatments, plant-derived natural compounds are of major interest. Plant phenolics are secondary metabolites that have gained importance as potential anti-cancer compounds. Phenolics display a great prospective as cytotoxic anti-cancer agents promoting apoptosis, reducing proliferation, and targeting various aspects of cancer (angiogenesis, growth and differentiation, and metastasis). Phenolic acids are a subclass of plant phenolics, furtherly divided into benzoic and cinnamic acids, that are associated with potent anticancer abilities in various in vitro and in vivo studies. Moreover, the therapeutic activities of phenolic acids are reinforced by their role as epigenetic regulators as well as supporters of adverse events or resistance associated with conventional anticancer therapy. Encapsulation of phyto-substances into nanocarrier systems is a challenging aspect concerning the efficiency of natural substances used in cancer treatment. A summary of phenolic acids and their effectiveness as well as phenolic-associated advances in cancer treatment will be discussed in this review.
Article
Full-text available
Colorectal cancer (CRC) is the second most common cancer in females and the third in males worldwide. Conventional therapy of CRC is limited by severe side effects and by the development of resistance. Therefore, additional therapies are needed in order to combat the problem of selectivity and drug resistance in CRC patients. Inula viscosa (IV) is a well-known medicinal perennial herb in traditional medicine. It is used for different therapeutic purposes, such as; topical anti-inflammatic, diuretic, hemostatic, antiseptic, antiphlogistic, and in the treatment of diabetes. Several studies attempted to reveal the anti-cancer activity of different extracts prepared by different organic solvents from different parts of the IV plant. The aim of the present study is to examine the potential beneficial effects of IV leaf aqueous extract on the growth of colon cancer cells in vitro and in vivo. The results indicated that exposure of colorectal cancer cells to IV extract, significantly reduced cell viability in a dose and time dependent manner. Moreover, treatment of cells with 300 μg/ml of IV extract induced apoptosis, as it was detected by Annexin V/FITC/PI, TUNEL assay, and the activation of caspases. In vivo studies revealed that treatment with 150 or 300 mg/kg IV extract inhibited tumor growth in mice transplanted with MC38 cells. Tumors' weight and volume were significantly (P < 0.001) reduced when compared to untreated-control group. Staining of the paraffin section of tumors revealed that IV treatment inhibited cell proliferation and induced apoptosis. Additionally, no side effects such as; weight loss, behavior changes, ruffled fur or changes in kidney, and liver functions were observed. These results may indicate that active doses of IV extract are not toxic. Further studies are needed in order to identify the structure of the active compounds. Results from this study may contribute to the development of new and efficient strategies for treatment of human colon cancer.
Chapter
Seaweeds represent some of the most important reservoirs of new remedial compounds for humans. The natural products obtained from seaweeds have received extensive consideration on account of their extraordinary dietary and pharmacology applications, having such antiviral, antifungal, antibacterial, and anticancer properties and so on. Above all, there are a number of natural products that have demonstrated attractive value for the development of novel anticancer agents. This book chapter draws attention on colorectal cancer which is one of the main causes of cancer-related demise of human beings. This chapter also illustrates a range of active natural products extracted from seaweeds that have shown to eliminate or slow the progression of cancer. This also covers the mechanism through which these compounds can induce apoptosis in vitro and in vivo. By considering the ability of compounds present in seaweeds to act against colorectal cancers, this chapter highlights the potential use of seaweeds as anticancer agents.
Article
Cell death was once believed to be the result of one of two distinct processes, apoptosis (also known as programmed cell death) or necrosis (uncontrolled cell death); in recent years however several other forms of cell death have been discovered highlighting that a cell can die via a number of differing pathways. Apoptosis is characterised by a number of characteristic morphological changes in the structure of the cell, together with a number of enzyme‐dependent biochemical processes. The result being the clearance of cells from the body, with minimal damage to surrounding tissues. Necrosis however, is generally characterised to be the uncontrolled death of the cell, usually following a severe insult, resulting in spillage of the contents of the cell into surrounding tissues and subsequent damage thereof. Failure of apoptosis and the resultant accumulation of damaged cells in the body can result in various forms of cancer. An understanding of the pathways is therefore important in developing efficient chemotherapeutics. It has recently become clear that there exists a number of subtypes of apoptosis and that there is an overlap between apoptosis, necrosis and autophagy. The goal of this review is to provide a general overview of the current knowledge relating to the various forms of cell death, including apoptosis, necrosis, oncosis, pyroptosis and autophagy. This will provide researchers with a summary of the major forms of cell death and allow them to compare and contrast between them. This article is protected by copyright. All rights reserved.