3-β-Εrythrodiol isolated from Conyza canadensis inhibits MKN‑45 human gastric cancer cell proliferation by inducing apoptosis, cell cycle arrest, DNA fragmentation, ROS generation and reduces tumor weight and volume in mouse xenograft model

Retraction in: /10.3892/or.2021.7980

  • Authors:
    • Kai Liu
    • Yue-Hong Qin
    • Jian-Yong Yu
    • Heng Ma
    • Xi-Lin Song
  • View Affiliations

  • Published online on: February 3, 2016     https://doi.org/10.3892/or.2016.4610
  • Pages: 2328-2338
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The objective of the present study was to investigate the in vitro and in vivo anticancer and apoptotic effects of 3-β-erythrodiol, a plant-derived triterpene against MKN-45 human gastric cancer cells. In addition, effects on cellular morphology, cell cycle phase distribution, DNA fragmentation, and ROS generation were also elucidated in the current research work. Cytotoxic activity of 3-β-erythrodiol was demonstrated by MTT cell viability and LDH assay. Cellular morphological study was carried out using phase contrast, fluorescence and scanning electron microscopy. Cell cycle analysis was evaluated by flow cytometry and gel electrophoresis was used to evaluate DNA fragmentation pattern. The results of the present study revealed that 3-β-erythrodiol induced dose-dependent as well as time-dependent anticancer effects in MKN-45 gastric cancer cells. Cellular morphological changes in MKN-45 cells as indicated by fluorescence and scanning electron microscopy were induced by 3-β-erythrodiol. This triterpene induced both early and late apoptotic features in these cancer cells. 3-β-Erythrodiol treatment led to sub-G1 cell cycle arrest with a corresponding decrease in S-phase cells and an increase in G2/M phase cells. DNA fragments were evident in gel electrophoresis experiment following 3-β-erythrodiol treatment. It was observed that 0.50 and 1.0 µg/g 3-β-erythrodiol injection reduced the tumor weight from 1.4 g in PBS-treated group (control) to 0.61 and 0.22 g, respectively. Similarly, 0.50 and 1.0 µg/g 3-β-erythrodiol injection reduced the tumor volume from 1.5 cm3 in PBS-treated group (control) to 0.91 and 0.31 cm3, respectively. The present investigation indicates that 3-β-erythrodiol exerts anti-proliferative effects in human gastric cancer by inducing early and late apoptosis, cell cycle arrest, and ROS generation. It also decreased the tumor volume and tumor weight in male Balb/c nude mice.

Introduction

Gastric cancer is the most common malignant cancer of the gastrointestinal tract in the world. Gastric cancer is the second leading cause of mortality in the world after lung cancer. It is estimated that more than 750,000 new cases of gastric cancer are diagnosed every year throughout the globe (1). Gastric cancer has been found to be more prevalent in people aged 60 years or above. In 2005, the incidence rate of gastric cancer (0,3 million deaths and 0,4 million new cases) ranked third among the most common cancers in China (2). The actual cause of gastric cancer is unknown but it has been linked with low vitamin intake and a high salt diet. A diet high in vegetables and fruits, citrus fruits, and fiber has been linked with lower risk of gastric cancer. Epigenetic fluctuation plays crucial roles in the initiation and progression of human gastric cancers. Gastric cancer is mostly asymptomatic with only non-specific symptoms in its initial stages (3,4). Consequently, by the time symptoms occur, the tumor has usually metastasized to other parts of the body. Regarding treatment of gastric cancer, surgery is still the best and last line of treatment for gastric cancer. Radiation therapy and chemotherapy as alternatives for surgery in the treatment of gastric cancer are not very promising. Despite the significant development of new surgical techniques, radiotherapy, chemotherapy, and targeted therapy, failures in gastric cancer treatment are still the most important challenges in oncology (5,6). The prognosis of gastric cancer is very poor, however, if detected at an early stage, long-term survival of gastric cancer is highly possible (7). As such there is an urgent need for the design and development of novel chemotherapeutic agents for the treatment of gastric cancer.

Among the new treatment regimens proposed for gastric cancer, is the use of complementary and alternative medicines. Numerous plant-derived substances, and their derivatives, are effective antitumor and chemopreventive agents. Naturally occurring plant derived molecules/extracts constitute a promising group of anticancer agents and have always played crucial roles for the treatment of numerous human cancers. In fact, more than 75% of the total number of commercially offered and clinically approved anticancer agents are either natural plant products or their semisynthetic derivatives. The well-known examples of plant based anticancer drugs are paclitaxel, vinblastine, vincristine, and camptothecin (811).

The aim of the present investigation was to assess the in vitro and in vivo anticancer and apoptotic activities of 3-β-erythrodiol isolated from Conyza canadensis in MKN-45 gastric cancer cells and a mouse xenograft model. Effects of 3-β-erythrodiol on cell cycle arrest, ROS generation and DNA fragmentation were also evaluated. No previous reports on the anticancer and apoptotic activities of 3-β-erythrodiol against MKN-45 gastric cancer cells have been reported, thus, the present study constitutes the first such report.

Materials and methods

Chemicals and reagents

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) were purchased from Sigma-Aldrich Inc. (St. Louis, MO, USA). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin, streptomycin, trypsin, phosphate-buffered saline (PBS) with calcium chloride and magnesium chloride were obtained from Hangzhou Sijiqing Biological Engineering Materials Co., Ltd. (Hangzhou, China). All other chemicals and solvents used were of the highest purity grade.

Plant material

The roots of Conyza canadensis were collected in May 2014 from Jinan City Shandong Province, China and identified by Professor Heng Song, a voucher specimen (voucher specimen no. 24-777-024-14) was deposited in the Herbarium of Southeast University (Nanjing, China).

Extraction, isolation and spectral data analysis

The air dried, finely powdered root material (2 kg) was extracted for 72 h with ethyl acetate in a soxhlet apparatus to afford the extract, which was concentrated under reduced pressure. The ethyl acetate extract (45 g) was charged on silica gel (60–120 mesh, 200 g) column and eluted with an increasing gradient of petroleum ether and ethyl acetate. Fractions of 50 ml volume each were collected and pooled according to TLC analysis. Five major fractions were collected (19:1, 9:1, 4:1, 1:1, 1:4). The fraction (petroleum ether:ethyl acetate, 19:1) yielded 3-β-erythrodiol as colorless crystalline solid, m.p. 229°C. The compound in its EIMS gave the molecular ion peak at m/z 442.3786 (calculated for C30H50O2, 442.3810). The IR spectrum displayed absorption at 3,580, 3,430 cm−1 due to hydroxyl group and 1,610 cm−1 due to double bond. The 1H NMR spectrum displayed resonance signals due to seven tertiary methyl groups at δ 1.15, 0.98, 0.93, 0.92, 0.87, 0.86, 0.78, besides, a resonance signal for an olefinic proton at δ 5.70 (1H, t, J=3.6 Hz, H-12). The 1H NMR spectrum of the compound displayed the resonance signal due to a carbinylic proton at δ 3.12 (1H, d, Jax-ax=11.2 Hz, Jax-eq= 4.5 Hz, H-3α) indicating that the hydroxyl group at C-3 was equatorially oriented. The 13C NMR spectral data of the compound indicated resonance for thirty carbons, resolved as, seven methyls, ten methylenes, five methines and seven quaternary carbons. EIMS m/z: 442 (M) +, 409, 234, 216, 207, 204, 203, 189, 107, 95, 69. IR (KBr) ν max cm−1: 3,580, 3,430 (O-H), 1,610 (C=C). 1H NMR (CDCl3, 500 MHz) δ: 5.70 (1H, t, J=3.6 Hz, H-12), 3.12 (1H, d, Jax-ax=11.2 Hz, Jax-eq=4.5 Hz, H-3), 1.15 (3H, s, H-27), 0.98 (3H, s, H-23), 0.93 (3H, s, H-26), 0.92 (3H, s, H-25), 0.87 (3H, s, H-30), 0.86 (3H, s, H-29), 0.78 (3H, s, H-24). 13C NMR (CDCl3, 125 MHz) δ: 38.6 (C-1), 27.2 (C-2), 79.0 (C-3), 38.8 (C-4), 55.1 (C-5), 18.4 (C-6), 32.6 (C-7), 39.8 (C-8), 47.6 (C-9), 36.9 (C-10), 23.6 (C- 11), 122.4 (C-12), 144.2 (C-13), 41.8 (C-14),25.6 (C-15), 22.0 (C-16), 36.9 (C-17), 42.3 (C-18), 46.5 (C-19), 30.9 (C-20), 34.1 (C-21), 31.0 (C-22), 28.1 (C-23), 15.6 (C-24), 15.5 (C-25), 16.6 (C-26), 25.9 (C-27), 69.5 (C-28), 33.2 (C-29), 23.11 (C-30). Comparison of physical characteristics and spectral data with that reported in literature (12), confirmed it to be 3-β-erythrodiol.

Cell line and culture conditions

The MKN-45 human gastric cancer cell line was purchased from the Shanghai Institute of Biochemistry and Cell Biology (Shanghai, China). These cells were cultured in DMEM supplemented with 10% (v/v) FBS (Hyclone, USA) under humidified atmosphere of 5% CO2 at 37°C. The medium was replaced every 2 days. Cells were subcultured every 4 days.

MTT assay for cell proliferation

The effects of 3-β-erythrodiol on MKN-45 cell viability were examined by MTT assay. Cells (2×105 cells/well in 100 µl medium) were seeded into 96-well plates for 24 h before drug treatment. Subsequent drug treatment with various doses of 3-β-erythrodiol (0, 2.5, 5, 25, 50, 75 and 100 µM) for 24 and 48 h, the cell plates were then treated with MTT solution (10 µl; 5 mg/ml in PBS solution) for an additional 2 h at 37°C. The formazan crystals in viable cells were solubilized with DMSO (150 µl) and the absorbance was measured on a microplate reader (Bio-Rad, Hercules, CA, USA) at a wavelength of 490 nm. The effects of 3-β-erythrodiol on cell viability were calculated as an inhibition ratio (1%) using the following equation (OD490, optical density at 490 nm):

Evaluation of LDH release

MKN-45 human gastric cancer cells were seeded into a 96-well plate and treated with 3-β-erythrodiol for 48 h, then 20 µl LDH was added to release the reagent. After 1 h, the cell culture plate was centrifuged at 500 g for 15 min and 150 µl supernatant from each well was collected into a new black 96-well plate. Next, 100 µl of LDH assay mixture was added to each well, and the plate was incubated at 37°C for 20 min. The absorbance was measured spectrophotometrically at 490 nm wavelength.

Phase contrast and fluorescence microscopic study of gastric cancer cell morphology

MKN-45 gastric cancer cells were seeded into 6-well plates at a density of 2×105 cells/well in 20 ml medium. The cells were treated with varying concentrations (0, 5, 50 and 100 µM) of 3-β-erythrodiol for 48 h. The morphological alterations were observed and the images were captured under an inverted light microscope (Nikon Corporation, Tokyo, Japan) after 48 h. The same spot of cells was noted and captured. The images were captured at a magnification of ×200.

The fluorescence microscopic staining images were recorded using a UV fluorescence microscope (Olympus; Olympus Optical Co., Ltd., Tokyo, Japan) using UV filter at ×200 magnification to detect morphological evidence of apoptosis. In brief, MKN-45 human gastric cancer cells were seeded into 12-well plates at a density of 2×105 cells/well and then treated with 0, 5, 50 and 100 µM dose of 3-β-erythrodiol for 48 h. The apoptotic changes were evaluated by Hoechst staining kit according to the instructions of the manufacturer. After drug treatment, the cells were fixed with 5% polyoxymethylene and then incubated in Hoechst solution for 10–15 min in the dark. The images were then captured by the fluorescence microscope.

Propidium iodide and acridine orange double staining assay for apoptosis quantification

The apoptotic cell death induced by 3-β-erythrodiol in MKN-45 human gastric cancer cells was quantified by using propidium iodide (PI) and acridine orange (AO) double staining as per the manufacturer guidelines and the cells were observed under fluorescence microscope (Olympus; Olympus Optical Co., Ltd.). Briefly, MKN-45 cells were plated at a density of 1×105 cells/ml, and treated with 0, 5, 50 and 100 µM of 3-β-erythrodiol for 48 h. The cells were incubated in 5% CO2 atmosphere at 37°C. The cells were then centrifuged at 12,000 rpm for 10 min, supernatant was discarded and the cells were washed twice using PBS. Ten microliters each of propidium iodide and acridine orange were added into the cell pellet. Freshly stained cell suspension was dropped into a glass slide and covered by a coverslip. The glass slides were examined under a UV-fluorescence microscope.

Scanning electron microscopy (SEM) studies of exterior ultra-structural cellular features

MKN-45 human gastric cancer cells at a density of 2×106 cells were seeded in 6-well microtitre plates. Various doses of 3-β-erythrodiol were added to the cell culture followed by incubation of 12 h. The cells were centrifuged at 10,000 rpm for 10 min followed by PBS washing. The supernatant was removed and resuspended in 10 ml of 0.1 M cacodylate buffer. The sample was again centrifuged at 10,000 rpm for 5 min. The supernatant was removed and the pellet was resuspended in 5 ml of 3.2% glutaraldehyde in 0.1 M cacodylate buffer. The cacodylate buffer was removed and 1.2% of osmium tetroxide (OsO4) in 0.2 M cacodylate buffer was added. The cover slips were washed three times with 0.2 M cacodylate buffer. Then 30% ethanol for 5 min was added. The coverslips were glued onto stubs with silver print and dried for about 20 min at room temperature. Then the samples were covered with gold using sputter coater. The images were captured by a scanning electron microscope (JOEL 64000, Japan) at accelerating voltage of 15–25 KV).

Measurement of intracellular ROS generation

Intracellular ROS generation was evaluated using f luorescent CM-DCFH2-DA. MKN-45 cells were seeded in 6-well plates and after adhesion, the cells were pretreated with 10 µM CM-DCFH2-DA for 30 min followed by co-incubation with various concentrations of 3-β-erythrodiol for another 3 h and washed with ice-cold PBS twice. The cells were collected and analyzed using a flow cytometry (FACSCanto™; Becton Dickinson, Franklin Lakes, NJ, USA) with wavelength of excitation and emission at 488 and 525 nm, respectively.

DNA fragmentation analysis by gel electrophoresis

MKN-45 human gastric cancer cells were seeded in a 100-mm cell culture dish for 24 h, and treated with 0, 5, 50 and 100 µM 3-β-erythrodiol for 48 h. The cells were harvested and washed with PBS, and the pellets were lysed with a 400-µl DNA lysis buffer (2% NP-40, 20 mM EDTA, 40 mM Tris-HCl) for 30 min. After centrifugation, the supernatants were prepared in an equal volume of 1.5% sodium-dodecyl sulphate, incubated with 2.5 mg/ml RNase A at 60°C for 2 h followed by digestion with 2.5 mg/ml proteinase K for 2 h at 20°C. Following the addition of 0.5 volumes of 10 M ammonium acetate, the DNA was precipitated with cold ethanol and collected by centrifugation at 15,000 rpm for 30 min. DNA was then dissolved in gel loading buffer, separated by electrophoresis in 1.5% agarose gel and visualized under UV light, following ethidium bromide staining.

Effect of 3-β-erythrodiol on cell cycle progression

Effect of 3-β-erythrodiol on cell cycle was analyzed by flow cytometry using a FACSCalibur instrument (BD Biosciences, San Jose, CA, USA), equipped with CellQuest 3.3 software. ModFit LT cell cycle analysis software was used to determine the percentage of cells in the various phases of the cell cycle. Briefly, MKN-45 human gastric cancer cells (1×105 cells) were treated with numerous doses of 3-β-erythrodiol (0, 5, 50 and 100 µM) for 48 h. Subsequently, the cells were collected, washed with ice cold PBS, fixed with 70% alcohol at 4°C for 12 h and stained with propidium iodide in the presence of 3% RNAase A at 37°C for 20 min prior to analysis using flow cytometry.

In vivo experiments

The effects of 3-β-erythrodiol on tumor development were examined using a nude mouse model. Male Balb/c nude mice (6 weeks old) were purchased from SLAC Laboratory Animal Co. (Shanghai, China), and all mice were maintained with water and food ad libitum in a pathogen free environment with a 12-h light and 12-h dark cycle in an animal care facility and according to Animal Welfare regulations and protocols approved by the Institutional Animal Care and Use Committee of Shandong Provincial Hospital (Jinan, Shandong, China). The MKN-45 human gastric cancer cells (1×105 cells/mouse) were subcutaneously injected into the right rear flank of each mouse (5–6 mice/group) to produce tumors in mice. After tumor development, the mice were divided into 3 groups and treated with 3-β-erythrodiol injected intraperitoneally. The control group in the study was treated with an equal amount of PBS. Afterwards, the mice were sacrificed after 24 days and the tumor weight and volume of each mouse were evaluated. Tumor length and width were measured using a caliper and the tumor volume was calculated using the formula: tumor volume = length × width × 0.5 width.

Statistical analysis

The results indicate values from three independent experiments with the data expressed as the means ± SD. Differences between the control and treatment groups were examined using the Student's t-test with SPSS 17.0 software. A p-value <0.05 was considered to indicate a statistically significant difference.

Results

Effects of 3-β-erythrodiol on the cytotoxicity of human gastric cancer cells (MKN-45)

The structure of 3-β-erythrodiol is shown in Fig. 1. The cytotoxic effects of the 3-β-erythrodiol in human gastric cancer cells were evaluated by MTT as well as LDH release assay. Initially we demonstrated the anticancer activity of 3-β-erythrodiol on MKN-45 cells by using MTT assay. The results revealed that 3-β-erythrodiol exerted potent anti-proliferative effects on these cancer cells. It showed both concentration-dependent as well as time-dependent growth inhibitory effects against these cells (Fig. 2). For determining the effectiveness of this triterpene compound, its IC50 value was also calculated to be 45.2 and 21.6 µM at 24 and 48 h, respectively. In addition, the lactate dehydrogenase (LDH) released to the culture medium was also increased in a concentration-dependent manner (Fig. 3). These two assays indicated that 3-β-erythrodiol induced potent cytotoxic effects in MKN-45 cells in a dose-dependent manner.

Morphological study of MKN-45 human gastric cancer cells using phase contrast and fluorescence microscopy

In this study, the morphological alterations of human gastric cancer cells (MKN-45) untreated and treated with 3-β-erythrodiol were identified under a phase contrast microscope. In comparison to the control-treated cells, the cells treated with 5, 50 and 100 µM of 3-β-erythrodiol exhibited a significant reduction in cell viability (Fig. 4). Untreated MKN-45 cells were tightly packed and organized multilayers, whereas after incubation with various concentrations of 3-β-erythrodiol for 48 h numerous cells became rounded and withered, and disconnected from each other or floated in the medium.

The process of apoptosis is characterized by certain morphological changes including reduction in cell volume, and chromatin condensation in the nucleus. To confirm whether 3-β-erythrodiol induces apoptosis in MKN-45 human gastric cancer cells, fluorescence microscopy using Hoechst 33342 as a staining agent was used. Following treatment with 5, 50 and 100 µM dose of 3-β-erythrodiol in MKN-45 cells, the most prominent morphological changes were observed in the treated cells as compared to the untreated cells. As shown by phase contrast microscopy (Fig. 4), the untreated control cells were morphologically normal. Reduction in the cell population and change in cellular morphology were observed with 3-β-erythrodiol treatment. Higher doses of 3-β-erythrodiol induced chromatin condensation, fragmented nuclei and nuclear shrinkage.

3-β-Erythrodiol induced early and late apoptosis (propidium iodide and acridine orange double staining assay)

Apoptotic, necrotic, and living MKN-45 gastric cancer cells were counted under a fluorescence microscope. Around 400 cells were randomly and differentially counted and it was observed that 3-β-erythrodiol initiated and induced apoptotic morphological features in a concentration-dependent manner (Fig. 6). Untreated cells (Fig. 6A) were shown to have green color showing normal nuclear structure. Early apoptosis is detected by intercalation of acridine orange within the fragmented DNA, which is indicated by fluorescent bright-green color (Fig. 6B). At higher doses of 3-β-erythrodiol, nuclear margination and blebbing was detected while as at very high doses of 3-β-erythrodiol, late stages of apoptosis were observed with appearance of apoptotic body formation and reddish-orange fluorescence (Fig. 6C and D). Differential counting of treated MKN-45 cells indicated that there is a statistically significant (P<0.05) difference in apoptosis positive cells, which indicates clearly that 3-β-erythrodiol has a concentration-dependent apoptogenic effect. The percentage of apoptotic cells increased from 5.2% in control cells to 26.5, 43.2 and 74.3% in 5, 50 and 100 µM 3-β-erythrodiol-treated cells, respectively.

MKN-45 cell surface analysis by scanning electron microscopy (SEM)

SEM was carried out indicating the cell surface morphology. The procedure of SEM in the analysis of apoptosis is primarily referred to the study of cell surface alterations such as smoothing, loss of microvillus structures, blebbing, and shrinking. As shown in Fig. 7A, SEM examination reveals that untreated MKN-45 human gastric cancer cells are spherical in shape with smooth surface. However as shown in Fig. 7B, 3-β-erythrodiol treatment to MKN-45 cells at 50 µM resulted in few surface projections and blebbing of the plasma membrane. Furthermore, when the concentration of the 3-β-erythrodiol was enhanced to 100 µM, a complete apoptotic body formation was observed. Overall, the SEM data clearly demonstrated that the atypical apoptotic phenomena occurred in MKN-45 cells treated with 3-β-erythrodiol. However, the relative appearance of apoptotic features were more evident at higher doses.

3-β-Erythrodiol induces ROS formation in MKN-45 human gastric cancer cells

The effect of 3-β-erythrodiol on intracellular ROS production was measured by flow cytometry with a fluorescent probe CM-DCFH2-DA. As shown in Fig. 8, after treating MKN-45 cells with 3-β-erythrodiol for 3 h, it profoundly induced ROS formation. A concentration dependent ROS generation was witnessed and 3-fold increase of ROS production was seen after 100 µM 3-β-erythrodiol treatment.

3-β-Erythrodiol induces DNA fragmentation in MKN-45 gastric cancer cells

In addition to the morphological changes of apoptosis in 3-β-erythrodiol-treated MKN-45 cells, DNA fragmentation was also studied by observation of the formation of DNA ladder. As shown in Fig. 9, DNA ladder seemed to be more marked with the increasing 3-β-erythrodiol concentration, however, no DNA fragments were observed in the control groups (Fig. 9, 0 µM). However, 5, 50 and 100 µM doses of the 3-β-erythrodiol after 48 h exposure led to a substantial increase in DNA fragmentation (Fig. 9, right panel). The DNA fragmentation is a hallmark of apoptosis, further confirming that the 3-β-erythrodiol induced cell death via apoptosis.

Effect of 3-β-erythrodiol on cell cycle phase distribution in MKN-45 human gastric cancer cells

Apoptosis and cell cycle are intimately connected biochemical processes, and any disruption in cell cycle progression may eventually result in apoptotic cell death. With the purpose of having a mechanistic indication of the growth inhibitory effect exerted by 3-β-erythrodiol in MKN-45 cancer cells, flow cytometry analysis was performed to identify whether the compound induces cell cycle arrest in this cell line. The results indicated that 3-β-erythrodiol induces sub-G1 cell cycle arrest and increases the portion of apoptotic cells. To determine the distribution of 3-β-erythrodiol-treated MKN-45 cells in different phases of the cell cycle, DNA content in cells was detected by propidium iodide (PI) staining and flow cytometry. Treatment with different concentrations of the compound for 48 h led to an increase in the population of cells in the sub-G0/G1 phase (apoptotic population) (P<0.05) (Fig. 10). This increase in sub-G1 population was accompanied by a corresponding reduction of the cells in S-phase and an increase in G2/M phase of the cell cycle. As compared to the control (Fig. 10A), 3-β-erythrodiol-treated (5 µM Fig. 10B, 50 µM C, and 100 µM D) cells showed a significant proportion of cells in apoptosis (sub-G1).

3-β-Erythrodiol reduces tumor volume and tumor weight in male Balb/c nude mice

In vitro findings reveal that 3-β-erythrodiol is a potent cytotoxic agent inhibiting cell proliferation and inducing apoptosis and cell cycle arrest. Next step was to demonstrate whether this compound induces the same anticancer effects also in vivo. Cancer was induced in the mice by injecting MKN-45 cancer cells (1×106 cells/mouse). Subsequent to the tumor development, the mice were sacrificed and tumors were removed and their weights and volumes were measured (Fig. 11). The findings revealed that 0.50 and 1.0 µg/g 3-β-erythrodiol injection decreased the tumor weight from 1.40 g in PBS-treated group (control) to 0.61 and 0.22 g, respectively (Fig. 11A and B). Similarly, 0.50 and 1.0 µg/g 3-β-erythrodiol injection reduced the tumor volume from 1.5 cm3 in PBS-treated group (control) to 0.91 and 0.31 cm3, respectively (Fig. 11A and C).

Discussion

Conyza canadensis, commonly referred to as Canada fleabane. The other names include horseweed, butterweed, Canadian horseweed (13). It is an annual plant native of Northern and Central America, but has spread over almost all parts of the world. Conyza canadensis is an annual plant growing to 1.5 m tall, with sparse hairy stems. The leaves are slender, 2–10 cm long and up to 1 cm broad, with a coarse toothed margin. The aerial parts of Conyza canedensis have been used in different parts of the world to treat several ailments, most commonly diarrhoea and dysentery, and as a diuretic agent. The petroleum ether and ethanolic extracts of aerial parts exhibit significant anti-inflammatory activity (14). In Chinese folk medicine, Conyza canadensis has also been applied for the treatment of wounds, swellings, and pain caused by arthritis (15). The whole plant is locally used for the treatment of edema, hematuria, hepatitis and cholecystitis (16). Moreover, a decoction of horse-weed has traditionally been used to treat cancerous diseases in North America (17). Numerous species of genus Conyza have been reported to be rich sources of diterpenes (18,19). Earlier phytochemical studies of C. canedensis revealed the presence of triterpenoids, sterols, sphingolipids (20,21), specific C-10 acetylenes 46–47, sesquiterpene hydrocarbons (22) and flavonoids (23). Recently, a triterpenoid ester, 3-β, 16-β, 20-β-trihydroxytaraxa-stane-3-O-palmitoyl ester, and phenylpropanoyl 2,7-anhydro-3-deoxy-2-octulosonic acid derivatives (24) were isolated. Phytochemical analysis of the essential oil of C. canedensis revealed constituents, including monoterpenes, sesquiterpenes, and acetylenes, among which d-limonene was the predominant constituent (25).

In this investigation, our aim was to evaluate the in vitro and in vivo anticancer and apoptotic effects of 3-β-erythrodiol against MKN-45 human gastric cancer cells and in a mouse xenograft model. 3-β-Erythrodiol was initially isolated from the ethyl acetate root extract of Conyza canadensis by column chromatography and then its structure was evaluated by different spectral techniques.

Many plant based compounds have been reported to curb cancer cell growth which paves the way for anticancer drug discovery. It has also been reported that reactive oxygen species (ROS)-mediated DNA damage after treatment with plant based chemotherapeutic agents is an essential contributing factor in the induction of apoptosis and cell death (26). Therefore, agents that can cause DNA damage and subsequent apoptosis will act as potent anticancer drugs to control the process of tumorigenesis and tumor recurrence (27,28). Furthermore, development of good leads for drug discovery should selectively be able to induce apoptosis in cancer cells (29) without causing excessive damage to normal cells. Apoptosis may be useful in the management and therapy of cancer. Apoptosis gives clues on effective anticancer therapy and many chemotherapeutic agents have been reported from the search of herbal and other natural products influencing apoptosis (30).

The current investigation revealed that 3-β-erythrodiol induced apoptosis in MKN-45 human gastric cancer cells. At lower concentration of 3-β-erythrodiol, nuclear margination and blebbing was detected while at very high doses of 3-β-erythrodiol, late stages of apoptosis were observed with appearance of apoptotic body formation with reddish-orange fluorescence. Differential counting of treated MKN-45 cells indicated that there is a statistically significant (P<0.05) difference in apoptosis positive cells, which indicates clearly that 3-β-erythrodiol has a concentration-dependent apoptogenic effect. The scanning electron microscopy data clearly demonstrated that the atypical apoptotic phenomena occurred in MKN-45 cells treated 3-β-erythrodiol with appearance of few surface projections and blebbing of the plasma membrane. After treating MKN-45 cells with 3-β-erythrodiol for 3 h, it strongly induced ROS formation. A dose-dependent ROS generation was witnessed and 3-fold increase of ROS production was seen after 100 µM 3-β-erythrodiol treatment. DNA ladder seemed to be more evident with the increasing 3-β-erythrodiol concentration, however, no DNA fragments were observed in the control groups. The DNA fragmentation is a hallmark of apoptosis, further confirming that the 3-β-erythrodiol induced cell death via apoptosis. 3-β-Erythrodiol also induced sub-G1 cell cycle arrest in these cancer cells. Disruption in cell cycle progression may also result in apoptotic cell death because there is an intimate relation between cell cycle and apoptosis. 3-β-Erythrodiol has been earlier reported to exhibit anti-proliferative and pro-apoptotic activities in astroglial tumor cells (1321N1) by inducing sub-G0/G1 cell cycle arrest (31).

In vivo studies revealed that 3-β-erythrodiol injection decreased the tumor weight and volume in male Balb/c nude mice. It was observed that 0.50 and 1.0 µg/g 3-β-erythrodiol injection decreased the tumor weight from 1.40 g in PBS-treated group (control) to 0.61 and 0.22 g, respectively, while the tumor volume was reduced from 1.5 cm3 in PBS-treated group (control) to 0.91 and 0.31 cm3 after treating with 0.50 and 1.0 µg/g 3-β-erythrodiol injection, respectively.

In conclusion, the present investigation reveals that 3-β-erythrodiol isolated from Conyza canadensis exerts potent anti-proliferative effects in human gastric cancer by inducing early and late apoptosis, cell cycle arrest, and ROS generation. It also decreased the tumor volume and tumor weight in male Balb/c nude mice.

Acknowledgments

This study was supported by the Traditional Chinese Medicine Science and Technology Development Plan of Shandong Province (no. 2013-210).

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar

2 

Yang L, Parkin DM, Ferlay J, Li L and Chen Y: Estimates of cancer incidence in China for 2000 and projections for 2005. Cancer Epidemiol Biomarkers Prev. 14:243–250. 2005.PubMed/NCBI

3 

Hwang H, Dwyer J and Russell RM: Diet, Helicobacter pylori infection, food preservation and gastric cancer risk: Are there new roles for preventative factors? Nutr Rev. 52:75–83. 1994. View Article : Google Scholar : PubMed/NCBI

4 

Rasul A, Yu B, Yang LF, Ali M, Khan M, Ma T and Yang H: Induction of mitochondria-mediated apoptosis in human gastric adenocarcinoma SGC-7901 cells by kuraridin and Norkurarinone isolated from Sophora flavescens. Asian Pac J Cancer Prev. 12:2499–2504. 2011.

5 

Baeza MR, Giannini TO, Rivera SR, González P, González J, Vergara E, del Castillo C, Madrid J and Vinés E: Adjuvant radiochemotherapy in the treatment of completely resected, locally advanced gastric cancer. Int J Radiat Oncol Biol Phys. 50:645–650. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Longley DB, Harkin DP and Johnston PG: 5-fluorouracil: Mechanisms of action and clinical strategies. Nat Rev Cancer. 3:330–338. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Jemal A, Center MM, DeSantis C and Ward EM: Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev. 19:1893–1907. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Newman DJ, Cragg GM and Snader KM: The influence of natural products upon drug discovery. Nat Prod Rep. 17:215–234. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Newman DJ and Cragg GM: Natural products as sources of new drugs over the last 25 years. J Nat Prod. 70:461–477. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Oberlies NH and Kroll DJ: Camptothecin and taxol: Historic achievements in natural products research. J Nat Prod. 67:129–135. 2004. View Article : Google Scholar : PubMed/NCBI

11 

van Der Heijden R, Jacobs DI, Snoeijer W, Hallard D and Verpoorte R: The Catharanthus alkaloids: Pharmacognosy and biotechnology. Curr Med Chem. 11:607–628. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Xue HZ, Lu ZZ, Konno C, Soejarto DD, Cordell GA, Fong HHS and Hodgson W: 3-Beta-(3,4-dihydroxycinnamoyl)-erythrodiol and 3-beta-(4-hydroxycinnamoyl)-erythrodiol from Larrea tridentata. Phytochemistry. 27:233–235. 1988. View Article : Google Scholar

13 

Arnold C: In A guide to Medicinal Plants of United States. The New York Times Book Co; 93. 1980

14 

Khare CP: Indian Medicinal Plants. An illustrated Dictionary. Springer; pp. p2422007

15 

Li TSC: Chinese and Related North American Herbs - Phytopharmacology and Therapeutic values. CRC Press; Boca Raton, London, New York, Washington: pp. p1792002

16 

Ling Y, Chen Y and Shih C: Flora Reipublicae Sinicae. Tomus 74. Science Press; Beijing: pp. p3481985

17 

Duke JA, Bogenschutz-Godwin MJ and Ottesen AR: Dukes Hand Book of Medicinal Plants of Latin America. CRC Press; Boca Raton, London, New York: pp. p2252009

18 

Bohlmann F and Wegner P: Three diterpenes from Conyza podocephala. Phytochemistry. 21:1693–1695. 1982. View Article : Google Scholar

19 

Xu LP, Guo DA, Liu JS and Zheng JH: A New transclerodane diterpene lactone from Conyza blinii. Heterocycles. 51:3605–3610. 1999.

20 

Mukhtar N, Iqbal K, Anis I and Malik A: Sphingolipids from Conyza canadensis. Phytochemistry. 61:1005–1008. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Mukhtar N, Iqbal K and Malik A: Novel sphingolipids from Conyza canadensis. Chem Pharm Bull (Tokyo). 50:1558–1560. 2002. View Article : Google Scholar

22 

Lenfeld J, Motl O and Trka A: Anti-inflammatory activity of extracts from Conyza canadensis. Pharmazie. 41:268–269. 1986.PubMed/NCBI

23 

Czeczot H, Tudek B, Kusztelak J, Szymczyk T, Dobrowolska B, Glinkowska G, Malinowski J and Strzelecka H: Isolation and studies of the mutagenic activity in the Ames test of flavonoids naturally occurring in medical herbs. Mutat Res. 240:209–216. 1990. View Article : Google Scholar : PubMed/NCBI

24 

Ding Y, Su Y, Guo H, Yang F, Mao H, Gao X, Zhu Z and Tu G: Phenylpropanoyl esters from Horseweed (Conyza canadensis) and their inhibitory effects on catecholamine secretion. J Nat Prod. 73:270–274. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Hrutfiord BF, Hatheway WH and Smith DB: Essential oil of Conyza canadensis. Phytochemistry. 27:1858–1860. 1988. View Article : Google Scholar

26 

Slater AF, Stefan C, Nobel I, van den Dobbelsteen DJ and Orrenius S: Signalling mechanisms and oxidative stress in apoptosis. Toxicol Lett. 82–83:149–153. 1995. View Article : Google Scholar

27 

Qiao L and Wong BC: Targeting apoptosis as an approach for gastrointestinal cancer therapy. Drug Resist Updat. 12:55–64. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Huang HL, Fang LW, Lu SP, Chou CK, Luh TY and Lai MZ: DNA-damaging reagents induce apoptosis through reactive oxygen species-dependent Fas aggregation. Oncogene. 22:8168–8177. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Wu K, Nie Y, Guo C, Chen Y, Ding J and Fan D: Molecular basis of therapeutic approaches to gastric cancer. J Gastroenterol Hepatol. 24:37–41. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Mao Y, Song G, Cai Q, Liu M, Luo H, Shi M, Ouyang G and Bao S: Hydrogen peroxide-induced apoptosis in human gastric carcinoma MGC803 cells. Cell Biol Int. 30:332–337. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Martín R, Ibeas E, Carvalho-Tavares J, Hernández M, Ruiz-Gutierrez V and Nieto ML: Natural triterpenic diols promote apoptosis in astrocytoma cells through ROS-mediated mitochondrial depolarization and JNK activation. PLoS One. 4:e59752009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2016
Volume 35 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu K, Qin Y, Yu J, Ma H and Song X: 3-β-Εrythrodiol isolated from Conyza canadensis inhibits MKN‑45 human gastric cancer cell proliferation by inducing apoptosis, cell cycle arrest, DNA fragmentation, ROS generation and reduces tumor weight and volume in mouse xenograft model Retraction in /10.3892/or.2021.7980. Oncol Rep 35: 2328-2338, 2016
APA
Liu, K., Qin, Y., Yu, J., Ma, H., & Song, X. (2016). 3-β-Εrythrodiol isolated from Conyza canadensis inhibits MKN‑45 human gastric cancer cell proliferation by inducing apoptosis, cell cycle arrest, DNA fragmentation, ROS generation and reduces tumor weight and volume in mouse xenograft model Retraction in /10.3892/or.2021.7980. Oncology Reports, 35, 2328-2338. https://doi.org/10.3892/or.2016.4610
MLA
Liu, K., Qin, Y., Yu, J., Ma, H., Song, X."3-β-Εrythrodiol isolated from Conyza canadensis inhibits MKN‑45 human gastric cancer cell proliferation by inducing apoptosis, cell cycle arrest, DNA fragmentation, ROS generation and reduces tumor weight and volume in mouse xenograft model Retraction in /10.3892/or.2021.7980". Oncology Reports 35.4 (2016): 2328-2338.
Chicago
Liu, K., Qin, Y., Yu, J., Ma, H., Song, X."3-β-Εrythrodiol isolated from Conyza canadensis inhibits MKN‑45 human gastric cancer cell proliferation by inducing apoptosis, cell cycle arrest, DNA fragmentation, ROS generation and reduces tumor weight and volume in mouse xenograft model Retraction in /10.3892/or.2021.7980". Oncology Reports 35, no. 4 (2016): 2328-2338. https://doi.org/10.3892/or.2016.4610