Antiproliferative activity of rosamultic acid is associated with induction of apoptosis, cell cycle arrest, inhibition of cell migration and caspase activation in human gastric cancer (SGC-7901) cells

Antiproliferative activity of rosamultic acid is associated with induction of apoptosis, cell cycle arrest, inhibition of cell migration and caspase activation in human gastric cancer (SGC-7901) cells

Accepted Manuscript Antiproliferative activity of rosamultic acid is associated with induction of apoptosis, cell cycle arrest, inhibition of cell mi...

3MB Sizes 1 Downloads 47 Views

Accepted Manuscript

Antiproliferative activity of rosamultic acid is associated with induction of apoptosis, cell cycle arrest, inhibition of cell migration and caspase activation in human gastric cancer (SGC-7901) cells Cheng-Guang Sui , Fan-Dong Meng , Yan Li , You-hong Jiang PII: DOI: Reference:

S0944-7113(15)00116-6 10.1016/j.phymed.2015.05.004 PHYMED 51828

To appear in:

Phytomedicine

Received date: Revised date: Accepted date:

5 February 2015 28 April 2015 11 May 2015

Please cite this article as: Cheng-Guang Sui , Fan-Dong Meng , Yan Li , You-hong Jiang , Antiproliferative activity of rosamultic acid is associated with induction of apoptosis, cell cycle arrest, inhibition of cell migration and caspase activation in human gastric cancer (SGC-7901) cells, Phytomedicine (2015), doi: 10.1016/j.phymed.2015.05.004

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

AC

CE

PT

ED

M

AN US

CR IP T

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Antiproliferative activity of rosamultic acid is associated with induction of apoptosis, cell cycle arrest, inhibition of cell migration and caspase activation in human gastric cancer (SGC-7901) cells Cheng-Guang Suia,*, Fan-Dong Menga, Yan Lia, You-hong Jianga

Molecular Oncology Department of Cancer Research Institution, The First Hospital of China

*

AN US

Medical University, Shengyang City 110001, China

CR IP T

a

Corresponding author:

Guang-Shui Jiang,

M

Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, No. 155 North of Nanjing Street, Heping District, Shenyang, Liaoning

ED

110001, China. Tel: 0086-024-83232354. fax: 0086-024-83282473.

CE

Abstract

PT

E-mail address: [email protected]

AC

Background: Gastric cancer is the second leading cause of cancer related deaths after lung cancer globally. Among natural products, natural triterpenes represent a structurally diverse group of organic compounds with potent antitumor activity. Purpose: The objective of the present research work was demonstrate the antiproliferative and apoptotic activity of rosamultic acid, a natural triterpenoid, in human gastric cancer (SGC-7901)

ACCEPTED MANUSCRIPT

cells. Its effect on cellular morphology, cell cycle arrest, DNA fragmentation and expression levels of caspase-3, caspase-8 and caspase-9 were also determined. Methods: Antiproliferative activity of rosamultic acid was evaluated by MTT assay. Phase

CR IP T

contrast, fluorescence microscopy as well as flow cytometry using Hoechst 33342, acridine orange/ethidium bromide and Annexin V-FITC as cellular probes were used to evaluate induction of apoptosis by rosamultic acid. Protein level expressions were analyzed by western blot analysis.

AN US

Results: The results revealed that rosamultic acid induced dose-dependent as well as time dependent cytotoxic effects in SGC-7901 gastric cancer cells. It also led to a reduction in clonogenic activity along with inhibiting the cell migration. Characteristic features of apoptosis induced by rosamultic acid were observed and quantified. Cell cycle arrest at sub-G1 phase was

M

induced by rosamultic acid along with downregulation of expression levels of CDK4, CDK6 and

ED

cyclin D1. Rosamultic acid also significantly led to the activation of caspase-3, -8 and -9 during the 48 h treatment along with cleaving PARP in a dose-dependent manner. DNA fragmentation

The current study strongly reveal that rosamultic acid inhibits gastric cancer

CE

Conclusion:

PT

following rosamultic acid treatment was also observed in these cells.

proliferation by inducing apoptosis mediated through cell cycle arrest, downregulation of cell

AC

cycle related protein expressions, inhibition of cell migration, DNA damage, and activation of caspases.

Keywords: Gastric cancer, Apoptosis, Rosamultic acid, Cell cycle, Caspases

Introduction

ACCEPTED MANUSCRIPT

Gastric cancer is the second leading cause of cancer related deaths after lung cancer, although the global incidence rate has now decreased. Despite the decreased global incidence of gastric cancer, it remains a prevalent burden in many Asian countries as compared to the western countries. China represents the country with the highest prevalence of gastric cancer, with an

CR IP T

estimated 380,000 new cases each year, accounting for more than 40% of the worldwide annual cancer incidence.In china, gastric cancer represents the third leading cause of cancer related deaths (Jiang, 2012; Kamangar et al., 2006; Chinese Ministry of Health, 2012). Gastric cancer represents the major cancer type in Asia, accounting for around 21% of malignant cancers. More

AN US

than 70% of the gastric cancer patients are considered to be incurable at the time of cancer diagnosis due to the already advanced stage and spreading of the tumor to other parts of the body. The recurrence rate is also very high in patients with resectable cancers resulting in

M

majority of the patients finally left with advanced cancer (Clarke et al., 1961). The etiology of gastric cancer remains to be defined as is the case with other cancers. The

ED

susceptibility of the people to gastric cancer depends on a number of factors including lifestyle,

PT

age, environmental and genetic factors. For example, vitamin C, carotenoids and green tea have been implied to have preventive effects in gastric cancer (Yoshida et al., 2010; Hwang et al.,

CE

1994). Gastric cancer is treated bythe surgical resection of the operable tumor, complemented by localized radiotherapy and chemotherapy with conventional chemotherapeutic drugs (Liu et al.,

AC

2013). Systemic chemotherapy is widely accepted as palliative treatment, leading to good responses, improved life quality, and increased survival rates. Chemotherapy has been vigorously investigated in gastric cancer. Several drugs have shown potent anticancer activity; however, single-drug chemotherapy has been unsuccessfulin increasing survival rates. Several combination treatments have been established with high activity in locally advanced and

ACCEPTED MANUSCRIPT

metastatic disease. Among these combinations are eplrubicln plus claptaUn plus 5-FU (ECF), etoposide plus leucovorin plus 5-FU (ELF), 5-fluorouracll (5-FU) plus high dose methotrexate plus doxorublcln (FAMTX), etoposide plus doxorubicin plus clsplatln (EAP) etc (Schipper and Wagener, 1996). Although the response rates of these combination regimens are encouraging,

CR IP T

the toxicity is immense. This is due to the non-specific toxicity of these drugs which also target normal cells in addition to cancer cells. This non-specific toxicity of these drugs necessitates the development of novel therapeutic agents to treat this deadly cancer (Cervanteset al., 2008;

AN US

Yoong et al., 2011).

A number of studies have drawn attention to natural products extracted from Chinese medicinal herbs as anticancer agents in gastric cancer therapy (Li et al., 2010; Chen et al., 2012). Over 60% of the current anticancer drugs have their origin in one way or another from natural sources.

M

Nature continues to be the most prolific source of biologically active and diverse chemotypes. Among natural products, natural triterpenes represent a structurally diverse group of organic

ED

compounds with potent bioactivity including antitumor effects. Several triterpenoids, including

PT

ursolic and oleanolic acid, betulinic acid, celastrol, pristimerin, lupeol, and avicins have been reported to possess anticancer activity (Petronelli et al., 2009).

CE

In the present research work, we report the anticancer and apoptotic inducing effects of

AC

rosamultic acid (A-ring contracted triterpenoid) isolated from the roots of Rosa multiflora, in human gastric cancer (SGC-7901) cells. We also demonstrated that these effects of rosamultic acid were mediated through cell cycle arrest, downregulation of cell cycle related protein expressions, inhibition of cell migration, DNA damage, and activation of caspases in these cells. The current report constitutes the first such report on this naturally occurring triterpene. Materials and methods

ACCEPTED MANUSCRIPT

Chemicals and other reagents MTT [3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide], DMSO, propidium iodide (PI) and trypsin were purchased from Sigma (St. Louis, MO, USA). RPMI-1640,

CR IP T

penicillin, streptomycin and other cell culture supplies were from Gibco BRL (Grand Island, NY, USA). Fetal bovine serum was from Hyclone (Logan, UT, USA).Acridine orange/ethidium bromide (AO/EtBr) and Hoechst 33342were obtained from Fluka (Ronkonkoma, NY, USA). Annexin V-FITC-Propidium Iodide Apoptosis Detection Kit was purchased from (Beyotime

AN US

Institute of Biotechnology, Shanghai, China). The cell-permeable pan-caspase inhibitor, the caspase-3 specific inhibitor, the caspase-9 specific inhibitor, the caspase-8 specific inhibitor, and the caspase-2 specific inhibitorwere purchased from (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). Therabbit polyclonal antibodies (Abs) against caspase-3, caspase-8, the mouse

M

monoclonal Abs (mAbs) againstpoly (ADP-ribose) polymerase (PARP), and the rabbit mAb against the active form ofcaspase-3 were purchased form PharMingen (Cell Signaling

ED

Technology, Inc.). All other chemicals and solvents used were of the highest purity grade.

PT

Plant material

CE

The roots of Rosa multiflora were collected in June 2014 from Guangzhou Province, China and identified by Prof. Liu-Xian Jiang, a voucher specimen (Voucher specimen number: 14-18-776-

AC

08) was deposited in the Herbarium of Southeast University, Nanjing, China. Extraction and isolation The root parts of the plant were thoroughly washed with water, shade dried and then grinded into small pieces. Dichloromethane/n-butanol (1:1) was used for cold extraction which was carried out for 72 hours in an extraction apparatus. The extract was then concentrated under reduced

ACCEPTED MANUSCRIPT

pressure in a rotary evaporator at 45 oC and was then kept in a refrigerator at 4 oC prior to use. Repeated chromatography of the extract on silica gel (50 g, 100–200mesh, Merck China) with chloroform: methanol (3:1) yielded rosamultic acid. White powder, ESI-MS m/z 509 (M+Na)+ , C30H46O5, 1 H-NMR (300 MHz, C5D5N): d 6.10 (1H, s, H-3), 5.57 (1H, s, H-12), 4.62 (1H, d,

CR IP T

J = 15 Hz, H-1b), 4.46 (1H, d, J = 15 Hz, H-1a), 4.03 (1H, d, J = 12 Hz, H-24b), 3.79 (1H, d, J = 12 Hz, H-24a), 3.02 (1H, s, H-18), 1.68 (3H, s, H-27), 1.45 (3H, s, H-23), 1.40 (3H, s, H-29), 1.21 (3H, s, H-25), 1.10 (3H, s, H-26), 1.09 (3H, d, J = 6 Hz, H- 30), 13C-NMR (75 MHz, C5D5N): d 61.3 (C-1), 158.4 (C-2), 131.3 (C-3), 49.4 (C-4), 63.9 (C-5), 18.3 (C-6), 35.3 (C-7),

AN US

42.2 (C-8), 44.2 (C-9), 51.2 (C-10), 27.4 (C-11), 128.3 (C-12), 140.5 (C-13), 42.6 (C-14), 30.0 (C-15), 26.6 (C-16), 48.6 (C-17), 55.1 (C-18), 72.9 (C-19), 42.6 (C-20), 27.2 (C-21), 38.8 (C22), 25.4 (C-23), 66.8 (C-24), 19.8 (C-25), 19.1 (C-26), 25.7 (C-27), 181.2 (C-28), 27.4 (C-29),

ED

Cell line and culture conditions

M

17.0 (C-30). It was characterized as rosamultic acid.

Human gastric cancer (SGC-7901) cancer cells were obtained from Cancer Research

PT

Institute of Beijing, China.These cells were cultivated in in RPMI-1640 medium supplemented with 10% fetal calf serum, 100 IU/ml penicillin, 100 μg/ml streptomycin. Cells were cultured in

CE

CO2 incubator (New Brunswick, Galaxy 170R, eppendroff) with an internal atmosphere of 95%

AC

air and 5% CO2 gas and the cell lines were maintained at 37 oC. MTT assay for cell viability evaluation Cell viability was measured using MTT assay. Gastric cancer (6 × 102) cells were seeded

in 200 μl of RPMI-1640 medium into 96-well plates, and cultured overnight. Then the medium was replaced with fresh RPMI-1640 or the same media containing different concentrations (0,

ACCEPTED MANUSCRIPT

2.5, 5, 25, 50, 75 and 100 µM) of rosamultic acid. After a further incubation for 24 or 48 h, 30 μl of MTT (2 mg/ml) was added to each well followed by 3 h incubation. The medium was discarded and 170 μl of dimethyl sulfoxide was added into each well, and incubated for 30 min.

(experimental OD value/control OD value) × 100%.

CR IP T

The OD490 nm was measured. The cell viability index was calculated according to the formula:

Cytotoxicity was expressed as the concentration of bergamottin inhibiting cell growth by 50% (IC50 value).

AN US

Colony formation assay

This assay was done as follows. Five ml complete medium containing 400 cells was added to a 60-mm dish. After cell culture with 5% CO2 for 14 days at 37 oC, the supernatant was

M

thrown away, and the cells were washed with PBS 3 times. The cells were then fixed with 5% paraformaldehyde for 20 min, and stained with GIMSA (Solarbio, Beijing, China) for 30 min.

ED

Colonies were numbered under an inverted microscope (Nikon, Tokyo, Japan). The whole assay

PT

was carried out in triplicate.

In vitro wound healing/migration assay

CE

Human gastric cancer (SGC-7901) cancer cells (1x 105 cells/ml) were seeded in a 6-well

AC

plate and incubated at 37 oC and 95 % full confluent monolayer was obtained. Upon 12 h of serum starvation, a 50 ml pipette tip was used to create a straight cell‑ free wound. Each well was washed 3 times with PBS and then subjected to various concentrations of rosamultic acid (0, 25, 50 and 100 µM for 48 and 72 h) in a medium. After incubation, cells were fixed and stainedwith 2.5% ethanol containing 0.3% crystal violet powder for 15 min, and

ACCEPTED MANUSCRIPT

arbitrarilyselected fields were photographed under a light microscope. The number of cells that migrated into the scratched area were counted. Morphological changes of gastric cancer cells observed by light microscopy

CR IP T

Human gastric cancer (SGC-7901) cancer cells were seeded in 96-well plates (2 × 105 cells/well) and incubated overnight to allow adhesion. The cells were treated with rosamultic acid at concentrations of 0, 25, 50 and 100 µM for 48 h. The morphological changes were observed under an inverted light microscope (Olympus, Center Valley, PA, USA) after 48 hours.

AN US

Morphological changes of gastric cancer cells observedby fluorescence microscopy using Hoechst 33342 and Acridine orange/ethidium bromide staining

Human gastric cancer (SGC-7901) cancer cells which were grown on coverslips in 12-

M

well plates were exposed to different concentrations (0, 25, 50 and 100 µM) of rosamultic acid for 48 hours, then the cells were washed twice with PBS and stained with Hoechst 33342

ED

(Hoechst Staining Kit, Beyotime, China) for 1 h at room temperature. Fluorescence microscopy was used to detect and measure cell shape captured from different random visual fields. The ratio

PT

of apoptotic cells to total cell number was calculated. The rosamultic acid-treated cells were also

CE

stained with acridine orange and ethidium bromide and visualized under the fluorescence microscope.

V-FITC/propidium iodide (PI) double-staining

AC

Annexin

assay for

quantification of

apoptotic/necrotic cells. Annexin V-FITC/propidium iodide (PI) double-staining was performed with an Annexin V-FITC Kit (BDBioscience, USA). Human gastric cancer (SGC-7901) cancer cells were treated with different concentrations (0, 25, 50 and 100 µM) of rosamultic acid for 48 hours. The cells

ACCEPTED MANUSCRIPT

were trypsinized, rinsed twice with PBS, and resuspended in 1× binding buffer. The cells were labeled with 10 μl of FITC-conjugated annexin V and 10 μl of propidium iodide. The cells were incubated for 20 min in dark at 37 oC and then 450 μl of binding buffer was added and the samples were immediately analyzed with a flow cytometer (Becton Dickinson, San Jose,

CR IP T

CA).The annexin V-FITC−/PI− cell population was considered as normal, while the annexin VFITC+/PI− and Annexin V-FITC+/PI+ cell populations were considered as indicators of early and late apoptotic cells, respectively.

AN US

DNA fragmentation analysis by gel electrophoresis

After subjecting human gastric cancer cells (SGC-7901) to rosamultic acid treatment for 48 h at various concentrations (0, 25, 50, and 100 µM), both adherent and floating cells were collected and washed with PBS. Pellets were then lysed with DNA lysis buffer (40 mM EDTA,

M

150 mM Tris, pH7.6, 0.8% SDS) at room temperature for 30 min. After centrifugation for 15

ED

min at 15 000 × g, the supernatants were collected and treated with RNase A (final concentration, 500 μg/ml) for 30 min at 37 oC, followed by digestion with proteinase K (final o

C. The DNA was extracted using the

PT

concentration 500 μg/ml) for 2.0 h at 55

phenol/chloroform/isoamylol (25:24:1), precipitated with ethanol, dissolved in TE buffer (10

CE

mM Tris, pH 8.0, 1 mM EDTA), and subjected to 2% agarose gel electrophoresis for DNA

AC

fragmentation analysis.

Cell cycle analysis by flow cytometry Briefly, human gastric cancer cells (1 × 106) were seeded into each well of 6-well plates

and incubated for 24 h for cell attachment and recovery. The cells were treated with different concentrations (0, 25, 50 and 100 µM) of rosamultic acid. Untreated cells (control) were also

ACCEPTED MANUSCRIPT

incorporated. After incubation for 24 h, the cells were harvested and fixed with ice-cold 70% ethanol (5 mL) at −20 °C for 2 h. Prior to analysis, the cells were washed with cold PBS and resuspended in 400 μl of PBS, 20 μl PI and 20 μl RNase A. The DNA contents were recorded by a

CR IP T

flow cytometer (Becton Dickinson, San Jose, CA) equipped with Cell Quest software.

Western Blot Analysis

Western blot assay was done as previously reported (Zhang et al., 2009) with slight

AN US

modifications. Bradford assay (Bio-Rad) was used to determine the protein content. After electrophoresing a total of 20-40 Ag ofprotein on 15% SDS-PAGE gels, it was transferred to nitrocellulose membranes. Membranes wereblocked, incubated with primary Abs at the

M

suitabledose, and consequently incubated withprimary antibody, washed and incubated with horseradish peroxidase conjugated secondary antibody (1:2500 dilution; Bio-Rad). Detection

Danvers, MA, USA).

PT

Statistical analysis

ED

was performed using a chemiluminescent western detection kit (Cell Signaling Technology, Inc.,

CE

Data are expressed as mean ± SD of three independent experiments. SPSS.13.0 software was used to perform statistical analysis. Differences were analyzed using one-way analysis of

AC

variance (ANOVA) or two-way ANOVA. P<0.05 was considered statistically significant. Results

Characterization of rosamultic acid isolated from the root extract of Rosa multiflora

ACCEPTED MANUSCRIPT

Rosamultic acid was isolated as white powder, White powder, ESI-MS m/z 509 (M+Na)+ , corresponding to the molecular formula C30H46O5.1H-NMR, and 13C-NMR data revealed that the data corresponds to rosamultic acid molecular architecture. HPLC chromatogram, chemical structure and

13

C-NMR of rosamultic acid are shown in Fig. 1A, Fig. 1B and Fig. 1C

CR IP T

respectively. The spectroscopic data was also compared with the literature data available.

Antiproliferative activity of rosamultic acidand its effect on colony formation in human gastric cancer cells (SGC-7901)

AN US

To examine the inhibitory effect of rosamultic acid on the proliferation of human gastric cancer cells, MTT assay was conducted. SGC-7901 cells were treated with different concentrations (0, 2.5, 5, 25, 50, 75 and 100 µM) of rosamultic acid dissolved in DMSO and the same volume of solvent was used as a control. The results showed that rosamultic acid exerted

M

potent and dose-dependent as well as time dependent antiproliferative effects on SGC-7901

ED

cancer cells (Fig. 2) after treatment for 24, 48 and 72 h. Further, we also evaluated the anticlonogenic effects of this triterpenoid which indicated that rosamultic acid decreased the number

PT

of colonies comprising of SGC-7901 cancer cells which also showed concentration-dependence. These results when taken together indicate that rosamultic acid suppresses both anchorage-

CE

dependent (antiproliferative) as well as anchorage-independent (colony formation) growth of

AC

SGC-7901 cancer cells (Fig. 3 and Fig. 4 A to D).

Effects of rosamultic acid on the migration of SGC-7901 gastric cancer cells

ACCEPTED MANUSCRIPT

We also evaluated the effect of rosamultic acid on the cell migration of SGC-7901 gastric cancer cells using wound-healing (scratch motility) assay. Confluent monolayers of cells were scratched to form wounds, then cultured in the absence or presence of various concentrations of rosamultic acid (25, 50 and 100 µM), and examined at different time intervals (48 and 72 h) after

CR IP T

cell monolayers had been wounded. Rosamultic acid exhibited potent inhibitory effects against cell migration (Fig. 5). As shown in Fig. 5, rosamultic acid-induced cells moved slowly compared with thecontrol group in a dose-dependent manner.The number of cells migrated into the scratched area were photographed and calculated as a percentage (%) of migration.

AN US

Rosamultic acid showed time-dependent as well as dose-dependent inhibitory effect against cell migration. Fig. 5 represents untreated (0 µM) control cells while Fig. 4 B, C and D represent effect of 25, 50 and 100 µM dose of rosamultic acid respectively.

M

Morphological evaluation of rosamultic acid-induced apoptosis in SGC-7901 cancer cells using

ED

phase contrast and fluorescence microscopy

Further, we studied the effect of rosamultic acid on the cellular morphology and

PT

apoptosis induction in SGC-7901 cancer cells using phase contrast and fluorescence microscopy. Rosamultic acid-treated cells showed characteristic morphological changes of apoptosis

CE

including detachment of the cells from substratum and cell shrinkage. As can be seen in Fig. 6

AC

A-D, Untreated SGC-7901 cells appeared as densely packed and disorganized multilayers, whereas after incubation with various concentrations of rosamultic acid for 48 h, several of the cells became irregularly shaped, some cells were broken with no content, some were rounded and shrunken, and detached from each other or floated in the medium. Further fluorescence microscopy using Hoechst 33342 and PI double staining and Acridine orange/ethidium bromide staining also revealed morphological features including

ACCEPTED MANUSCRIPT

chromatin condensation, fragmented nuclei and nuclear shrinkage which increased with the increasing dose of rosamultic acid (Fig. 7 A-D and Fig. 8 A-D) after 48 h. Bright nuclear condensation was noted by Hoechst 33342 and PI staining, and apoptotic cells were observed as masses in the cells (Fig. 7). The numbers of broken and necrotic/apoptotic cells increased in

CR IP T

conjunction with increasing concentrations of rosamultic acid. Similarly, in case of acridine orange/ethidium bromide staining, green intact nuclei indicated normal cells, while as red staining indicated apoptosis. The number of cells with red staining increased with increasing

AN US

dose of rosamultic acid (Fig. 8). Apoptosis quantification by Annexin V-FITC/PI assay

Annexin V/PI double staining was used to detect apoptosis in human gastric cancer cells (SGC-7901) (Fig. 9A-D). SGC-7901 cells were treated with different concentration (0, 25, 50

M

and 100 µM) of rosamultic acid for 48 h. Rosamultic acid induced both early and late apoptosis

ED

in a concentration-dependent manner (Fig. 9B-D) as compared to the untreated control cells (Fig. 9A). When the cells were treated with 25, 50 and 100 µM for 48 h, the average proportion of

PT

Annexin V-staining positive cells (total apoptotic cells) significantly increased from 3.2% in control to 34.3%, 46.8% and 65.9% respectively. Fig. 10 shows the graphical representation of

AC

CE

the increase in the apoptotic cells with increase in the dose of rosamultic acid.

Effect of Rosamultic acid on DNA fragmentation in SGC-7901 human gastric cancer cells Besides the morphological changes of apoptosis in Rosamultic acid-treated SGC-7901 cells, DNA fragmentation of wasalso examined by observation of the formation of DNA ladder. As shown in Fig. 11, DNA ladder appeared to be more evident with the increasing Rosamultic

ACCEPTED MANUSCRIPT

acid concentration, however, no DNA fragments were observed in the control groups (Fig. 11, 0 µM). However, 25, 50 and 100 µM doses of rosamultic acid after 48 h exposure led to a substantial increase in DNA fragmentation (Fig. 11, right panel).

CR IP T

Rosamultic acid induced sub-G1 (G0/G1) cell cycle arrest in SGC-7901 gastric cancer cells. To determine the distribution of rosamultic acid -treated SGC-7901 cells in different phases of the cell cycle, DNA content in cells was detected by propidium iodide (PI) staining and flow cytometry. The results showed that treatment with different concentrations of rosamultic

AN US

acid for 48 h led to an increase in the population of cells in the sub-G0/G1 phase (apoptotic population) (P < 0.01) (Fig. 12A-D). This increase in sub-G1 population was also accompanied by a corresponding increase of the cells in G2/M phase of the cell cycle. As compared to the control (Fig. 12A), where only 1.5% of the cells were in sub-G1 phase, 25, 50 and 100 µM

M

rosamultic acid-treated cells showed 6.2% (Fig. 12B) , 32.6% (Fig. 12C) and 45.8% (Fig. 12D)

ED

of the cells in the sub-G1 phase (apoptotic phase) of the cell cycle. Rosamultic acid-induced downregulation of the cell cycle related protein expressions

PT

Since rosamultic acid induced cell cycle arrest in sub-G1 (G0/G1) phase, therefore we

CE

also evaluated the effect of this triterpenes on the expression levels of the cell cycle related proteins including CDK4, CDK6 and cyclin D1 in SGC-7901 gastric cancer cells. The results

AC

showed that rosamultic acid induced a considerable downregulation in the expression levels of proteins such as CDK4, CDK6 and cyclin D1that are known to control the cell cycle (Fig. 13 A). This decrease in the expression level of these proteins showed concentration dependence of rosamultic acid (Fig. 13 B). Thus we can safely conclude that rosamultic acid induced sub-G1 arrest due to downregulation of these proteins.

ACCEPTED MANUSCRIPT

Rosamultic acid-induced apoptosis was mediated through caspase-3, caspase-9 andcaspase-8 activation To demonstrate the relation between caspase activation and apoptosis induction by

CR IP T

rosamultic acid in SGC-7901 cancer cells, we evaluated the activities of caspase-3, -9 and -8 using the colorimetric assay. The results revealed that rosamultic acid significantly led to the activation of caspase-3, -8 and -9 during the 48 h treatment (Fig. 14). Western blot analysis showed that full-length procaspase-9, procaspase-8 and procaspase-3 decreased with the

AN US

increased dose of rosamultic acid, while as their cleaved form increased especially at higher doses. The results also showed that PARP, which is a hallmark of caspase-3 activation during apoptosis, was also cleaved at 50 and 100 µM dose of rosamultic acid, but not at lower doses.

M

Discussion

Rosa multiflora, commonly known as multiflora rose is a species of rose native to China, eastern

ED

Asia, Korea and Japan. The plant is a scrambling shrub climbing over other plants and is grown as an ornamental plant (Roger and Martyn, 2004). The plant contains a diverse kind of

PT

phytochemicals including pentacyclic triterpenes (tormentic acid and its glucoside known as

CE

rosamultin), flavonoids, phenolic acids, volatile fragrance molecules, carotenoids, tannins etc (Bhandari et al., 2007; Pang et al., 2009). Different parts of the plant are used as herbal medicine

AC

for the treatment of a variety of diseases including trauma, cold, flu, inflammation, management of pain. Recent studies have shown that Rosa multiflora extracts also possess high antioxidant and antimutagenic effects in all tested assays (Chrubasik et al., 2006; Chrubasik et al., 2007; Shan et al., 2005).

ACCEPTED MANUSCRIPT

Apoptosis is a cellular suicide program that exterminates unwanted, faulty and potentially dangerous cells during the development and maintenance of cell homeostasis. Inducing apoptosis is a key tactic to eliminate cancer cells without stimulating an inflammatory reaction. Regulation of apoptotic signaling pathways encompasses a complicated system consisting of several

CR IP T

elements. Several conventional drugs are currently used in anticancer chemotherapy which are believed to induce cell apoptosis via activation of these elements (Earnshaw, 1995; Kundu et al., 2005). Therefore, the ability of cancer cells to induce the apoptotic program has been recognized as one of the major mechanisms which might serve for the development of novel approaches to

AN US

treat cancer.Caspases are the vital machineries in the implementation of apoptosis (Kerr et al., 1972; Chiarugi and Giannoni, 2008).

Usually, caspases associated with apoptosis can be divided into the initiator caspases and

M

the executioner caspases. Caspase-8 and -9 are the initiator caspases in the death receptor and the mitochondrial pathways, respectively. Caspase-3, is the crucialexecutioner caspase in apoptosis

ED

pathway (Li and Yuan, 2008; Kantari and Walczak, 2011). Our results showed that rosamultic

PT

acid induces apoptosis which is mediated through the activation of caspase-3, -8 and -9. Procaspase-9, procaspase-8 and procaspase-3 decreased with the increased dose of rosamultic

CE

acid, while as their cleaved form increased especially at higher doses. PARP was also cleaved at

AC

50 and 100 µM dose of rosamultic acid, but not at lower doses. Dysregulation in the cell division and apoptosis are connected to the development of

most cancers. Many anticancer drugs function primarily to induce apoptosis in cancer cells and prevent tumor development (Li et al., 2011; Khoo et al., 2010). The morphological changes of apoptosis observed in most cell types initially start with a reduction in cell volume and condensation of the nucleus (Moongkarndi et al., 2004). In many cases extensive DNA damage

ACCEPTED MANUSCRIPT

leads to activation of cell cycle check points and results in cell cycle arrest and apoptosis (Pietenpol and Stewart, 2002). In the present study, we found that rosamultic acid induced apoptosis in SGC-7901 gastric cancer cells as revealed by fluorescence microscopy as well as Annexin V-FITC assay. When the cells were treated with 25, 50 and 100 µM for 48 h, the

CR IP T

average proportion of Annexin V-staining positive cells (total apoptotic cells) significantly increased from 3.2% in control to 34.3%, 46.8% and 65.9% respectively. Further, we evaluated the effect of rosamultic acid on cell cycle phase distribution using flow cytometry. It was observed that rosamultic acid induced cell cycle arrest in the sub-G1 phase. The expression

AN US

levels of the cell cycle related proteins includingCDK4, CDK6 and cyclin D1 in SGC-7901 gastric cancer cells were also demonstrated. Rosamultic acid induced a considerable downregulation in the expression levels of proteins such as CDK4, CDK6 and cyclin D1that are

M

known to control the cell cycle. In addition, rosamultic acid also led to a significant DNA fragmentation. Higher doses of (50 and 100 µM) rosamultic acid after 48 h exposure led to a

ED

substantial increase in DNA fragmentation.

PT

Tumor invasion is a collective feature of many malignant and deadly tumors resulting in high morbidity and death on account of their high growth rate, invasive potential, and their

CE

resistance towards the drug treatment. Migration and invasion are the key features of cancer progression and metastasis (Lee et al., 2008; Neudauer and McCarthy, 2003; Goncharova et al.,

AC

2002). As a result, therapeutic approaches for preventing or suppressing cancer invasion, migration and metastasis can significantly improve the survival of the patients.We also evaluated the effect of rosamultic acid on the cell migration of SGC-7901 cancer cells. The wound healing assay revealed that rosamultic acid significantly reduced the migration of SGC-7901 cells in a

ACCEPTED MANUSCRIPT

dose-dependent manner. The compound also induced anticlonogenic effects against the colony formation tendency in these gastric cancer cells. Various plant triterpenes have been reported to induce cell cycle arrest including the pentacyclic

CR IP T

triterpenes. These include asiatic acid, odoratol, ursolic acid, oleanolic acid, boswellic acid etc Triterpenoids are highly multifunctional compounds and as a result have promise as agents in the treatment of cancer because of their ability to block the NF-κB activation, induce apoptosis, and prevent proliferation, invasion, metastasis and angiogenesis (Hsu et al., 2005; Cazal et al., 2010).

AN US

As far as rosamultic acid is concerned, we could not find reports of its anticancer action or its effects on the cell cycle arrest. To the best of our knowledge, the current research work on this molecule has not been reported earlier and as such constitutes the first such report. In conclusion, we can summarize that rosamultic acid exhibits antiproliferative effects in

M

SGC-7901 cancer cells by inducing apoptosis which is mediated by activation of caspase-3,

ED

caspase-8 and caspase-9. Rosamultic acid also induced cell cycle arrest at sub-G1 phase along with downregulation of expression levels of proteins such as CDK4, CDK6 and cyclin D1 which

PT

are key controllers of cell cycle. The triterpene also inhibited cell migration as well as colony

CE

formation tendency of SGC-7901 cancer cells. Conflict of interest

AC

The authors declare that there is no conflict of interest to reveal.

References Cervantes, A., Rosello, S., Roda, D., Rodriguez-Braun, E., 2008. The treatment of advanced gastric cancer: current strategies and future perspectives. Ann. Oncol. 19, v103-v107.

ACCEPTED MANUSCRIPT

Bhandari, P., Kumar, N., Gupta, A.P., Singh, B., and Kaul, V.K., 2007. A rapid RP‐HPTLC densitometry method for simultaneous determination of major flavonoids in important medicinal plants. J. sep. sci. 30, 2092-2096.

CR IP T

Cazal, C.M., Choosang, K., Severino, V.G., Soares, M.S., Sarria, A.L., Fernandes, J.B., Silva, M.F., Vieira, P.C., Pakkong, P., Almeida, G.M., Vasconcelos, M.H., Nascimento, M.S., Pinto, M.M., 2010. Evaluation of effect of triterpenes and limonoids on cell growth, cell cycle and apoptosis in human tumor cell line. Anticancer Agents Med Chem. 10,769-76. Chen, J., Shi, D.Y., Liu, S.L., Zhong, L., 2012. Tanshinone IIA induces growth inhibition and apoptosis in gastric cancer in vitro and in vivo. Oncol. Rep. 27, 523-528. Chiarugi, P., Giannoni, E., 2008. Anoikis: a necessary death program for anchorage-dependent cells. Biochem. Pharmacol. 76, 1352-1364.

AN US

Chrubasik, C., Duke, R., and Chrubasik, S., 2006. The evidence for clinical efficacy of rose hip and seed: a systematic review. Phyto. Res. 20, 1-3. Chrubasik, J.E., Roufogalis, B.D., and Chrubasik, S., 2007. Evidence of effectiveness of herbal anti-inflammatory drugs in the treatment of painful osteoarthritis and chronic low back pain. Phyto. Res. 21, 675-683.

M

Clarke, J.S., Cruze, K., El Farra, S., Longmire, W.P. Jr., 1961.The natural history and results of surgical therapy for carcinoma of the stomach. An analysis of 250 cases. Am. J. Surg. 102,143152.

ED

Earnshaw, W.C., 1995. Nuclear changes in apoptosis. Curr. Opin. Cell Biol. 7, 337-343.

PT

Gastric Cancer Diagnosis and Treatment Expert Panel of the Chinese Ministry of Health. Chinese guidelines for diagnosis and treatment of gastric cancer (2011edition)., 2012. Transl. Gastrointest. Cancer 1,103-14.

CE

Goncharova, E.A., Ammit, A.J., Irani, C., Carroll, R.G., Eszterhas, A.J., Panettieri, R.A., Krymskaya, V.P., 2002. PI3K is required for proliferation and migration of human pulmonary vascular smooth muscle cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 283, L354-L363.

AC

Hsu, Y.L., Kuo, P.L., Lin, L.T., Lin, C.C., 2005. Asiatic acid, a triterpene, induces apoptosis and cell cycle arrest through activation of extracellular signal-regulated kinase and p38 mitogenactivated protein kinase pathways in human breast cancer cells. J Pharmacol Exp Ther. 313, 33344. Hwang, H., Dwyer, J., Russell, R.M., Diet, 1994. Helicobacter pylori infection, food preservation and gastric cancer risk: are there new roles for preventative factors? Nutr. Rev. 52, 75-83.

ACCEPTED MANUSCRIPT

Jiang, Y., 2012. Multidisciplinary approach Transl.Gastrointest. Cancer 1,175-7.

for the treatment

of

gastric

cancer.

Kamangar, F., Dores, G.M., Anderson, W.F., 2006. Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J. Clin. Oncol. 24,2137-50.

CR IP T

Kantari, C., Walczak, H., 2011. Caspase-8 and bid: caught in the act between death receptors and mitochondria. Biochim. Biophys. Acta 1813, 558-63. Kerr, J.F., Wyllie, A.H., Currie, A.R., 1972. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 26, 239-257. Khoo, B.Y., Chua, S.L., Balaram, P., 2010. Apoptotic effects of chrysin in human cancer cell lines. Int. J. Mol. Sci. 11,2188–2199.

AN US

Kundu, T., Dey, S., Roy, M., Siddiqi, M., Bhattacharya, R.K., 2005. Induction of apoptosis in human leukemia cells by black tea andits polyphenol theaflavin. Cancer Lett. 230,111-121. Lee, W.J., Chen, W.K., Wang, C.J., Lin, W.L., Tseng, T.H., 2008. Apigenin inhibits HGFpromoted invasive growth and metastasis involving blocking PI3K/Akt pathway and beta 4 integrin function in MDA-MB-231 breast cancer cells. Toxicol. Appl. Pharmacol. 226,178–191.

M

Li, J., Yuan, J., 2008. Caspases in apoptosis and beyond. Oncogene. 27, 6194-206.

ED

Li, N., Fan, L.L., Sun, G.P., Wan, X.A., Wang, Z.G., Wu, Q., Wang, H., 2010. Paeonol inhibits tumor growth in gastric cancer in vitro and in vivo. World J. Gastroenterol. 16, 4483-4490.

PT

Li, W., Wang, J., Jiang, H.R., Xu, X.L., Zhang, J., Liu, M.L., Zhai, L.Y., 2011. Combined effects of cyclooxygenase-1 and cyclooxygenase-2 selective inhibitors on ovarian carcinoma in vivo. Int. J. Mol. Sci. 12, 668–681.

CE

Liu, Y.P., Ling, Y., Qi, Q.F., Zhang, Y.P., Zhang, C.S., Zhu, C.T., Wang, M.H., Pan, Y.D., 2013. The effects of ERCC1 expression levels on the chemo sensitivity of gastric cancer cells to platinum agents and survival in gastric cancer patients treated with oxaliplatin-based adjuvant chemotherapy. Oncol. Lett. 5,935-942.

AC

Moongkarndi, P., Kosem, N., Kaslungka, S., Luanratana, O., Pongpan, N., Neungton, N.,2004. Antiproliferation, antioxidation and induction of apoptosis by Garcinia mangostana (mangosteen) on SKBR3 human breast cancer cell line. J. Ethnopharmacol., 90,161–166. Neudauer, C.L., McCarthy, J.B., 2003. Insulin-like growth factor I-stimulated melanoma cell migration requires phosphoinositide 3-kinase but not extracellular-regulated kinase activation. Exp. Cell Res. 286, 128-137.

ACCEPTED MANUSCRIPT

Pang, N., Malike, D., and Liu, H., 2009. Simultaneous determination of main bioactive components in Rosa multiflora Thunb. and their fragmentation study by LC–MS. Chromatographia. 70, 1253-1257. Petronelli, A., Pannitteri, G., Testa, U., 2009. Triterpenoids as new promising anticancer drugs. Anticancer Drugs 20,880-92.

CR IP T

Pietenpol, J.A., Stewart, Z.A., 2002. Cell cycle checkpoint signaling: cell cycle arrest versus apoptosis. Toxicology 181–182, 475–481. Roger, P., Martyn, R., 2004. The Ultimate Guide to Roses. Pan Macmillan Ltd. p. 262. ISBN 1405049200. Schipper, D.L., Wagener, D.J.T., 1996. Chemotherapy of gastric cancer. Anticancer Drugs 7, 137-232.

AN US

Shan, B., Cai, Y.Z., Sun, M., and Corke, H., 2005. Antioxidant capacity of 26 spice extracts and characterization of their phenolic constituents. J. Agric. Food Chem. 53, 7749-7759. Yoong, J., Michael, M., Leong, T., 2011. Targeted therapies for gastric cancer: current status. Drugs 71, 1367-1384.

M

Yoshida, T., Ono, H., Kuchiba, A., Saeki, N., Sakamoto, H., 2010. Genome-wide germline analyses on cancer susceptibility and GeMDBJ database: Gastric cancer as an example. Cancer Sci. 7, 1582-1589.

PT

ED

Zhang, X.D., Franco, A., Myers, K., Gray, C., Nguyen, T., Hersey, P., 1999. Relation of TNFrelated apoptosis-inducing ligand (TRAIL) receptor and FLICE-inhibitory protein expression to TRAIL-induced apoptosis of melanoma. Cancer Res. 59, 2747–53.

Fig. 1. (A) HPLC chromatogram, (B) molecular structure and (C) 13C- NMR of rosamultic acid isolated from the roots of Rosa multiflora.

CE

Fig. 2. Dose-dependent and time-dependent antiproliferative effect of rosamultic acid in human gastric cancer cells (SGC-7901). Data are shown as the mean ± SD of three independent experiments. *, P < 0.05, **, P < 0.01, vs 0 µM (control).

AC

Fig. 3. Anticlonogenic activity of rosamultic acid on human gastric cancer cells (SGC-7901). The anti-colony formation effect showed strong dose-dependence. Data are shown as the mean ± SD of three independent experiments. *, P < 0.05, **, P < 0.01, vs 0 µM (control). Fig. 4. Effect of rosamultic acid on colony formation in SGC-7901 cancer cells. The cells were treated with 0 µM (control, A), 25 µM (B), 50 µM (C) and 100 µM (D) concentration of rosamultic acid respectively. Fig. 5. Effect of rosamultic acid on human gastric cancer (SGC-7901) cell migration in vitro. Photographs of wound of cells treated with 0, 25, 50 and 100 μM of rosamultic acid for 48 and

ACCEPTED MANUSCRIPT

72 h. The number of cells migrated into the scratched area was calculated as a percentage of migration. Fig. 6. Rosamultic acid-induced morphological changes in SGC-7901 gastric cancer cells as detected by inverted phase contrast microscopy (magnification 200X). Rounded and shrunk cells were observed in rosamultic acid-treated cells (Red colored arrows). A, represents control (untreated cells), B,C and D represent effect of 25, 50 and 100 µM of rosamultic acid on cell morphology of SGC-7901 cells.

CR IP T

Fig. 7. Rosamultic acid-induced apoptosis in SGC-7901 gastric cancer cells. (A) Untreated SGC7901 control cells, (B) SGC-7901 cells treated with 25 μM, (C) 50 μM and (D) 100 μM concentration of Rosamultic acid respectively. The cells were treated with Rosamultic acid for 48 h, stained with Hoechst 33258/PI and observed by fluorescence microcopy at a magnification of × 200. Bright nuclear condensation/apoptotic cells is shown by orange arrows.

AN US

Fig. 8. Rosamultic acid-induced apoptosis in SGC-7901 gastric cancer cells. (A) Untreated SGC7901 control cells, (B) SGC-7901 cells treated with 25 μM, (C) 50 μM and (D) 100 μM concentration of Rosamultic acid respectively. The cells were treated with Rosamultic acid for 48 h, stained with acridine orange/ethidium bromide and observed by fluorescence microcopy at a magnification of × 200. Red staining (orange arrows) shows apoptotic cells.

M

Fig. 9. Quantification of rosamultic acid-induced apoptosis in human gastric cancer cells (SGC7901). The cells were subjected to different doses of rosamultic acid (0, 25, 50 and 100 µM) for 48 h and analyzed by flow cytometry with annexin V-FITC/PI staining. Percentage of apoptotic cells increases from 3.2% in control cells (A), to 34.3%, 46.8% and 65.9% in 25 µM (B), 50 µM (C) and 100 µM (D) rosamultic acid-treated cells respectively.

ED

Fig. 10. Increase in the number of Annexin V positive cells with increase in the dose of rosamultic acid. The results are mean ± SEM and mean values of three independent experiments. * p < 0.05, ** p < 0.01 versus the 0 μM of rosamultic acid-treated cells.

PT

Fig. 11. Rosamultic acid induces DNA fragmentation in human gastric cancer cells (SGC-7901). The cells were treated with 0, 25, 50 and 100 μM Rosamultic acid for 48 h. Cells from each sample were harvested for DNA gel electrophoresis as described in Materials and methods.

AC

CE

Fig. 12. Effect of Rosamultic acid on the cell cycle arrest in human gastric cancer (SGC-7901) cells. Cells were treated with (B) 25 μM, (C) 50 μM and (D) 100 μM of rosamultic acid for 48 h. (A) shows the control (untreated) group. The percentage of cells in the sub-G1 phase increased significantly with increase in the rosamultic acid dose from 0 μM (A) to 25, 50 and finally to 100 μM. The DNA histogram shows the distribution and the percentage of cells in phases of the cell cycle. Results are the mean ± SD of 3 independent experiments. Fig. 13 A and B. Effect of Rosamultic acid on the expression of proteins that regulate the cell cycle (Fig. 13A) of gastric cancer cells. Following treatment of SGC-7901 cells with different doses of Rosamultic acid (0, 25, 50 and 100 µM) for 48 h, (A) western blot analysis was used to analyze changes in the expression levels of CDK4, CDK6 and cyclin D1 proteins. (Fig. 13B) The histogram shows the quantification (%) of the western blot presented in A. * P < 0.05, **P < 0.01, when compared with the control group. Fig. 14. Effects of Rosamultic acid treatment on caspases activation and PARP cleavage in SGC7901 cancer cells. Caspase activities in Rosamultic acid-treated SGC-7901 cells. The lysates of rosamultic acid treated cells were adjusted to equal protein amounts and the enzymatic activities

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

of caspases-8, -9, and -3 were measured using the colorimetric assay kits. The cells were treated with 0, 25, 50 and 100 µM concentration of rosamultic acid then lysed and subjected to western blot analysis to detect total and activated caspase-8, -9, -3 and PARP. The data shown are representative for three independent experiments

ACCEPTED MANUSCRIPT

Figures

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 1.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 2.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 3.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 4.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 5.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 6.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 7.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 8.

ACCEPTED MANUSCRIPT

Fig. 9.

B

AN US

A

C

ED

M

D

PT CE AC

CR IP T

Fig. 9.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 10.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 11.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 12.

ACCEPTED MANUSCRIPT

M

AN US

CR IP T

Fig. 13A.

AC

CE

PT

ED

Fig. 13 B.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN US

CR IP T

Fig. 14.