Octyl Ester of Ginsenoside Rh2 Induces Apoptosis and G1 Cell Cycle

Sep 26, 2016 - Octyl Ester of Ginsenoside Rh2 Induces Apoptosis and G1 Cell Cycle Arrest in Human HepG2 Cells by Activating the Extrinsic Apoptotic Pa...
0 downloads 3 Views 3MB Size
Subscriber access provided by CORNELL UNIVERSITY LIBRARY

Article

Octyl Ester of Ginsenoside Rh2 Induces Apoptosis and G1 Cell Cycle Arrest in Human HepG2 Cells by Activating the Extrinsic Apoptotic Pathway and Modulating the Akt/p38 MAPK Signaling Pathway Fang Chen, Shi-Lian Zheng, Jiang-Ning Hu, Yong Sun, Yue-Ming He, Han Peng, Bing Zhang, David Julian McClements, and Ze-Yuan Deng J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.6b03519 • Publication Date (Web): 26 Sep 2016 Downloaded from http://pubs.acs.org on September 30, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Agricultural and Food Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 42

Journal of Agricultural and Food Chemistry

Octyl Ester of Ginsenoside Rh2 Induces Apoptosis and G1 Cell Cycle Arrest in Human HepG2 Cells by Activating the Extrinsic Apoptotic Pathway and Modulating the Akt/p38 MAPK Signaling Pathway Fang Chen,†,‡ Shi-Lian Zheng,†,‡ Jiang-Ning Hu,*,†,‡ Yong Sun,†,‡ Yue-Ming He,†,‡ Han Peng,†,‡ Bing Zhang,† David Julian McClements,& Ze-Yuan Deng*,†,‡ †

State Key Laboratory of Food Science and Technology, Nanchang University,

Nanchang, Jiangxi 330047, China ‡

College of Food Science, Nanchang University, Nanchang, Jiangxi 330047, China

&

Department of Food Science, University of Massachusetts Amherst, Amherst, MA

01003, USA

Corresponding authors: *Jiang-Ning Hu: Telephone No: +86 88304449-8226, E-mail address: [email protected] *Ze-Yuan Deng: Telephone/ Fax No: +86 791 88304402, E-mail address: [email protected]

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

1

ABSTRACT

2

Ginsenoside Rh2 is a potential active metabolite of ginseng that has antitumor activity

3

against a variety of tumor cells. Previously, we reported that Rh2-O, an octyl ester

4

derivative of ginsenoside Rh2, had a higher anti-cancer activity than Rh2 through

5

activating the intrinsic apoptotic pathway. In this study, we found that the extrinsic

6

apoptotic pathway was also involved in Rh2-O-induced HepG2 cells apoptosis as

7

evidenced by the up-regulation of Fas, FasL, TNFR1 and TNF-α, as well as the

8

cleavage of caspase 8. Moreover, flow cytometric analysis demonstrated that Rh2-O

9

induced G1 cell cycle arrest in HepG2 cells. Rh2-O-induced G1 phase arrest was

10

accompanied by the down-regulation of cyclin D3 and cyclin E and cyclin-dependent

11

kinase (CDK) 4 and 6, and the up-regulation of p21WAF1/CIP1 and p27KIP1. In addition,

12

Rh2-O down-regulated the phosphorylation of Akt, and its inhibitor LY294002

13

promoted Rh2-O-induced G1 phase arrest. Rh2-O treatment also activated p38

14

MAPK, JNK and ERK expression. Inhibitors of p38 MAPK (SB203580), but not

15

those of JNK (SP600125) or ERK (PB98095), promoted Rh2-O-induced G1 phase

16

arrest in HepG2 cells. These results indicated that the disruption of Akt and p38

17

MAPK cascades played a pivotal role in Rh2-O-induced G1 phase arrest.

18

KEYWORDS: Ginsenoside Rh2, octyl ester derivative, cell cycle, HepG2 cells, Akt,

19

MAPKs

ACS Paragon Plus Environment

Page 2 of 42

Page 3 of 42

Journal of Agricultural and Food Chemistry

20

INTRODUCTION

21

Ginseng, well known as “renshen” in Chinese, is one of the most popular tonics in

22

oriental countries. Ginsenoside Rh2 is a major bioactive ingredient in ginseng that has

23

been shown to have antitumor activities on a variety of tumor cells, including human

24

hepatoma SK-HEP-1 cells, human astroglioma U87MG cells, and human cervical

25

carcinoma Hela cells.1-4 However, Rh2 currently has a limited application as a

26

bioactive ingredient in the food, supplement, and pharmaceutical industries because

27

of its poor oral bioavailability, which is associated with its low membrane

28

permeability.5-7 The main reason for the low permeability of Rh2 is the fact that it is a

29

hydrophilic substance that is not easily solubilized and transported through the

30

phospholipid bilayers in cell membranes.

31

has been increased by synthesizing an octyl ester (Rh2-O) from Rh2 and octanoyl

32

chloride.8 Structural analysis indicated that the fatty acid ester substituent connected

33

to the hydroxyl group at C-12 of Rh2.8 A recent study in our laboratory showed that

34

Rh2-O improved the absorption of Rh2 in Caco 2 cells, which suggested that

35

esterification of Rh2 might lead to an improvement in its bioavailability.9 Moreover,

36

it was confirmed that Rh2-O had a higher anti-cancer activity than Rh2.8 However,

37

the exact molecular mechanisms responsible for this increased activity need to be

38

elucidated before Rh2-O can be used as an effective anticancer agent.

Recently, the lipophilic character of Rh2

39

Apoptosis, a process of programmed cell death, is a highly regulated form of cell

40

death.10, 11 Two main cysteine-aspartate proteases (caspase) activation cascades have

41

been characterized.12,

42

receptors, such as Fas, TNFR, and DR4.14, 15 The death receptors are activated when

43

they bind with their corresponding natural ligands, which is followed by activation of

13

The extrinsic apoptotic pathway is mediated by death

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 42

44

caspase 8 to induce cell apoptosis.16 The intrinsic apoptotic pathway, driven by the

45

Bcl-2 family proteins, involves the depolarization of the mitochondrial outer

46

membrane, which subsequently leads to the release of cytochrome c and the activation

47

of caspase 9.17, 18 Our previous study demonstrated that Rh2-O induced HepG2 cells

48

apoptosis by stimulating the production of reactive oxygen species (ROS),

49

mitochondrial dysfunction, and caspase activation.8 However, the signal transduction

50

pathways involving the extrinsic apoptotic pathway in Rh2-O-induced HepG2 cells

51

apoptosis are not yet fully understood.

52

Controlling the growth and proliferation of cancer cells is another main

53

molecular mechanism responsible for the anti-tumor effects of phytochemicals.

54

Eukaryotic

55

cyclin-dependent kinases (CDKs), which is dependent upon association with

56

cyclins.19 In addition, some CDK inhibitors (CDKIs) are also involved in Eukaryotic

57

cell-cycle progression.20 Rh2 has been shown to induce G1 cell cycle arrest by

58

down-regulations of the protein levels and kinase activities of cyclin D1, cyclin E, and

59

CDK 6, and up-regulation of pRb2/p130 in human lung adenocarcinoma A549 cells.21

60

Another study has shown that Rh2-induced G1 cell cycle arrest was accompanied by

61

the down-regulation of CDK4, CDK6, cyclin D1/2/3, and cyclin E at protein level and

62

the gradual up-regulation of p21WAF1/CIP1 and p27KIP1 in HL-60 promyelocytic

63

leukemia and U937 human histocytic lymphoma cells.22 However, the specific

64

mechanisms of action and underlying signaling pathways in the inhibition of the

65

proliferation of HepG2 cells by Rh2-O have not yet been elucidated.

66

cell-cycle

progression

involves

the

sequential

activation

of

A network of interacting proteins mediates the transmission of signals in cells a

large

number

of cellular processes.23

67

that govern

68

3-kinase/protein kinase B (PI3K/Akt) and mitogen-activated protein kinases (MAPKs)

ACS Paragon Plus Environment

Phosphatidylinositol

Page 5 of 42

Journal of Agricultural and Food Chemistry

69

play critical roles in controlling the growth and proliferation of cancer cells through

70

different extracellular stimulations.24,

71

MAPKs has been reported in several cancers, including H1299 lung cancer cell lines,

72

HSC-3 oral carcinoma cell lines, and MDA-MB-468 breast cancer cell lines.26-28

73

Therefore, it is important to verify the relationship between Rh2-O and PI3K/Akt or

74

MAPKs in carcinogenesis for cancer prevention or treatment. The primary objective

75

of the current study was therefore to identify the molecular mechanisms responsible

76

for the anticancer activity of Rh2-O using HepG2 as a representative cell line model.

77

The results obtained are expected to support the development of a strong scientific

78

foundation for using Rh2-O as a chemopreventive and/or chemotherapeutic agent

79

against liver cancer.

80

MATERIALS AND METHODS

81

Materials and Reagents. Rh2-O was synthesized using the method described in our

82

recent study.8 Normal growth media (MEM) medium and fetal bovine serum (FBS)

83

were

84

diphenyltetrazolium bromide (MTT), proteinase K, phenylmethanesulfonyl fluoride

85

(PMSF) as well as all inhibitors utilized in this study were procured from Sigma

86

Chemical Co. (St. Louis, MO, USA). The cell cycle detection kit was obtained from

87

B.D. Clontech Laboratories (Mountain View, CA, USA). Antibodies against cyclin

88

D3, cyclin E, CDK2, CDK4, CDK6, p21WAF1/CIP1, p27KIP1, Fas, FasL, TNF-α, TNFR1,

89

DR4, cleaved-caspase 8, pro-caspase 8, ERK1/2, p-ERK1/2, p38 MAPK, p-p38

90

MAPK, JNK, p-JNK, Akt and p-Akt were obtained from Cell Signaling Technology

91

(Beverly, MA, USA). Antibodies against β-actin and all the second antibodies were

obtained

from

25

Gibco-BRL

The proliferative role of PI3K/Akt and

Co.

3-(4,5-dimethylthiazole-2-yl)-2,5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

92

purchased from Trans Transgen Biotechnology Co. (Beijing, China). All other

93

compounds were of analytical grade.

94

Cell Culture and Treatment. The human liver hepatocellular carcinoma cell line

95

HepG2 was obtained from the National Centre for Cell Sciences (NCCS, Shanghai,

96

China). HepG2 cells were grown in MEM medium supplemented with 10% (v/v) FBS,

97

100 units/mL penicillin, and 100 µg/mL streptomycin. Cells were incubated in a

98

humidified incubator with 95% air and 5% CO2 at 37 °C. In additional experiments,

99

cells were pretreated with different inhibitors: PI3K inhibitor (LY294002, 10-30 µM),

100

p38 MAPK inhibitor (SB203580, 10-30 µM), ERK inhibitor (PD98059, 50 µM), or

101

JNK inhibitor (SP600125, 50 µM) for 1 h followed by incubation with Rh2-O

102

(dissolved in DMSO) or the vehicle (DMSO, < 0.1%) for 12 h.

103

Cell Viability Assay. The effects of inhibitors on Rh2-O-induced HepG2 cell death

104

were monitored using MTT assays.29 Briefly, 0.2 mL of cells (1 × 104 cells per well)

105

was placed onto 96-well microassay plates. Following incubation for 12 h, cells were

106

pretreated with various inhibitors (LY294002, SB203580, PD98059, or SP600125)

107

for 1 h and then treated with 17.5 µM of Rh2-O. After treatment, 0.2 mL of MTT

108

solution (2 mg/mL in culture media) was added into each well of the plates and then

109

incubated for 4 h in the dark. The supernatant was then removed, and incubated with

110

0.15 mL of DMSO to dissolve the purple MTT-formazan crystals with shaking for 10

111

min. The absorbance was determined at 490 nm in a microplate reader. Cells

112

incubated with 0.1% DMSO were used as controls. In MTT assays, every sample was

113

performed at least 3 times.

114

PI Staining Assay. Cell cycle distribution was measured by flow cytometric

115

analysis.27 HepG2 cells were seeded on culture dishes for 12 h, and a double

116

thymidine block procedure was used to synchronize the cell cycle. The cell cycle

ACS Paragon Plus Environment

Page 6 of 42

Page 7 of 42

Journal of Agricultural and Food Chemistry

117

synchronized HepG2 cells were cleansed twice with PBS and then re-stimulated to

118

enter the G1 phase together by treatment with Rh2 (17.5 µM) or various

119

concentrations of Rh2-O (10-17.5 µM) for 12 h. The cells were collected and washed

120

with pre-cooling PBS. One mL 70% ethanol was added to fix the collected cells at

121

-20 °C overnight. After centrifugation, the collected cells were incubated with 50.1

122

µg/mL PI (1.21 mg/mL Tris, 700 U/mL RNase, pH 8.0) for 1-2 h. The DNA contents

123

were further determined by flow cytometry (FACSCalibur; Becton Dickinson

124

Biosciences, San Jose, CA). The number of cells for each sample was at least 20,000.

125

RNA Isolation and Real-Time Quantitative Reverse Transcription-PCR. Total

126

RNA of the cells was extracted using total RNA extraction reagent (TaKaRa)

127

according to the manufacturer’s instruction. RNA was confirmed by running on a 1.5%

128

agarose gel. Reverse transcription was performed with 1 µg of total RNA using

129

TransScript first-strand cDNA synthesis supermix kit (TaKaRa) according to the

130

supplier’s instruction. The mRNA expression levels of genes were determined by

131

real-time PCR using SYBR premix ex TagTM (TaKaRa) in the ABI 7900HT

132

real-time PCR system (Applied Biosystems, USA). Simultaneously, the quantitative

133

analysis of mRNA levels was normalized to the endogenous reference gene GAPDH.

134

The reaction was carried out under the following conditions: 95 °C for 30 s, 40 cycles

135

at 95 °C for 5 s, and 58 °C for 30 s. The primers used are shown in Table 1. Values of

136

each group mRNA level was calculated as 2-∆∆Ct levels and performed at least 3 times.

137

Preparation of Whole-Cell Lysates. HepG2 cells (1 × 105 cells) were plated in a 100

138

mm dish and treated with 17.5 µM of Rh2 or various concentrations of Rh2-O

139

(10-17.5 µM) for 12 h. After treatment, cells were cleansed twice with cold PBS and

140

then lysed with iced-cold radio-immunoprecipitation assay (RIPA) buffer. PMSF (10

141

mg/mL), leueptin (17 mg/mL), and sodium orthovanadate (10 mg/mL) were then

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

142

added. After incubation for 30 min on ice, the lysates were centrifuged at 1000g for

143

10 min, and then the supernatants were collected as whole-cell lysates.

144

Western Blot Analysis. The concentrations of protein in the samples were quantified

145

using a BCA Protein Assay kit (Beyotime Biotechnology, Shanghai, China). Forty µg

146

of cell lysate protein was mixed with SDS loading buffer (6 ×) and then treated by

147

boiling for 10 min. The obtained protein samples were separated on the basis of their

148

size using 12% gradient SDS-PAGE gel, and then transferred onto a nitrocellulose

149

membrane. After that, the membrane was blocked with 5% nonfat milk containing 1%

150

(v/v) Tween-20 for 1 h at ambient temperature, and then incubated with specific

151

primary antibodies at 4 °C for more than 16 h. After washing 5 times with

152

Tris-buffered saline containing 1% (v/v) Tween-20 (TBST), the membrane was

153

incubated with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse

154

secondary antibodies at ambient temperature for 1 h. Finally, protein bands were

155

detected using an enhanced chemiluminescence (ECL) detection system (GE

156

Healthcare).

157

Statistical Analysis. Study data are reported as means ± the standard deviations.

158

Statistical analyses were carried out by the student’s t-test or the one-way analysis of

159

variance (ANOVA) using a statistical software package (SPSS, USA). p-values of

160