Ginsenoside Re Ameliorates Brain Insulin Resistance and Cognitive

Mar 17, 2017 - Dysfunction in High Fat Diet-Induced C57BL/6 Mice ... KEYWORDS: cognitive impairment, diabetes mellitus, high-fat diet, ginsenoside Re,...
0 downloads 0 Views 3MB Size
Subscriber access provided by HACETTEPE UNIVERSITESI KUTUPHANESI

Article

Ginsenoside Re Ameliorates Brain Insulin Resistance and Cognitive Dysfunction in High-fat Diet-induced C57BL/6 Mice Jong Min Kim, Chang Hyeon Park, Seon Kyeong Park, Tae Wan Seung, Jin Yong Kang, Jeong Su Ha, Du Sang Lee, Uk Lee, Dae Ok Kim, and Ho Jin Heo J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.7b00297 • Publication Date (Web): 17 Mar 2017 Downloaded from http://pubs.acs.org on March 18, 2017

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 41

Journal of Agricultural and Food Chemistry

1

Running Title: Ginsenoside Re attenuated cognitive impairment in C57BL/6 mice

2

3

Ginsenoside Re Ameliorates Brain Insulin Resistance and

4

Cognitive Dysfunction in High-fat Diet-induced C57BL/6 Mice

5 6

☆ ☆ Jong Min Kim†,☆ , Chang Hyeon Park†,☆ , Seon Kyeong Park†, Tae Wan Seung†,

7

Jin Yong Kang†, Jeong Su Ha†, Du Sang Lee†, Uk Lee§, Dae-Ok Kim#, Ho Jin Heo†,*

8

-----------------------------------------------------------------------------------------------------------------------------------

9

*

Corresponding author at: Division of Applied Life Science(BK21 plus), Institute of

10

Agriculture & Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea.

11

Tel: +82 55 772 1907; Fax: +82 55 772 1909. E-mail address: [email protected] (H. J. Heo)

12

Author detail

13



14

Gyeongsang National University, Jinju 52825, Republic of Korea

15

§

16

Republic of Korea

17

#

18

Republic of Korea

19

CHP: [email protected], JMK: [email protected], SKP: [email protected],

20

TWS: [email protected], JYK: [email protected], JSH: [email protected],

21

DSL: [email protected], UL: [email protected], DOK: [email protected], HJH:

22

[email protected]

Division of Applied Life Science (BK21 plus), Institute of Agriculture and Life Science,

Division of Special Purpose Trees, National Institute of Forest Science, Suwon 16631,

Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104,

23 24



These authors contributed equally to this work as the co-first author. 1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

25

ABSTRACT

26

The ameliorating effects of ginsenoside Re (G Re) on high-fat diet (HFD)-induced insulin

27

resistance in C57BL/6 mice were investigated to assess its physiological function. In the

28

results of behavioral tests, G Re improved cognitive dysfunction on diabetic mice using Y-

29

maze, passive avoidance and Morris water maze tests. G Re also significantly recovered

30

hyperglycemia and fasting blood glucose level. In the results of serum analysis, G Re

31

decreased triglyceride (TG), total cholesterol (TCHO), low density lipoprotein cholesterol

32

(LDLC), glutamic-oxaloacetic transaminase (GOT) and glutamic-pyruvic transaminase

33

(GPT), and increased the ratio of high density lipoprotein cholesterol (HDLC). G Re

34

regulated the acetylcholine (ACh), acetylcholinesterase (AChE), malondialdehyde (MDA),

35

superoxide dismutase (SOD) and oxidized glutathione (GSH)/total GSH by regulating the c-

36

Jun N-terminal protein kinase (JNK) pathway. These findings suggest that G Re could be

37

used to improve HFD-induced insulin resistance condition by ameliorating hyperglycemia via

38

protecting the cholinergic and antioxidant system in the mice brains.

39 40

Keywords: Cognitive impairment, diabetes mellitus, high-fat diet, ginsenoside Re, JNK

41

pathway

42 43 44 45 46 47 48

2

ACS Paragon Plus Environment

Page 2 of 41

Page 3 of 41

Journal of Agricultural and Food Chemistry

49

INTRODUCTION

50

Diabetes mellitus (DM) is a chronic disease abnormally progressed by

51

hyperglycemia, and also, may damage almost all organs in the body, such as the central and

52

peripheral nerves, retinas, liver, and kidneys. Recently, there have been increasing issues

53

associated with DM and its complications, in particular, diabetes-associated cognitive

54

impairment (DACM).1 Although the DM-induced mechanisms of cognitive deficiency are

55

not fully understood, hyperglycemia and insulin resistance seem to be markedly associated

56

with DM. In addition, it has been consistently suggested that oxidative stress is connected

57

with the progression of diabetic complications.2 Oxidative stresses also play an important role

58

in DACM. Simultaneously, chronic consumption of a high-fat diet (HFD) may cause the

59

production of reactive oxygen species (ROS) and mitochondrial impairment.3 The damaged

60

mitochondrial function in a neuronal cell leads to a deficit of insulin signaling, which may

61

trigger insulin resistance and hyperinsulinemia. Recent studies have considered the role of

62

insulin resistance in the cognitive impairment and pathological progress of Alzheimer’s

63

disease (AD).4 Insulin resistance is also associated with phosphorylated c-Jun N-terminal

64

kinase (p-JNK), which leads to phosphorylation of a serine site in the insulin receptor

65

substrate (IRS), and continuously impedes serine/threonine kinase (Akt) signaling.5

66

Consequently, inhibited Akt signaling is caused by decrease of phosphorylated Akt (p-Akt),

67

the activated form, and induce the activation of GSK-3β, which expedites the

68

phosphorylation of tau attached in microtubules and aggregation of phosphorylated tau (p-

69

tau).6 Moreover, an abnormal insulin signal and activated GSK-3β precipitate

70

neurodegeneration promoting the formation of neurofibrillary tangles (NFT), leading to

71

neuronal death.7 Finally, in neurons, p-JNK can trigger the impairment of energy production

72

processes and cause neuronal inflammation, cognitive dysfunction and signaling associated 3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 41

73

with AD.5 Also, p-JNK up-regulates the expression of Bax and Bad as pro-apoptosis factors,

74

and impedes the Bcl-2 as survival factor. An increase in these pro-apoptotic factors stimulates

75

cytochrome c release from the mitochondria to cytosol, and consistently, expedites the

76

activation and cleavage of caspase-9, caspase-3, and poly (ADP-ribose) polymerase (PARP)

77

related with apoptosis in the neuronal cell.8

78

Ginsenoside Re (PubChem CID: 441921, G Re) is a triterpenoid saponin compound

79

typically isolated from Panax ginseng. Recently, our research showed that the extract of

80

Panax ginseng berry had an anti-amnesic effect, and G Re was identified as the main

81

compound by ultra-performance liquid chromatography/quadrupole time of flight tandem

82

mass spectrometry (Q-TOF UPLC/MS2).9 G Re improves cognitive impairment against

83

scopolamine, NaNO2 and ethanol-induced oxidative stress.10 G Re also has an anti-diabetic

84

effect including the protection of the kidney and eyes against diabetic oxidative stress, and

85

remarkably ameliorates DACM in streptozotocin-induced type 1 diabetic rats.11-12

86

Nevertheless, there is relatively limited information on the cognitive ability associated with

87

type 2 diabetes mellitus (T2DM) induced by the HFD. For this reason, this study aimed to

88

evaluate whether G Re can improve HFD-induced T2DM associated cognitive dysfunction,

89

using various in vivo tests, ex vivo biochemical changes and the JNK pathway.

90 91

MATERIALS AND METHODS

92

Materials. Ginsenoside Re was purchased from Aktin Chemicals, Inc. (Chengdu,

93

China).

94

metaphosphoric acid, 4-vinylpyridine, bovine serum albumin (BSA), glucose, dimethyl

95

sulfoxide (DMSO), hydrochloride, iron (Ш) chloride hexahydrate, sodium hydroxide,

96

superoxide dismutase (SOD) determination kit, and all other chemicals used in this study

Acetylthiocholine

iodide,

thiobarbituric

acid

4

ACS Paragon Plus Environment

(TBA),

phosphoric

acid,

Page 5 of 41

Journal of Agricultural and Food Chemistry

97

were purchased from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA). The total

98

glutathione (GSH) kit was purchased from Enzo Life science Inc. (Farmingdale, NY, USA).

99

Animals and treatment. C57BL/6 mice (4 weeks-old, male) were purchased from

100

supplier (Samtako, Osan, Korea). Mice were housed 3 per cage, and freely allowed access to

101

water and feed, and controlled in standard laboratory conditions (steady temperature, 23±1°C;

102

humidity, 55±5% and 12 h light/dark cycle). All experiments followed the guidelines

103

produced by the Animal Care and Use Committee of Gyeongsang National University

104

(certificate: GNU-131105-M0067). All mice excluding the control group were fed with the

105

HFD for 8 weeks. After induction of high glucose status on mice, the G Re was mixed in

106

purified water (5, 10 and 20 mg/kg of body weight; G Re 5, G Re 10 and G Re 20,

107

respectively), and administered intragastrically once per day for 4 weeks. Daily food intake

108

and weekly body weight were measured. The overall design of the experiment is shown in

109

Figure 1.

110 111

Behavioral tests for cognitive evaluation. Y-maze is constituted as triple opaquely

112

white plastic arms. Each mouse which was located at the end of designated arm is allowed to

113

freely explore the maze for 8 min. The sequences of arm entries were assessed using a video

114

system (Smart 3.0 video tracking system, Panlab, Barcelona, Spain). The ratio of spatial

115

perception was measured as the ratio of the number of entries.13

116

Passive avoidance test chamber is separated to a light compartment and a dark

117

compartment, with a gate between the two compartments. When the mice entered the dark

118

zone, an electrical shock was provide to entered mice (0.5 mA, 3 sec). Mice will learn to

119

associate certain properties of the chamber with the foot shock. Next day, the mice were put

120

into non-dark area, and the latency times until entering to the dark compartment were 5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

121

measured for 5 min.14

122

Morris water maze consists of a round pool divided into quadrant (N, S, E and W

123

zones, respectively), and each quadrant zones was indicated to different visual cues. This

124

pool was filled with opaque water using skim milk, depth of water was 30 cm, and

125

temperature was maintained at 20±2°C. The platform was fixed into the steady location in the

126

W zone during training session (day 1-4). In the training test, mice freely were swam until

127

they reached the platform to escape from the opaque water during maximum 60 sec. Latency

128

time to arrive in the hidden platform was recorded in each training trial using the video

129

system (Panlab). After training test, the probe test was conducted. In the probe test, the

130

platform was removed from the test zone, and mice allowed to freely swimming for 90 sec.15

131

Intraperitoneal glucose tolerance test (IPGTT) and measurement of fasting

132

blood glucose. To measure the IPGTT, mice were fasted during 4 h. After that, D-glucose (2

133

g/kg of body weight) was injected in the abdominal cavity of mice, and blood samples were

134

taken at 0, 30, 60, 90, and 120 min after the intraperitoneal injection. Glucose levels in the

135

fasting state, which collected from tip of the tail vein, was measured at every week using

136

Accu-Chek glucose meter (Roche Diagnostics GmbH, Mannheim, Germany).

137

Collection of serum and brain tissues. After behavioral test, mice which had

138

undergone fasting for 12 h were sacrificed by CO2 inhalation for biochemical studies. The

139

serum was collected from abdominal aorta. The collected brain tissue was homogenized

140

using a bullet blender (Nextadvance Inc., Averill Park, NY, USA) with 10-fold volumes of

141

phosphate buffered saline (PBS) for acetylcholine (ACh), acetylcholinesterase (AChE),

142

malondialdehyde (MDA) and SOD assay, and 20-fold volumes of 5% metaphosphoric acid

143

for GSH assay in an ice bath. The concentration of cerebral protein was determined through a

144

Bradford protein assay.13 6

ACS Paragon Plus Environment

Page 6 of 41

Page 7 of 41

Journal of Agricultural and Food Chemistry

145

Serum biochemicals. The collected blood sample was stored at room temperature for

146

20 min in heparin tube. And then, the supernatant was collected from obtained blood sample

147

centrifuged at 10,000×g for 15 min at 4°C, and has conducted serum analysis. The glutamic

148

oxaloacetic transaminase (GOT), glutamine pyruvic transaminase (GPT), blood urea nitrogen

149

(BUN), creatine (CRE), total cholesterol (TCHO), triglyceride (TG), and high density

150

lipoprotein cholesterol (HDLC) concentrations of serum were measured using clinical

151

chemistry analyzer (Fuji dri-chem4000i; Fuji film Co., Tokyo, Japan). Low density

152

lipoprotein cholesterol (LDLC) was determined as follow; LDLC (mg/dl) = TCHO - (HDLC

153

+ TG/5).16

154

AChE activity and ACh level in brain tissue. The homogenized brain tissues at the

155

PBS were centrifuged to collect the supernatant to measure AChE activity and ACh level,

156

respectively at 14,000×g for 30 min at 4°C. AChE activity was determined using the

157

colorimetric method.17 The supernatant with 50 mM sodium phosphate buffer (pH 8.0) was

158

reacted at 37°C for 15 min. Acetylthiocholine iodide solution (0.5 mM) and buffered

159

Ellman's reagent [1 mM 5,50-dithio-bis (2-nitrobenzoic acid) in 50 mM sodium phosphate

160

buffer (pH 8.0)] were reacted at room temperature for 10 min. Absorbance was measured at

161

405 nm. ACh level was estimated as described by the colorimetric method.18 The supernatant

162

was added to alkaline hydroxylamine reagent [2 M hydroxylamine in HCl and 3.5 N sodium

163

hydroxide] at room temperature for 1 min. Then, 0.5 N HCl and 0.37 M FeCl3 in 0.1N HCl

164

(pH 1.2) was added. The absorbance measured at 540 nm.

165

MDA level, SOD content, and ratio of oxidized GSH/total GSH in brain tissue.

166

The MDA levels were implemented in accordance with colorimetric way by combining

167

between the MDA in the brain tissues in mice and TBA. The homogenized tissues at the PBS

168

were centrifuged at 2,500×g for 10 min at 4°C. After that, the acquired supernatants were 7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

169

mixed with 1% phosphoric acid and 0.67% TBA in water bath for 1 h at 95°C. And then, this

170

reactants were centrifuged at 600×g for 10 min to discard the insoluble substances. This

171

supernatants were measured at 532 nm, and the MDA contents were expressed as nmole/mg

172

of protein.13

173

To measure the SOD content, homogenized brain tissues at the PBS were centrifuged

174

at 400×g for 10 min at 4°C, and after these supernatants were removed. 1×Cell Extraction

175

Buffer containing the 10% SOD buffer, 0.4% (v/v) Triton X-100, and 200 µM

176

phenylmethane sulfonylfluoride in distilled water were added into pellet, and incubated for

177

30 min on the ice bath. This reactant was centrifuged at 10,000×g for 10 min at 4°C. The

178

supernatant was acquired, and used according to guided protocol of the commercial kit.

179

To measure the total GSH and oxidized GSH ratio, the homogenized brain tissues at

180

5% metaphosphoric acid were centrifuged at 14,000×g for 15 min at 4°C. And then,

181

supernatant and 2 M 4-vinylpyridine were incubated at room temperature for 1 h to measure

182

the oxidized GSH. The evaluation of GSH was performed using the commercial kit.

183

Western blot analysis. The brain tissues were homogenized for 15 min with

184

ProtinExTM Animal cell/tissue (GeneAll Biotechnology, Seoul, Korea) with 1% protease

185

inhibitor cocktail (Thermo Fisher Scientific, Rockford, IL, USA). After that, homogenates

186

were centrifuged at 13,000×g for 10 min at 4oC, and these supernatants were collected and

187

stored at -80oC. The supernatant was loaded into sodium dodecyl sulfate polyacrylamide

188

(SDS-PAGE) gel electrophoresis, and electro-transferred to a poly-vinylidene difluoride

189

(PVDF) membrane (Millipore, Billerica, MA, USA). The membranes were reacted in tris-

190

buffered saline (TBS) containing 0.1% Tween 20 (TBST) containing 1:4000 diluted solutions

191

of mouse monoclonal primary antibody to bind β-actin, p-JNK (Thr183/Tyr185), p-tau

192

(Ser404), and cleaved PARP (Asp214; c-PARP) or 1:4000 diluted solutions of rabbit 8

ACS Paragon Plus Environment

Page 8 of 41

Page 9 of 41

Journal of Agricultural and Food Chemistry

193

monoclonal primary antibody to combine phosphorylated IRS (Ser307; p-IRS) at 4oC. After

194

that, membrane was reacted with horserdish peroxide-conjugated anti-mouse IgG (1:2000) or

195

anti-rabbit IgG (1:5000) secondary antibody solution for 1 h in TBST including 5% skim

196

milk. For chemiluminescence detection, the immune complexes were incubated by

197

chemiluminescence with ECL reagent (Bionote, Hwaseong, Korea). The density of band was

198

measured by Image J software (National Institutes of Health, Bethesda, MD, USA).

199

Statistical analysis. All data was presented as the mean±standard deviation (SD).

200

The statistical significance of differences between the groups was measured by a one-way

201

analysis of variance (ANOVA). The significance of differences was conducted by the

202

Duncan's new multiple-range test (p‹0.05) of SAS ver. 9.1 (SAS Institute Inc., Cary, NC,

203

USA).

204 205

RESULTS

206

Body weight and food intake. To evaluate the ameliorating effect of G Re on HFD-

207

induced weight regulation, the weight gain, and food intake values were measured (Table 1.).

208

The final body weight of the control, HFD, and G Re 5, 10, and 20 groups showed 33.25 g,

209

50.50 g, 48.25 g, 47.33 g, and 45.23 g, respectively. The food intake of the control, HFD, and

210

G Re 5, 10 and 20 groups was 2.36 g/day, 3.18 g/day, 2.74 g/day, 2.56 g/day, and 2.46 g/day,

211

respectively.

212

Blood glucose and insulin tolerance. The fasting blood glucose during the sample

213

intake period is presented in Figure 2A. After starting the HFD, the fasting blood glucose

214

level of the HFD group was significantly higher than the control group. However, G Re

215

administrations improved the abnormal fasting blood glucose level. To confirm the regulation

216

of insulin tolerance on HFD-induced T2DM, the intraperitoneal glucose tolerance (IPGT) 9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

217

was also measured at 0, 15, 30, 60, and 90 min (Figure 2B).

218

Before glucose injection (0 min), the groups treated with the HFD (312.47 mg/dL)

219

showed a hyperglycemic state compared to the control group (185.00 mg/dL), and all groups

220

showed the highest glucose level at 15 min. The glucose level of the control group returned to

221

a normal state after 90 min (190.42 mg/dL). In contrast, the HFD group presented a lower

222

width of decreased at a faster rate between 15 and 90 min (528.37 mg/dL and 395.11 mg/dL,

223

respectively) than the other groups. However, the sample groups (G Re 5, 357.23 mg/dL; G

224

Re 10, 320.23 mg/dL; and G Re 20, 281.32 mg/dL, respectively) showed improved glucose

225

tolerance compared to the HFD group.

226

in vivo behavioral tests. The learning and memory effects against diabetes-

227

associated cognitive dysfunction were sequentially evaluated by the Y-maze, passive

228

avoidance, and Morris water maze tests (Figure 3A-F). Alternation behavior of the HFD

229

group (23.56%) was decreased compared to the control group (52.23%). However, those of

230

the sample groups (G Re 5, 41.76%; G Re 10, 44.33%, and G Re 20, 46.89%, respectively)

231

were statistically improved (Figure 3A). All groups, except the HFD group, showed a similar

232

number of arm entries. However, the HFD group showed only a decreased movement caused

233

by HFD-induced obesity in the arms (Figure 3B).

234

The HFD group had a significantly shortened latency time before entering the dark

235

room in the retention trial (182 .00 sec) compared to that of the control group (300.00 s).

236

Whereas, the G Re 10 (300.00 sec) and G Re 20 group (300.00 sec) showed similar

237

improvement in short-term memory compared to the HFD group (Figure 3C).

238

The escape latency time of the HFD group (51.03 sec at day 1 to 37.12 sec at day 4)

239

was relatively higher than the control group (38.15 sec at day 1 to 13.69 sec at day 2).

240

However, those of the G Re groups (G Re 5, 44.40 sec to 23.92 sec; G Re 10, 43.38 sec to 10

ACS Paragon Plus Environment

Page 10 of 41

Page 11 of 41

Journal of Agricultural and Food Chemistry

241

21.83 sec, and G Re 20, 39.43 sec to 16.84 sec, respectively) were significantly decreased

242

compared to the HFD group (Figure 3D). In the probe test, long-term memory and the

243

spontaneous behavioral ability of the HFD group was significantly decreased (37.18% stay

244

ratio in the target area) compared to that of the control group (56.18%). However, the G Re

245

treatment groups (G Re 5, 40.51%; G Re 10, 45.05%, and G Re 20, 49.95%, respectively)

246

showed improved long-term cognitive function (Figure 3E) compared to that of the HFD

247

group. In addition, the path tracing showed the mice of the control group circled around the

248

area marked with a circle (W zone) on the platform, relatively more than the other groups,

249

whereas, the mice of the HFD group circled around all zones (Figure 3F). These tracks made

250

by the mice also present learning and memory ability regarding eliminated platforms in the

251

area marked with a circle (W zone).

252

Serum biochemicals. Effects of G Re on changes of serum biochemicals in the

253

HFD-induced mice were analyzed (Table 2). The contents of the GOT and GPT have

254

generally been used as hepatotoxic indicators.19 The contents of both indicators in the G Re

255

groups (G Re 5, 110.33 U/L and 80.13 U/L; G Re 10, 103.33 U/L and 50.32 U/L, and G Re

256

20, 86.32 U/L and 39.24 U/L, respectively) were decreased compared to those of the HFD

257

group (130.67 U/L and 110.25 U/L). The levels of the TG, TCHO, HDLC and LDLC are

258

commonly used to assess the serum lipid state, such as hyperlipidemia and

259

hypercholesterolemia.20 The levels of these lipid indicators, excluding the HDLC in the HFD

260

group, was considerably increased compared to that of the control group. In contrast, the

261

levels of these indicators in the G Re groups were significantly decreased compared to those

262

in the HFD groups, and the increased HDLC level contrasted with the decreased TG, TCHO,

263

and LDLC levels.

264

AChE activity and ACh level in brain tissue. In the current study, the AChE 11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

265

activity was increased in the HFD group (110.14%) compared to that of the control group

266

(100.00%). However, that of the G Re groups (G Re 5, 102.56%; G Re 10, 98.36%, and G Re,

267

91.55%, respectively) was dose-dependently decreased (Figure 4A). The ACh level showed

268

the opposite result to the AChE activity (Figure 4B). The ACh content of HFD group (1.77

269

mmole/mg of protein) was increased compared to that of the control group (1.99 mmole/mg

270

of protein). However, that of the G Re groups were represented as G Re 5, 1.78 mmole/mg of

271

protein; G Re 10, 1.89 mmole/mg of protein, and G Re, 2.03 mmole/mg of protein,

272

respectively.

273

MDA level, SOD content, and oxidized GSH /total GSH ratio. The protective

274

effect of G Re against lipid peroxidation was measured by the MDA level (Figure 5A). The

275

MDA level of the HFD group (4.51 nmol/mg of protein) increased compared to the control

276

group (3.45 nmol/mg protein). However, the sample groups (G Re 5, 4.23 nmol/mg of protein;

277

G Re 10, 3.71 nmol/mg of protein, and G Re 20, 3.35 nmol/mg of protein, respectively)

278

gradually decreased the MDA level compared to that of the HFD group. The SOD content of

279

the HFD group (3.18 U/mg of protein) was decreased in the brain because of the increased

280

oxidative stress compared to the control group (4.50 U/mg). However, the sample groups (G

281

Re 5, 3.51 U/mg of protein; G Re 10, 4.12 U/mg of protein; and G Re 20, 4.46 U/mg of

282

protein, respectively) statistically increased the SOD content by decreasing the oxidative

283

stress compared to that of the HFD group (Figure 5B). In addition, the ratio of oxidized

284

GSH/total GSH of the HFD group (50.23%) was drastically increased compared to that of the

285

control group (34.25%). In contrast, the administration of G Re (G Re 5, 43.33%; G Re 10,

286

39.12%, and G Re 20, 35.13%, respectively) showed a dose-dependent improvement in the

287

ratio of oxidized GSH/total GSH (Figure 5C).

288

Cerebral apoptotic signaling. The ameliorating effect of G Re on cerebral insulin 12

ACS Paragon Plus Environment

Page 12 of 41

Page 13 of 41

Journal of Agricultural and Food Chemistry

289

signaling was examined in the brain of the HFD mice via western blot assay (Figure 6). The

290

level of p-JNK expression in the brain of the HFD mice was significantly up-regulated

291

compared to the control group (Figure 6B). In contrast, in the G Re treatment groups, it was

292

down-regulated compared to the HFD group. Also, p-IRS and p-tau expression levels in the

293

HFD group were considerably increased compared to the control group, while, in the

294

respective G Re groups the levels were remarkably decreased compared to the HFD group

295

(Figure 6C, D). While, c-PARP expression of HFD group is not significant compared to that

296

of control group. However, that of G Re group is considerably reduced compared to that of

297

HFD group (Figure 6E).

298 299

DISCUSSION

300

The two primary forms of DM are known to type 1 and 2 diabetes. Both types are

301

connected with progressive β-cell impairment.21 Type 1 DM (T1DM) results from mainly

302

impairment of pancreatic β-cell due to an autoimmune-mediated impairment.22 And also,

303

these β-cell loss is induced by activated macrophages and T-cells, and cytokines, nitric oxide

304

(NO), and free radicals secreted by these cells23. Whereas, T2DM, one of the most common

305

metabolic disorders, is related to impaired insulin signaling and insulin resistance.21 T2DM is

306

related to dysfunction of carbohydrate, lipid and protein metabolism with serious effects

307

resulting in long-term complications. These complications are associated with the

308

physiological abnormalities and problems in organ, and may cause cerebrovascular disease

309

and dementia.4 The causes of these DM are known to excess of glucorticoids and growth

310

hormone, pregnancy, Lipodystrophy, mutations of insulin receptor, and obesity/overweight

311

through high calorie diet.24 Especially, the administration of HFD is associated with the

312

T2DM, obesity, and diverse metabolic syndromes, and HFD can induce the collapse of 13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

313

antioxidant system in brain as well as increased hepatocellular TG contents and lipid

314

peroxidation in liver. 1, 2 And, these factors lead to insulin resistance in brain, and ultimately

315

cause the cognitive dysfunction and AD.5 Therefore, we studied whether G Re can ameliorate

316

the HFD-induced metabolic disease related with T2DM such as the insulin resistance, the

317

memory impairment, hyperglycemia, hypercholesterolemia, dysfunction of the antioxidant

318

system and cholinergic system, and impaired metabolic pathway in diabetic mice brain.

319

HFD is closely related to weight gain. When consuming more than a certain amount

320

of calories during long-term intake, excess carbohydrates promote de novo lipogenesis, which

321

promotes fat accumulation, particularly in the liver and adipocyte. And also, because insulin

322

stimulates de novo lipogenesis, when insulin resistance in developed, it can not only

323

gradually increase insulin levels, but also make it easier to accumulate fat. Thus, increased fat

324

in liver and adipocyte will increase insulin resistance, body weight and glucose level in

325

blood.25 To determine the effect of G Re on weight gain, body weight and calorie intake were

326

measured (Table 1). The HFD group showed increased body weight compared with that of

327

control group. However, G Re groups showed reduced body weight than HFD group.

328

Previous

329

phosphoenolpyruvate carboxykinase (PEPCK), glucose-6-phosphatase (G6Pase), fatty acid

330

synthase (FAS) and stearoyl-CoA desaturase-1 (SCD1) in HepG2 cells and HFD-induced

331

C57BL/6J mice, and ultimately it inhibited hyperglycemia and hepatic steatosis.26

research reported

G Re regulated

fat accumulation factors including

332

Abnormal fasting glucose and glucose tolerance are associated with the development

333

of insulin resistance and signaling.27 Moreover, insulin resistance negatively influences

334

signaling between the brain, insulin, and cognitive functioning.28 To evaluate the

335

improvement effect of insulin resistance through the consumption of the G Re, blood glucose

336

level during sample intake periods, and IPGTT were conducted (Figure 2). The HFD groups 14

ACS Paragon Plus Environment

Page 14 of 41

Page 15 of 41

Journal of Agricultural and Food Chemistry

337

showed hyperglycemia status related with insulin resistance through the long-term

338

administration of the HFD (Figure 1A). However, the G Re groups showed improved blood

339

glucose level through the consumption of the G Re. And also, the G Re groups presented

340

ameliorated insulin resistance compared with the HFD group (Figure 2B). The Panax ginseng

341

berry extract containing the G Re improved the blood glucose level and the insulin

342

resistance.9 Also, these results suggest that G Re administration may have a positive effect on

343

HFD-induced T2DM by regulating the hyperglycemic state and insulin tolerance.

344

The consumption of the G Re considerably improved the HFD-induced cognitive

345

deficit in the Y-maze, passive avoidance and Morris water maze tests (Figure 3). In the Y-

346

maze test, when a mouse is exposed to a new environment, instinctively they prefer to

347

navigate a new environment rather than return to the previously visited arms.29 However,

348

HFD-induced diabetic mouse show impaired structures of hippocampal synaptic plasticity

349

and abnormal neurotransmitter, so they tend to re-enter the previous visited arm.30 The HFD

350

group showed spatial working and behavioral memory deficit through reduced spontaneous

351

alternative behavior and number of arm entries compared with the control group (Figure 3A,

352

B). In contrast, the G Re groups presented ameliorated spatial and behavioral memory

353

function. Short-term learning and memory ability associated with activation of N-methyl-D-

354

aspartic acid (NMDA) receptors and amygdala in the brain tissue were assessed using the

355

passive avoidance test.31 The HFD groups showed lowered steps-through latency in the

356

retention trial compared with that of the control group (Figure 3C). However, the G Re

357

groups presented ameliorated cognitive function similar to that of the control group. The

358

Morris water maze test is based on the reward principle. The Morris water maze test

359

examined the long-term memory and spontaneous behavioral ability. Long-term learning and

360

memory ability is usually trained across four trials, and spontaneous alternation behavior is 15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

361

measured by the probe test.15 In this results, the HFD group showed impaired long-term

362

learning and cognitive function compared with the control group (Figure 3D). Escape latency

363

time of all groups is decreased. However, the HFD group showed lower change of escape

364

latency time against that of the control group in the final trail. And also, the retention time in

365

target zone of the HFD group was reduced (Figure 3E, F). Contrastively, G Re groups

366

showed increased escape latency time and retention time in the target zone. Previous research

367

reported that mice treated with HFD for 1-8 mouths might lead to abnormalities of learning

368

and memory functions associated with hippocampal-dependent performance.32 Exclusive

369

oxidative stress appearing in the hyperglycemic state in the cerebral region also leads to nerve

370

damage and cell death.33 Consequently, activation of NMDA receptors was decreased, and

371

continuous deactivation of NMDA receptors could cause the deficit of post-achieving

372

memory and long-term memory.31 A recent study reported that G Re remarkably alleviated

373

the cognitive dysfunction in streptozotocin-induced diabetic mice by decreasing oxidative

374

stress and inflammation.12 Considering these studies, learning and memory abilities could be

375

decreased by HFD-induced insulin resistance and oxidative stress, and G Re may be used to

376

improve DM-associated cognitive defects.

377

Hypercholesteremia, hyperlipidemia, polyphagia and hyperglycemia are typical

378

diabetic features.2 Administration of HFD increases the serum TG and free fatty acid (FFA)

379

levels, expedites adipose development, and subsequently increases the body weight of mice.

380

The fat absorbed from the diet is dissolved by bile acid and permeated into the blood as TGs,

381

and FFAs are cleaved from the TGs by lipoprotein lipase.34 The FFAs are transported into the

382

blood as albumin complexes. This complex is extensively carried to the internal tissues, such

383

as the liver. The accumulated FFA and TG in the liver promote the excessive production of

384

very-low-density lipoprotein (VLDL) cholesterol in the liver and blood circulation.35 16

ACS Paragon Plus Environment

Page 16 of 41

Page 17 of 41

Journal of Agricultural and Food Chemistry

385

Previous research showed that G Re-rich Panax ginseng berry particularly G Re, improved

386

the serum lipid state and down-regulated the blood glucose levels, TG, TCHO, and LDLC. In

387

addition, it improved the HDLC levels in HFD-induced mice.36 Our results suggest that the

388

serum lipid balance may be affected by the G Re treatment by reducing the hepatic injury

389

indicators (GOT and GPT), and improving the serum lipid status (TG, TCHO, HDLC, and

390

LDLC) in HFD-induced mice (Table 2), and ultimately, it may have an ameliorating effect on

391

HFD-induced hyperlipidemia in the mice.

392

The cholinergic system is associated with cognitive function. AChE activity and ACh

393

level in the cholinergic neuronal system are also associated with behaviorally sensory

394

signaling events. ACh is separated to acetate and choline by the AChE in the cerebral

395

synaptic cleft.37 AChE is fixed in the lipid bilayer and tends to be altered by lipid

396

peroxidation. ACh is a chemically stable compound which can exist in a stable state for a

397

long time after being secreted into the neuronal synaptic cleft.38 However, AChE from the

398

lipid membrane damaged by lipid peroxidation related factors such as MDA and 4-

399

hydroxynonenal (4-HNE) increases the activity. They accelerate the degradation of ACh,

400

which normally plays a role, and break down into acetic acid and choline.37 Excessive

401

activation of the AChE is associated with abnormal deformation of the cell membrane, and

402

this causes abnormal neurotransmission and cognitive dysfunction.39 An increased AChE

403

activation, which lead to ACh degradation and neurotransmitter disorder was found in

404

diabetic rats.37 Therefore, HFD-induced impaired cholinergic system ultimately induces

405

cognitive impairment originating from synaptic dysfunction. When HFD-induced oxidative

406

stress was excessively generated, AChE activity was increased.37 G Re stimulates the release

407

of neurotransmitters, such as dopamine and ACh, by passing through the BBB, and normally

408

promotes neurotransmission in the neuronal system.40 In this study, G Re groups showed 17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

409

reduced activation of AChE and increased ACh content in cerebral tissue compared with that

410

of the HFD group (Figure 4), and G Re seemed to improve the HFD-induced cholinergic

411

defects via inhibition of AChE activity and decrease of ACh decomposition.

412

SOD is an enzyme that catalyzes the partitioning of the superoxide radical to either

413

non-toxic molecular oxygen or hydrogen peroxide.41 Hydrogen peroxide is continuously

414

converted to H2O by glutathione peroxidase (GPx) using cytosolic GSH. When these

415

constituents of antioxidant system are reduced, ROS is not sufficiently scavenged, and it

416

causes oxidative stress, which can induce the lipid peroxidation and cell membrane

417

deformation. And, these stress is known to continuously cause cell death by producing the

418

MDA and numerous free radicals.42 Alteration of the ratio between total GSH and oxidized

419

GSH, the form that scavenges the radicals, is biologically used to assess oxidative cell

420

damage.43 MDA is a secondary metabolite of lipid peroxidation, and it is used as a biomarker

421

of oxidative stress during the onset of numerous diseases.42 Long-term HFD feed generates

422

the ROS in cerebral mitochondria, and impairs the antioxidant system, which can scavenge

423

the these ROS, including SOD and GSH.44 And, Kuhad et al.45 reported that DM increases

424

the MDA and nitrite levels, and also, decreases the GSH levels, SOD contents and catalase

425

activity in the brain of rats. In addition, the reduction of MDA may be associated with the

426

activity of AChE (figure 4A). G Re inhibited lipid peroxidation and ultimately may suppress

427

the activity of AChE by protecting the neuronal lipid bilayer, and reduce the decomposition

428

of ACh. Kim et al. 44 showed that HFD-induced diabetic condition led to impairment of the

429

antioxidant system, and accompanied cognitive dysfunction. However, Panax ginseng berry,

430

including G Re, treatment improved the lipid status, SOD contents, and ratio of oxidized

431

GSH/total GSH by lowering the oxidative stress in HFD-induced mice.9 In this study, G Re

432

groups showed increased SOD contents and reduced oxidized GSH/total GSH ratio and MDA 18

ACS Paragon Plus Environment

Page 18 of 41

Page 19 of 41

Journal of Agricultural and Food Chemistry

433

content against HFD group (Figure 5). Consequently, our results suggest that the G Re could

434

have an ameliorating effect on HFD-induced cognitive deficits through the inhibition of lipid

435

peroxidation and down-regulation of antioxidant enzymes caused by insulin resistance.

436

Insulin resistance, a typical feature of T2DM, impedes the insulin signal in neuronal

437

cells, and this deficit of cerebral insulin signal and insulin secretion was associated with AD

438

neurodegeneration.24 In the normal status, insulin binds to insulin receptors, thereby

439

regulating the insulin signal, which expedites glucose uptake by the neurons. Also, activation

440

of the insulin receptors leads to phosphorylation of IRS (tyrosine site), which promotes PI3-K

441

phosphorylation and glucose uptake.46 If the insulin signaling is abnormally impaired,

442

however, insulin resistance is developed in most tissues. Particularly, a high-calorie diet

443

induces insulin resistance, and inhibits the reaction of insulin and its receptors. This abnormal

444

signal leads to JNK phosphorylation, which phosphorylates the serine site of IRS instead of

445

the tyrosine site.47 This increased p-IRS with its phosphorylated serine site impedes PI3-K

446

phosphorylation and significantly impedes Akt activation associated with cell survival. The

447

inhibited Akt induces GSK-3β activation, and expedites p-tau aggregation and development

448

of the NFT. This formation of p-tau and NFT ultimately accelerates abnormal structural

449

modification of cell and neuronal death.7 Recent research also showed that the expression of

450

the insulin receptor, p-IRS (tyrosine site), Akt, and PI3-K in the brain was considerably

451

decreased in the cerebral insulin signaling mechanism of diabetes-induced models.48 A

452

number of apoptotic cascades can be induced by the release of cytochrome c from the

453

mitochondria into the cytosol, where it binds to the apoptotic protease activating factor 1

454

(Apaf-1), leading to binding of procaspase-9 and activation of procaspase-9 by proteolytic

455

processes.49 When Akt is inactivated by mediators of oxidative stress, caspase-9 is activated

456

by inducing the leakage of cytochrome c.50 Therefore, the release of cytochrome c to the 19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 20 of 41

457

cytosol also activates the caspase pathway and cleaves the PARP. As a result of this, decrease

458

of NAD and ATP was presented in the cerebral tissue of diabetic models.51,

459

stimulates apoptosis through DNA fragmentation and ultimately leads to cell death.51

460

Polyphenolic compounds could down-regulate the dysfunctions of insulin signaling in brain

461

tissue by alleviating the expression of signaling proteins associated with the JNK pathway.44

462

Our research suggests that G Re may improve insulin resistance by regulating the associated

463

protein expression levels, such as p-JNK, p-IRS, p-tau, and c-PARP levels. Finally, G Re may

464

alleviate diabetes associated cognitive dysfunctions in the HFD-induced mice.

52

c-PARP

465

Ginsenoside Re has an ameliorating effect on HFD-induced hyperglycemic C57BL/6

466

mice by improving insulin tolerance, including blood glucose. G Re significantly regulated

467

food intake associated with weight gain and serum lipid statuses, such as TG, TCHO, HDLC,

468

and LDLC levels. The administration of G Re also improved the learning and memory

469

dysfunction in HFD-induced cognitive impairment mice. The anti-amnesic effect of G Re

470

may be produced by cholinergic recovery (AChE inhibition and ACh increase) and protection

471

of antioxidant systems (SOD contents and ratio of oxidized GSH/total GSH) in the brain

472

tissue damaged by HFD-induced oxidative stress. An abnormal insulin signal via the

473

regulation of the JNK pathway in brain tissue was ameliorated by G Re administration.

474

Consequently, the ameliorating effect of G Re on the DACM could be induced by cholinergic

475

recovery, activation of antioxidant systems, and improved insulin tolerance and signaling in

476

brain tissue. Therefore, G Re can be used as a natural potential resource to ameliorate

477

learning and memory dysfunction associated with HFD-induced hyperglycemic status.

478 479

AUTHOR INFORMATION

480

Corresponding author 20

ACS Paragon Plus Environment

Page 21 of 41

481

Journal of Agricultural and Food Chemistry

*(H.J.H.) Phone: +82 55 772 1907. Fax: +82 55 772 1909. Email: [email protected].

482 483

Funding Information

484

This study was supported by Basic Science Research Program through the National

485

Research Foundation of Korea (NRF-2015R1D1A3A01015931) funded by the Ministry of

486

Education, Republic of Korea. C.H.P., J.M.K., S.K.P., T.W.S., J.Y.K., J.S.H., and D.S.L. were

487

supported by the BK21 Plus program, Ministry of Education, Republic of Korea.

488 489 490

Notes The authors declare no competing financial interest.

491

21

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

492

REFERENCES

493

1. Mijnhout, G. S.; Scheltens, P.; Diamant, M.; Biessels, G. J.; Wessels, A. M.; Simsek, S.;

494

Snoek, F. J.; Heine, R. J. Diabetic encephalopathy: a concept in need of a definition.

495

Diabetologia 2006, 49, 1447-1148

496

2. Costa, R. R. S.; Villela, N. R.; Maria, das Graças, C. S.; Boa, B. C. S.; Cyrino, F. Z. G. A.;

497

Silva, S. V.; Lisboa, P. C.; Moura, E. G.; Barja-Fidalgo, T. C.; Bouskela, E. High fat diet

498

induces central obesity, insulin resistance and microvascular dysfunction in hamsters.

499

Microvasc. Res. 2011, 82, 416-422

500

3. Ma, W.; Yuan, L.; Yu, H.; Xi, Y.; Xiao, R. Mitochondrial dysfunction and oxidative

501

damage in the brain of diet-induced obese rats but not in diet-resistant rats. Life Sci. 2014,

502

110, 53–60

503

4. Kothari, V.; Luo, Y.; Tornabene, T.; O'Neill, A. M.; Greene, M. W.; Thangiah, G.; Babu, J.

504

R. High fat diet induces brain insulin resistance and cognitive impairment in mice. BBA-

505

Mol. Basis Dis. 2016, 1863, 499-508

506

5. Petrov, D.; Pedrós, I.; Artiach, G.; Sureda, F. X.; Barroso, E.; Pallàs, M.; Casadesús, G.;

507

Beas-Zarate, C.; Carro, E.; Ferrer, I.; Vazquez-Carrera, M.; Folch, J.; Camins, A. High-fat

508

diet-induced deregulation of hippocampal insulin signaling and mitochondrial homeostasis

509

deficiences contribute to Alzheimer disease pathology in rodents. BBA-Mol. Basis Dis.

510

2015, 1852, 1687-1699

511

6. Bhat, R.; Xue, Y.; Berg, S.; Hellberg, S.; Ormo, M.; Nilsson, Y.; Radesater, A. C.; Jerning,

512

E.; Markgren, P. O.; Borgegard, T.; Nylöf, M.; Giménez-Cassina, A.; Hernández, F.;

513

Lucas, J. J.; Díaz-Nido, J.; Avila, J. Structural insights and biological effects of glycogen

514

synthase kinase 3-specific inhibitor AR-A014418. J. Biol. Chem. 2003, 278, 45937–45945

515

7. Ittner, L. M.; Ke, Y. D.; Delerue, F.; Bi, M.; Gladbach, A.; van Eersel, J.; Wölfing, H.; 22

ACS Paragon Plus Environment

Page 22 of 41

Page 23 of 41

Journal of Agricultural and Food Chemistry

516

Chieng, B. C.; Christie, M. J.; Napier, I. A.; Eckert, A.; Staufenbiel, M.; Hardeman, E.;

517

Götz, J. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease

518

mouse models. Cell 2010, 142, 387–397

519

8. Wang, M.; Zhang, L.; Han, X.; Yang, J.; Qian, J.; Hong, S.; Samaniego, F.; Romaguera, J.;

520

Yi, Q. Atiprimod inhibits the growth of mantle cell lymphoma in vitro and in vivo and

521

induces apoptosis via activating the mitochondrial pathways. Blood 2007, 109, 5455-5462

522

9. Park, C. H.; Park, S. K.; Seung, T. W.; Jin, D. E.; Guo, T.; Heo, H. J. Effect of ginseng

523

(Panax ginseng) berry EtOAc fraction on cognitive impairment in C57BL/6 mice under

524

high-fat diet inducement. Evid-Based. Compl. Alt. 2015, 316527

525 526

10. Jiang, H. L.; Yang, Z.; Meng, Q.; Hong, T. Improvement and mechanism of ginsenoside Re on learning and memory deficits of mice. Chin. Pharm. Bull. 2008, 24, 1399-1400

527

11. Cho, W. C. S.; Chung, W. S.; Lee, S. K. W.; Leung, A. W. N.; Cheng, C. H. K.; Yue, K.

528

K. M. Ginsenoside Re of Panax ginseng possesses significant antioxidant and

529

antihyperlipidemic efficacies in streptozotocin-induced diabetic rats. Eur. J. Pharmacol.

530

2006, 550, 173-179

531

12. Liu, Y. W.; Zhu, X.; Li, W.; Lu, Q.; Wang, J. Y.; Wei, Y. Q.; Yin, X. X. Ginsenoside Re

532

attenuates diabetes-associated cognitive deficits in rats. Pharmacol. Biochem. Be. 2012,

533

101, 93-98

534

13. Heo, H. J.; Kim, M. J.; Lee, J. M.; Choi, S. J.; Cho, H. Y.; Hong, B.; Kim, H. K.; Kim, E.;

535

Shin, D. H. Naringenin from Citrus junos has an inhibitory effect on acetylcholinesterase

536

and a mitigating effect on amnesia. Dement. Geriatr. Cogn. Disord. 2004, 17, 151-157

537

14. Choi, S. J.; Kim, M. J.; Heo, H. J.; Kim, J. K.; Jun, W. J.; Kim, H. K.; Kim, E. K.; Kim,

538

M. O.; Cho, H. Y.; Hwang, H. J.; Kim, Y. J.; Shin, D. H. Ameliorative effect of 1,2-

539

benzenedicarboxylic acid dinonyl ester against amyloid beta peptide-induced neurotoxicity. 23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

540 541 542

Amyloid 2009, 16, 15-24 15. Morris, R. Development of water maze procedure for studying spatial learning in the rat. J. Neurosci. Meth. 1984, 11, 47-60

543

16. Friedewald, W. T.; Levy, R. I.; Fredrickson, D. S. Estimation of the concentration of low-

544

density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge.

545

Clin. Chem. 1972, 18, 499-502

546

17. Ellman, G. L.; Courtney, K. D.; Andres, V.; Featherstone, R. M. A new and rapid

547

colorimetric determination of acetylcholinestrase activity. Biochem. Pharmacol. 1961, 7,

548

88-95

549

18. Vincent, D.; Segonzac, G.; Vincent, M. C. Colorimetric determination of acetylcholine by

550

the Hestrin hydroxylamine reaction and its application in pharmacy. Ann. Pharm. Fr. 1958,

551

16, 179-185

552

19. Lin, J. D.; Lin, P. Y.; Chen, L. M.; Fang, W. H.; Lin, L. P.; Loh, C. H. Serum glutamic-

553

oxaloacetic transaminase (GOT) and glutamic-pyruvic transaminase (GPT) levels in

554

children and adolescents with intellectual disabilities. Res. Dev. Disabil. 2010, 31. 172-177

555

20. Stone, N. J. Secondary causes of hyperlipidemia. Med. Clin. North. Am. 1994, 78, 117-

556

141

557

21. Gavin III, J. R.; Alberti, K. G. M. M.; Davidson, M. B.; DeFronzo, R. A. Report of the

558

expert committee on the diagnosis and classification of diabetes mellitus. Diabetes care

559

1997, 20, 1183

560 561 562 563

22. Eizirik D. L.; Mandrup-Poulsen T. A choice of death: the signal-transduction of immunemediated β -cell apoptosis. Diabetologia 2001, 44, 2115–2133 23. Cnop, M.; Welsh, N.; Jonas, J. C.; Jörns, A.; Lenzen, S.; Eizirik, D. L. Mechanisms of pancreatic β-cell death in type 1 and type 2 diabetes. Diabetes 2005, 54, 97-107 24

ACS Paragon Plus Environment

Page 24 of 41

Page 25 of 41

Journal of Agricultural and Food Chemistry

564

24. Ozougwu, J. C.; Obimba, K. C.; Belonwu, C. D.; Unakalamba, C. B. The pathogenesis

565

and pathophysiology of type 1 and type 2 diabetes mellitus. J. Physiol. Pathophysiol. 2013,

566

4, 46-57

567

25.

Adiels, M.; Taskinen, M. R.; Packard, C.; Caslake, M. J.; Soro-Paavonen, A.;

568

Westerbacka, J.; Vehkavaara, S.; Häkkinen, A.; Olofsson, S. O.; Yki-Järvinen, H.; Boren, J.

569

Overproduction of large VLDL particles is driven by increased liver fat content in man.

570

Diabetologia 2006; 49: 755–765

571

26. Quan, H. Y.; Yuan, H. D.; Jung, M. S.; Ko, S. K.; Park, Y. G.; Chung, S. H. Ginsenoside

572

Re lowers blood glucose and lipid levels via activation of AMP-activated protein kinase in

573

HepG2 cells and high-fat diet fed mice. Int. J. Mol. Med. 2012, 29. 73

574

27. Zahn, D.; Tug, S.; Wenzel, M.; Simon, P.; Kubiak, T. Glucose metabolism and self-

575

regulation—Is insulin resistance a valid proxy of self-control?. Pers. Individ. Dif. 2016, 99,

576

38-45

577

28. McNay, E. C.; Recknagel, A. K. Brain insulin signaling: A key component of cognitive

578

processes and a potential basis for cognitive impairment in type 2 diabetes. Neurobiol.

579

Learn. Mem. 2011, 96, 432-442

580

29. Dellu, F.; Mayo, W.; Cherkaoui, J.; Le Moal, M.; Simon, H. A two-trial memory task

581

with automated recording: study in young and aged rats. Brain Res. 1992, 588, 132–139

582

30. Tuzcu, M.; Baydas, G. Effect of melatonin and vitamin E on diabetes-induced learning

583

and memory impairment in rats. Eur. J. Pharmacol. 2006, 537, 106–110

584

31. Tsuji, M.; Takeda, H.; Matsumiya, T. Modulation of passive avoidance in mice by the 5-

585

HT1A receptor agonist flesinoxan: comparison with the benzodiazepine receptor agonist

586

diazepam. Neuropsychopharmacol. 2003, 28, 664–674

587

32. Sharma, S.; Taliyan, R. Neuroprotective role of indirubin-3′-monoxime, a GSKβ inhibitor 25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

588

in high fat diet induced cognitive impairment in mice. Biochem. Bioph. Res. Co. 2014, 452,

589

1009–1015

590

33. Xiang, Q.; Zhang, J.; Li, C. Y.; Wang, Y.; Zeng, M. J.; Cai, Z. X.; Tian, R. B.; Jia, W.; Li,

591

X. H. Insulin resistance-induced hyperglycemia decreased the activation of Akt/CREB in

592

hippocampus neurons: Molecular evidence for mechanism of diabetes-induced cognitive

593

dysfunction. Neuropeptides 2015, 54, 9-15

594 595

34. Zechner, R.; Strauss, J. G.; Haemmerle, G.; Lass, A.; Zimmermann, R. Lipolysis: Pathway under construction. Curr. Opin. Lipidol. 2005, 16, 333–340

596

35. Jensen, M. D.; Haymond, M. W.; Rizza, R. A.; Cryer, P. E.; Miles, J. M. Influence of

597

body fat distribution on free fatty acid metabolism in obesity. J. Clin. Invest. 1989, 83,

598

1168–1173

599

36. Attele, A. S.; Zhou, Y. P.; Xie, J. T.; Wu, J. A.; Zhang, L.; Dey, L.; Pugh, W.; Rue, P. A.;

600

Polonsky, K. S.; Yuan, C. S. Antidiabetic effects of Panax ginseng berry extract and the

601

identification of an effective component. Diabetes 2002, 51, 1851-1858

602 603 604 605

37. Soreq, H.; Seidman, S. Acetylcholinesterase — new roles for an old actor. Nat. Rev. Neurosci. 2001, 2, 294–302 38. Rotundo, R.L. Expression and localization of acetylcholinesterase at the neuromuscular junction. J. Neurocytol. 2003, 32, 743–766

606

39. Kaizer, R. R.; Correa, M. C.; Spanevello, R. M.; Morsch, V. M.; Mazzanti, C. M.;

607

Goncalves, J. F.; Schetinger, M. R. C. Acetylcholinesterase activation and enhanced lipid

608

peroxidation after long-term exposure to low levels of aluminum on different mouse brain

609

regions. J. Inorg. Biochem. 2005, 99, 1865–1870

610

40. Shi, J.; Xue, W.; Zhao, W. J.; Li, K. X. Pharmacokinetics and dopamine/acetylcholine

611

releasing effects of ginsenoside Re in hippocampus and mPFC of freely moving rats. Acta 26

ACS Paragon Plus Environment

Page 26 of 41

Page 27 of 41

612 613 614 615 616 617 618

Journal of Agricultural and Food Chemistry

Pharmacol. Sin. 2013, 34, 214-220 41. Misra, H. P.; Fridovich, I. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J. Biol. Chem. 1972, 247, 3170-3175 42. Moore, K.; Roberts, L. J. Measurement of lipid peroxidation. Free Radical Res. 1998, 28, 659–671 43. Chance, B.; Sies, H.; Boveris, A. Hydroperoxide metabolism in mammalian organs. Physiol. Rev. 1979, 59, 527-605

619

44. Kim, J. M.; Park, S. K.; Guo, T. J.; Kang, J. Y.; Ha, J. S.; Lee, U.; Heo, H. J. Anti-

620

amnesic effect of Dendropanax morbifera via JNK signaling pathway on cognitive

621

dysfunction in high-fat diet-induced diabetic mice. Behav.Brain Res. 2016, 312, 39-54

622 623

45. Kuhad, A.; Chopra, K. Effect of sesamol on diabetes-associated cognitive decline in rats. Exp. Brain Res. 2008, 185, 411-420

624

46. Lee, B. H.; Hsu, W. H.; Pan, T. M. Inhibitory effects of Dioscorea polysaccharide on

625

TNF-induced insulin resistance in FL83B cells. J. Agr. Food Chem. 2011, 59, 5279–5285

626

47. Tanti, J. F.; Jager, J. Cellular mechanisms of insulin resistance: role of stress-regulated

627

serine kinases and insulin receptor substrates (IRS) serine phosphorylation. Curr. Opin.

628

Pharmacol. 2009, 9, 753-762

629

48. Battu, C. E.; Rieger, D.; Loureiro, S.; Furtado, G. V.; Bock, H.; Saraiva-Pereira, M. L.;

630

Pessoa-Pureur, R.; Gonçalves, C. A.; Perry, M. L. S. Alterations of PI3K and Akt

631

signaling pathways in the hippocampus and hypothalamus of Wistar rats treated with

632

highly palatable food. Nutr. Neurosci. 2012, 15, 10–17

633

49. Cardone, M. H.; Roy, N.; Stennicke, H. R.; Salvesen, G. S.; Franke, T. F.; Stanbridge, E.;

634

Frisch, S.; Reed, J. C. Regulation of Cell Death Protease Caspase-9 by Phosphorylation.

635

Science 1998, 282, 1318–1321 27

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

636

50. Woo, H. J.; Jun, D. Y.; Lee, J. Y.; Woo, M. H.; Yang, C. H.; Kim, Y. H. Apoptogenic

637

activity of 2α, 3α-dihydroxyurs-12-ene-28-oic acid from Prunella vulgaris var. lilacina is

638

mediated via mitochondria-dependent activation of caspase cascade regulated by Bcl-2 in

639

human acute leukemia Jurkat T cells. J. Ethnopharmacol. 2011, 135, 626–635

640

51. Looi, C. Y.; Arya, A.; Cheah, F. K.; Muharram, B.; Leong, K. H.; Mohamad, K.; Wong,

641

W. F.; Rai, N.; Mustafa, M. R. Induction of apoptosis in human breast cancer cells via

642

caspase pathway by vernodalin isolated from Centratherum anthelminticum (L.) seeds.

643

PloS one 2013, 8, e56643

644

52. Manna, P.; Das, J.; Ghosh, J.; Sil, P. C. Contribution of type 1 diabetes to rat liver

645

dysfunction and cellular damage via activation of NOS, PARP, IκBα/NF-κB, MAPKs, and

646

mitochondria-dependent pathways: Prophylactic role of arjunolic acid. Free Radical Biol.

647

Med. 2010, 48, 1465-1484

648

28

ACS Paragon Plus Environment

Page 28 of 41

Page 29 of 41

Journal of Agricultural and Food Chemistry

Table 1. Effect of ginsenoside Re on body weight and food intake Group

Control

HFD

G Re 5

G Re 10

G Re 20

Wight gain (g)

33.25±1.25e

50.50±3.32a

48.25±2.22b

47.33±0.57c

45.23±0.41d

Food intake (kcal/day)

9.09±4.43e

16.66±1.15a

14.36±0.84b

13.41±0.73c

12.89±0.94d

Results shown are mean±SD (݊=8). Data were statistically considered at ‫