Leaves of Persimmon (Diospyros kaki Thunb ... - ACS Publications

Aug 11, 2015 - The purpose of the study was to investigate the protective effects of the ethanol extract of Diospyros kaki (EEDK) persimmon leaves to ...
0 downloads 0 Views 3MB Size
Subscriber access provided by RUTGERS UNIVERSITY

Article

The Leaves of Persimmon (Diospyros kaki Thunb.) Ameliorate Nmethyl-N-nitrosourea (MNU)-Induced Retinal Degeneration in Mice Kyung-A Kim, Suk Woo Kang, Hong Ryul Ahn, Youngwoo Song, Sung Jae Yang, and Sang Hoon Jung J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.5b02578 • Publication Date (Web): 11 Aug 2015 Downloaded from http://pubs.acs.org on August 17, 2015

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 33

Journal of Agricultural and Food Chemistry

301x176mm (72 x 72 DPI)

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

The Leaves of Persimmon (Diospyros kaki Thunb.) Ameliorate N-methyl-N-nitrosourea (MNU)-Induced Retinal Degeneration in Mice Kyung-A Kim,†,§ Suk Woo Kang,† Hong Ryul Ahn,† Youngwoo Song,† Sung Jae Yang,‡ and Sang Hoon Jung†,§*



Natural Products Research Center, Korea Institute of Science and Technology (KIST),

Gangneung, Korea ‡

Department of Ophthalmology, University of Ulsan, Gangneung Asan Hospital, Gangneung,

Korea §

Department of Biological Chemistry, University of Science and Technology (UST), Daejeon,

Korea.

ACS Paragon Plus Environment

Page 2 of 33

Page 3 of 33

Journal of Agricultural and Food Chemistry

1

ABSTRACT

2

The purpose of the study was to investigate the protective effects of the ethanol extract of

3

Diospyros kaki (EEDK) Persimmon leaves to study N-methyl-N-nitrosourea (MNU)-induced

4

retinal degeneration in mice. EEDK was orally administered after MNU-injection. Retinal

5

layer thicknesses were significantly increased in the EEDK-treated group, compared with the

6

MNU-treated group. The outer nuclear layer was preserved in the retinas of EEDK-treated

7

mice. Moreover, EEDK treatment reduced the MNU-dependent up-regulation of glial

8

fibrillary acidic protein (GFAP) and nestin expression in Müller and astrocyte cells. EEDK

9

treatment also inhibited MNU-dependent down-regulation of rhodopsin expression.

10

Quercetin exposure significantly attenuated the negative effects of H2O2 in R28 cells,

11

suggesting that quercetin can act in an anti-oxidative capacity. Thus, EEDK may be

12

considered as an agent for treating or preventing degenerative retinal diseases, such as

13

retinitis pigmentosa and age-related macular degeneration.

14 15

Keywords

16

Diospyros kaki; N-methyl-N-nitrosourea; persimmon; photoreceptor; retinal degeneration

17 18

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

19

INTRODUCTION

20

Retinitis pigmentosa and age-related macular degeneration (AMD) are leading

21

worldwide causes of retinal degeneration and blindness.1, 2 Several important risk factors for

22

retinal degeneration have been identified, and substantial evidence indicates that retinal

23

degeneration is associated with reactive oxygen species (ROS).3 In the signaling mechanism

24

of vision, light is converted into electronic signals via retinal photoreceptors, a process that

25

requires a large supply of oxygen.4 Oxygen is continuously consumed during the visual

26

process; therefore, retinas are highly exposed to ROS, such as superoxide anions, hydroxyl

27

radicals, and hydrogen peroxide. Although anti-oxidative defenses exist to protect the retina

28

from ROS, the excessive accumulation of ROS causes photoreceptor cell death and,

29

subsequently, retinal damage and visual loss.5 This model is supported by epidemiological

30

studies wherein patients with retinal degeneration exhibit high ROS levels and low levels of

31

anti-oxidative proteins, including glutathione, superoxide dismutase, and catalase, compared

32

to healthy controls.6 These findings explain how antioxidants may protect against retinal

33

degeneration, and it has been reported in epidemiological studies that the consumption of

34

foods containing high concentrations of antioxidants are associated with a reduced risk for

35

developing retinal degeneration.7, 8 Therefore, the consumption of antioxidants from natural

36

foods or nutritional supplements can be helpful in intervention against retinal degeneration.

37

N-methyl-N-nitrosourea (MNU) is known to be a potent alkylating agent that interacts

38

with DNA, and leads to the loss of retinal photoreceptor cells.9, 10 Although the underlying

39

mechanism has not been clearly elucidated, Prater et al. reported that MNU induced the

40

production of ROS via protein alkylation, which promoted apoptosis.11

41

ACS Paragon Plus Environment

Page 4 of 33

Page 5 of 33

Journal of Agricultural and Food Chemistry

42

It has also been reported that NADPH oxidase plays a critical role in the generation of

43

the oxidative stress, which was shown to promote cone cell death in retinitis pigmentosa,

44

using a genetic model (rd1 mice).12

45

Persimmon (Diospyros kaki Thunb.) belongs to the Ebenaceae and is widely cultivated

46

in Korea, China, Japan, and Eastern Asia. The leaves of D. kaki are rich in bioactive

47

compounds, such as polyphenols, flavonoids, and vitamins13 and it has been reported that

48

these compounds, which contain gallic acid derivatives and glucose units linked together via

49

glycosidic bonds, have potent radical-scavenging and anti-oxidative activities.14,

50

compounds have also been used for medicinal purposes as remedies to treat a wide variety of

51

conditions, including neuronal injury and neurodegeneration, due to their anti-oxidative

52

properties.16, 17

15

Such

53

Antioxidants have been shown to have beneficial effects in human eyes.18, 19 The goal of

54

this study was therefore to determine whether D. kaki has protective effects on MNU-induced

55

retinal degeneration.

56 57

MATERIALS AND METHODS

58

Chemicals

59

Anti-nestin and anti-rhodopsin antibodies were purchased from Novus Biologicals

60

(Littleton, CA, USA). Anti-glial fibrillary acidic protein (GFAP), anti-superoxide dismutase

61

(SOD)-3, and anti-chicken IgG antibodies were purchased from Millipore (Billerica, MA,

62

USA). Anti-SOD-1, anti-SOD-2, anti-mouse IgG, anti-rabbit IgG, and anti-sheep IgG

63

antibodies were purchased from Santa Cruz (Santa Cruz, CA, USA). Anti-GAPDH and

64

glutathione peroxidase (GPx-1) antibodies were purchased from Cell Signaling Technology

65

(Beverly, MA, USA). Alexa 488-conjugated anti-sheep, Alexa 594-conjugated anti-mouse

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

66

and Alexa 633-conjugated anti-sheep antibodies were purchased from Invitrogen (Carlsbad,

67

CA, USA). A blocking solution was purchased from Dako (Santa Clara, CA, USA), and

68

mounting medium was purchased from Vector Laboratories (Burlingame, CA, USA). Zoletil

69

and Rumpun were purchased from Virbac Laboratories (Fort Worth, TX, USA) and Bayer

70

(Newbury, UK), respectively, and used as anesthesia. All other chemicals and reagents were

71

purchased from Sigma-Aldrich (St. Louis, MO, USA).

72 73

Plant materials and sample preparation

74

D. kaki was collected in Gangneung, Gangwon Province of Korea in August 2013. A

75

voucher specimen (D-521) was deposited at the KIST Gangneung Institute. Eight hundred

76

grams of the dried D. kaki leaves were extracted 3 times with 7 L of ethanol at room

77

temperature for 3 h in an ultrasonic cleaning bath (model RK 158s, Bandelin, Germany) and

78

filtered through Whatman No. 1 filter paper. The combined filtrate was concentrated to

79

dryness by rotary evaporation at 40 °C to obtain 33 g (yield: 4.13%) of the ethanol extract of

80

D. kaki leaves (EEDK). HPLC-PDA-MS analysis was applied using a Thermo Accela HPLC

81

system and LCQ FLEET ion trap mass spectrometer (Thermo Fisher Scientific Inc., SanJose,

82

CA, USA) for compound determinations in EEDK. The mobile phase consisted of 0.1%

83

formic acid in water (Solvent A) and 0.1% formic acid in acetonitrile (Solvent B), which was

84

used with a YMC-Triart C18 column (3-µm particle size, 150 mm × 4.6 mm I.D., YMC Co.,

85

Kyoto, Japan). A linear gradient (from 10 to 60% solvent B for 30 min) at a flow rate of 1

86

mL/min was used at 254 nm. The mass spectrometer conditions were as follows: negative ion

87

mode; mass range, m/z 200−800; capillary voltage, 10.0 V; capillary temperature, 350 °C.

88

The NMR spectra were recorded on a Varian 500 MHz NMR system (Varian, Palo Alto, CA,

ACS Paragon Plus Environment

Page 6 of 33

Page 7 of 33

Journal of Agricultural and Food Chemistry

89

USA) at 500 MHz for 1H NMR and 125 MHz for 13C NMR in DMSO-d6. Compounds 1–9

90

were purified by semi-preparative HPLC.

91 92

Animals

93

Six-week-old male C57BL/6J mice weighing 20–25 g (Central Lab. Animal Inc., Seoul,

94

Korea) were housed at 23 ± 0.5 °C and 10% humidity, with a 12-h light-dark cycle. All

95

animals were acclimated for at least 1 week, caged in groups of 5 or less, and fed with a diet

96

of animal chow and water ad libitum.

97

All animal studies were performed in a pathogen-free barrier zone at the KIST

98

Gangneung Institute in accordance with the procedure outlined in the ARVO Statement for

99

the Use of Animals in Ophthalmic and Vision Research. Procedures used in this study were

100

approved by the Animal Care and Use Committee of the KIST Gangneung Institute (No.

101

2014-011)

102

MNU was used as a negative insult for retinal degeneration and was induced by single

103

intraperitoneal (IP) injection of 50 mg/kg MNU (50 mg/kg body weight) in C57BL/6J mice.20

104

After the injection of MNU, EEDK was orally administrated at doses of 10, 50, or 100

105

mg/(kg·day) for 4 weeks. Animals were examined at 1 and 4 weeks after the injection of

106

MNU. Eyes were dissected and examined either for histological or western blot analysis.

107

Fixed eyes were cut into 4-µm slices and were then studied by staining with hematoxylin and

108

eosin (H&E) or by immunofluorescence.

109 110

Spectral-domain optical coherence tomography (SD-OCT)

111

SD-OCT imaging was performed at 1 and 4 weeks after 50-mg/kg MNU treatment. The

112

animals were anaesthetized using a mixture of Zoletil (1.6 µg/g, Verbac Laboratories, USA)

113

and Rumpun (0.05 µL/g, Bayer, UK). Anesthetized animals were placed in front of the

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

114

Spectral OCT system (Heidelberg Engineering, Germany) and analyzed as described

115

previously.21

116 117

Hematoxylin & eosin (H&E) staining

118

H&E staining of the retina slides was performed using a standard procedure. Briefly, the

119

retina slides were treated with hematoxylin buffer (0.1% hematoxylin and 10% ammonium)

120

at room temperature. The slides were then washed 3 times with distilled water and dipped in

121

1% eosin Y solution. Next, the slides were washed twice with 95% alcohol and mounted.

122

Histological analysis was evaluated under a CKX41 inverted phase contrast microscope

123

(Olympus, Tokyo, Japan).

124 125

Western blot analysis

126

Proteins were separated on 10% or 12% SDS-PAGE gels and then transferred onto

127

polyvinylidene difluoride (PVDF) membranes. The membranes were blocked at room

128

temperature in 5% skim milk before overnight incubation at 4 °C with antibodies (diluted as

129

indicated in PBS) against nestin (1:1000), GFAP (1:1000), GAPDH (1:2000), rhodopsin

130

(1:1000), SOD-1 (1:1000), SOD-2 (1:1000), SOD-3 1 (1:1000), or GPx-1 (1:1000). Binding

131

of the primary antibodies was detected following incubation with appropriate horseradish

132

peroxidase-conjugated IgG secondary antibodies (diluted 1:5000) for 2 h at room temperature.

133

The membranes were developed using an enhanced chemiluminescence detection kit

134

(Thermo Scientific, Waltham, MA, USA) and measured by densitometry using an LAS-4000

135

image reader and Multi Gauge 3.1 software (Fujifilm, Tokyo, Japan). GAPDH expression

136

was detected as a loading control.

137 138

Immunofluorescence staining

ACS Paragon Plus Environment

Page 8 of 33

Page 9 of 33

Journal of Agricultural and Food Chemistry

139

Paraffin-embedded retinal sections were deparaffinized and blocked in normal goat serum

140

overnight at 4 °C. Subsequently, the sections were incubated with specific primary antibodies

141

against nestin (diluted 1:200), GFAP (diluted 1:200), and rhodopsin (diluted 1:200) overnight

142

at 4 °C. The sections were washed 3 times with PBST buffer (8 g/L NaCl, 0.2 g/L KCl, 1.44

143

g/L, Na2HPO4, 0.24 g/L, NaH2HPO4, and 0.1% Triton X-100) and incubated with matching

144

secondary antibodies (Alexa 488-conjugated anti-sheep, Alexa 594-conjugated anti-mouse,

145

and Alexa 633-conjugated anti-chicken antibodies; diluted 1:500) for 2 h at room temperature

146

in the dark. The sections were washed with PBST, mounted, and covered with a glass

147

coverslip. Staining images were examined using a Leica TCS SP5 confocal microscope

148

system (Leica, Wetzlar, Germany).

149 150

Cell culture

151

Immortalized rat retinal precursor cells (R28 cells) were purchased from Kerafast

152

(Kerafast, Inc., MA, USA). R28 cells were grown in 75-cm2 culture flasks in low-glucose

153

Dulbecco's Modified Eagle's Medium (DMEM, HyClone, Logan, UT, USA) supplemented

154

with 10% (v/v) heat inactivated fetal bovine serum (FBS; HyClone, Logan, UT, USA) and

155

100 U/mL penicillin/streptomycin (HyClone, Logan, UT, USA). Cultures were maintained at

156

37 °C in a humidified atmosphere containing 5% CO2. The cells were passaged twice a week.

157

R28 cells were pretreated with the indicated compounds for 1 h after 300 µM hydrogen

158

peroxide (H2O2) was added to cultures for 24 h.

159 160

Cell viability

161

R28 cell viabilities were determined by performing 3-(4,5-dimethylthiazol-2-yl)-2,5-

162

diphenyltetrazolium bromide (MTT) assays. Briefly, MTT assays were performed by treating

163

cells with MTT solution (0.5 mg/mL final concentration) for 1 h at 37 °C. After a 1-h

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

164

incubation, the blue formazan crystals that formed in intact cells were solubilized with

165

DMSO, and absorbance values at 570 and 590 nm were measured with a microplate reader

166

(BioTek Instruments, Winooski, VT, USA). Results are expressed as the percent of MTT

167

reduction.

168 169

Assessment of ROS production

170

To quantify intracellular ROS production, we used DCFH-DA as a probe for free radicals

171

22

. The R28 cells were seeded at a density of 5.0 × 103 cells per well into 96-well plates and

172

incubated in a humidified atmosphere of 95% air and 5% CO2 at 37 °C. Twenty-four hours

173

later, the cell culture medium was replaced with DMEM containing 1% FBS. Superoxide

174

radicals effectively oxidize nonfluorescent dichlorofluorescein (DCFH) to fluorescent

175

dichlorofluorescein (DCF). The cells were loaded with the radical probe DCFH-DA (10 µM)

176

through incubation for 20 min at 37 °C. Then, the cell culture medium was replaced to

177

remove the excess probe. To generate radical species, we added KO2 at 1 mM (O2·−) to the

178

radical probe-loading medium. Fluorescence was then measured after various durations,

179

using excitation and emission wavelengths of 485 and 535 nm, respectively (Luminescence

180

Spectrometer LS50B, Perkin-Elmer Ltd., England).

181 182

Statistical analysis

183

Experimental values were analyzed by Kruskal–Wallis and Mann–Whitney tests using

184

the Bonferroni correction and are presented as mean ± standard deviation. A result of p < 0.05

185

was considered to be statistically significant. Quantification of western blotting results was

186

performed using Fujifilm Multi Gauge software, version 3.0 (Tokyo, Japan). All statistical

187

analyses were performed using SPSS, version 16.0 (IBM Corporation, Armonk, NY).

188

ACS Paragon Plus Environment

Page 10 of 33

Page 11 of 33

Journal of Agricultural and Food Chemistry

189

RESULTS AND DISCUSSION

190

The chromatogram of EEDK showed 9 major peaks (Figure 1). Peak 1 was identified as p-

191

salicylic acid by mass (Figure 1) and NMR spectra. The major NMR peaks detected by 1H-

192

NMR were 12.33 (1H, s, OH-7), 10.22 (1H, s, OH-4), 7.77 (2H, d, J = 8.7 Hz, H-2, 6), and

193

6.80 (2H, d, J = 8.7 Hz, H-3, 5), while the major peaks detected with 13C-NMR were 167.6

194

(C-7), 162.0 (C-4), 132.0 (C-2, 6), 121.9 (C-1), and 115.6 (C-3, 5). Peaks 2–9 were identified

195

by MS as quercetin, kaempferol, or their glycoside, galactoside, or galloylated derivatives

196

(Figure 1). The chromatograms and spectral data of Peaks 2–9 were consistent with results

197

from a previous study. 23

198

The concentrations of peaks 1–9 in EEDK were measured as 0.8 ± 0.02 mg/g (1), 4.8 ±

199

0.08 mg/g (2), 10.6 ± 0.18 mg/g (3), 12.7 ± 1.24 mg/g (4), 9.4 ± 0.15 mg/g (5), 17.3 ± 0.27

200

mg/g (6), 17.2 ± 0.23 mg/g (7), 2.1 ± 0.03 mg/g (8) and 5.0 ± 0.08 mg/g (9).

201

In order to observe retinal degeneration caused by MNU treatment, we performed H&E

202

staining of retinal tissues, as shown in Figure 2a and 2b. The total thickness of the retinal

203

layer and outer nuclear layer significantly decreased in the mouse group treated with MNU

204

alone (Figure 2). However, the retinas of C57BL/6J mice administrated EEDK were

205

protected against MNU-induced retinal degeneration (Figure 2a and 2b). During the

206

observation period, the inner nuclear layer and ganglion cell layer appeared to be unaffected

207

except for in MNU-treated group at 4 weeks post-MNU treatment. Our results also revealed

208

that EEDK administration protected the outer retina against MNU-induced retinal

209

degeneration. These results are consistent with those from a previous study demonstrating

210

that MNU affects the outer layer by causing a reduction in the number of photoreceptors in

211

the retina,24 and demonstrate protective effects of EEDK against retinal degeneration.

212 213

We observed further morphological changes in the retinas of live mice via SD-OCT scanning, as shown in Figure 3. 25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 12 of 33

214

SD-OCT is a non-invasive scanning technique that has been widely adopted clinically to

215

assess retinal disorders involving changes in retinal thickness, structural changes, and edema,

216

among others. SD-OCT enables accurate analysis because it can provide cross-sectional

217

images of retinas with high resolution. Recently, OCT has been used in many studies to

218

evaluate changes occurring during retinal degeneration in small animal-model studies. 21

219

Figure 3a shows an OCT image of a normal retina (Figure 3a). The thickness of outer

220

retinal layer was slightly reduced in the MNU-treated group at 1 week, compared to the

221

control group (Figure 3b). After 4 weeks, significant differences were found in retinal

222

thickness between the control- and MNU-treated groups (Figure 3b). However, MNU-

223

induced retinal thinning was significantly attenuated by pre-treatment with EEDK (Figure 3b).

224

The reference drug Lutein also showed a protective effect against MNU-induced retinal

225

thinning (Figure 3c).

226

Western blot analysis was conducted to investigate the effects of EEDK treatment on

227

nestin, GFAP, and rhodopsin expression in mouse retinal tissues after MNU-induced retinal

228

degeneration. Nestin, a type-VI intermediate filament protein is expressed in stem cells and

229

progenitor cells, as well as the related Müller cells that differentiate in the central nerve

230

system.26 The observed configuration of Müller cells and astrocytes has demonstrated that

231

these glial cells help maintain the structural integrity of the retina.

232

expression is often observed during late-stage pathology of neurodegenerative diseases.

233

As shown in Figure 4, nestin protein expression was considerably increased in the MNU-

234

treated group at 1 week compared to the control group, but nestin expression was not

235

increased at 4 weeks post-MNU treatment. This result may have been due to low number of

236

cells preserved in the whole retina.

237 238

27

Up-regulated nestin 28, 29

Importantly, the elevation in nectin protein expression was significantly attenuated by EEDK administration (Figure 4a and 4b).

ACS Paragon Plus Environment

Page 13 of 33

Journal of Agricultural and Food Chemistry

239

The results of recent studies have indicated that GFAP, a type-III intermediate filament

240

protein, is present in astrocytes and Müller cells in the retina. Increased GFAP expression is a

241

reliable biomarker of retinal degeneration.

242

time-dependent manner after mice were treated with MNU alone, compared to the control

243

group; this up-regulation was inhibited by EEDK administration (Figure 4a and 4b). The dose

244

of 50 mg/kg EEDK showed a significant inhibition, compared with the MNU-treated group.

245

30

GFAP protein expression was up-regulated in a

Rhodopsin is a photosensitive pigment in the rod-containing outer retinal layer and plays 24

246

a key role in the visual system.

247

protein was down-regulated in a time-dependent manner compare to the control group.

248

However, rhodopsin protein expression was maintained by EEDK administration (Figure 4a

249

and 4b), and the photoreceptor tissue layer was well preserved by EEDK treatment. These

250

results demonstrated that the protective effects of EEDK treatment on retinal degeneration

251

might be related to the expression of retinal factors including nestin, GFAP, and rhodopsin

252

during MNU-induced retinal degeneration.

In the MNU-treated group, expression of the rhodopsin

253

To further evaluate the protective effects of EEDK on retinal degeneration, we analyzed

254

nestin, GFAP, and rhodopsin expression by immunofluorescence staining, as shown in Figure

255

4c and 4d. Immunofluorescence staining revealed that the expression levels of nestin and

256

GFAP, which are related to early signs of gliosis, were significantly increased in the MNU-

257

treated group compared with the control group (Figure 4c and 4d). However, the 50 and 10

258

mg/kg EEDK-treated groups showed decreased nestin and GFAP protein expression

259

compared with that of the MNU-treated group.

260

The loss of photoreceptor cells was evaluated by immunofluorescence staining for

261

rhodopsin. The expression of rhodopsin was considerably decreased in the MNU-treated

262

group relative to that observed in the control group (Figure 4c and 4d). In contrast, rhodopsin

263

protein expression was preserved in the 50 and 10 mg/kg EEDK-treated groups (Figure 4c

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

264

and 4d). As shown in Figures 2, 3, and 4, the 100 mg/kg EEDK-treated groups showed no

265

significant differences compared with the MNU-treated group. Thus, administration of EEDK

266

can potentially confer a protective benefit, but a high dose may elicit retinotoxicity.

267

As observed by western blot analysis, immunofluorescence staining demonstrated the

268

capacity of EEDK to inhibit retinal degeneration by down-regulating nestin and GFAP

269

expression, and by up-regulating rhodopsin expression.

270

Cell viability assays were performed to confirm the protective effects of compounds 1–9

271

(Figure 5). As shown in Figure 5a, only quercetin (10 µM; compound 8) significantly

272

inhibited H2O2-induced cell death (~65%) among compounds 1–9. The inhibition of H2O2-

273

induced cell death by quercetin occurred in a dose-dependent manner (Figure 5b).

274

Recently, Keiichi et al. reported that the administration of exogenous antioxidants such

275

as α-tocopherol, ascorbic acid, Mn (III) tetrakis (4-benzoic acid) porphyrin, and α-lipoic acid

276

protected against oxidative damages in an rd1 mouse model. 31 Quercetin is known to possess

277

antioxidant and free radical-scavenging activity. 32

278

DCFH-DA was used as a radical probe to quantify intracellular ROS production. The

279

intracellular ROS levels caused by O2·− were increased relative to the control by up to 360%.

280

However, pre-treating cells with EEDK and quercetin decreased the ROS levels in a dose-

281

dependent manner (Figure 6a and 6b).

282

The expression levels of antioxidant proteins such as SOD 1–3 and Gpx-1 induced by

283

MNU in mouse retina were evaluated by western blot analysis. Our results showed that SOD-

284

1, SOD-3, and Gpx-1 expression levels were significantly inhibited by EEDK treatment

285

(Figure 6c).

286

Endogenous antioxidant enzymes, including SODs, GPx, and catalase, served to reduce

287

the oxidative stress of photoreceptors in retinas. Previous findings have shown that

288

endogenous antioxidant enzymes up-regulated under oxidative stress conditions decreased

ACS Paragon Plus Environment

Page 14 of 33

Page 15 of 33

289 290 291

Journal of Agricultural and Food Chemistry

ROS production and prevented cone cell death in several retinitis pigmentosa models. 33, 34 Therefore, the protective effects of EEDK in MNU-induced retinal degeneration may be due to its direct or indirect anti-oxidative properties.

292

In summary, our results showed that EEDK has protective effects against oxidative

293

stress-induced cell death in vitro and considerably alleviated MNU-induced retinal

294

degeneration in vivo. In particular, compound 8 (quercetin) could possibly be the acting

295

compound due to anti-oxidative capacity. Quercetin glycosides, Compounds 2–4, did not

296

show significant activity. However, it was known that flavonoid mono-glucosides such as

297

quercetin-3-O-β-glucoside are hydrolyzed by lactase phlorizin hydrolase

298

glucosidase

299

that compounds 2–4 (quercetin-3-O-β-galactoside, quercetin-3-O-β-glucoside, quercetin-3-O-

300

β-2″galloylglucoside) may be absorbed from the small intestine of humans as aglycone forms

301

to exhibit anti-oxidative activity. Collectively, our findings indicate that EEDK may be useful

302

as a potential agent for the prevention and treatment of retinal degeneration, such as with

303

retinitis pigmentosa and AMD.

36, 37

35

and cytosolic β-

to be absorbed in the small intestine as aglycone forms. This demonstrates

304 305

ABBREVIATIONS USED

306

AMD, age related macular degeneration; CAT, catalase; CNS, central nerve system; EEDK,

307

ethanol extract of Diospyros kaki; ELM, external limiting membrane; GFAP, glial fibrillary

308

acidic protein; GCL, ganglion cell layer; GSH, glutathione; GPx, glutathione peroxidase; INL,

309

inner nuclear layer; IP, intraperitoneal; IPL, inner nuclear layer; MNU, N-methyl-N-

310

nitrosourea; ONL, outer nuclear layer; RD-1, retinal degeneration-1 mice; RNFL/GC, retinal

311

nerve fiber layer/ganglion cell layer; ROS, reactive oxygen species; RPE, retinal pigment

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

312

epithelium; SD-OCT, spectral-domain optical coherence tomography; SOD, superoxide

313

dismutase

314

AUTHOR INFORMATION

315

Corresponding Authors

316

*Telephone: +82-33-650-3653. Fax: +82-33-650-3679. E-mail: [email protected].

317

Funding

318

This work was financially supported by an intramural grant (2Z04381) from the Korea

319

Institute of Science and Technology (KIST), Republic of Korea.

320

Notes

321

The authors declare no competing financial interest.

322 323

ACS Paragon Plus Environment

Page 16 of 33

Page 17 of 33

324

Journal of Agricultural and Food Chemistry

REFERENCES

325

(1) Cuenca, N.; Fernandez-Sanchez, L.; Campello, L.; Maneu, V.; De la Villa, P.; Lax, P.;

326

Pinilla, I., Cellular responses following retinal injuries and therapeutic approaches for

327

neurodegenerative diseases. Prog. Retin. Eye Res. 2014, 43, 17-75.

328

(2) Fletcher, A. E., Free radicals, antioxidants and eye diseases: evidence from

329

epidemiological studies on cataract and age-related macular degeneration. Ophthalmic Res.

330

2010, 44, 191-8.

331 332 333 334

(3) Kiang, A. S.; Humphries, M. M.; Campbell, M.; Humphries, P., Antioxidant therapy for retinal disease. Adv. Exp. Med. Biol. 2014, 801, 783-9. (4) Anderson, B., Jr., Ocular effects of changes in oxygen and carbon dioxide tension. Trans. Am. Ophthalmol. Soc. 1968, 66, 423-74.

335

(5) Brennan, L. A.; Kantorow, M., Mitochondrial function and redox control in the aging

336

eye: role of MsrA and other repair systems in cataract and macular degenerations. Exp. Eye

337

Res. 2009, 88, 195-203.

338 339 340 341 342 343 344

(6) Danulescu, R.; Costin, D., Use of blood markers in early diagnosis of oxidative stress in age related macular degeneration. Rev. Med. Chir. Soc. Med. Nat. Iasi 2012, 116, 1136-42. (7) Zampatti, S.; Ricci, F.; Cusumano, A.; Marsella, L. T.; Novelli, G.; Giardina, E., Review of nutrient actions on age-related macular degeneration. Nutr.Res. 2014, 34, 95-105. (8) Klein, R.; Peto, T.; Bird, A.; Vannewkirk, M. R., The epidemiology of age-related macular degeneration. Am. J. Ophthalmol. 2004, 137, 486-95. (9) Tsubura, A.; Lai, Y. C.; Miki, H.; Sasaki, T.; Uehara, N.; Yuri, T.; Yoshizawa, K., Review:

345

Animal models

346

degeneration with special emphasis on therapeutic trials. In Vivo 2011, 25, 11-22.

347

of

N-Methyl-N-nitrosourea-induced mammary

cancer and

retinal

(10) Zulliger, R.; Lecaude, S.; Eigeldinger-Berthou, S.; Wolf-Schnurrbusch, U. E.; Enzmann,

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

348

V., Caspase-3-independent photoreceptor degeneration by N-methyl-N-nitrosourea (MNU)

349

induces morphological and functional changes in the mouse retina. Graefes Arch. Clin. Exp.

350

Ophthalmol. 2011, 249, 859-69.

351

(11) Prater, M. R.; Zimmerman, K. L.; Pinn, L. C.; Keay, J. M.; Laudermilch, C. L.;

352

Holladay, S. D., Role of maternal dietary antioxidant supplementation in murine placental

353

and fetal limb development. Placenta 2006, 27, 502-9.

354

(12) Usui, S.; Oveson, B. C.; Lee, S. Y.; Jo, Y. J.; Yoshida, T.; Miki, A.; Miki, K.; Iwase, T.;

355

Lu, L.; Campochiaro, P. A., NADPH oxidase plays a central role in cone cell death in retinitis

356

pigmentosa. J. Neurochem. 2009, 110, 1028-37.

357

(13) Izuchi, R.; Takahashi, H.; Inada, Y., Preparing a carotenoid polyphenol-enriched extract

358

from the peel of persimmon, Diospyros kaki L.f. Biosci. Biotechnol. Biochem. 2009, 73,

359

2793-5.

360 361

(14) Ozen, A.; Colak, A.; Dincer, B.; Guner, S., A diphenolase from persimmon fruits (Diospyros kaki L., Ebenaceae). Food Chem. 2004, 85, 431-437.

362

(15) Ahn, H. S.; Jeon, T. I.; Lee, J. Y.; Hwang, S. G.; Lim, Y.; Park, D. K., Antioxidative

363

activity of persimmon and grape seed extract: in vitro and in vivo. Nutr. Res. 2002, 22, 1265-

364

1273.

365

(16) Katsube, T.; Tabata, H.; Ohta, Y.; Yamasaki, Y.; Anuurad, E.; Shiwaku, K.; Yamane, Y.,

366

Screening for antioxidant activity in edible plant products: comparison of low-density

367

lipoprotein oxidation assay, DPPH radical scavenging assay, and Folin-Ciocalteu assay. J.

368

Agric. Food Chem. 2004, 52, 2391-6.

369

(17) Bei, W.; Zang, L.; Guo, J.; Peng, W.; Xu, A.; Good, D. A.; Hu, Y.; Wu, W.; Hu, D.; Zhu,

370

X.; Wei, M.; Li, C., Neuroprotective effects of a standardized flavonoid extract from

371

Diospyros kaki leaves. J. Ethnopharmacol. 2009, 126, 134-42.

372

(18) Age-Related Eye Disease Study Research, G., A randomized, placebo-controlled,

ACS Paragon Plus Environment

Page 18 of 33

Page 19 of 33

Journal of Agricultural and Food Chemistry

373

clinical trial of high-dose supplementation with vitamins C and E and beta carotene for age-

374

related cataract and vision loss: AREDS report no. 9. Arch. Ophthalmol. 2001, 119, 1439-52.

375

(19) Age-Related Eye Disease Study Research, G., A randomized, placebo-controlled,

376

clinical trial of high-dose supplementation with vitamins C and E, beta carotene, and zinc for

377

age-related macular degeneration and vision loss: AREDS report no. 8. Arch. Ophthalmol.

378

2001, 119, 1417-36.

379

(20) Jeong, E.; Paik, S. S.; Jung, S. W.; Chun, M. H.; Kim, I. B., Morphological and

380

functional evaluation of an animal model for the retinal degeneration induced by N-methyl-N-

381

nitrosourea. Anat. Cell Biol. 2011, 44, 314-23.

382

(21) Huber, G.; Beck, S. C.; Grimm, C.; Sahaboglu-Tekgoz, A.; Paquet-Durand, F.; Wenzel,

383

A.; Humphries, P.; Redmond, T. M.; Seeliger, M. W.; Fischer, M. D., Spectral domain optical

384

coherence tomography in mouse models of retinal degeneration. Invest. Ophthalmol. Vis. Sci.

385

2009, 50, 5888-95.

386

(22) Shimazawa, M.; Nakajima, Y.; Mashima, Y.; Hara, H., Docosahexaenoic acid (DHA)

387

has neuroprotective effects against oxidative stress in retinal ganglion cells. Brain Res. 2009,

388

1251, 269-75.

389

(23) Kawakami, K.; Shibukura, Y.; Kanno, T.; Furuki, T.; Aketa, S.; Hirayama, M.,

390

Identification of 2''-galloylated flavonol 3-o-glycosides accumulating in developing leaves of

391

persimmon. Phytochem. Anal. 2011, 22, 403-10.

392

(24) Wan, J.; Zheng, H.; Chen, Z. L.; Xiao, H. L.; Shen, Z. J.; Zhou, G. M., Preferential

393

regeneration of photoreceptor from Muller glia after retinal degeneration in adult rat. Vision

394

Res. 2008, 48, 223-34.

395

(25) Rosch, S.; Johnen, S.; Mataruga, A.; Muller, F.; Pfarrer, C.; Walter, P., Selective

396

photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea. Invest.

397

Ophthalmol. Vis. Sci. 2014, 55, 1711-23.

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

398

(26) Kawaguchi, A.; Miyata, T.; Sawamoto, K.; Takashita, N.; Murayama, A.; Akamatsu, W.;

399

Ogawa, M.; Okabe, M.; Tano, Y.; Goldman, S. A.; Okano, H., Nestin-EGFP transgenic mice:

400

visualization of the self-renewal and multipotency of CNS stem cells. Mol. Cell. Neurosci.

401

2001, 17, 259-73.

402

(27) Bignami, A.; Dahl, D., The radial glia of Muller in the rat retina and their response to

403

injury. An immunofluorescence study with antibodies to the glial fibrillary acidic (GFA)

404

protein. Exp. Eye Res. 1979, 28, 63-9.

405

(28) Xue, L.; Ding, P.; Xiao, L.; Hu, M.; Hu, Z., Nestin, a new marker, expressed in Muller

406

cells following retinal injury. The Canadian journal of neurological sciences. Le journal

407

canadien des sciences neurologiques 2010, 37, 643-9.

408

(29) Valamanesh, F.; Monnin, J.; Morand-Villeneuve, N.; Michel, G.; Zaher, M.; Miloudi, S.;

409

Chemouni, D.; Jeanny, J. C.; Versaux-Botteri, C., Nestin expression in the retina of rats with

410

inherited retinal degeneration. Exp. Eye Res. 2013, 110, 26-34.

411

(30) Kohno, H.; Sakai, T.; Kitahara, K., Induction of nestin, Ki-67, and cyclin D1 expression

412

in Muller cells after laser injury in adult rat retina. Graefes Arch. Clin. Exp. Ophthalmol.

413

2006, 244, 90-5.

414 415

(31) Komeima, K.; Rogers, B. S.; Lu, L.; Campochiaro, P. A., Antioxidants reduce cone cell death in a model of retinitis pigmentosa. Proc. Natl. Acad. Sci. U. S. A. 2006, 103, 11300-5.

416

(32) Hollman, P. C. H.; vanTrijp, J. M. P.; Buysman, M. N. C. P.; VanderGaag, M. S.;

417

Mengelers, M. J. B.; deVries, J. H. M.; Katan, M. B., Relative bioavailability of the

418

antioxidant flavonoid quercetin from various foods in man. Febs Lett. 1997, 418, 152-156.

419

(33) Usui, S.; Komeima, K.; Lee, S. Y.; Jo, Y. J.; Ueno, S.; Rogers, B. S.; Wu, Z.; Shen, J.;

420

Lu, L.; Oveson, B. C.; Rabinovitch, P. S.; Campochiaro, P. A., Increased expression of

421

catalase and superoxide dismutase 2 reduces cone cell death in retinitis pigmentosa. Mol.

422

Ther. 2009, 17, 778-86.

ACS Paragon Plus Environment

Page 20 of 33

Page 21 of 33

Journal of Agricultural and Food Chemistry

423

(34) Usui, S.; Oveson, B. C.; Iwase, T.; Lu, L.; Lee, S. Y.; Jo, Y. J.; Wu, Z.; Choi, E. Y.;

424

Samulski, R. J.; Campochiaro, P. A., Overexpression of SOD in retina: need for increase in

425

H2O2-detoxifying enzyme in same cellular compartment. Free Rad. Biol. Med. 2011, 51,

426

1347-54.

427

(35) Day, A. J.; Canada, F. J.; Diaz, J. C.; Kroon, P. A.; Mclauchlan, R.; Faulds, C. B.;

428

Plumb, G. W.; Morgan, M. R. A.; Williamson, G., Dietary flavonoid and isoflavone

429

glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase. Febs Lett. 2000,

430

468, 166-170.

431

(36) Day, A. J.; DuPont, M. S.; Ridley, S.; Rhodes, M.; Rhodes, M. J. C.; Morgan, M. R. A.;

432

Williamson, G., Deglycosylation of flavonoid and isoflavonoid glycosides by human small

433

intestine and liver beta-glucosidase activity. Febs Lett. 1998, 436, 71-75.

434

(37) Ioku, K.; Pongpiriyadacha, Y.; Konishi, Y.; Takei, Y.; Nakatani, N.; Terao, J., beta-

435

glucosidase activity in the rat small intestine toward quercetin monoglucosides. Biosci.

436

Biotech. Bioch. 1998, 62, 1428-1431.

437

438

439

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

440

Figure legends

441

Figure 1. (a) HPLC-UV chromatogram of EEDK. Retention time for compound 1: 7.665 min;

442

compound 2: 11.350 min; compound 3: 11.514 min; compound 4: 11.828 min; compound 5:

443

12.441 min; compound 6: 12.889 min; compound 7: 13.305 min; compound 8: 17.918 min;

444

compound 9: 21.222 min. (b) MS spectra of major peaks (1–9). (c) Identified compounds.

445 446

Figure 2. Histological evaluation in hematoxylin and eosin (H&E) staining. Panel (a) shows

447

retinal cross sections of control (A1, A2), MNU-treated (B1, B2), MNU plus EEDK (100

448

mg/kg)-treated (C1, C2), MNU plus EEDK (50 mg/kg)-treated (D1, D2), and MNU plus

449

EEDK (10 mg/kg)-treated (E1, E2) mice at 1 and 4 weeks, with or without MNU and/or

450

EEDK treatment. Panel (b) shows total retinal thicknesses and outer layer thicknesses at the

451

indicated time points. Scale bar = 50 µm. Experimental values are expressed as mean ±

452

S.E.M. (**) p < 0.01, (***) p < 0.001 from 3 independent experiments (n = 9 mice per group).

453 454

Figure 3. Evaluation of retinal thicknesses by optical coherence tomography. (a) SD-OCT

455

cross-sectional image of a control retina. The following retinal layers were labeled:

456

RNFL/GC, retinal nerve fiber layer/ganglion cell layer; IPL, inner plexiform layer; INL,

457

inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; ELM, external

458

limiting membrane; RPE, retinal pigment epithelium. (b) Representative images were

459

obtained from the experimental animal retina at 1 and 4 weeks after, with or without MNU

460

and/or EEDK treatment; control (A1, A2), MNU-treated (B1, B2), MNU plus EEDK (100

461

mg/kg)-treated (C1, C2), MNU plus EEDK (50 mg/kg)-treated (D1, D2), and MNU plus

462

EEDK (10 mg/kg)-treated (E1, E2) (c) Lutein plus vitamin A supplement was used as

ACS Paragon Plus Environment

Page 22 of 33

Page 23 of 33

Journal of Agricultural and Food Chemistry

463

reference drug (Bright lutein, Ildong Pharmaceutical, Korea). Reference drug group was

464

received lutein plus vitamin A supplement (0.2%/kg/day). Scale bar = 50 µm.

465 466

Figure 4. (a) Western blot analysis showing the effects of EEDK treatment on protein-

467

expression levels of nestin (Müller cell marker; ~220–240 kDa), glial fibrillary-acidic protein

468

(GFAP; Müller cell and astrocyte marker; 51 kDa), rhodopsin (photoreceptor marker; 40

469

kDa), and GAPDH (loading control; 37 kDa) in C57BL/6J mice treated with 50 mg/kg MNU,

470

at 1 and 4 weeks. (b) Quantification of relative protein levels. (c, d) Representative

471

immunostaining images were stained anti-netin (purple), anti-rhodopsin (green), and anti-

472

GFAP (red) in the retina (Nuclei were stained with DAPI) and obtained by confocal

473

microscopy (original magnification, 630×). Retinal cross section from control, MNU-treated,

474

MNU plus EEDK (100 mg/kg)-treated, MNU plus EEDK (50 mg/kg)-treated, and MNU plus

475

EEDK (10 mg/kg)-treated mice at 1 and 4 weeks, with or without MNU and/or EEDK

476

treatment. Scale bar = 50 µm. Experimental values are expressed as mean ± S.E.M. (*) p