Potentiation of in Vivo Anticancer Efficacy of Selenium Nanoparticles

6 days ago - However, different decoration altered the tumor selectivity of the SeNPs, while gastric adenocarcinoma AGS cells showed relative highest ...
0 downloads 0 Views 1MB Size
Subscriber access provided by MIDWESTERN UNIVERSITY

Bioactive Constituents, Metabolites, and Functions

Potentiation of In Vivo Anticancer Efficacy of Selenium Nanoparticles by Mushroom Polysaccharides Surface Decoration Delong Zeng, Jianfu Zhao, Kar-Him Luk, Siu-To Cheung, Ka Hing Wong, and Tianfeng Chen J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.9b00193 • Publication Date (Web): 20 Feb 2019 Downloaded from http://pubs.acs.org on February 21, 2019

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 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 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.

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

1

Potentiation of In Vivo Anticancer Efficacy of Selenium Nanoparticles

2

by Mushroom Polysaccharides Surface Decoration

3 4

Delong Zeng a,#, Jianfu Zhao a,#, Kar-Him Luk b, Siu-To Cheung b, Ka-Hing Wong b,* and

5

Tianfeng Chen a,*

6 7

a

8

China.

9

b Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University,

10

The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632,

Hong Kong, China.

11 12

Corresponding Author

13

* E-mail addresses: [email protected].

14

* E-mail: [email protected] (K. W.)

15 16

Author Contributions

17

#

These authors contributed equally to this work.

18

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 2 of 33

19

ABSTRACT: Selenium nanoparticles (SeNPs) are recently emerging as promising anticancer agents

20

because of their high bioavailability, low toxicity and remarkable anticancer activities. However, the

21

effects of surface physicochemical properties on the biological actions remain elusive. Herein we

22

decorated SeNPs with various water-soluble polysaccharides extracted from various mushrooms, to

23

compare physical characteristics and anticancer profile of these SeNPs. The results showed that the

24

prepared spherical SeNPs displayed particle sizes at 91-102 nm, and kept stable in aqueous solution

25

for up to 13 weeks. However, different decoration altered the tumor selectivity of the SeNPs, while

26

gastric adenocarcinoma AGS cells showed relative highest sensitivity. Moreover, PTR-SeNPs

27

demonstrated potent in vivo antitumor, by inducing caspases- and mitochondria-mediated apoptosis,

28

but showed no obvious toxicity to nomal organs. Taken together, this study offers insights into how

29

surface decoration can tune the cancer selectivity of SeNPs and provides a basis for engineering

30

particles with increased anticancer efficacy.

31 32

Keywords: selenium nanoparticles; mushroom polysaccharide; surface decoration; in vivo anticancer

33

efficacy

34

2

ACS Paragon Plus Environment

Page 3 of 33

35 36

Journal of Agricultural and Food Chemistry

INTRODUCTION Cancer has become a leading cause of death worldwide.1-2 Chemotherapy is one of the major

37

treatments of cancer. Although effective, it is limited by the side effects and drug-resistance

38

developed by cancer cells. Novel high efficacy and low toxicity drugs are still needed for further

39

control of cancers. Selenium is a multifunction essential trace element in human and animal

40

bodies,3-4 which also displays potent activities in cancer prevention and treatment.5 Large amount of

41

selenium-containing compounds have been synthesized and evaluated for their anticancer

42

activities.6-14 Selenium nanoparticles (SeNPs) are recently emerging as promising anticancer agents

43

because of their high bioavailability, much lower toxicity than selenium compounds and remarkable

44

anticancer activities. In our previous studies, we found that SeNPs showed potent anticancer

45

efficacy,15 which could be significantly enhanced by conjugation of targeting molecules such as

46

RGD peptide,16 folic acid,17 and transferrin. 18 SeNPs may also be effective drug carriers that

47

enhance the efficacy of the loaded drugs.19-21 Additionally, SeNPs displayed strong synergism with

48

radiotherapy by increasing ROS production.17, 22-23 A multifunction selenium nanosystem we

49

recently designed, in which ultra-small SeNPs were combined with bevacizumab and then coated

50

with erythrocyte membrane, showed simultaneous cancer radiosensitization and anti-angiogenesis

51

activities.24

52

However, SeNPs are poor in stability and easy to aggregate and precipitate, which will greatly

53

reduce their anti-tumor activities. In addition, unmodified SeNPs cannot selectively target tumor

54

cells, causing side effects. In order to increase the stability of nanoparticles in preparation, storage

55

and application, stabilizers are often used. Common nanoparticle stabilizers include polyvinyl

56

pyrrolidone (PVP),25 sodium methyl cellulose,26 polyvinyl alcohol,27 chitosan,28 and so on. We

57

previously found that glucose, sucrose, chitosan, polysaccharide sulfate, degradable 3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 33

58

polysaccharides, cyanobacteria protein, amino acids, ATP and other biomolecules could effectively

59

regulate the morphology and particle size of SeNPs 29-30 In recent years, increasing studies reported

60

the use of biological macromolecules to regulate and stabilize inorganic nanoparticles.31-32

61

However, research on the regulation and preparation of SeNPs by biomolecules and its application

62

in biomedicine is rare. Mushroom polysaccharides have significant immunomodulatory and anti-

63

tumor activities and have been used as natural adjuvants for tumor chemotherapy in Asia for

64

decades.33 Mushroom polysaccharide has a large number of hydroxyl groups, this unique chemical

65

structure endows it with a strong physical adsorption onto SeNPs, avoiding their accumulation and

66

precipitation.31 We had previously constructed several mushroom polysaccharide-decorated SeNPs

67

and evaluated their bioactivities. Decoration of mushroom polysaccharides not only increased their

68

uptake by cancer cells in cancer therapy,34-35 but also showed strong promotion of bone formation in

69

vitro and in vivo.36

70

Although the SeNPs decorated with different mushroom polysaccharides showed potent

71

activities against cancer cells, whether the physical characteristics, anti-tumor efficacy and tumor

72

specificity of SeNPs were affected by the polysaccharides decorated is unknown and their in vivo

73

anti-tumor activities have not been explored. In this study, we decorated SeNPs with

74

polysaccharides extracted from Pleurotus tuber-regium (PTR), Polyporus rhinoceros (PR),

75

Coriolus versicolor (CV) and Ganoderma lucidum (GL), respectively, and evaluated their anti-

76

tumor efficacy and tumor specificity against a panel of cancer cell lines. In addition to compare

77

their physical characteristics and tumor specific, we established nude mouse model to evaluate their

78

in vivo anti-tumor activities and toxicities. Taken together, this study offers insights into how

79

surface decoration can tune the cancer selectivity of SeNPs and provides a basis for engineering

80

particles with increased anticancer efficacy. 4

ACS Paragon Plus Environment

Page 5 of 33

81

82

Journal of Agricultural and Food Chemistry

MATERIALS AND METHODS

Materials and Chemicals. Sodium selenite (Na2SeO3), propidium iodide (PI), 4’,6-

83

Diamidino-2-phenyindole (DAPI), bicinchoninic acid (BCA) kit, 5,5’,6,6’-

84

tetraethylimidacarbocyanine iodide (JC-1) were purchased from Sigma (Shanghai, China). [3-(4,5-

85

dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt

86

(MTS) was purchased from Promega (Madison, WI). Terminal deoxynucleotidyl transferase dUTP

87

nick end labeling (TUNEL) assay kit was purchased from Roche Applied Science (Mannheim,

88

Germany). Vitamin C was purchased from Guangzhou chemical reagent factory (Guangzhou,

89

China).

90

Preparation of Different Mushroom Polysaccharides-decorated SeNPs. The extraction of

91

water soluble polysaccharides from Pleurotus tuber-regium, Polyporus rhinoceros, Coriolus

92

versicolor and Ganoderma lucidum and the preparation of polysaccharide encapsulated SeNPs were

93

conducted as described previously.34-35, 37 Briefly, the polysaccharides stock solution (0.25%) of

94

different mushrooms were mixed with sodium selenite solution (25 mM). And then freshly prepared

95

ascorbic acid solution (100 mM) was added dropwise into the mixtures under magnetic stirring. The

96

mixtures were protected from light and allowed to react at room temperature for 24 h. The solutions

97

were then dialyzed against ultra-pure water in the dark with intermittent changes of water until no

98

Se could be detected in the outer solution by ICP-AES analysis. Coumarin-6 loaded PTR-SeNPs

99

were prepared in the similar procedures with exception that 4 μg/mL coumarin-6 was added to the

100

101 102

reaction system after the addition of polysaccharides. Characterization of the Different Encapsulated SeNPs. The obtained polysaccharideencapsulated SeNPs were characterized by transmission electron microscopy (TEM), high-

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 6 of 33

103

resolution TEM (HR-TEM), energy dispersive X-ray (EDX), selected area electron diffraction

104

(SAED), particle size analysis and Fourier transform infrared spectroscopy (FT-IR). TEM samples

105

were prepared by dropping the particle solutions onto holey carbon film on copper grids. The

106

images were obtained on an H-7650 Transmission Electron Microscope (Hitachi, Tokyo, Japan) at

107

an acceleration voltage of 80 kV. The HR-TEM images and the corresponding SAED patterns were

108

taken on a JEOL 2010 high-resolution TEM at a voltage of 200 kV. Elemental composition analysis

109

were conducted on an EX-250 system (Horiba, Kyoto, Japan). The size distribution and stability of

110

the nanoparticles in aqueous solution were measured on a NanoSight NS300 Instrument (Malvern

111

Panalytical, Malvern, UK). FT-IR was recorded on a FT-IR spectrometer (Equinox 55, Bruker,

112

Ettlingen, Germany) in the range 4000–500 cm-1.

113

Cell Lines and Cell Culture. Human malignant melanoma A375, lung carcinoma A549,

114

gastric adenocarcinoma AGS, cervix adenocarcinoma HeLa, hepatocellular carcinoma HepG2,

115

foreskin fibroblast Hs68, breast adenocarcinoma MCF-7, osteosarcoma MG-63, pancreatic

116

epithelioid carcinoma PANC-1 and prostrate adenocarcinoma PC-3 were gift from Professor Ming-

117

Chiu Fung, Division of Life Science of the Chinese University of Hong Kong. A375, A549, Hs68,

118

MG63 and PANC-1 were cultivated in DMEM medium. HeLa, HepG2 and MCF-7 were cultivated

119

in MEM medium. AGS was cultivated in ATCC-formulated RPMI-1640 medium while PC-3 was

120

cultivated in DMEM/F12 medium. All human cancer cell lines and normal cell were supplemented

121

with 10% fetal bovine serum and 1% penicillin-streptomycin, except that 10 μg/mL insulin was

122

additionally added for MCF-7, in a 37 °C humidified incubator with 5% CO2.

123

Cell Viability Assay. The anti-proliferation effects of different encapsulated SeNPs on

124

different human cancer cells and normal cell were determined by MTS assay as previously

125

described.38 Cell at desired cell density was seeded into 96-well plate and incubated at 37 °C in CO2 6

ACS Paragon Plus Environment

Page 7 of 33

Journal of Agricultural and Food Chemistry

126

incubator overnight. Then a serial of concentrations of different SeNPs were added for various time

127

points. At the designated time-point, 20 ul of MTS/PMS (2mg/ml MTS, 150 μM PMS) mixture was

128

added into each well of 96-well plates and incubated for 4 hours. Absorbance was measured by a

129

microplate reader at 490nm. Results were expressed as the percentage of absorbance of treated

130

groups relative to the control group. Cytotoxicity Assay. The cytotoxicity of PTR/PR-SeNPs on AGS cancer cells were

131 132

determined by the Cytotoxicity Detection KitPLUS (LDH) (Roche, Mannheim, Germany) and done

133

according to the manufacturer’s manual. Briefly, the cells were seeded in 96-well plate and treated

134

with different SeNPs as in the cell viability assay. Two control groups, low control and high

135

control, were set for each experiment. Low control contained cells and media only and high control

136

contained cells and lysis buffer. At the end of treatment, the plates were centrifuged at 250 g for

137

10min. Then transfered 100ul/well supernatant carefully into corresponding wells of another 96-

138

well plate. Added 100ul reaction mixture to each well and incubated for up to 30min at room

139

temperature (protected from light) and stopped the reaction by adding 50uL stop solution. Measured

140

the absorbance of the samples at 492nm in a microplate reader. The percentage of cytotoxicity was

141

calculated as: Cytotoxicity (%) = (experiment value - low control) / (high control - low control) ×

142

100.

143

Cell Cycle Pattern Analysis. The effect of PTR/PR-SeNPs on the cell cycle distribution of

144

AGS cancer cells were analyzed by the flow cytometry analysis as previously described.39 Cells at

145

desired density were seeded into 6-well plate. After incubation at 37 °C CO2 incubator overnight,

146

the cells were treated with desired concentrations of different SeNPs for various time. The cells

147

were harvested and fixed overnight at -20 °C with 70% ice-cold ethanol. Fixed cells were washed

148

twice with PBS (pH7.4) and centrifuge to remove ethanol. Cells were further washed with 1% BSA 7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 8 of 33

149

and incubated in dark at 4 °C with propidium iodide (PI) staining mixture (1.21mg/ml Tris,

150

700U/ml RNase, 50.1ug/ml PI, pH8.0) overnight. Stained cells were analyzed using flow cytometer.

151

DNA Damage Analysis. The effect of PTR/PR-SeNPs on the DNA damage of AGS cancer

152

cell was assessed by the DNA fragmentation assay.40 Cell at desired cell density was seeded into 6-

153

well plate and incubated at 37 °C CO2 incubator overnight. After treatment with desired

154

concentrations of different SeNP for various time, the cells were harvested and lysed with DNA

155

lysis buffer (200mM Tris-HCl pH8.3, 100mM EDTA & 1%SDS). Proteinase K (10 mg/ml) was

156

used to remove proteins. DNA were then precipitated by adding ice-cold ethanol and RNA were

157

removed by RNase A (0.2 mg/ml). Finally, equal amount of DNA was loaded to 1.5% TAE agarose

158

gel and the gel were ran at 90V for 30 min. The DNA were imaged after ethidium bromide (EB)

159

staining.

160 161

162

Confirmation of Apoptosis. The apoptotic effect of PTR/PR-SeNPs on AGS cancer cell was confirmed by TUNEL-DAPI co-staining assay 18 and Annexin-V-FITC assay.41 For TUNEL assay, cells were seed at desired cell density into 2-cm confocal dishs and treated

163

with desired concentration of SeNPs. Then fixed the cell samples with a freshly prepared Fixation

164

solution (4% Paraformaldehyde in PBS, pH7.4) for 1h at room temperature. The sample were

165

washed twice with PBS and incubated in Permeabilisation solution (0.1% Trition X-100 in 0.1%

166

sodium citrate) for 2 min on ice. After wash with PBS again, 50ul TUNEL reaction mixture were

167

added to the samples and samples were incubated in a humidified atmosphere for 60 min at 37 °C in

168

the dark. Then washed out the buffer and stained with DAPI solution. Images of the samples were

169

captured by fluorescence microscopy.

8

ACS Paragon Plus Environment

Page 9 of 33

170

Journal of Agricultural and Food Chemistry

For Annexin-V-FITC assay, cells were seeded at desired density into 6-well plate and treated

171

with desired concentration of SeNPs, then harvested the cells. After wash with ice-cold PBS, the

172

cell pellets were resuspended with 1 × binding buffer adjusting the cell density to 2-5 × 105

173

cells/mL. The cells were stained with Annexin V-FITC at room temperature for 15 min under

174

darkness. Next, washed the cells with 1 × binding buffer then stained with PI (20 ug/mL) and

175

analyzed by flow cytometer (BD Accuri C6, San Jose, CA).

176

Apoptotic Signaling Pathway Analysis. Cells were seeded in 10-cm dishes. After treatment,

177

whole cell protein lysate was extracted with lysis buffer (Cell Signaling Technology, Inc) and the

178

cytosolic protein was extracted with the ice-cold buffer A (20 mM Hepes, pH 7.5, 1.5 mM MgCl2,

179

10 mM KCl, 1mM EDTA, 1 mM EGTA, 1 mM DTT, 0.5 mM PMSF, 10 μg/mL leupeptin,

180

aprotinin, pepstatin and 250 mM sucrose). Protein concentration were determined by BCA assay.

181

Equal amounts of protein were subjected to Western blot analysis 9 for caspase family and Bcl-2

182

family proteins.

183

Mitochondrial Membrane Potential (ΔΨm) Measurement. The change of mitochondrial

184

membrane potential of AGS cancer cell induced by PTR/PR-SeNPs was measured by JC-1 staining

185

as described before.42 Briefly, cells were seeded at desired cell density into 6-well plate and treated

186

with desired concentration of SeNPs. Then harvested the cells and washed with ice-cold PBS (pH

187

7.4). Resuspended the cells with 1 × JC-1 working solution in the dark at 37 °C for 15 min. After

188

wash twice with assay buffer, cells were analyzed by flow cytometer (BD Accuri C6).

189

In Vitro Cellular Uptake. The uptake of PTR-SeNPs by AGS cancer cell was quantified by

190

the cellular uptake of coumarin-6-loaded PTR-SeNPs as mentioned earlier.34 The efficiency of the

191

cellular uptake of PTR-SeNPs was expressed as the percentage of the tested wells over that of the

9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 33

192

positive wells. The intracellular activity of coumarin-6-loaded PTR-SeNPs in AGS cancer cell was

193

indicated with LysoTracker® Deep Red and DAPI as mentioned previously.18

194

Cellular uptake Mechanism. The cellular uptake mechanism of PTR-SeNPs by AGS cancer

195

cell was studied with several endocytosis inhibitors including nystatin, sucrose and 2-deoxy-D-

196

glucose.18

197

Animal Study. Animal studies were approved by and followed the guidelines of the

198

Laboratory Animal Ethics Committee of Jinan University. The in vivo anticancer activities of PTR-

199

SeNPs and PR-SeNPs were evaluated in nude mouse model. Three to four-week old Balb/c nude

200

mice were inoculated subcutaneously with 2 × 106 MGC-803 gastric cancer cells. The mice were

201

divided randomly into 5 groups (n = 10), after the average volume of tumor reached ~ 100 mm3.

202

The mice of treated groups were administrated with PTR-SeNPs or PR-SeNPs through intravenous

203

injection at dosages of 750 or 2500 μg/kg BW/day, and the mice of control group received vehicle

204

(PBS) only. The tumor volume was recorded during the treatment by measuring the tumor length (l)

205

and width (w) and calculating by the formula: volume = l × w2/2. The treatment lasted for 27 d. At

206

the end of experiment, the mice were euthanized and tumors were dissected and weighed. Major

207

organs including heart, liver, spleen, lung, and kidney were isolated and fixed with tumors in

208

formalin for hematoxylin and eosin (H&E) staining and immunohistochemical analysis, as

209

previously described.12 Hematological analysis were conducted in Blood Test Center of the First

210

Affiliated Hospital of Jinan University.

211

Statistical Analysis. All results were expressed as mean ± SD from at least three independent

212

experiments. Significant differences were tested using Student’s t-test or one-way ANOVA with the

213

aid of statistical program Graphpad Prism version 5.0.

10

ACS Paragon Plus Environment

Page 11 of 33

214

Journal of Agricultural and Food Chemistry

RESULTS

215

Synthesis of Mushroom Polysaccharides-decorated SeNPs and Comparison on the

216

Physicochemical Characteristics. The preparation of polysaccharide-decorated SeNPs were

217

depicted as in Figure 1. Water-soluble polysaccharides were first extracted from four mushrooms,

218

Pleurotus tuber-regium, Polyporus rhinoceros, Coriolus versicolor and Ganoderma lucidum,

219

respectively. Decoration of SeNPs with polysaccharides were done by dropping ascorbic acid

220

solution to the mixture of corresponding polysaccharide and sodium selenite solution. To compare

221

the physical and chemical characteristics of the four SeNPs, the morphology, stability and element

222

composition were tested. The TEM images showed that the four type of SeNPs were well

223

monodispered and nearly spherical particles (Figure 1A-D). PTR-SeNPs had the smallest diameter

224

(~12.5 nm), CV-SeNPs were the largest (~20 nm), and PR- and GL-SeNPs were almost equal in

225

size (~17 nm). Clear lattice fringes could be observed in all the SeNPs in HR-TEM images. These

226

data demonstrated that there was no significant difference of the morphology between the different

227

mushroom polysaccharides-decorated SeNPs. The hydrodynamic size of the CV-SeNPs (~102 nm)

228

and PR-SeNPs were slightly larger (107 nm, Figure S1 Aa-Da) in aqueous solution. These particle

229

sizes were suitable for biological applications.43 We next compared the stability of the four SeNPs

230

by measuring their particle sizes in aqueous solution intermittently for up to 13 weeks. As showed

231

in Figure S1 Ab-Db, no significant change in particle size of all the SeNPs during the measurement.

232

The sizes were comparable to those at the beginning, without aggregation and precipitation. The

233

results implied that the four different mushroom polysaccharides could stabilize SeNPs in aqueous

234

solution with a similar degree. The SeNPs were further analyzed by SAED (Figure S1Ac-Dc) and

235

element composition (Figure S1Ad-Dd). The FT-IR spectra demonstrated the abundance of −OH

236

and −NH groups on the polysaccharides (Figure S1Ae-De) which could make them effectively bind 11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

237

Page 12 of 33

onto the SeNPs. Comparison on Anticancer Activities of Different SeNPs. To compare the anticancer

238 239

profile of the four different SeNPs, a panel of cancer cell lines were treated with various

240

concentrations of SeNPs, and the cell viabilities were measured by MTS assay. As shown in Figure

241

2A, the four functionalized SeNPs could strongly inhibit the growth of cancer cells. However, the

242

inhibition effects of different SeNPs on different cancer cells varied, indicating the tumor specificity

243

of the SeNPs were affected by the surface decorating polysaccharides. It was worth noting that

244

among the 9 cancer cell lines, gastric cancer cell AGS showed relative high sensitivity to all four

245

SeNPs (IC50 = 3.12- 5.58 μM), especially to PR-SeNPs and PTR-SeNPs (Figure S2 and Table S1).

246

In contrast, a much lower cytotoxicity of PR-SeNPs and PTR-SeNPs against the normal cells Hs68

247

could be observed with the IC50 values of 225 μM and 113 μM, respectively (Figure 2B, Figure S2

248

and Table S1). Additionally, PR-SeNPs and PTR-SeNPs also inhibited the growth of AGS cells in a

249

time-depend manner (Figure S3A). According to these results, PR-SeNPs and PTR-SeNPs were

250

used for further studies exploring the mechanisms by which SeNPs inhibit the proliferation of AGS

251

cells.

252

Cellular Uptake of PTR-SeNPs by AGS Cells. Cellular uptake is an important factor

253

affecting the anti-tumor activity of nanomedicines.44 To determine the cellular uptake profile of the

254

encapsulated SeNPs, AGS cells were incubated with a series of concentrations of couramin-6 (green

255

fluorescent)-loaded PTR-SeNPs for different time. The uptake of PTR-SeNPs was monitored by

256

fluorescent imaging. As shown in Figure 3A, the fluorescent intensity of couramin-6 in AGS cells

257

was increased with time lapse, and after internalization, the PTR-SeNPs were co-localized with

258

lysosomes. The uptake of PTR-SeNPs were quantified by determining the fluorescent intensity of

259

the cell lysates. Figure 3B showed that the uptake of SeNPs by AGS cells were increased dose- and 12

ACS Paragon Plus Environment

Page 13 of 33

Journal of Agricultural and Food Chemistry

260

time-dependently. We next explored the internalization pathway of PTR-SeNPs. Endocytosis of

261

AGS cells were blocked by pre-treatment with different inhibitors before incubation of PTR-SeNPs.

262

Quantification of cell lysate fluorescent intensity showed that the uptake of PTR-SeNPs by AGS

263

cells was significantly inhibited when pre-treated with high concentration of nystatin, sucrose and

264

2-deoxy-D-glucose (Figure 3C), implying that endocytosis plays an important role in the uptake of

265

PTR-SeNPs.

266

Induction of Apoptosis of AGS Cell by PR-SeNPs and PTR-SeNPs. To understand the

267

anticancer mechanism of PR-SeNPs and PTR-SeNPs, we analyzed the cell cycle distribution of

268

AGS cells treated with these SeNPs for different time. As shown in Figure 4, treatment with PR-

269

SeNPs and PTR-SeNPs resulted in a dose- and time-dependent increase of the sub-G1 population in

270

AGS cells, which suggested the induction of apoptosis by the SeNPs. The apoptosis was further

271

confirmed by Annexin V/PI double staining and TUNEL assay. Flow cytometry analysis of

272

Annexin-V/PI double staining showed that the apoptotic cells, including early (Annexin V+/PI-)

273

and late apoptotic (Annexin V+/PI+) subsets, were increased up to more than 53% and 46% when

274

AGS cell were treated with high concentration of PTR-SeNPs or PR-SeNPs, respectively (Figure

275

5A). In comparison, staurosporine (STS), as a positive apoptosis inducer, resulted in about 10% of

276

apoptosis at the concentration of 10 μM. Similar results were observed in TUNEL assay (Figure 5B

277

and C) and the DNA fragmentation assay (Figure S4). Taken together, these data demonstrated that

278

PR-SeNPs and PTR-SeNPs were effective in inducing apoptotic cell death of AGS cells.

279

Apoptotic Signaling Pathways Induced by PR-SeNPs and PTR-SeNPs. Mitochondrion is

280

the central regulator of the intrinsic apoptosis pathway.45 Apoptotic signals trigger the loss of

281

mitochondrial membrane potential (ΔΨm) and release of pro-apoptotic molecules to activate

282

caspase cascade.46 To examine the role of mitochondrion in PR-SeNPs and PTR-SeNPs induced 13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 14 of 33

283

apoptosis, we used JC-1 to detect the change of mitochondrial membrane potential of AGS cells

284

treated with PR-SeNPs or PTR-SeNPs. As shown in Figure 6A and B, PTR-SeNPs and PR-SeNPs

285

caused substantial loss of mitochondrial membrane potential in a dose- and time-dependent manner,

286

implying the involvement of mitochondrion during the PR-SeNPs and PTR-SeNPs caused

287

apoptosis.

288

Caspase family proteins are activated sequentially and then play a central roles in the process

289

of apoptosis.47 To examine the activation of caspases in AGS cells treated with PR-SeNPs and

290

PTR-SeNPs, we measured their cleavage by Western blot. As shown in Figure 7A, the full-length

291

caspase-3, -7 and -9 were decreased when AGS cells were treated with the SeNPs, and the active

292

(cleaved) forms were increased correspondently. The substrate of caspases PARP was also cleaved,

293

which is considered as a marker of apoptosis. Caspase-8, which mediates the death signal from

294

outside the cells, were not activated, implying that the extrinsic pathway was not involved in the

295

PR-SeNPs and PTR-SeNPs induced apoptosis. These results were in line with the loss of

296

mitochondrial membrane potential, which is the upstream signal of the intrinsic apoptosis pathway.

297

The mitochondrial outer membrane integrity is regulated by the Bcl-2 family proteins.48 We

298

therefore measured the expression levels of a panel of Bcl-2 family proteins. As showed in Figure

299

7B, the expression levels of anti-apoptotic members, Bcl-2 and Bcl-XL, were significantly decreased

300

while Mcl-1 and phosphorylated Bcl-2 were nearly unchanged after treatment of PR-SeNPs or

301

PTR-SeNPs. The activation of apoptotic signal was also promoted by the upregulation of the pro-

302

apoptotic proteins, including Bad, Bax, Bim, Bid, Puma and Bak (Figure 7B). Taken together, these

303

data demonstrated that PR-SeNPs and PTR-SeNPs could induce apoptosis of AGS cells by

304

regulating the Bcl-2 family proteins to decrease the integrity of mitochondrial outer membrane and

305

activation of caspases. The possible apoptosis signaling pathway induced by PR- and PTR-SeNPs 14

ACS Paragon Plus Environment

Page 15 of 33

306

307

Journal of Agricultural and Food Chemistry

were depicted in Figure7C. In vivo Anticancer Activities of PR-SeNPs and PTR-SeNPs. The in vivo anticancer

308

activities of mushroom polysaccharides-decorated SeNPs haven’t been evaluated before. Therefore,

309

we tested the in vivo efficacy of PR-SeNPs and PTR-SeNPs by using nude mouse xenograft model.

310

Because AGS cells failed to graft on nude mice in our pre-experiments, another gastric cancer cells

311

MGC-803 were used. The cells were inoculated subcutaneously on nude mice which then received

312

treatment with PTR-SeNPs, PR-SeNPs or vehicle. Tumor growth curves were shown in Figure 8A

313

which demonstrated potent tumor inhibition effects of PTR-SeNPs and PR-SeNPs. Images of

314

tumors dissected from mice and representative images of mice in different treatment groups were

315

shown in Figure 8B and C respectively. Tumor sections were further analyzed by

316

immunohistochemical staining (Figure 8D). Ki67 is a proliferation marker, which significantly

317

decreased in the SeNPs treated groups, especially in the high dose groups. In the contrast, the tumor

318

suppressor p53 were strongly induced by the treatment of SeNPs. VEGFR2, which plays a critical

319

role in angiogenesis, displayed similar inhibition tendency to Ki67. Moreover, results of TUNEL

320

assay showed that high dose of SeNPs treatment induced remarkable apoptosis in tumor tissues. In

321

H&E staining, larger areas of necrosis were found in the groups of high dose of SeNPs treatment. In

322

summary, the results above clearly showed that PR-SeNPs and PTR-SeNPs were effective in

323

inhibiting the growth of tumors in vivo.

324

Furthermore, the toxicity of PR-SeNPs and PTR-SeNPs to the mice was evaluated by

325

examination of the H&E-staining sections of major organs and blood biochemical indexes. As

326

shown in Figure 9A, no obvious damage could be observed in the organ sections, including lung,

327

liver, spleen, kidney and heart. Blood biochemical indexes, LDLC and GLB, reflecting the level

328

blood fat and liver function, were increased in the model group, which were alleviated by the 15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 16 of 33

329

treatment of SeNPs (Figure 9B). These data together demonstrated potent anti-tumor activity of PR-

330

SeNPs and PTR-SeNPs in vivo and a relative low toxicity to the mice.

331

DISCUSSION

332

To increase the stability and efficacy of nanoparticles in medical use, surface decorators were

333

commonly used. We had previously found that decoration of polysaccharides from certain

334

mushrooms significantly increase the stability, cellular uptake and in vitro anti-tumor potency of

335

SeNPs.34-35 However, whether there is difference of physical characteristics and cancer type

336

selectivity between different mushroom polysaccharides-decorated SeNPs were unclear. Thus, we

337

synthesized and compared the physical characteristics and anticancer activities of four mushroom

338

polysaccharides-decorated SeNPs in the present study. And the in vivo efficacy of mushroom

339

polysaccharide-decorated SeNPs were evaluated for the first time.

340

Stability is an important characteristic affecting the application of nanoparticles. Naked

341

SeNPs are very unstable and will precipitate in the solution soon after synthesized. 34-35 Due to the

342

abundant hydroxyl groups of the mushroom polysaccharides, they had strong physical adsorption to

343

SeNPs, which could effectively avoid their accumulation and regulate their particle size. The

344

modified SeNPs in our study were stable in aqueous solution for up to 13 weeks. Another

345

interesting result found in the present study is that SeNPs decorated with different mushroom

346

polysaccharides inhibited the growth of different cancer cells in varied degrees. As the four

347

mushroom polysaccharide-modified SeNPs were similar in physical characteristics, we assumed

348

that the difference of anti-cancer profiles of these SeNPs were derived from the chemical

349

differences of the polysaccharides. The chemical component and molecular structure of the

350

polysaccharides covering the SeNPs may affect the interaction of the nanoparticles and cell

351

membranes and therefore affect their cellular uptake behavior and the signalling inside the cells. It 16

ACS Paragon Plus Environment

Page 17 of 33

Journal of Agricultural and Food Chemistry

352

would be interested for us to further explorer the chemical structure of the mushroom

353

polysaccharides and their relation with anti-cancer efficacy in the future.

354

The in vivo anti-tumor activity of polysaccharide-modified SeNPs had not been evaluated

355

before. Here, we used xenograft model to demonstrate the tumor inhibition efficacy in nude mice.

356

The tumor growth of the PT/PTR-SeNPs treated group was significantly slower than the control

357

group, confirming the in vivo activity of polysaccharide-modified SeNPs for the first time. In

358

additionally, the SeNPs showed no evident toxicity to the mice, implied by the results of blood

359

biochemical indexes and histochemical staining of organ tissues. These data demonstrated an

360

effective way to change cancer selectivity of SeNPs by decoration of mushroom polysaccharides

361

and suggested their potential of further usage in clinical.

362

Overall, comparing with previous study on SeNPs, this study makes improvement on the

363

following issues. Firstly, we have compared the difference in physical characteristics and anticancer

364

efficacy of SeNPs decorated with polysaccharides from different mushrooms, while the previous

365

papers were intended to investigate whether the decoration of mushroom polysaccharides could

366

stabilize SeNP and increase its uptake by cancer cells. The results of present study demonstrated the

367

different species of polysaccharide determined the cancer selectivity of SeNPs they decorated. It

368

would be a pioneer work that guide the future design of tumor selective SeNPs for cancer therapy

369

based on surface decoration. Secondly, the present study is a pioneer study on the in vivo anticancer

370

activity and action mechanisms of polysaccharide-decorated SeNPs. However, previous studies just

371

focused on in vitro anticancer activity of PTR-SeNPs. Herein we have examined the changes of

372

Bcl-2 family proteins which are central regulators in the process of endogenous apoptosis pathway.

373

In addition, the in vivo anti-cancer efficacy of polysaccharide-decorated SeNPs had been evaluated

374

in the present study, which haven’t been reported before. Indeed, not all the agents that display 17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

375

good anticancer efficacy in vitro show similar potency in vivo. So, our present study could provide

376

solid data demonstrating the in vivo tumor inhibition effect and the underlying molecular

377

mechanisms of the mushroom polysaccharide-decorated SeNPs. Taken together, this study offers

378

insights into how surface decoration can tune the cancer selectivity of SeNPs and provides a basis

379

for engineering particles with increased anticancer efficacy.

Page 18 of 33

380 381

AUTHOR INFORMATION

382

Corresponding Authors

383

* E-mail addresses: [email protected]. [email protected].

384

Author Contributions

385

#

These authors contributed equally to this work.

386 387

Funding

388

This work was supported by Natural Science Foundation of China (21877049), National Program for

389

Support of Top-notch Young Professionals (W02070191), YangFan Innovative & Entepreneurial

390

Research Team Project (201312H05), Fundamental Research Funds for the Central Universities,

391

Shenzhen's strategic emerging industries development fund (JCYJ20130401152508660).

392

Notes

393

The authors declare no competing financial interest.

394

Supplementary Information

395

The Supporting Information is available free of charge on the ACS Publications website at DOI:

396

10.1021/acs.jafc.XXXXX.

18

ACS Paragon Plus Environment

Page 19 of 33

Journal of Agricultural and Food Chemistry

397

Characterization of the four mushroom polysaccharide-decorated SeNPs; the dose-response

398

curves of the PTR- and PR-SeNPs against cancer and normal cells; time-dependent inhibition of

399

growth of AGS cells by PRT- and PR-SeNPs; the IC50 values of different polysaccharides-

400

decorated SeNPs against different cell lines; agarose gel electrophoresis of DNA extracted from

401

PR/PTR-SeNPs treated AGS cells(PDF).

402 403

References

404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435

(1) Torre, L. A.; Bray, F.; Siegel, R. L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A., Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65 (2), 87-108. (2) Siegel, R. L.; Miller, K. D.; Jemal, A., Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68 (1), 7-30. (3) Duntas, L. H.; Benvenga, S., Selenium: an element for life. Endocrine 2015, 48 (3), 756-775. (4) Rayman, M. P., Selenium and human health. The Lancet 2012, 379 (9822), 1256-1268. (5) Bartolini, D.; Sancineto, L.; Fabro de Bem, A.; Tew, K. D.; Santi, C.; Radi, R.; Toquato, P.; Galli, F., Selenocompounds in Cancer Therapy: An Overview. Adv. Cancer. Res. 2017, 136, 259-302. (6) Fernandes, A. P.; Gandin, V., Selenium compounds as therapeutic agents in cancer. Biochim. Biophys. Acta. 2015, 1850 (8), 1642-60. (7) Zhao, Z.; Gao, P.; You, Y.; Chen, T., Cancer-Targeting Functionalization of Selenium-Containing Ruthenium Conjugate with Tumor Microenvironment-Responsive Property to Enhance Theranostic Effects. Chem-Eur. J. 2018, 24 (13), 3289-3298. (8) Deng, S.; Zeng, D.; Luo, Y.; Zhao, J.; Li, X.; Zhao, Z.; Chen, T., Enhancement of cell uptake and antitumor activity of selenadiazole derivatives through interaction and delivery by serum albumin. Rsc Adv. 2017, 7 (27), 16721-16729. (9) Lai, H.; Fu, X.; Sang, C.; Hou, L.; Feng, P.; Li, X.; Chen, T., Selenadiazole Derivatives Inhibit AngiogenesisMediated Human Breast Tumor Growth by Suppressing the VEGFR2-Mediated ERK and AKT Signaling Pathways. Chem-Asian J. 2018, 13 (11), 1447-1457. (10) Lai, H.; Zhang, X.; Feng, P.; Xie, L.; Chen, J.; Chen, T., Enhancement of Antiangiogenic Efficacy of Iron(II) Complex by Selenium Substitution. Chem-Asian J. 2017, 12 (9), 982-987. (11) Liang, Y.; Huang, W.; Zeng, D.; Huang, X.; Chan, L.; Mei, C.; Feng, P.; Tan, C.-H.; Chen, T., Cancer-targeted design of bioresponsive prodrug with enhanced cellular uptake to achieve precise cancer therapy. Drug Deliv. 2018, 25 (1), 1350-1361. (12) Zeng, D.; Deng, S.; Sang, C.; Zhao, J.; Chen, T., Rational Design of Cancer-Targeted Selenadiazole Derivative as Efficient Radiosensitizer for Precise Cancer Therapy. Bioconjug. Chem. 2018, 29 (6), 2039-2049. (13) Zhang, X.; Dai, C.; You, Y.; He, L.; Chen, T., Tea regimen, a comprehensive assessment of antioxidant and antitumor activities of tea extract produced by Tie Guanyin hybridization. Rsc Adv. 2018, 8 (21), 11305-11315. (14) Zhao, J.; Zeng, D.; Liu, Y.; Luo, Y.; Ji, S.; Li, X.; Chen, T., Selenadiazole derivatives antagonize hyperglycemiainduced drug resistance in breast cancer cells by activation of AMPK pathways. Metallomics 2017, 9 (5), 535-545. (15) Luo, H.; Wang, F.; Bai, Y.; Chen, T.; Zheng, W., Selenium nanoparticles inhibit the growth of HeLa and MDAMB-231 cells through induction of S phase arrest. Colloid. Surface. B 2012, 94, 304-308. (16) Fu, X.; Yang, Y.; Li, X.; Lai, H.; Huang, Y.; He, L.; Zheng, W.; Chen, T., RGD peptide-conjugated selenium nanoparticles: antiangiogenesis by suppressing VEGF-VEGFR2-ERK/AKT pathway. Nanomed-Nanotechnol. 2016, 12 19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481

Page 20 of 33

(6), 1627-39. (17) Yang, Y.; Xie, Q.; Zhao, Z.; He, L.; Chan, L.; Liu, Y.; Chen, Y.; Bai, M.; Pan, T.; Qu, Y.; Ling, L.; Chen, T., Functionalized Selenium Nanosystem as Radiation Sensitizer of 125I Seeds for Precise Cancer Therapy. ACS Appl. Mater. Inter. 2017, 9 (31), 25857-25869. (18) Huang, Y.; He, L.; Liu, W.; Fan, C.; Zheng, W.; Wong, Y. S.; Chen, T., Selective cellular uptake and induction of apoptosis of cancer-targeted selenium nanoparticles. Biomaterials 2013, 34 (29), 7106-7116. (19) Liu, T.; Zeng, L.; Jiang, W.; Fu, Y.; Zheng, W.; Chen, T., Rational design of cancer-targeted selenium nanoparticles to antagonize multidrug resistance in cancer cells. Nanomed-Nanotechnol. 2015, 11 (4), 947-958. (20) He, L.; Zeng, L.; Mai, X.; Shi, C.; Luo, L.; Chen, T., Nucleolin-targeted selenium nanocomposites with enhanced theranostic efficacy to antagonize glioblastoma. J. Mater. Chem. B 2017, 5 (16), 3024-3034. (21) Fang, X.; Li, C.; Zheng, L.; Yang, F.; Chen, T., Dual-Targeted Selenium Nanoparticles for Synergistic Photothermal Therapy and Chemotherapy of Tumors. Chem-Asian J. 2018, 13 (8), 996-1004. (22) Chan, L.; He, L.; Zhou, B.; Guan, S.; Bo, M.; Yang, Y.; Liu, Y.; Liu, X.; Zhang, Y.; Xie, Q.; Chen, T., CancerTargeted Selenium Nanoparticles Sensitize Cancer Cells to Continuous gamma Radiation to Achieve Synergetic ChemoRadiotherapy. Chem-Asian J. 2017, 12 (23), 3053-3060. (23) Yu, B.; Liu, T.; Du, Y.; Luo, Z.; Zheng, W.; Chen, T., X-ray-responsive selenium nanoparticles for enhanced cancer chemo-radiotherapy. Colloid. Surface. B 2016, 139, 180-189. (24) Liu, T.; Shi, C.; Duan, L.; Zhang, Z.; Luo, L.; Goel, S.; Cai, W.; Chen, T., A highly hemocompatible erythrocyte membrane-coated ultrasmall selenium nanosystem for simultaneous cancer radiosensitization and precise antiangiogenesis. J. Mater. Chem. B 2018, 6 (29), 4756-4764. (25) Koczkur, K. M.; Mourdikoudis, S.; Polavarapu, L.; Skrabalak, S. E., Polyvinylpyrrolidone (PVP) in nanoparticle synthesis. Dalton T. 2015, 44 (41), 17883-17905. (26) Tan, J. J.; Liu, R. G.; Wang, W.; Liu, W. Y.; Tian, Y.; Wu, M.; Huang, Y., Controllable Aggregation and Reversible pH Sensitivity of AuNPs Regulated by Carboxymethyl Cellulose. Langmuir 2010, 26 (3), 2093-2098. (27) Chen, W.; Achazi, K.; Schade, B.; Haag, R., Charge-conversional and reduction-sensitive poly(vinyl alcohol) nanogels for enhanced cell uptake and efficient intracellular doxorubicin release. J. Control. Release 2015, 205, 15-24. (28) Hu, Y. W.; Du, Y. Z.; Liu, N.; Liu, X.; Meng, T. T.; Cheng, B. L.; He, J. B.; You, J.; Yuan, H.; Hu, F. Q., Selective redox-responsive drug release in tumor cells mediated by chitosan based glycolipid-like nanocarrier. J. Control. Release 2015, 206, 91-100. (29) Yu, B.; Chen, T.; Yang, F.; Liu, W.; Li, Y. H.; Zheng, W., Chitosan as morphology-directing agent for the preparation of multiarmed selenium/carbon coaxial nanorods. Chem. Lett. 2011, 40 (3), 242-243. (30) Li, Q.; Chen, T.; Yang, F.; Liu, J.; Zheng, W., Facile and controllable one-step fabrication of selenium nanoparticles assisted by l-cysteine. Mater. Lett. 2010, 64 (5), 614-617. (31) Zhang, Y.; Wang, J.; Zhang, L., Creation of highly stable selenium nanoparticles capped with hyperbranched polysaccharide in water. Langmuir 2010, 26 (22), 17617-23. (32) Semenova, M., Protein-polysaccharide associative interactions in the design of tailor-made colloidal particles. Curr. Opin. Colloid. In. 2017, 28, 15-21. (33) Wasser, S. P., Medicinal mushrooms as a source of antitumor and immunomodulating polysaccharides. Appl. Microbiol. Biotechnol. 2002, 60 (3), 258-74. (34) Wu, H.; Li, X.; Liu, W.; Chen, T.; Li, Y.; Zheng, W.; Man, C. W. Y.; Wong, M. K.; Wong, K. H., Surface decoration of selenium nanoparticles by mushroom polysaccharides-protein complexes to achieve enhanced cellular uptake and antiproliferative activity. J. Mater. Chem. 2012, 22 (19), 9602-9610. (35) Wu, H.; Zhu, H.; Li, X.; Liu, Z.; Zheng, W.; Chen, T.; Yu, B.; Wong, K. H., Induction of apoptosis and cell cycle arrest in A549 human lung adenocarcinoma cells by surface-capping selenium nanoparticles: An effect enhanced by polysaccharide-protein complexes from Polyporus rhinocerus. J. Agric. Food Chem. 2013, 61 (41), 9859-9866. (36) Yu, S.; Luk, K.-H.; Cheung, S.-T.; Kwok, K. W.-H.; Wong, K.-H.; Chen, T., Polysaccharide-Protein Complex20

ACS Paragon Plus Environment

Page 21 of 33

482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506

Journal of Agricultural and Food Chemistry

Decorated Selenium Nanosystem as Efficient Bone-Formation Therapeutics. J. Mater. Chem. B 2018, 6 (32), 5215-5219. (37) Huang, W.; Huang, Y.; You, Y.; Nie, T.; Chen, T., High-Yield Synthesis of Multifunctional Tellurium Nanorods to Achieve Simultaneous Chemo-Photothermal Combination Cancer Therapy. Adv. Funct. Mater. 2017, 27 (33). (38) Malich, G.; Markovic, B.; Winder, C., The sensitivity and specificity of the MTS tetrazolium assay for detecting the in vitro cytotoxicity of 20 chemicals using human cell lines. Toxicology 1997, 124 (3), 179-92. (39) Huang, W.; Wu, H.; Li, X.; Chen, T., Facile One-Pot Synthesis of Tellurium Nanorods as Antioxidant and Anticancer Agents. Chem-Asian J. 2016, 11 (16), 2301-11. (40) Walker, P. R.; Smith, C.; Youdale, T.; Leblanc, J.; Whitfield, J. F.; Sikorska, M., Topoisomerase II-reactive chemotherapeutic drugs induce apoptosis in thymocytes. Cancer Res. 1991, 51 (4), 1078-85. (41) Hollville, E.; Martin, S. J., Measuring Apoptosis by Microscopy and Flow Cytometry. Curr. Protoc. Immunol. 2016, 112, 14 38 1-14 38 24. (42) Esner, M.; Graifer, D.; Lleonart, M. E.; Lyakhovich, A., Targeting cancer cells through antibiotics-induced mitochondrial dysfunction requires autophagy inhibition. Cancer Lett. 2017, 384, 60-69. (43) Thorek, D. L.; Tsourkas, A., Size, charge and concentration dependent uptake of iron oxide particles by nonphagocytic cells. Biomaterials 2008, 29 (26), 3583-90. (44) Nie, S., Understanding and overcoming major barriers in cancer nanomedicine. Nanomedicine-UK 2010, 5 (4), 5238. (45) Desagher, S.; Martinou, J. C., Mitochondria as the central control point of apoptosis. Trends Cell Biol. 2000, 10 (9), 369-377. (46) Elmore, S., Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol. 2007, 35 (4), 495-516. (47) Shalini, S.; Dorstyn, L.; Dawar, S.; Kumar, S., Old, new and emerging functions of caspases. Cell Death Differ. 2015, 22 (4), 526-39. (48) Chen, H. C.; Kanai, M.; Inoue-Yamauchi, A.; Tu, H. C.; Huang, Y.; Ren, D.; Kim, H.; Takeda, S.; Reyna, D. E.; Chan, P. M.; Ganesan, Y. T.; Liao, C. P.; Gavathiotis, E.; Hsieh, J. J.; Cheng, E. H., An interconnected hierarchical model of cell death regulation by the BCL-2 family. Nat. Cell Biol. 2015, 17 (10), 1270-81.

507 508

21

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 22 of 33

Figure Captions Figure 1. Preparation of Pleurotus tuber-regium (A), Polyporus rhinoceros (B), Coriolus versicolor (C) and Ganoderma lucidum (D) polysaccharide-decorated SeNPs and characterization of their morphology by TEM and HR-TEM. Figure 2. Tumor growth inhibition effects of mushroom polysaccharide-decorated SeNPs. (A) The IC50 of PTR-, PR-, CV- and GL-SeNPs against nine different cancer cells measured by MTS assay. (B) The IC50 of PTR- and PR-SeNPs against normal fibroblasts cells Hs68 measured by MTS assay. Figure 3. The uptake mechanism and intracellular localization of coumarin-6-loaded PTR-SeNPs. (A) Localization of PTR-SeNPs in AGS cells were visualized by staining the cells with DAPI, LysoTracker® Deep Red after treatment with coumarin-6-loaded-PTR-SeNPs. (B) The uptake of coumarin-6-loaded PTR-SeNPs by AGS cells were quantified by measuring the fluorescent intensity of lysates from cells treated with a serial of concentrations of PTR-SeNPs for the indicated time. (C) The uptake of coumarin-6-loaded PTR-SeNPs in AGS cells pre-treated with the indicated endocytosis inhibitors. Figure 4. Flow cytometry analysis of AGS cells treated with PTR- and PR-SeNPs. (A) Representative PI staining flow cytometry histograms of the AGS cells treated with IC50 (3 μM) or 10 × IC50 (30 μM) of PTR- or PR-SeNPs for different time. (B) Statistic bar charts of three replicated flow cytometry analyses showing the distribution of cells in different cell cycle phase. Sub-G1 population was considered as the apoptotic subset. Figure 5. Validation of apoptosis in AGS cells treated with PTR- or PR-SeNPs. (A and B) Apoptosis of AGS cells treated with PTR- or PR-SeNPs was detected by Annexin-V/PI double staining (A) and TUNEL assay (B). (C) Statistics bar chart of the percent of TUNEL-positive cells in (B). Staurosporine (STS) was used as a positive control of apoptosis inducer. 22

ACS Paragon Plus Environment

Page 23 of 33

Journal of Agricultural and Food Chemistry

Figure 6. Mitochondrial member potential is decreased in PR-SeNPs and PTR-SeNPs-caused apoptosis. Loss of mitochondrial member potential in AGS cells treated with PTR- or PR-SeNPs was examined by JC-1 staining. Shown are representative flow cytometry dot plots (A) and statistic bar charts (B). Figure 7. Apoptotic pathways triggered by PTR- or PR-SeNPs. (A) Activation of caspases in AGS cells treated with PTR- or PR-SeNPs was examined by Western blot. (B) The changes of expression of Bcl-2 family proteins in AGS cells after treatment of PTR- or PR-SeNPs were examined by Western blot. (C) The hypothetic signaling pathway by which the polysaccharide-decorated SeNPs induced apoptosis of AGS cells. Figure 8. In vivo anticancer efficacy of PTR- and PR-SeNPs tested in nude mice xenograft model. (A) The growth curves of xenograft tumor inoculated on nude mice treated with 750 μg/kg (L) or 2500 μg/kg (H) of PTR-/PR-SeNPs or vehicle (PBS). (B and C) The images of tumors and representative mice in each group taken at the end of treatment. (D) Tumor inhibition effects of PTRand PR-SeNPs were examined by immunohistochemical and H&E analysis. Figure 9. Toxicities of PR- and PTR-SeNPs to mice were evaluated by H&E staining of tissue sections (A) and hematological analysis (B).

23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 24 of 33

Figure 1. Preparation of Pleurotus tuber-regium (A), Polyporus rhinoceros (B), Coriolus versicolor (C) and Ganoderma lucidum (D) polysaccharide-decorated SeNPs and characterization of their morphology by TEM and HR-TEM.

24

ACS Paragon Plus Environment

Page 25 of 33

Journal of Agricultural and Food Chemistry

Figure 2. Tumor growth inhibition effects of mushroom polysaccharide-decorated SeNPs. (A) The IC50 of PTR-, PR-, CV- and GL-SeNPs against nine different cancer cells measured by MTS assay. (B) The IC50 of PTR- and PR-SeNPs against normal fibroblasts cells Hs68 measured by MTS assay.

25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 26 of 33

Figure 3. The uptake mechanism and intracellular localization of coumarin-6-loaded PTR-SeNPs. (A) Localization of PTR-SeNPs in AGS cells were visualized by staining the cells with DAPI, LysoTracker® Deep Red after treatment with coumarin-6-loaded-PTR-SeNPs. (B) The uptake of coumarin-6-loaded PTR-SeNPs by AGS cells were quantified by measuring the fluorescent intensity of lysates from cells treated with a serial of concentrations of PTR-SeNPs for the indicated time. (C) The uptake of coumarin-6-loaded PTR-SeNPs in AGS cells pre-treated with the indicated endocytosis inhibitors.

26

ACS Paragon Plus Environment

Page 27 of 33

Journal of Agricultural and Food Chemistry

Figure 4. Flow cytometry analysis of AGS cells treated with PTR- and PR-SeNPs. (A) Representative PI staining flow cytometry histograms of the AGS cells treated with IC50 (3 μM) or 10 × IC50 (30 μM) of PTR- or PR-SeNPs for different time. (B) Statistic bar charts of three replicated flow cytometry analyses showing the distribution of cells in different cell cycle phase. Sub-G1 population was considered as the apoptotic subset.

27

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 28 of 33

Figure 5. Validation of apoptosis in AGS cells treated with PTR- or PR-SeNPs. (A and B) Apoptosis of AGS cells treated with PTR- or PR-SeNPs was detected by Annexin-V/PI double staining (A) and TUNEL assay (B). (C) Statistics bar chart of the percent of TUNEL-positive cells in (B). Staurosporine (STS) was used as a positive control of apoptosis inducer.

28

ACS Paragon Plus Environment

Page 29 of 33

Journal of Agricultural and Food Chemistry

Figure 6. Mitochondrial member potential is decreased in PR-SeNPs and PTR-SeNPs-caused apoptosis. Loss of mitochondrial member potential in AGS cells treated with PTR- or PR-SeNPs was examined by JC-1 staining. Shown are representative flow cytometry dot plots (A) and statistic bar charts (B).

29

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 30 of 33

Figure 7. Apoptotic pathways triggered by PTR- or PR-SeNPs. (A) Activation of caspases in AGS cells treated with PTR- or PR-SeNPs was examined by Western blot. (B) The changes of expression of Bcl-2 family proteins in AGS cells after treatment of PTR- or PR-SeNPs were examined by Western blot. (C) The hypothetic signaling pathway by which the polysaccharide-decorated SeNPs induced apoptosis of AGS cells.

30

ACS Paragon Plus Environment

Page 31 of 33

Journal of Agricultural and Food Chemistry

Figure 8. In vivo anticancer efficacy of PTR- and PR-SeNPs tested in nude mice xenograft model. (A) The growth curves of xenograft tumor inoculated on nude mice treated with 750 μg/kg (L) or 2500 μg/kg (H) of PTR-/PR-SeNPs or vehicle (PBS). (B and C) The images of tumors and representative mice in each group taken at the end of treatment. (D) Tumor inhibition effects of PTRand PR-SeNPs were examined by immunohistochemical and H&E analysis.

31

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 32 of 33

Figure 9. Toxicities of PR- and PTR-SeNPs to mice were evaluated by H&E staining of tissue sections (A) and hematological analysis (B).

32

ACS Paragon Plus Environment

Page 33 of 33

Journal of Agricultural and Food Chemistry

The table of contents

Herein we decorate selenium nanoparticles (SeNPs) with water soluble polysaccharides extracted from mushrooms to compare their physical characteristics and evaluate their effects on in vivo anticancer efficacy and the underlying action mechanisms. Taken together, this study offers insights into how surface decoration can tune the cancer selectivity of SeNPs and provides a basis for engineering particles with increased anticancer efficacy.

33

ACS Paragon Plus Environment