Physicochemical Properties of Whey-Protein-Stabilized Astaxanthin

Jan 18, 2018 - ... University, Harbin, Heilongjiang 150030, People,s Republic of China. § ... 351 Carrigan Wing, Burlington, Vermont 05405, United St...
0 downloads 0 Views 756KB Size
Subscriber access provided by READING UNIV

Article

Physicochemical properties of whey protein-stabilized astaxanthin nanodispersion and its transport via Caco-2 monolayer Xue Shen, Changhui Zhao, Jing Lu, and Mingruo Guo J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.7b05284 • Publication Date (Web): 18 Jan 2018 Downloaded from http://pubs.acs.org on January 19, 2018

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 34

Journal of Agricultural and Food Chemistry

Physicochemical properties of whey protein-stabilized astaxanthin nanodispersion and its transport via Caco-2 monolayer

Xue Shena, Changhui Zhaoa, Jing Lua, Mingruo Guo*a b c a College of Food Science and Engineering, Jilin University, Changchun, 130062, China b Department of Food Science, Northeast Agricultural University, Harbin, 150030, China c Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington, Vermont, 05405, USA

* Corresponding author: Mingruo Guo E-mail: [email protected] Mailing address: University of Vermont, 109 Carrigan Drive, 351 Carrigan Wing, Burlington, VT 05405, USA Tel: (802) 656-8168 Fax: 802-656-0001

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

1

Page 2 of 34

ABSTRACT

2

Astaxanthin nanodispersion was prepared using whey protein isolate (WPI) and

3

polymerized whey protein (PWP) through an emulsification–evaporation technique.

4

The physicochemical properties of the astaxanthin nanodispersion were evaluated and

5

the transport of astaxanthin was assessed using a Caco-2 cell monolayer model. The

6

astaxanthin nanodispersions stabilized by WPI and PWP (2.5%, w/w) had a small

7

particle size (121 ± 4.9 and 80.4 ± 5.9 nm), negative zeta potential (-19.3 ± 1.5 and

8

-35.0 ± 2.2 mV), and high encapsulation efficiency (92.1 ± 2.9% and 93.5 ± 2.4%).

9

Differential scanning calorimetry (DSC) curves indicated that amorphous astaxanthin

10

existed in both astaxanthin nanodispersions. Whey protein-stabilized astaxanthin

11

nanodispersion showed resistance to pepsin digestion but readily released astaxanthin

12

after trypsin digestion. The nanodispersions showed no cytotoxicity to Caco-2 cells at

13

a protein concentration below 10 mg/mL. WPI and PWP stabilized nanodispersions

14

improved the apparent permeability coefficient (Papp) of Caco-2 cells to astaxanthin

15

by 10.3- and 16.1- fold, respectively. The results indicated that whey

16

protein-stabilized nanodispersion is a good vehicle to deliver lipophilic bioactive

17

compounds like astaxanthin and to improve their bioavailability.

18 19

Keywords: Astaxanthin;

20

Bioavailability

Whey

protein;

Nanodispersion;

21 22 2

ACS Paragon Plus Environment

Caco-2

cells;

Page 3 of 34

Journal of Agricultural and Food Chemistry

23

Introduction

24

Astaxanthin is a xanthophyll carotenoid that has antioxidant activity (1) and can

25

be used to reduce the risk of cancer (2), cardiovascular diseases (3), Helicobacter

26

pylori infections (4), and age-related diseases (5-8). Astaxanthin cannot be

27

synthesized de novo by mammals and thus must be acquired from the diet (4).

28

Utilization of carotenoids as nutraceutical ingredients for foods is therefore

29

recommended (8). Unfortunately, the applications of astaxanthin in different food

30

formulations are currently limited because of its poor water solubility, high melting

31

point, low bioavailability, and susceptibility to chemical degradation under certain

32

conditions, such as an acidic environment, heating, and exposure to light and oxygen

33

(9).

34

To use astaxanthin in functional foods, different strategies have been developed to

35

improve its stability and bioavailability in foodstuffs, including microencapsulation

36

(10, 11), liposomes (12), emulsions (13), and nanodispersions (14). The

37

nanodispersion system has attracted much attention for increasing the bioavailability

38

of lipophilic bioactive agents in water-based food products due to its special features,

39

including very small particle size (20 to 200 nm), relatively high physical stability,

40

enhanced chemical stability and improved water solubility. Nanodispersions also have

41

thermodynamic stability and optical transparency, which is particularly useful for

42

transparent beverages (15, 16). Stabilizers can affect the physicochemical and

43

bioavailability properties of nanodispersions depending on the type and nature of the

44

stabilizers (17). Various stabilizing agents have been used to prepare astaxanthin 3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

45

nanodispersions including gelatin (18), polysaccharides (19), Tween 20 (16), sodium

46

caseinate (20, 21), and mixture stabilizers (22-24). Anarjan et al. reported that

47

astaxanthin was more bioavailable when administered with protein or dietary fat

48

ingredients such as milk (14). Therefore, milk protein should be one of the best

49

candidates to stabilize astaxanthin nanodispersion.

50

Whey protein is a valuable byproduct of cheese making. In addition to their

51

desirable nutritional quality, whey proteins are amphiphilic molecules with high

52

surface activity and emulsifying properties. Whey proteins can form a protective layer

53

around fat droplets and protect them from aggregation or coalescence due to a

54

combination of electrostatic effects and hydrophobic interactions. Another advantage

55

of whey protein as an emulsifier or stabilizer is its antioxidant activity (25). The

56

emulsifying properties of WPI can be increased by polymerized whey protein soluble

57

aggregates also called PWP (26). However, information about WPI and PWP used as

58

emulsifiers to stabilize astaxanthin nanodispersions is very limited.

59

Therefore, the objective of this study was to prepare astaxanthin

60

nanodispersions using an emulsification–evaporation method with WPI and/or PWP

61

as stabilizers. The astaxanthin nanodispersions were characterized by their particle

62

size distribution, zeta potential, encapsulation efficiency, flow behavior, viscosity, and

63

thermal properties. Caco-2 cells were used to evaluate the cytotoxicity and

64

bioavailability of the whey protein-astaxanthin nanodispersions.

65

Materials and methods

66

Chemicals 4

ACS Paragon Plus Environment

Page 4 of 34

Page 5 of 34

Journal of Agricultural and Food Chemistry

67

WPI, containing 93.1% protein, 0.36% fat, 4.79% moisture, 1.60% ash and

68

0.70% lactose, was purchased from Fonterra (Auckland, New Zealand).

69

Dimethylsulfoxide (DMSO) was purchased from Beijing Solarbio Science and

70

Technology Co., (China). Astaxanthin and its standard were purchased from Sigma

71

(St. Louis, MO, USA). Methanol, methyl tert-butyl ether (MTBE), acetone, and

72

dichloromethane were HPLC grade and purchased from Thermo Fisher Scientific Co.

73

(Waltham, MA, USA). High-glucose and L-glutamine Dulbecco's Modified Eagle

74

Medium (DMEM) and fetal bovine serum (FBS) were purchased from Thermo Fisher

75

Scientific Co. (Waltham, MA, USA). All other chemicals used were of reagent grade

76

and purchased through Sigma (St. Louis, MO, USA). Purified water used in this study

77

was filtered using a Millipore Milli-Q™ water purification system (Millipore Corp.,

78

Milford, MA, USA).

79

Preparation of astaxanthin nanodispersions

80

Astaxanthin (25 mg) was dissolved in 100 mL acetone/ dichloromethane (3:1, v/v)

81

at room temperature as an organic phase. As previously reported, 0.10, 0.25, 0.50, 1.0,

82

2.5, and 5.0% (w/w) WPI solution and PWP solution (85°C for 20 min) was prepared

83

(26). Sodium azide (0.022%, w/w) was added to the aqueous phase to inhibit the

84

growth of microorganisms. The organic phase and aqueous phase were mixed at ratio

85

of 1:9 (v/v), and a coarse emulsion was formed using an Ultra-Turrax T25 high-speed

86

blender (IKA, Staufen, Germany) at 12,000 rpm for 2 min. The coarse emulsion was

87

then homogenized through an ultrasonic processor (VCX800, Vibra-Cell, Sonics,

88

USA) with a 13-mm high grade titanium alloy probe in an ice bath at 40% amplitude 5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

89

for 2 min (10 s:5 s work/rest cycles). The solvent was subsequently removed from the

90

emulsion by rotary evaporation (EYELA N-1100,Tokyo, Japan) at 37°C and 100 rpm

91

(27). The container was wrapped in foil to keep astaxanthin protected from light

92

during the treatment.

93

Determination of particle size and zeta potential

94

The mean particle size and zeta-potential (ζ) of nanodispersions were determined

95

by dynamic light scattering (DLS) and electrophoretic mobility (UE) using laser

96

Doppler velocimetry and phase analysis light scattering by a Zetasizer Nano ZS 90

97

(Malvern Instruments, UK). The samples were diluted 100-fold using deionized water.

98

Then 1 mL of diluted samples was transferred into the measuring cell. The refractive

99

index (RI) values for oil droplets and water were 1.45 and 1.33, respectively. Particle

100

size values were reported as Z-average (Dz), which is the intensity-weighted mean

101

hydrodynamic size of the particles. ζ was calculated based on the Henry equation. All

102

measurements were conducted at 25°C.

103

Determination of rheological property

104

Rheological property analyses were performed using a rheometer (DHR-1, TA

105

Instrument, USA). A steel parallel plate geometry (diameter = 40 mm, sample,

106

thickness = 1 mm) was used. The sample temperature was controlled by a Peltier unit

107

attached to a water circulation system.

108 109 110

Flow ramp analyses were performed for shear rate from 0.1 to 1000 s-1 at 25°C. Apparent viscosity was recorded as a function of shear rate. Peak hold analyses were performed at 200 s-1 for 60 s at 25°C. 6

ACS Paragon Plus Environment

Page 6 of 34

Page 7 of 34

Journal of Agricultural and Food Chemistry

111

Differential Scanning Calorimetry

112

The thermal properties of the samples were evaluated using a Differential

113

Scanning Calorimeter (Q2000, TA Instrument, USA). Approximately 5 mg of

114

freeze-dried samples were placed in aluminum pans and weighed accurately, and then

115

the pans were sealed. The temperature was programmed to rise from 25 to 250°C at

116

5°C /min. An empty pan was used as a reference. The TA Universal Analysis 2000

117

was used to determine the melting temperature, peak temperature, and enthalpy (∆H).

118

Release of astaxanthin in vitro

119

Freeze-dried samples (2 mg/ml) were added to simulated gastric fluid (SGF, 3.2

120

mg/ml pepsin in 0.035 M NaCl, 0.084 N HCl, pH 1.2) and simulated intestinal fluid

121

(SIF, 1.0 mg/ml trypsin in 0.1 M PBS, pH 7.0), respectively. This mixture was

122

incubated in a glass tube at 37oC in a shaking water bath. An aliquot of reactant was

123

removed, and the hydrolysis was terminated by adjusting the pH at set intervals

124

during 120 min of digestion time. The astaxanthin content was determined by the

125

HPLC method described below.

126

Extraction and determination of astaxanthin in nanodispersions

127

Astaxanthin was extracted from nanodispersions using methanol and

128

dichloromethane as described (14) with some modifications. Briefly, 0.5 mL of

129

sample was mixed with 2 mL methanol/dichloromethane (1:1, v/v), vortexed for 10

130

min, and then centrifuged at 1000 g for 10 min. The upper supernatant containing

131

astaxanthin was transferred to a 10-mL brown volumetric flask. The extraction was

132

repeated with 1 mL dichloromethane three times until the aqueous layer was clear. 7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

133

The extracts were brought up to 10 mL. The encapsulation efficiency of astaxanthin

134

was calculated by comparing the astaxanthin content encapsulated in nanodispersions

135

with that of the total content added.

136

High Performance Liquid Chromatography (HPLC)

137

Astaxanthin analysis was performed using an UHPLC system (Nexera UHPLC

138

LC-30A, Shimadzu, Japan) with a PDA UV-vis absorption detector and a C30

139

reverse-phase analytical column (250 × 4.6 mm i.d., 5µm, YMC, Co., LTD). The flow

140

rate was set at 1.0 mL/min at room temperature. The injection volume was 20 µL, and

141

the detection wavelength was 474 nm. The chromatography conditions were as

142

follows: solvent A, methanol; solvent B, MTBE; solvent C, phosphoric acid/H2O

143

(1:99, v/v). The solvent gradient program was 81% A/15% B/4% C at 0 min and was

144

changed linearly to 66% A/30% B/4% C at 5 min, maintained for 10 min and was

145

subsequently changed linearly to 16% A/80% B/4% C at 16-23 min, maintained for 4

146

min, and followed by a linear return to 81% A/15% B/4% C at 28-30 min, maintained

147

for 5 min. The calibration of peak area versus the astaxanthin standard concentration

148

was linear in the measured concentration ranging from 0.1 to 50 µg/mL for all-trans

149

astaxanthin (R2 > 0.99, n=3).

150

Cytotoxicity of astaxanthin nanodispersions on Caco-2 Cells

151

To determine the cell viability after incubation with astaxanthin nanodispersions,

152

a methyl thiazolyl tetrazolium (MTT) assay was performed using a Caco-2 cell line

153

(passages 30-40). Caco-2 cells were incubated in high-glucose and L-glutamine

154

DMEM medium supplemented with 1% (v/v) penicillin/streptomycin and 10% (v/v) 8

ACS Paragon Plus Environment

Page 8 of 34

Page 9 of 34

Journal of Agricultural and Food Chemistry

155

FBS. The cells were seeded at a density of 2.5 × 104 cells/well in 96-well plates and

156

incubated at 37°C and 5% CO2 for 72 h. Culture medium was removed, and

157

astaxanthin nanodispersions diluted with cell culture medium at different protein

158

concentrations (1.0 - 10.0 mg/mL) were added to the wells. DMEM was used as a

159

control. After 24 h of incubation, the cells were washed with 200 µL PBS/well three

160

times. Then, 200 µL MTT containing medium (5 mg/mL MTT in DMEM) was added

161

to each well. After 4 h of incubation, the medium was removed and 150 µL DMSO

162

was added to dissolve the formed formazan crystals. Absorbance measurement was

163

performed at 570 nm using a microplate reader (Synergy HT, BioTek, USA). Relative

164

cell viability (%) was calculated by comparing the absorbance of nanoparticle cells

165

with that of control cells (28).

166

Caco-2 cell monolayer incubation

167

Caco-2 cells were seeded at a density of 1.5 × 106 cells/well onto polyethylene

168

terephthalate (PET) filters with a 0.4-µm pore size (Corning 6-well transwell, Corning,

169

Massachusetts, USA) for 21 days to achieve a differentiated intestinal cell monolayer.

170

Both the transepithelial electrical resistance (TEER) value and the phenol red flux (29,

171

30) were measured to ensure the monolayer integrity.

172

Astaxanthin transport via Caco-2 monolayers

173

The differentiated Caco-2 monolayers were washed with Hanks buffer three

174

times. Both astaxanthin nanodispersions and free astaxanthin (in DMSO) were diluted

175

with DMEM to a final concentration of 12.5 µg/mL and added to the chambers. The

176

transportation experiments were performed at 37°C. Next, 1 mL of sample was added 9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 34

177

to the apical side and 2.5 mL DMEM was added to basal side, which was sampled at

178

intervals (0.5, 1, 1.5, 2, 3, and 4 h), extracted and analyzed for astaxanthin content by

179

HPLC. The TEER (0, 0.5, 1, 2, 3, and 4 h) and phenol red flux (0, 4 h) were also

180

measured to evaluate the integrity of Caco-2 monolayers. The apparent permeability

181

coefficient (Papp, cm s−1) was calculated as follows (31): Papp =

182

dQ 1 · dt AC0

183

where dQ/dt = the permeability rate (µg s−1), A= the surface area of the filter (4.67

184

cm-2), C0= the initial concentration in the chamber (µg mL−1).

185

Statistical analyses

186

All experiments were performed in triplicate. Statistical analyses were performed

187

using the statistical program SPSS Version 17.0 SPSS (SPSS Inc. Chicago, IL, USA).

188

Comparisons among data of different groups were performed with one-way ANOVA,

189

where the LSD method and Dunnet’s C were used on the basis of the homogeneity

190

test. The results were presented as the mean ± standard deviation (SD) and considered

191

significantly different when P