Bioactive Peptides Isolated from Casein Phosphopeptides Enhance

Feb 20, 2017 - College of Food Science, South China Agricultural University, Guangzhou 510642, People,s Republic of China. ‡. Department of Food Sci...
0 downloads 0 Views 636KB Size
Subscriber access provided by University of Newcastle, Australia

Article

Bioactive Peptides Isolated from Casein Phosphopeptides Enhance Calcium and Magnesium Uptake in Caco-2 Cell Monolayers Yong Cao, Jian-yin Miao, Guo Liu, Zhen Luo, Zumeng Xia, Fei Liu, Mingfei Yao, Xiaoqiong Cao, Shengwei Sun, Yanyin Lin, Yaqi Lan, and Hang Xiao J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.6b05711 • Publication Date (Web): 20 Feb 2017 Downloaded from http://pubs.acs.org on February 23, 2017

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

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

Page 1 of 32

Journal of Agricultural and Food Chemistry

TITLE RUNNING HEADER Bioactive Peptides Enhancing Calcium and Magnesium Transport

Bioactive Peptides Isolated from Casein Phosphopeptides Enhance Calcium and Magnesium Uptake in Caco-2 Cell Monolayers Yong Cao†*, Jianyin Miao†,‡, Guo Liu†, Zhen Luo§, Zumeng Xia§, Fei Liu¶, Mingfei Yao ‡, Xiaoqiong Cao ‡, Shengwei Sun†, Yanyin Lin†, Yaqi Lan#, †, Hang Xiao‡*

†College of Food Science, South China Agricultural University, Guangzhou 510642, People’s Republic of China ‡Department of Food Science, University of Massachusetts, Amherst, MA 01003, United States §Infinitus (China) Company Ltd ¶Guangzhou Greencream Biotech Co., Ltd # Department of Food Science, Rutgers University, 65 Dudley Road, New Brunswick, NJ 08901, USA

Corresponding Authors: *Tel.: +86 020 85286234. Fax: +86 020 85286234. Email:[email protected] (Y. Cao) *Tel.: +1 413 545 2281; fax: +1 413 545 1262. Email: [email protected] (H. Xiao)

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

1

ABSTRACT:The ability of casein phosphopeptides (CPPs) to bind and transport

2

minerals has been previously studied. However, the single bioactive peptides

3

responsible for the effects of CPPs have not been identified. This study was to purify

4

calcium-binding peptides from CPPs and to determine their effects on calcium and

5

magnesium uptake by Caco-2 cell monolayers. Five monomer peptides designated P1

6

to P5 were isolated and the amino acid sequences were determined using LC-MS/MS.

7

Compared with the CPP-free control, all five monomeric peptides exhibited

8

significant enhancing effects on the uptake of calcium and magnesium (P < 0.05).

9

Interestingly, when calcium and magnesium were presented simultaneously with P5,

10

magnesium was uptaken with priority over calcium in the Caco-2 cell monolayers.

11

For example, at 180 min, the amount of transferred magnesium and calcium was 78.4

12

±0.95 μg/well and 2.56 ±0.64 μg/well, respectively, showing a more than 30-fold

13

difference in the amount of transport caused by P5. These results provide novel

14

insight into the mineral transport activity of phosphopeptides obtained from casein.

15

KEYWORDS: casein phosphopeptides, identification, calcium transport,

16

magnesium transport, Caco-2 cell monolayers.

17

18

19

20

21

22

2

ACS Paragon Plus Environment

Page 2 of 32

Page 3 of 32

23

Journal of Agricultural and Food Chemistry

INTRODUCTION Calcium and magnesium are crucial elements to maintaining human health,1

24 25

particularly in aging individuals. Given the current and future size of global elderly

26

populations, research on calcium and magnesium supplements has been an area with

27

increasing interest.2, 3 Calcium accounts for about 2 % of adult human body weight. 4,

28

5

29

and osteoporosis. 6 Magnesium is also a necessary component for proper bone health

30

and plays an important role in the physiological functioning of the brain, heart, and

31

skeletal muscles. 3 However, approximately 60% of all Americans receive less than

32

the recommended daily amount of magnesium. 3

33

The effects of calcium deficiency on health include increased risks of bone fracture

Currently, calcium, magnesium, and other elemental mineral supplements are

34

mainly composed of inorganic ionic forms. 7 However, numerous studies have

35

focused on using organic supplements due to their increased bioavailability. 8-10 To

36

this end, many organic supplements improve the absorptive ability of mineral

37

elements and include oligophosphopeptide, 11 chicken eggshell matrix proteins, 12

38

tilapia protein hydrolysate, 13 whey protein hydrolysates, 14 hen egg yolk phosvitin 15

39

and desalted duck egg white peptides. 16

40

Casein phosphopeptides (CPPs) are derived from trypsin-mediated casein cleavage

41

and are considered mineral carriers with potential roles in improving elemental

42

mineral absorption. 17 Several factors affect the activity and mechanisms of mineral

43

uptake of CPPs. These include what phosphate residues are present on serine, 15 amino

44

acid composition 18, 19 and molecular structures. 16 The effects of CPP-mediated

45

calcium absorption have been in-depth studied by previous researchers and obtained

46

some significant results on the activity and mechanisms. 20, 21 However, the effects of

47

CPPs on regulating magnesium absorption are few reported. At the same time, though 3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

48

the peptide mixture in CPPs has the effects of mineral transport, the activity and

49

mechanisms of single peptide monomer may be different. It is significant to study the

50

pure mineral element transport active peptide that has been isolated from the CPPs.

51

With this in mind, the work presented here sought to purify and identify specific

52

CPP-derived peptides that exhibited excellent calcium transport capacity. Additionally,

53

magnesium transport capacity and the difference in transport activity between calcium

54

and magnesium was also investigated. Thus, this study allowed for the further

55

exploration of the exact activity and mechanism of action of CPP-derived calcium and

56

magnesium transport peptides.

57

MATERIALS AND METHODS

58

Materials. A commercial CPP mixture was provided by Guangzhou Greencream

59

Biotech Co., Ltd (20% CPPs, Guangzhou, China) was used.

60

Chemicals. Cell culture media, fetal bovine serum (FBS), antibiotic, Hank’s balanced

61

salt solution (HBSS, without calcium and magnesium), and nonessential amino acids

62

and all other chemicals were purchased from Sigma (St. Louis, MO, USA).

63

Calcium-binding assay. The final concentration of 1.0 mg/mL of lyophilized sample

64

dissolved in deionized water was mixed with 5 mM CaCl2 in 0.2 M sodium phosphate

65

buffer (pH 8.0). The solution was stirred at 37 °C for 2 h and the pH was maintained

66

at 8.0. The reaction mixture was centrifuged at 10,000×g at room temperature for 10

67

min. The calcium content of the supernatant was determined using inductively

68

coupled plasma mass spectrometry (ICP-MS) (NexION 300X ICP, MA, USA). 22

69

Purification of active peptides. CPPs were first isolated by preparative

70

high-performance liquid chromatography (LC-8, Shimadzu, Japan) with a 4

ACS Paragon Plus Environment

Page 4 of 32

Page 5 of 32

Journal of Agricultural and Food Chemistry

71

reversed-phase (RP) column (20 mm × 450 mm) of C18 (10 μm, 300 Å, Macherey

72

Nagel, France). Solvent A was 0.1% trifluoroacetic acid in double-distilled water, and

73

solvent B was 100% acetonitrile. A linear gradient of solvent B at a flow rate of 10

74

mL/min for 55 min was applied as follows: 10%–20% of solvent B for 10 min and

75

20%-26% of solvent B for 45 min. Fractions were collected, concentrated, and tested

76

using a calcium-binding assay. The active fractions were then further purified using

77

HPLC in order to search for monomeric peptides that had an increased calcium

78

transport ability. The active fraction solution collected in the first step was filtered

79

through a 0.45-μm membrane filter and separated using a Diamosil C18 column (5

80

μm, 4.6 mm × 250 mm) (Dikma Technologies, Beijing, China). The mobile phases

81

were solvent A (double-distilled water with 0.1% trifluoroacetic acid) and solvent B

82

(acetonitrile with 0.1% trifluoroacetic acid). Elution was performed using a gradient

83

of 5-18% of solvent B (0-19.5 min) then 18-30% of solvent B (19.5-55 min)

84

at a flow rate of 1 mL/min. The main peaks were collected, concentrated, and

85

lyophilized. All elutions were monitored at 215 nm. Calcium transport activity of the

86

eluted peaks was tested using the Caco-2 cell model.

87

Transport studies in Caco-2 cell monolayer. Cell culture. Caco-2 cell lines (passage

88

50−60) were the model for the intestinal epithelium that was used in the calcium

89

transport experiments. Cells were cultured in complete Dulbecco’s modified essential

90

medium (DMEM) with 10% fetal bovine serum (FBS), 1% antibiotic, and 1%

91

nonessential amino acids as we described previously. 23 Cells were seeded at a density

92

of 3 × 105 cells/mL on transwell culture plates (Corning Inc., MA, USA) with a

93

polycarbonate membrane (24 mm diameter inserts, 0.4 μm pore size). The culture

94

medium was added to both the basolateral (3.5 mL) and to the apical sides (2.5 mL).

95

The medium was changed every other day for three weeks. 5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

96

Calcium transport studies. Before the calcium transport experiments began,

97

transepithelial electrical resistance (TEER) was measured using a Millicell ERS-2

98

epithelial voltammeter (World Precision Instruments, FL, USA). TEER values of the

99

cell monolayers were about 260 Ω/cm2. 23 Culture medium was discarded and

100

monolayers were immediately washed twice with Hank’s balanced salt solution

101

(HBSS, without calcium and magnesium). Monolayers were then transferred to a new

102

cluster plate containing 2 mL of HBSS buffer. Prior to the start of the calcium

103

transport experiments, the apical side was also added to 2 mL of HBSS buffer and

104

incubated for 30 min at 37 °C with 5% CO2. After 30 min of incubation, the CPPs

105

(300 µg/well) and the HPLC-isolated peaks (P1 to P5, 100 µg/well each) were added

106

to the apical sides in a calcium solution (300 µg/well). The samples and calcium

107

solution were pre-mixed before the transport studies began. CPP-free controls had

108

only the addition of a calcium solution (300 µg/well) and no CPP or HPLC-isolated

109

peaks. At various time periods (20, 40, 60, 90, 120, 180, and 240 min), 1 mL of HBSS

110

buffer was collected from the basolateral side to measure calcium ion concentration.

111

One mL of fresh HBSS buffer was simultaneously added to the basolateral side to

112

keep a constant volume.

113

Resulting calcium concentrations at each time point were determined using

114

ICP-MS (NexION 300X ICP, MA, USA). 22 The total quantity of calcium transported

115

to the basolateral side of each well was calculated according to the following formula:

116

B n  2  An  1 

n 1

 Ak k

(1)

1

117

in which Bn represents the total quantity of transported calcium in 2-mL of HBSS

118

buffer in the basolateral side of each well at each selected time point (20, 40, 60, 90,

119

120, 180, and 240 min) (unit: μg/well). Two is a constant, representing 2-mL of HBSS

120

buffer in the basolateral side of each well. An represents the calcium ion concentration 6

ACS Paragon Plus Environment

Page 6 of 32

Page 7 of 32

Journal of Agricultural and Food Chemistry

121

of the HBSS buffer in the basolateral side of each well at different time points,

122

measured by ICP-MS (unit: μg/mL). One is also a constant and represents the 1-mL of

123

HBSS buffer collected from the basolateral side of each well in order to measure the

124

calcium ion concentration. n is an independent variable. For the present study, it could

125

be the number of 1, 2, 3, 4, 5, 6, or 7, representing time points 20, 40, 60, 90, 120, 180,

126

and 240 min, respectively).

127 128 129

All assays were carried out in triplicate and the results were expressed as mean values ±standard deviation (SD). Magnesium transport studies. CPPs (300 µg/well) and the HPLC-isolated peaks

130

(P1 to P5, 100 µg/well each) were added to the apical sides using a magnesium

131

solution (150 µg/well). The samples and magnesium solution were pre-mixed before

132

the transport studies began. CPP-free controls had just the magnesium solution (150

133

µg/well) added. All other conditions (e.g. time points) and analysis (e.g. Equation 1)

134

in the magnesium transport studies were the same as those described in the calcium

135

transport studies.

136

Calcium and magnesium transport studies. CPPs (300 µg/well) and the

137

HPLC-isolated peaks (P1 to P5, 100 µg/well each) were each added to the apical sides

138

in a solution containing both calcium and magnesium (calcium: 300 µg/well,

139

magnesium: 150 µg/well). The samples as well as the combined calcium and

140

magnesium solution were pre-mixed before the transport studies began. CPP-free

141

controls had just the combined calcium and magnesium solution (calcium: 300

142

µg/well, magnesium: 150 µg/well) added. All other conditions and analysis in the

143

combined calcium and magnesium transport studies were the same as those in the

144

calcium transport studies.

145

Amino acid sequence analysis of active peptides. The amino acid sequence of the 7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

146

active peptides that displaying calcium transport activity were analyzed using

147

nanospray LC-MS/MS with an HPLC instrument coupled to an LTQ-Velos-Orbitrap

148

mass spectrometer. The active peptides were solubilized in 0.1% TFA, then 1 μL

149

solution was loaded on the nano-LC column. The data were acquired using

150

LTQ-Velos-Orbitrap mass spectrometry. Peak lists were generated using Proteome

151

Discoverer software. The resulting data were searched against Swiss-Prot database

152

using Mascot 2.2. 24

153

Statistical Analysis. All experiments were carried out at least three times and the data

154

are expressed as means ± standard deviations (SD). The differences among samples

155

were analyzed by ANOVA with a significance level of p< 0.05.

156

RESULTS AND DISCUSSION

157

Purification of fractions with calcium binding activity. Several different

158

purification procedures have been reported for calcium transport peptides. For

159

instance, a novel oligophosphopeptide with high calcium binding activity was

160

obtained from carp egg hydrolysate. This was achieved by way of a three-step

161

purification that consisted of ultrafiltration, size exclusion chromatography, and

162

high-performance liquid chromatography. 11 In another instance, Chen et al. purified a

163

calcium-binding peptide from tilapia scale protein hydrolysate by hydroxylapatite

164

affinity, gel filtration and RP-HPLC. 8 In a third example, a histidine-containing

165

peptide was initially fractionated by an ultrafiltration membrane, then purified using

166

ion-exchange chromatography, gel filtration, and RP-HPLC. 25 Our study featured

167

active fractions capable of calcium transport that were then separated through a

168

two-step purification process.

169

In the first step, CPP fractions with high calcium binding activity were collected 8

ACS Paragon Plus Environment

Page 8 of 32

Page 9 of 32

Journal of Agricultural and Food Chemistry

170

between 20 and 55 min and were termed F1, F2, and F3 (Figure 1). The collected

171

fractions between 20 and 55 min were further separated into different fractions

172

(Figure 2). The numerous peaks in Figure 2 suggested that there were more than ten

173

different calcium-binding peptides present in the sample. The five main peaks were

174

designated as P1 to P5. They were subsequently collected and lyophilized prior to

175

activity analysis of calcium and magnesium transport (Figure 2).

176

Calcium transport studies in Caco-2 cell monolayer. Caco-2 cells have been

177

previously used as an effective model of human intestinal epithelial cells in vitro.

178

They have been successfully applied to simulative absorption studies involving drugs,

179

26, 27

180

activity (300 µg/well) and the HPLC-isolated peaks (100 µg/well each) were

181

evaluated using Caco-2 cell monolayers. The resulting effects on calcium transport

182

are shown in Figure 3. CPPs showed significant calcium transport activity across all

183

Caco-2 monolayers when compared with CPP-free control across all values from the

184

20 to 240 min time points (P