Contribution of the Individual Small Intestinal α-Glucosidases to

Aug 2, 2016 - Department of Biology, University of Waterloo, Waterloo, Ontario ... Universidad Autonoma de San Luis Potosi, San Luis Potosi 78360, Mex...
0 downloads 0 Views 1MB Size
Subscriber access provided by CORNELL UNIVERSITY LIBRARY

Article

Contribution of the Individual Small Intestinal #-Glucosidases on Digestion of Unusual #-Linked Glycemic Disaccharides Byung-Hoo Lee, David R Rose, Amy Hui-Mei Lin, Roberto Quezada-Calvillo, Buford L. Nichols, and Bruce R. Hamaker J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.6b01816 • Publication Date (Web): 02 Aug 2016 Downloaded from http://pubs.acs.org on August 5, 2016

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 27

Journal of Agricultural and Food Chemistry

Contribution of the Individual Small Intestinal α-Glucosidases on Digestion of Unusual αLinked Glycemic Disaccharides

Byung-Hoo Lee,†, § David R. Rose,‡ Amy Hui-Mei Lin,§,# Roberto Quezada-Calvillo,∆ Buford L. Nichols,⊥ Bruce R. Hamaker§,∥,*



Department of Food Science & Biotechnology, College of BioNano Technology, Gachon

University, Seongnam, Gyeonggi-do 13120, Republic of Korea §

Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University,

West Lafayette, IN 47907, USA ‡

Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada

#

Bi-State School of Food Science, University of Idaho and Washington State University,

Moscow, ID 83844, USA ∆

Department of Chemistry, Universidad Autonoma de San Luis Potosi, SLP 78360, Mexico



USDA, Agricultural Research Service, Children’s Nutrition Research Center and Department

of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA ∥

Department of Food Science & Technology, Sejong University, Gunja-Dong, Gwangjin-Gu,

Seoul 05006, Republic of Korea *

Corresponding Author:

Tel: +1-765- 494-5668; Fax: +1-765-494-7953 E-mail: [email protected]

1 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

1

2

Page 2 of 27

ABSTRACT The mammalian mucosal α-glucosidase complexes, maltase-glucoamylase (MGAM) and

3

sucrase-isomaltase (SI), have two catalytic subunits (N- and C-terminals). Concurrent with the

4

desire to modulated glycemic response, there has been a focus on di-/oligo-saccharides with

5

unusual α-linkages that are digested to glucose slowly by these enzymes. Here, we look at

6

disaccharides with various possible α-linkages and their hydrolysis. Hydrolytic properties of the

7

maltose and sucrose isomers were determined using rat intestinal and individual recombinant α-

8

glucosidases. The individual α-glucosidases had moderate to low hydrolytic activities on all α-

9

linked disaccharides, except trehalose. Maltase (N-terminal MGAM) showed a higher ability to

10

digest α-1,2 and -1,3 disaccharides, as well as α-1,4, making it the most versatile in α-hydrolytic

11

activity. These findings apply to the development of new glycemic oligosaccharides based on

12

unusual α-linkages for extended glycemic response. It also emphasizes that mammalian mucosal

13

α-glucosidases must be used in in vitro assessment of digestion of such carbohydrates.

14

Keywords: α-glucosidases, carbohydrate digestion, disaccharides, glycemic, slowly digestible carbohydrates

15 16

17

2 ACS Paragon Plus Environment

Page 3 of 27

18 19

Journal of Agricultural and Food Chemistry

INTRODUCTION Digestible carbohydrate-based diets such as those containing starch or sucrose depend

20

upon the activities of small intestinal mucosal α-glucosidases to produce glucose and fructose.

21

After hydrolysis, the released monosaccharides are absorbed into the blood stream via the

22

sodium-dependent glucose transporter 1 (SGLT1) and glucose transporter 2 (GLUT2) for glucose

23

and glucose transporter 5 (GLUT 5) for fructose. The absorbed monosaccharides are utilized as

24

carbon and energy sources.1-3 The mucosal α-glucosidases consist of two complexes: maltase-

25

glucoamylase [MGAM; EC 3.2.1.20 for maltase (N-terminal MGAM, or ntMGAM) and EC

26

3.2.1.3 for glucoamylase (C-terminal MGAM, or ctMGAM)] and sucrase-isomaltase [SI; EC

27

3.2.1.48 for sucrase (ctSI) and EC 3.2.1.10 for isomaltase (ntSI)]. Both MGAM and SI consist of

28

two catalytic enzymes: the C-terminal luminal domains (ctMGAM and ctSI) and N-terminal

29

membrane-proximal domains (ntMGAM and ntSI). The N-terminal α-glucosidases are connected

30

to the brush-border membrane via an O-glycosylated linker in the small intestine (Figure 1).4, 5

31

Based on the carbohydrate-active enzymes (CAZY) classification,6 mucosal α-glucosidases are

32

classified as Glycoside Hydrolase Family 31 (GH31) enzymes due to structural and functional

33

similarities of their proteins. Furthermore, all four individual mucosal α-glucosidases include the

34

signature amino acid sequences “WIDMNE” in the catalytic site, and share 40-60% protein

35

sequence identity.7-9

36

While all four enzymes have α-1,4 hydrolytic activity, the individual α-glucosidases also

37

exhibit a variety of specific α-hydrolysis properties on different substrates.10, 11 MGAM has high

38

α-1,4 hydrolytic activity for digestion of maltose and larger starch-based oligomers and

39

polymers.12-14 The SI complex accounts for the majority of mucosal maltase activity, because the

40

amount of SI protein is 40-50 times higher than MGAM in the human intestine.12, 13 The

3 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 27

41

hydrolytic activity of ctMGAM, unlike the other subunits, is inhibited during α-1,4 linkage

42

hydrolysis by concentration of its products, termed the “brake” effect.15-17 Both N-terminal α-

43

glucosidases (ntMGAM and ntSI) have α-1,6 hydrolytic activity with ntSI having much higher

44

hydrolytic activity than ntMGAM.18 Using powdered rat intestine (containing both MGAM and

45

SI α-glucosidase complexes), it was previously shown that kojibiose (α-1,2) and nigerose (α-1,3)

46

were digested at about 65 and 86% of maltose hydrolysis.19, 20 Prior to that study using pig

47

intestinal MGAM complex showed hydrolysis of all types of α-glycosidic linkages between two

48

glucose molecules, except trehalose.10

49

Low glycemic index foods are associated with reduced risk of common chronic health

50

diseases,21 and this has led to interest in glycemic carbohydrates with slow digestion rate for

51

modulated postprandial glycemic response. Apart from efforts to understand how to slow starch

52

digestion, unusual α-linked carbohydrates, such as isomaltulose (α-1,6 sucrose isomer) and

53

sucromalt (α-glucan containing α-1,3 and α-1,6 linkages) have been commercially developed as

54

slowly digestible carbohydrates.22, 23 Even so, it is still difficult to devise precise strategies to

55

produce specific slowly digestible carbohydrates based on linkage type and monosaccharide

56

composition. In this research, digestion properties of a range of disaccharides of different linkage

57

and monosaccharide composition were investigated using rat intestinal α-glucosidases and the

58

four individual recombinant mucosal α-glucosidases with the aim of finding new ways to control

59

the rate of glucogenesis from added dietary carbohydrates.

60 61

MATERIALS AND METHODS

62

Materials. Trehalose (1-O-(α-D-glucopyranosyl)-α-D-glucopyranose), kojibiose (2-O-(α-D-

63

glucopyranosyl)-D-glucose), nigerose (3-O-(α-D-glucopyranosyl)-D-glucose), maltose (4-O-(α-D-

4 ACS Paragon Plus Environment

Page 5 of 27

Journal of Agricultural and Food Chemistry

64

glucopyranosyl)-D-glucose), and isomaltose (6-O-(α-D-glucopyranosyl)-D-glucose) were used as

65

substrates (Sigma-Aldrich Co., St. Louis, MO) to test the hydrolysis properties of α-glycosidic

66

linkages between two glucose molecules. Sucrose (α-D-glucopyranosyl-(1,2)-β-D-

67

fructofuranose), turanose (3-O-(α-D-glucopyranosyl)-D-fructose), maltulose (4-O-(α-D-

68

glucopyranosyl)-D-fructose), leucrose (5-O-(α-D-glucopyranosyl)-D-fructose), and isomaltulose

69

(6-O-(α-D-glucopyranosyl)-D-fructose) were used to study the α-hydrolytic properties on sucrose

70

isomers (Carbosynth, Berkshire, UK). The reagents for the glucose assay were purchased from

71

Megazyme (Wicklow, Ireland). Other chemicals were purchased from Sigma-Aldrich Co. (St.

72

Louis, MO).

73 74

Optimal pH range of rat intestinal α-glucosidases on different substrates. To test optimal pH

75

range of mucosal α-glucosidases on different substrates, rat intestinal powder (Sigma-Aldrich

76

Co., St. Louis, MO) was used as a source of mucosal α-glucosidases. Rat intestinal powder (2 g)

77

was mixed with 20 mL of sodium phosphate buffer (100 mM, pH 6.9) and left at 4°C for 24 h.

78

The mixture was centrifuged at 9000 × g for 15 min, and the supernatant was filtered through a

79

0.8 µm Nylon membrane.24 The filtered extract was applied as a crude α-glucosidase solution.

80

Maltodextrin DE 1 (dextrose equivalent), maltose, sucrose, and isomaltulose were applied as

81

substrates (10 mg/mL, w/v) for the mucosal α-glucosidase assay. Optimum pH for individual α-

82

glucosidase activities was determined using a phosphate-based universal buffer (pH 3-8).25 The

83

mixture, containing each substrate solution (100 uL), 100 mM universal buffer (800 uL), and rat

84

intestinal enzyme (100 uL), was incubated at 37˚C for 10 min for maltodextrin and maltose and

85

60 min for sucrose and isomaltulose. The enzyme solution was inactivated by placing tubes in

86

boiling water and the amount of released glucose was determined by the GOPOD method.26

5 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 6 of 27

87 88

Hydrolysis properties of rat intestinal α-glucosidases on different α-linked disaccharides.

89

Maltose isomers (trehalose, kojibiose, nigerose, and isomaltose) and sucrose isomers (turanose,

90

maltulose, leucrose, and isomaltulose) were applied as substrates (100 mg/mL) for mucosal α-

91

glucosidases. Maltose isomers were hydrolyzed in pH 6.5 and sucrose isomers were hydrolyzed

92

in pH 6.0 sodium phosphate buffers (100 mM). The mixture, containing each substrate solution

93

(100 uL), 100 mM sodium phosphate buffer (800 uL), and prepared-rat intestinal enzyme (100

94

uL), was incubated at 37˚C for 120 min for maltose isomers and 360 min for sucrose isomers.

95

The enzyme solution was inactivated by placing tubes in boiling water and the amount of

96

released glucose was determined by the GOPOD method.26

97 98

Purification of the individual recombinant mucosal α-glucosidases. The individual

99

recombinant mucosal α-glucosidases were expressed via the baculovirus system through

100

different insect cell as a host as described by Jones, et al. 27 The recombinant baculovirus for ctSI

101

and ctMGAM (both C-terminal genes from mouse mRNA) were transfected into the Sf9 insect

102

cell, and the recombinant ntMGAM and ntSI (both N-terminal genes from human mRNA) were

103

transfected to Drosophila S2 cells to express recombinant protein.28 The expressed individual

104

protein was purified with nickel-nitrilotriacetic acid (Ni-NTA) affinity column chromatography

105

(Qiagen, Hilden, Germany), and then concentrated with Microcon YM-30 (MWCO 30,000;

106

Millipore, CA, USA). Protein amount was measured by the Bradford method and compared with

107

a bovine serum albumin (BSA) standard.29

108 109

Enzyme kinetics on different types of linkage and glucose composition. To study the enzyme

6 ACS Paragon Plus Environment

Page 7 of 27

Journal of Agricultural and Food Chemistry

110

kinetics of individual mucosal α-glucosidases on different substrates, 5 µg of each enzyme was

111

reacted at 37°C in 10 mM PBS buffer (pH 6.9) with 1-200 mM of different concentrations of

112

substrate. The amount (mM) of released glucose per 1 min (mM/min) from the different

113

concentrations of the substrates (maltose and sucrose isomers) by the individual recombinant α-

114

glucosidase reaction was determined by the GOPOD method.26 Kinetic values (Km and kcat)

115

were calculated by the Michaelis-Menten equation using SigmaPlot 12 (Systat Software Inc., San

116

Jose, CA, USA). Analyses were done in duplicate.

117 118

Analysis of hydrolysis properties on different α-linkages on sucrose, isomaltose,

119

isomaltulose. Different types of α-linked disaccharide solution [1.0%, w/v, (sucrose,

120

isomaltulose and isomaltose)] as a substrate were reacted with 100 U (one unit enzyme activity

121

was arbitrarily defined as the amount of enzyme that releases 1 µg of glucose from 1% maltose

122

per 10 min at 37°C) of recombinant mucosal α-glucosidases at 37˚C with 10 mM PBS (pH 6.9)

123

containing 0.02% of sodium azide. For testing α-hydrolytic activity, the mixture was reacted

124

during 72 h. The resulting product was reacted with GOPOD solution to analyze the released

125

glucose by enzyme reaction. Then, the amount of released glucose was divided by the number of

126

glucose units to calculate glucose recovery ratio. All reactions were performed in triplicate.

127 128

RESULTS AND DISCUSSION

129

Mucosal α-glucosidases have different optimal pH range depending on substrates. In the

130

small intestine, luminal pH increases from the acidic environment of the stomach to the neutral

131

range, but still has a slightly increasing gradient from the proximal to distal parts.30, 31 Depending

132

on the α-linkage type of the glycemic carbohydrate, optimal pHs of small intestinal α-

7 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 8 of 27

133

glucosidase activities on different substrate were somewhat different (Figure 2). The optimal pH

134

in vitro for maltose and maltodextrin digestion, which are connected by α-1,4 linkages, was pH

135

6.5, while for sucrose (α-1,2) and isomaltulose (α-1,6) were pH 6.0, and all four substrates show

136

relatively broad optimal pH ranges. The optimal pH data suggests that sucrose and α-1,6 linked

137

digestible carbohydrates (e.g., isomaltose, isomaltulose, and branched α-limit dextrins from

138

starch hydrolyzates by α-amylase) may be favorably hydrolyzed in the upper level of small

139

intestine due to relatively low pH conditions.32, 33 Different optimal pH range is one of the factors

140

that may affect the hydrolysis rate of digestible carbohydrates in the small intestine. The catalytic

141

sites between the rat and human α-glucosidases are conserved,4 thus it is expected that the pH

142

dependent-hydrolysis properties would be similar.

143 144

Hydrolytic properties of rat intestinal α-glucosidases on maltose and sucrose isomers. A

145

number of studies have focused on specific linkage-based small molecule slowly digestible

146

carbohydrates to decrease postprandial blood glucose level.34-36 In the present study, various

147

disaccharides bound by different linkages between α-D-glucosyl-D-glucose (Figure 3A) and α-D-

148

glucosyl-D-fructose (Figure 3B) were tested using mammalian (rat) intestinal mucosal α-

149

glucosidases to determine relative hydrolysis rates. Table 1 shows that all available types of two

150

glucose α-linked disaccharides (maltose isomers) were hydrolyzed by crude rat intestinal α-

151

glucosidases to glucose, but with different hydrolysis rates. Compared to maltose (α-1,4 linkage),

152

the maltose isomers were slowly hydrolyzed (p < 0.05); and large differences were observed with

153

relative hydrolysis rate of nigerose (α-1,3) at about 50%, followed by kojibiose (α-1,2),

154

isomaltose (α-1,6), and trehalose (α-1,1). Thus, different types of α-linked maltose isomers might

155

be applied, and in variations as larger oligomers, to make slowly digestible carbohydrates due to

8 ACS Paragon Plus Environment

Page 9 of 27

156 157

Journal of Agricultural and Food Chemistry

their slower glucose generation rates. The available sucrose isomers were also hydrolyzed to monosaccharides (glucose and

158

fructose) by rat intestinal α-glucosidases at the optimal pH (6.0) of sucrose hydrolysis (Table 2).

159

Maltulose (α-1,4) and leucrose (α-1,5) produced a similar amount of glucose as sucrose, while

160

turanose (α-1,3) and isomaltulose (α-1,6) were more slowly hydrolyzed (p < 0.05). Glucose

161

generation rates (µg/min) from the sucrose isomers were much lower than all of the maltose

162

isomers except trehalose (Tables 1 and 2). Interestingly, sucrose is considered a rapidly digestible

163

carbohydrate,37 though its glucose generation rate is lower than α-amylase products from starch

164

digestion (e.g., maltose and maltotriose).38

165 166

Enzyme kinetics on different α-glycosidic linkages between α-D-glucosyl-D-glucose.

167

Glycemic substrates with different α-glycosidic-linked disaccharides [trehalose (α-1,1), kojibiose

168

(α-1,2), nigerose (α-1,3), maltose (α-1,4), and isomaltose (α-1,6)] were investigated to

169

understand their hydrolytic properties related to the individual recombinant α-glucosidases. As

170

shown in the literature,17, 39, 40 all mucosal α-glucosidases have α-1,4 linkage (maltose)

171

hydrolysis activity with different rates of glucogenesis; and ntSI is mainly responsible for α-1,6

172

linkage hydrolysis (isomaltose).

173

Trehalose (α-1,1), which does not have a reducing end, was not hydrolyzed to glucose by

174

the individual recombinant mucosal α-glucosidases (Table 3), although it was hydrolyzed by rat

175

intestinal α-glucosidases (Table 1). This is because the digestion of trehalose in the

176

gastrointestinal tract is accomplished by trehalase,41 which has a different catalytic mechanism

177

compared to mucosal MGAM and SI enzymes.

178

For the maltose isomers, all four mucosal α-glucosidases hydrolyzed the α-1,2 (kojibiose)

9 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 27

179

and α-1,3 (nigerose) glucosyl-glucose linkages, though with a significantly reduced rates of

180

glucogenesis compared to maltase (α-1,4) activity (Table 3). Further, the α-1,2 linkage was more

181

slowly hydrolyzed than the α-1,3 linkage, as it was with the rat intestinal enzymes. For α-1,2

182

linkage hydrolysis, enzyme efficiency of glucose generation (kcat/Km) was approximately 8 to 56

183

times lower than that of maltase activity for the four enzymes (kcat/Km of ctMGAM: 0.91;

184

ntMGAM: 1.52; ctSI: 0.03; and ntSI: 0.06). Under the same conditions, glucose production rate

185

(kcat/Km) from the α-1,3 linkage was 3 to 15 times lower than that of maltase activity (kcat/Km of

186

ctMGAM: 2.73; ntMGAM: 4.45; ctSI: 0.17; and ntSI: 0.56). Notably, maltase (ntMGAM) had

187

higher hydrolytic property on α-1,2 and α-1,3 linkages compared to the other α-glucosidases.

188

This would be due to the comparatively open binding site of ntMGAM that can better

189

accommodate glycosidic linkages to the C-2, -3 and -4 positions than the other enzymes tested.18,

190

42

191

Isomaltose was not digested well by the ctMGAM and ctSI which can be explained by

192

the fact that the (α-1,6) glycosidic linkage of isomaltose is longer and more flexible than those of

193

kojibiose, nigerose and maltose (Figure 3A). This requires a narrower opening of the +1 sugar

194

binding site to accommodate isomaltose, as is observed in the ntSI (isomaltase) structure.18

195

Because ntMGAM has minor hydrolytic activity towards the α-1,6 bond, it can be considered

196

intermediate between ntSI and the C-terminal enzymes and thus is more promiscuous in

197

specificity. The catalytic site is sufficiently close in structure to have low activity towards

198

isomaltose, but sufficiently open to hydrolyze kojibiose and nigerose as well as its primary

199

activity towards maltose.

200 201

Hydrolysis of sucrose, isomaltose, and isomaltulose by individual mucosal α-glucosidases.

10 ACS Paragon Plus Environment

Page 11 of 27

Journal of Agricultural and Food Chemistry

202

Sucrase activity was unexpectedly found in ctMGAM, though with 5 times lower activity than

203

ctSI (Table 4). Considering the amount of SI in the small intestine is 40-50 times higher than

204

MGAM,12, 13 it is reasonable that ctSI is considered as the only major α-glucosidase related to

205

sucrose hydrolysis. Contrary to our findings, previous investigations using pig and human

206

intestine revealed no sucrase activity in MGAM.11 The clear, but low, sucrose hydrolysis activity

207

of ctMGAM found here might be explained by the fact that the protein structure of ctSI is closer

208

to that of ctMGAM (with 60% protein similarity) than to ntSI (around 40% sequence identity)

209

(Figure 1).42

210

Isomaltulose (the α-1,6 isomer of sucrose), which is recognized as a slowly digestible

211

sweetener,43 was hydrolysable by each recombinant α-glucosidase, but at markedly different

212

rates. ntSI and ntMGAM had major hydrolysis activities on isomaltulose with the ntSI showing

213

substantially higher activity (91.6 versus 29.4%, Table 4). The high hydrolysis rate of

214

isomaltulose by ntSI confirms previous research.10 Both ctMGAM and ctSI had some, though

215

quite low, hydrolytic activity for isomaltulose (4.2 and 8.5%, respectively), while a previous

216

study by Lina, Jonker et al. (2002) showed no isomaltulose activity in the MGAM complex in rat

217

and human small intestine.23

218

Expectedly, isomaltose was almost fully hydrolyzed by the action of ntSI (Table 4).

219

Notably, all mucosal α-glucosidases had some degree of isomaltase (α-1,6 linkage) activity,

220

particularly at longer reaction times. While 28.4 % of isomaltose was hydrolyzed to glucose by

221

the action of ntMGAM after 72 h reaction, ctMGAM and ctSI hydrolyzed only 8.8 and 18.1% of

222

the substrate (Table 4). Considering the amount of protein and hydrolysis activity in the small

223

intestine, ntSI by far represents the major hydrolyzing enzyme for isomaltose and the branched

224

α-limit dextrins from α-amylase degraded starch.44

11 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 12 of 27

225

It is interesting that isomaltulose has both structural similarity with sucrose (an α-

226

glycosyl-fructose disaccharide) and isomaltose (α-1,6 linkage), but is hydrolyzed mainly by ntSI.

227

Considering that sucrose is digested only by ctSI and isomaltose is primarily digested by ntSI, it

228

appears that linkage type is the more important factor, rather than sugar composition, in

229

determining which of the individual α-glucosidases will digest isomaltulose. A further

230

investigation with changing carbohydrate structures by modifying the α-linkage could better

231

relate structures with binding properties on enzyme active sites.

232

In this study, there were clear differences in hydrolytic properties of the enzymes to

233

disaccharide substrates differing in linkages that appeared to parallel structural homologies of the

234

proteins. These investigations provide insight into the role of individual mucosal α-glucosidases

235

in the digestion of unusual linkage glycemic disaccharides for guidance in the development of

236

slowly digestible carbohydrate (i.e. oligosaccharide) ingredients based on α-linkage type. For the

237

first time, maltase (ntMGAM) was shown to have a particular capacity to digest α-1,2 and -1,3

238

disaccharides of glucose, ascribing a broader specificity to the catalytic site of this enzyme than

239

known before. Lastly, our findings imply that glycemic carbohydrates such as studied here must

240

be tested using mammalian mucosal α-glucosidases for evaluating digestion rate differences, and

241

not the glucoamylases from fungal or microbial sources commonly used in in vitro starch

242

digestion assays.

243 244

12 ACS Paragon Plus Environment

Page 13 of 27

Journal of Agricultural and Food Chemistry

245

REFERENCES

246

1.

247

Cummings, R. D.; Esko, J. D.; Freeze, H. H.; Stanley, P.; Bertozzi, C. R.; Hart, G. W.; Etzle, M.

248

E., Eds. Cold Spring Harbor Laboratory Press: Cold Spring Harbor, New York, 1999.

249

2.

Holmes, R., Carbohydrate digestion and absorption. J. Clin. Pathol. 1971, s3-5, 10-13.

250

3.

Leturque, A.; Brot-Laroche, E.; Le Gall, M.; Stolarczyk, E.; Tobin, V., The role of

251

GLUT2 in dietary sugar handling. J. Physiol. Biochem. 2005, 61, 529-537.

252

4.

253

Primary structure, membrane-orientation, and evolution of a stalked, intrinsic brush border

254

protein. Cell 1986, 46, 227-234.

255

5.

256

intestinal maltase-glucoamylase cDNA cloning. homology to sucrase-isomaltase. J. Biol. Chem.

257

1998, 273, 3076-3081.

258

6.

259

hydrolases. Curr. Opin. Chem. Biol. 1997, 7, 637-644.

260

7.

261

of the Sulfolobus solfataricus α-glucosidase: Implications for domain conservation and substrate

262

recognition in GH31. J. Mol. Biol. 2006, 358, 1106-1124.

263

8.

264

different binding patterns for malto- and isomaltooligosaccharides. Biol. Chem. Hoppe-Seyler.

265

1995, 376, 249-253.

266

9.

267

clinical and molecular aspects. Clin. Gastroenterol. Hepatol. 2006, 4, 276-287.

Freeze, H. H., Monosaccharide metabolism. In Essentials of Glycobiology, Varki, A.;

Hunziker, W.; Spiess, M.; Semenza, G.; Lodish, H. F., The sucrase-isomaltase complex:

Nichols, B. L.; Eldering, J.; Avery, S.; Hahn, D.; Quaroni, A.; Sterchi, E., Human small

Henrissat, B.; Davies, G., Structural and sequence-based classification of glycoside

Ernst, H. A.; Lo Leggio, L.; Willemoës, M.; Leonard, G.; Blum, P.; Larsen, S., Structure

Heymann, H.; Breitmeier, D.; Günther, S., Human small intestinal sucrase-isomaltase:

Robayo-Torres, C. C.; Quezada-Calvillo, R.; Nichols, B. L., Disaccharide digestion:

13 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 14 of 27

268

10.

Dahlqvist, A.; Auricchio, S.; Semenza, G.; Prader, A., Human intestinal disaccharides and

269

herediatry disaccharide intolerance. The hydrolysis of sucrose, isomaltose, palatinose

270

(isomaltulose) and a 1,6-α-oligosaccharide (isomalto-oligosaccharide) preparation. J. Clin.

271

Invest. 1963, 42, 556-562.

272

11.

273

microvillus maltase/glucoamylase. Eur. J. Biochem. 1982, 126, 559-568.

274

12.

275

basis of inherited disease 8ed.; Scriver, C. R.; Sly, A. L. S.; Beaudet, W.; David, V., Eds.

276

McGraw-Hill: New York, 2001; Vol. II, pp 1623-1650.

277

13.

278

I. Chromatographic separation of maltases and of two lactases. Biochim. Biophys. Acta. 1965, 96,

279

487-497.

280

14.

281

Hamaker, B. R., Mucosal C-terminal maltase-glucoamylase hydrolyzes large size starch

282

digestion products that may contribute to rapid postprandial glucose generation. Mol. Nutr. Food

283

Res. 2014, 58, 1111-1121.

284

15.

285

glucoamylase-maltase. Biol. Chem. Hoppe-Seyler. 1994, 375, 451-455.

286

16.

287

Brayer, G. D.; Sterchi, E. E.; Baker, S. S.; Nichols, B. L., Luminal substrate "Brake" on mucosal

288

maltase-glucoamylase activity regulates total rate of starch digestion to glucose. J. Pediatr. Gastr.

289

Nutr. 2007, 45, 32-43

290

17.

Sørensen, S. H.; Norén, O.; Sjöström, H.; Danielsen, E. M., Amphiphilic pig intestinal

Semenza, G.; Auricchio, S.; Mantei, N., Small intestinal disaccharidases. In Metabolic

Semenza, G.; Auricchio, S.; Rubino, A., Multiplicity of human intestinal disaccharidases

Lee, B.-H.; Lin, A. H.-M.; Nichols, B. L.; Jones, K.; Rose, D. R.; Quezada-Calvillo, R.;

Heymann, H.; Günther, S., Calculation of subsite affinities of human small intestinal

Quezada-Calvillo, R.; Robayo-Torres, C. C.; Ao, Z.; Hamaker, B. R.; Quaroni, A.;

Quezada-Calvillo, R.; Sim, L.; Ao, Z.; Hamaker, B. R.; Quaroni, A.; Brayer, G. D.;

14 ACS Paragon Plus Environment

Page 15 of 27

Journal of Agricultural and Food Chemistry

291

Sterchi, E. E.; Robayo-Torres, C. C.; Rose, D. R.; Nichols, B. L., Luminal starch substrate

292

"Brake" on maltase-glucoamylase activity is located within the glucoamylase subunit. J. Nutr.

293

2008, 138, 685-692.

294

18.

295

basis for substrate selectivity in human maltase-glucoamylase and sucrase-isomaltase N-terminal

296

domains. J. Biol. Chem. 2010, 285, 17763-17770.

297

19.

298

nigerosylmaltooligosaccharides-supplemented syrup. J. Appl. Glycosci. 1999, 46, 475-482.

299

20.

300

Enzymatic synthesis of kojioligosaccharides using kojibiose phosphorylase. J. Biosci. Bioeng.

301

2001, 92, 177-182.

302

21.

303

Jenkins, A. L.; Axelsen, M., Glycemic index: overview of implications in health and disease. Am.

304

J. Clin. Nutr. 2002, 76, 266S-273S.

305

22.

306

responses in human subjects. Eur. J. Clin. Nutr. 2007, 62, 1364-1371.

307

23.

308

biological and toxicological studies. Food Chem. Toxicol. 2002, 40, 1375-1381.

309

24.

310

Activity Inhibitor and Food and Feed Containing the Same. 2008.

311

25.

312

Germany, 1974.

313

26.

Sim, L.; Willemsma, C.; Mohan, S.; Naim, H. Y.; Pinto, B. M.; Rose, D. R., Structural

Takeshi, Y.; Takehiro, U.; Masayoshi, S.; Toshinao, G., Properties of a nigerose and

Chaen, H.; Nishimoto, T.; Nakada, T.; Fukuda, S.; Kurimoto, M.; Tsujisaka, Y.,

Jenkins, D. J.; Kendall, C. W.; Augustin, L. S.; Franceschi, S.; Hamidi, M.; Marchie, A.;

Grysman, A.; Carlson, T.; Wolever, T. M. S., Effects of sucromalt on postprandial

Lina, B. A. R.; Jonker, D.; Kozianowski, G., Isomaltulose (Palatinose®): a review of

Kashimura, J.; Nagai, Y.; Ebashi, T.; Goda, T. Sucrase Activity Inhibitor, Glucoamylase

D. D. Perrin, B. D., Buffers for pH and Metal Ion Control. Springer: Heidelberg,

Vasanthan, T., Enzymatic quantitation of total starch in plant products. In Current

15 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 16 of 27

314

Protocols in Food Analytical Chemistry, Wrolstad, R. E.; Acree, T. E.; Decker, E. A.; Penner, M.

315

H.; Reid, D. S.; Schwartz, S. J.; Shoemaker, C. F.; Smith, D. M.; Sporns, P., Eds. John Wiley &

316

Sons, Inc.: Hoboken, New Jersey, 2001; pp E2.2.1-E2.2.

317

27.

318

Calvillo, R.; Nichols, B. L.; Mario Pinto, B.; Rose, D. R., Mapping the intestinal alpha-

319

glucogenic enzyme specificities of starch digesting maltase-glucoamylase and sucrase-

320

isomaltase. Bioorg. Med. Chem. 2011, 19, 3929-3934.

321

28.

322

Expression and purification of sea raven type II antifreeze protein from Drosophila melanogaster

323

S2 cells. Protein Express. Purif. 2006, 47, 374-383.

324

29.

325

quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 1976, 72, 248.

326

30.

327

Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 1988, 29,

328

1035-1041.

329

31.

330

1999, 46, 183-196.

331

32.

332

hydrolysis and absorption. J. Clin. Invest. 1965, 44, 390-398.

333

33.

334

hydrolysis and monosaccharide product absorption. J. Clin. Invest. 1966, 45, 388-398.

335

34.

336

responses in human subjects. Eur J Clin Nutr 2007, 62, 1364-1371.

Jones, K.; Sim, L.; Mohan, S.; Kumarasamy, J.; Liu, H.; Avery, S.; Naim, H. Y.; Quezada-

Scotter, A. J.; Kuntz, D. A.; Saul, M.; Graham, L. A.; Davies, P. L.; Rose, D. R.,

Bradford, M. M., A rapid and sensitive method for the quantitation of microgram

Evans, D. F.; Pye, G.; Bramley, R.; Clark, A. G.; Dyson, T. J.; Hardcastle, J. D.,

Fallingborg, J., Intraluminal pH of the human gastrointestinal tract. Dan. Med. Bull.

Gray, G. M.; Ingelfinger, F. J., Intestinal absorption of sucrose in man: the site of

Gray, G. M.; Ingelfinger, F. J., Intestinal absorption of sucrose in man: interrelation of

Grysman, A.; Carlson, T.; Wolever, T. M. S., Effects of sucromalt on postprandial

16 ACS Paragon Plus Environment

Page 17 of 27

Journal of Agricultural and Food Chemistry

337

35.

van Can, J. G. P.; IJzerman, T. H.; van Loon, L. J. C.; Brouns, F.; Blaak, E. E., Reduced

338

glycaemic and insulinaemic responses following isomaltulose ingestion: implications for

339

postprandial substrate use. Br. J. Nutr. 2009, 102, 1408-1413.

340

36.

341

isomaltulose is lower than that of sucrose during exercise in men. J. Nutr. 2007, 137, 1143-1148.

342

37.

343

digestible energy content and energy utilization. Anim. Sci. 1990, 51, 343-355.

344

38.

345

R.; Quaroni, A.; Brayer, G. D.; Wattler, S.; Nehls, M. C.; Sterchi, E. E.; Nichols, B. L.,

346

Contribution of mucosal maltase-glucoamylase activities to mouse small intestinal starch alpha-

347

glucogenesis. J. Nutr. 2007, 137, 1725-1733.

348

39.

349

hereditary disaccharide intolerance. J. Clin. Invest. 1962, 41, 463-470.

350

40.

351

glucoamylase gene: common ancestry to sucrase-isomaltase with complementary starch

352

digestion activities. Proc. Natl. Acad. Sci. 2003, 100, 1432-1437.

353

41.

354

Pediatric Gastointestinal Disease, Walker, W. A., Ed. BC Decker INC: Lewiston, 2004; Vol. 1,

355

pp 880-897.

356

42.

357

intestinal maltase-glucoamylase: Crystal structure of the N-terminal catalytic subunit and basis

358

of inhibition and substrate specificity. J. Mol. Biol. 2008, 375, 782-792.

359

43.

Achten, J.; Jentjens, R. L.; Brouns, F.; Jeukendrup, A. E., Exogenous oxidation of

Beech, S. A.; Elliott, R.; Batterham, E. S., Sucrose as an energy source for growing pigs:

Quezada-Calvillo, R.; Robayo-Torres, C. C.; Opekun, A. R.; Sen, P.; Ao, Z.; Hamaker, B.

Dahlqvist, A., Specificity of the human intestinal disaccharidases and implications for

Nichols, B. L.; Avery, S.; Sen, P.; Swallow, D. M.; Hahn, D.; Sterchi, E., The maltase-

Naim, H. Y.; Zimmer, K.-P., Congenital disease of dysfunction and absorption. In

Sim, L.; Quezada-Calvillo, R.; Sterchi, E. E.; Nichols, B. L.; Rose, D. R., Human

Kawai, K.; Yoshikawa, H.; Murayama, Y.; Okuda, Y.; Yamashita, K., Usefulness of

17 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

360

palatinose as a caloric sweetener for diabetic patients. Horm. Metab. Res. 1989, 21, 338-340.

361

44.

362

free monomers on an alpha-limit dextrin. J. Biol. Chem. 1979, 254, 6038-6043.

Page 18 of 27

Gray, G. M.; Lally, B. C.; Conklin, K. A., Action of intestinal sucrase-isomaltase and its

363 364 365

ACKNOWLEDGMENTS This research was supported by Basic Science Research Program through the National

366

Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning

367

(NRF-2015R1C1A1A02036467) (BHL). We are thankful for support from the United States

368

Department of Agriculture (USDA) Agriculture and Food Research Initiative competitive grant

369

program, no. 08-555-03-18793 (BRH), Whistler Center for Carbohydrate Research at Purdue

370

University, Indiana, USA, and the Canadian Institutes for Health Research and Natural Science

371

and Engineering Research Council (DRR). Also, supported in part by federal funds from the

372

USDA, Agricultural Research Service, under Cooperative Agreement Number 58-6250-1-003

373

with Baylor College of Medicine (BLN, RQC).

374

18 ACS Paragon Plus Environment

Page 19 of 27

375

Journal of Agricultural and Food Chemistry

FIGURE CAPTIONS

376 377

Figure 1. Maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI) protein complex

378

structures of the small intestinal lumen linked to the transmembrane domain via an O-

379

glycosylated linkage. Percentage comparisons between individual enzymes indicate sequence

380

identity. AA: amino acids

381 382

Figure 2. Optimal pH conditions for reaction of crude rat intestinal powder on different types of

383

α-linked substrates determined as the amount of glucose release at 37°C. The optimal pH in vitro

384

for maltose and maltodextrin digestion, which are α-1,4 linked, was pH 6.5; while for sucrose (α-

385

1,2) and isomaltulose (α-1,6) were pH 6.0. MD (DE1): maltodextrin dextrose equivalent 1

386 387

Figure 3. Various disaccharides based on different α-glycosidic linkages and monosaccharide

388

composition. A: α-linked disaccharides between two glucose molecules B: α-linked

389

disaccharides between glucose and fructose molecules

390

19 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 20 of 27

Table 1. Glucose Production (µg/min) from Maltose Isomers (10 mg/mL, w/v) and Relative Hydrolysis Rate (%) Compared to Maltose (at 100%) by Crude Rat Intestinal αGlucosidases.a Trehalose (α-1,1)

Kojibiose (α-1,2)

Nigerose (α-1,3)

Maltose (α-1,4)

Isomaltose (α-1,6)

Glucose production rate (µg/min)

7.9 ± 1.3

21.4 ± 1.3

30.9 ± 0.7

59.3 ± 2.3

14.4 ± 1.4

Relative hydrolysis rate (%)

13 ± 2e

36 ± 2c

52 ± 1b

100 ± 4a

24 ± 2d

a

Enzymatic reactions were carried out in 100 mM sodium phosphate buffer (pH 6.5) at 37°C. Values represent the mean ± standard deviation and were performed in tripliicate. Values denoted by different letters (a – e) are significantly different from each other at p < 0.05 as determined using ANOVA and Tukey’s HSD multiple comparison tests.

20 ACS Paragon Plus Environment

Page 21 of 27

Journal of Agricultural and Food Chemistry

Table 2. Glucose Production (µg/min) from Sucrose Isomers (10 mg/mL, w/v) and Relative Hydrolysis rate (%) Compared to Sucrose (at 100%) by Crude Rat Intestinal αGlucosidases.a Sucrose (α-1,2)

Turanose (α-1,3)

Maltulose (α-1,4)

Leucrose (α-1,5)

Isomaltulose (α-1,6)

Glucose production rate (µg/min)

9.6 ± 0.1

6.6 ± 0.2

9.5 ± 0.3

9.0 ± 0.3

3.7 ± 0.1

Relative hydrolysis rate (%)

100 ± 1a

69 ± 3b

99 ± 3a

94 ± 4a

39 ± 1c

a

Enzymatic reactions were carried out in 100 mM sodium phosphate buffer (pH 6.5) at 37°C. Values represent the mean ± standard deviation and were performed in tripliicate. Values denoted by different letters (a – e) are significantly different from each other at p < 0.05 as determined using ANOVA and Tukey’s HSD multiple comparison tests.

21 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 22 of 27

Table 3. Kinetic Parameters of Each Recombinant Mucosal α-Glucosidase on Differently αLinked Disaccharides with Two Glucoses. ctMGAM (glucoamylase)

ntMGAM (maltase)

ctSI (sucrase)

ntSI (isomaltase)

Km (mM)

nd*

nd

nd

nd

Kcat (s-1)

nd

nd

nd

nd

Kcat/Km

nd

nd

nd

nd

12.7 ± 2.5

11.6 ± 1.2

17.3 ± 2.2

53.7 ± 13.7

Kcat (s )

11.5 ± 0.6

17.6 ± 0.5

0.5 ± 0.0

3.2 ± 0.3

Kcat/Km

0.9 ± 0.2

1.5 ± 0.4

0.3 ± 0.0

0.1 ± 0.0

35.2 ± 3.6

27.1 ± 2.6

63.6 ± 13.0

44.4 ± 6.4

Kcat (s )

96.0 ± 3.3

120.9 ± 3.7

11.0 ± 1.0

24.8 ± 1.3

Kcat/Km

2.7 ± 0.9

4.4 ± 1.4

0.2 ± 0.1

0.6 ± 0.2

2.6 ± 0.6

8.7 ± 1.3

4.2 ± 1.4

11.1 ± 1.5

Kcat (s )

133.9 ± 4.3

110.2 ± 3.8

11.4 ± 0.8

18.3 ± 0.6

Kcat/Km

51.0 ± 7.0

12.7 ± 3.0

2.7 ± 0.5

1.7 ± 0.4

nda

128.0 ± 8.4

nd

15.2 ± 2.0

-1

Kcat (s )

nd

8.9 ± 0.3

nd

18.1 ± 0.7

Kcat/Km

nd

0.1 ± 0.0

nd

1.2 ± 0.3

Trehalose

Kojibiose Km (mM) -1

Nigerose Km (mM) -1

Maltose Km (mM) -1

Isomaltose Km (mM)

a

nd, not detected.

22 ACS Paragon Plus Environment

Page 23 of 27

Journal of Agricultural and Food Chemistry

Table 4. Hydrolysis (%) of Different Types of Substrates (sucrose, isomaltulose, and isomaltose; 10 mg/mL, w/v) to Glucose by Individual Recombinant Mucosal α-Glucosidases (100 U) after 72 h.a,b

ctMGAM (glucoamylase) ntMGAM (maltase) ctSI (sucrase) ntSI (isomaltase)

Sucrose (α-1,2; G+F) c

Isomaltulose (α-1,6; G+F)

Isomaltose (α-1,6; G+G)

14.8 ± 0.6

4.2 ± 0.6

8.8 ± 0.2

0.6 ± 0.1

29.4 ± 0.4

28.4 ± 1.0

73.1 ± 0.1

8.5 ± 0.4

18.1 ± 0.1

0.9 ± 0.0

91.6 ± 3.1

98.4 ± 3.6

a

One unit enzyme activity arbitrarily defined as the amount of enzyme that released 1 µg of glucose from 1% maltose per 10 min at 37°C b Mean value ± standard deviation of measurement of experiments performed in triplicate. c G: glucose and F: fructose

23 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 24 of 27

Figure 1

24 ACS Paragon Plus Environment

Page 25 of 27

Journal of Agricultural and Food Chemistry

Figure 2

25 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 26 of 27

Figure 3 26 ACS Paragon Plus Environment

Page 27 of 27

Journal of Agricultural and Food Chemistry

Table of Contents (TOC) 72x45mm (300 x 300 DPI)

ACS Paragon Plus Environment