Maternal N-Carbamylglutamate supply during early pregnancy

May 27, 2018 - ... previous study demonstrates that maternal N-Carbamylglutamate (NCG) supply during early pregnancy enhances embryonic survival in gi...
0 downloads 0 Views 980KB Size
Subscriber access provided by University of Massachusetts Amherst Libraries

Bioactive Constituents, Metabolites, and Functions

Maternal N-Carbamylglutamate supply during early pregnancy enhanced pregnancy outcomes in sows through modulations of targeted genes and metabolism pathways Shuang Cai, Jinlong Zhu, Xiangzhou Zeng, Qianhong Ye, Changchuan Ye, Xiangbing Mao, Shihai Zhang, Shiyan Qiao, and Xiangfang Zeng J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.8b01637 • Publication Date (Web): 27 May 2018 Downloaded from http://pubs.acs.org on May 27, 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 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 36

Journal of Agricultural and Food Chemistry

1

Maternal N-Carbamylglutamate supply during early pregnancy enhanced

2

pregnancy outcomes in sows through modulations of targeted genes and

3

metabolism pathways

4

Shuang Cai,†,§ Jinlong Zhu,† Xiangzhou Zeng,† Qianhong Ye,† Changchuan Ye,†

5

Xiangbing Mao,‡ Shihai Zhang,|| Shiyan Qiao,† and Xiangfang Zeng†,*

6 7



8

Centre, China Agricultural University, Beijing 100193, China;

9



State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry

Animal

Nutrition

Institute,

Sichuan

Agricultural

University,

No.

10

Gongpinghuimin Road, Wenjiang District, Chengdu 611130, China;

11

||

12

Animal Science, South China Agricultural University, Guangzhou 510642, China

211,

Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

13

ABSTRACT: Reducing pregnancy loss is important for improving reproductive

14

efficiency for both human and mammalian animals. Our previous study demonstrates

15

that maternal N-Carbamylglutamate (NCG) supply during early pregnancy enhances

16

embryonic survival in gilts. However whether maternal NCG supply improves the

17

pregnancy outcomes is still not known. Here, we found maternal NCG supply during

18

early pregnancy in sows significantly increased the numbers of total piglets born alive

19

per litter (P < 0.05), and significantly changed the levels of metabolites in amniotic

20

fluid and serum involved in metabolism of energy, lipid, and glutathione, and

21

immunogical regulation. The expression of endometrial progesterone receptor

22

membrane component 1 (PGRMC1) were significantly increased by NCG

23

supplementation (P < 0.05), as well as the expression of PGRMC1, endothelial nitric

24

oxide synthesases (eNOS), and lamin A/C in fetuses and placentae (P < 0.05). Among

25

the NCG associated amino acids, arginine and glutamine markedly increased

26

PGRMC1 and eNOS expression in porcine trophectoderm cells (P < 0.05). While

27

glutamate could stimulate the expression of vimentin and lamin A/C in porcine

28

trophectoderm (pTr) cells (P < 0.05), and proline stimulated lamin A/C expression (P

29

< 0.05). Collecetively, these data reveal mechanisms of NCG in reducing early

30

embryo loss. These findings have important implications that NCG has great potential

31

to improve pregnancy outcomes in human and mammalian animals.

32

KEYWORDS: embryo, metabolome, N-carbamylglutamate, pregnancy outcome,

33

sows

2

ACS Paragon Plus Environment

Page 2 of 36

Page 3 of 36

Journal of Agricultural and Food Chemistry

34

INTRODUCTION

35

Embryo implantation is a critically important biological process in pregnancy in

36

mammals, which, to a large degree, has a direct impact on the pregnancy outcome.1

37

Early pregnancy loss, accounting for 30% - 50% in mammals, profoundly restricts

38

reproductive performance.2 Embryo implantation failure is the major cause of these

39

early pregnancy losses.1 In women, about 15% of all pregnancies end due to early

40

pregnancy loss.3 Even though in vitro fertilization (IVF) has become a successful

41

technology in treating infertility, the pregnancy rate of IVF is still relatively low at

42

around 25% - 30%, because of the failure of the IVF embryo to implant in the uterus.4

43

Therefore, reducing early embryo implantation failure is a crucially important issue in

44

improvement of the reproductive efficiency for both humans and mammalian animals.

45

Although numerous studies have demonstrated that nutrition plays important roles

46

in fetal development and subsequent pregnancy outcomes, only limited studies have

47

focused on the impacts and roles of specific nutrients or general nutritional status on

48

embryo development and embryo implantation.5-7 Arginine is a conditionally essential

49

amino acid for most mammals. Many studies have shown that arginine regulates

50

embryo development and improves embryo quality subsequently enhancing embryo

51

implantation.8-10 Arginine supplementation during early pregnancy is demonstrated to

52

improve embryo implantation through the PI3K/PKB/mTOR/NO signaling pathway,

53

ultimately enhancing the pregnancy outcome in rats.11,12 As an activator of

54

endogenous arginine synthesis, NCG has been reported to improve early embryonic

55

implantation and pregnancy outcome in rats.13,14 In gilts, maternal NCG supply during

56

early pregnancy also improves embryonic survival and development through

57

modulation of endometrial proteomes.15,16 However, whether maternal NCG

58

supplementation during early pregnancy improves pregnancy outcomes in sows is still 3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

59

unknown, as well as the specific roles of NCG in regulation of targeted genes’

60

expression in fetuses, placentae and endometriums, and the metabolic changes in

61

serum and amniotic fluid. Additionally, pig, which shares high similarity with humans

62

in anatomic and physiologic characteristics, becomes one of the best models for

63

human reproduction studie.17 Therefore, approaches to enhance sows’ reproductive

64

performance have important implication for improving human reproduction. The

65

objective of this study was to investigate whether maternal NCG supplementation

66

during early pregnancy improved pergancy outcomes in sows and to examine the

67

targeted genes’ expression in fetuses, placentae andendometriums, as well as the

68

metabolic changes in serum and amniotic fluid.

69

MATERIALS AND METHODS

70

Ethics statement

71

This study was carried out in strict accordance with the Chinese guidelines for animal

72

welfare. Animal handling procedures reported herein were approved by the

73

Institutional Animal Care and Use Committee of China Agricultural University.

74

Animals and protocols

75

In order to examine whether maternal NCG supplementation during early pregnancy

76

improved pergancy outcomes in sows, a total of 80 sows (parity 3 or 4; Landrace ×

77

Yorkshire) with an initial body weight of 210.00 ± 10.91 kg were used in this study.

78

Sows were housed individually in gestation stalls (2.2 m × 0.65 m) with concrete

79

floors. Sows were checked daily for estrus using direct boar exposure in the morning

80

and artificially inseminated three times with fresh semen during estrus (10-12 h apart).

81

Immediately after breeding, sows were allocated based on body weight, back fat, and

82

parity into 4 groups. Sows in the control group were fed the basal diet for the entire

83

gestation period. Sows in the three N-carbamylglutamate groups were fed the basal 4

ACS Paragon Plus Environment

Page 4 of 36

Page 5 of 36

Journal of Agricultural and Food Chemistry

84

diet supplemented with 0.05% NCG (wt:wt) during d 1-8, d 9-28, or d 1-28.

85

Thereafter sows were fed the basal diet until delivery (n = 20). Here, we designed

86

three NCG supply phases to determine the optimal one. All sows were fed 1.25 kg

87

diets during d 1-28 of pregnancy and 1.75 kg diets from d 29 of pregnancy to delivery

88

twice daily at 06:00 h and 16:00 h. Sows had free access to water throughout the

89

experiment. Diets were formulated to meet the recommended nutrient requirements of

90

NRC (2012) for gestating sows (Table 1). NCG (purity, 97%) was obtained from

91

National Feed Engineering Technology Research Center (Beijing, China) and 0.05%

92

NCG level was based on results from our previous study.15 Litter size, live litter size,

93

litter weight, number of stillborn and mummified piglets, and birth weight were

94

recorded soon after birth.

95

To investigate whether NCG improving pregnancy outcome was associated with

96

the role of NCG in regulation of targeted genes’ expression in fetuses, placentae and

97

endometriums, as well as the metabolic changes in serum and amniotic fluid, sixteen

98

gilts (Duroc × Yorkshire × Landrace) with average initial body weight of 132.0 kg

99

were randomly assigned to the the basal diet (control) or fed the basal diet

100

supplemented with 0.05% NCG between d 1 and d 28 of pregnancy (0.05% NCG, n =

101

8). This NCG supply phase was chosen based on the results of the first animal study.

102

Diets were the same as in Exp. 1. On d 28 of gestation, blood samples were collected

103

via jugular venepuncture into 5 mL tubes after overnight starvation before being

104

centrifuged at 3,000 × g for 10 min at 4°C to obtain serum samples. Gilts were killed

105

by electrocution to obtain uteri, which were transported on ice to the lab. A volume of

106

5 mL amniotic fluid from each fetus was harvested with syringes. Placentae and

107

endometriums were carefully isolated from the individual fetus. All the samples were

108

immediately snap frozen in liquid nitrogen and stored at -80°C until further analysis. 5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

109

Chemicals and reagents

110

HPLC-grade methanol and ACN were obtained from Fisher Scientific International

111

(Hampton, NH, USA). HPLC-grade formic acid was purchased from Dikma

112

Technology (RichmondHill, Ontario, Canada). Arginine, glutamine, glutamate, and

113

proline were purchased from Sigma Aldrich (St. Louis, Missouri, USA). Ultrapure

114

water (Milli-Q, Millipore Corporation, Bedford, MA, USA) was used to prepare all

115

aqueous solutions.

116

Cell culture and treatment

117

To further examine the contributions of arginine, glutamine, glutamate, and proline,

118

the concentrations of which are changed in maternal serum in response to maternal

119

NCG supply during early pregnancy, pTr cells were used as the in vitro model. Cells

120

were isolated originally from d 12 conceptuses and was kindly provided by Dr.

121

Zhenlong Wu from China Agricultural University. Cells were cultured in 6-well plates

122

in DMEM/F-12 medium with 10% FBS, 1% insulin–transferrin–selenium (ITS,

123

Sigma–Aldrich, St. Louis, Missouri, USA) until reaching 80% confluence. Cells were

124

serum starved overnight in customized medium, deprived of either arginine, proline,

125

glutamate or glutamine, and then treated with different doses (0, 0.25, 0.50, 1.00 mM)

126

of Arg, Pro, Glu or Gln for 24 h. All experiments were repeated at least 2-3 times.

127

Non-targeted metabolic fingerprinting analysis

128

Metabolite extraction

129

Methanol was used to extract both intracellular and extracellular metabolites. Serum

130

and amniotic fluid samples (200 µL) were centrifuged at 14,000 × g for 20 min at 4°C.

131

The supernatant were transferred and mixed with 4 volumes of methanol. The samples

132

were incubated at -80°C for 2 h and then centrifuged at 14,000 × g for 10 min at 4°C.

133

The metabolites containing supernatant were transferred to new tubes on dry ice and 6

ACS Paragon Plus Environment

Page 6 of 36

Page 7 of 36

Journal of Agricultural and Food Chemistry

134

then dried to pellets using speedvax/lyophilizer with no heat. The dried metabolite

135

samples were stored at -80°C until further processing.

136

HPLC methods for polar metabolites

137

Ultimate 3000 UHPLC (Dionex) coupled with Q Exactive was used to perform LC

138

separation. In positive mode, Waters Atlantis HILIC Silica column (2.1 × 100 nm)

139

was used. Mobile phase A was prepared by dissolving 0.63 g of ammonium formate in

140

50 mL of HPLC-grade water, then adding 950 mL of HPLC-grade acetonitrile and 1

141

µL of formic acid. Mobile phase B was prepared by dissolving 0.63 g of ammonium

142

formate in 500 mL of HPLC-grade water, followed by 500 mL of HPLC-grade

143

acetonitrile and 1 uL of formic acid. Linear gradient was as follows: 0 min, 1% B; 2

144

min, 1% B; 3.5 min, 20% B; 17 min, 80% B; 17.5 min, 99% B; 19 min, 99% B; 19.1

145

min, 1% B; 22 min, 1% B. In negative mode, Waters BEH Amide column was applied

146

(2.1 × 100 mm). Mobile phase A was prepared by dissolving 0.77 of ammonium

147

acetate in 50 mL of HPLC-grade water, then 950 mL of HPLC-grade acetonitrile was

148

added. pH was adjusted to 9.0 with ammonium hydroxide solution. Mobile phase B

149

was prepared by dissolving 0.77 g of ammonium acetate in 500 mL of HPLC-grade

150

water, subsequently, adding 500 mL of HPLC-grade acetonitrile and adjusting pH to

151

9.0 with ammonium hydroxide solution. Linear gradient was as follows: 0 min, 5% B;

152

2 min, 5% B; 4 min, 20% B; 18 min,85% B;19 min, 95% B; 21 min, 95% B; 21.1 min,

153

5% B; 25 min, 5% B.

154

MS methods for polar metabolites

155

Polar metabolite analysis was performed on Q Exactive Orbitrap mass spectrometer.

156

The detailed mass spectrometer parameters are as follows: spray voltage, 3.5 kV for

157

positive and 2.5 kV for negative; capillary temperature, 275 °C for positive and 320°C 7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

158

for negative; sheath gas flow rate (arb), 35; aux gas flow rate (arb), 8; mass range

159

(m/z), 70-1,050 for positive and 80-1,200 for negative; full MS resolution, 70,000;

160

MS/MS resolution, 17,5000; topN, 10; NCE, 15/30/45; duty cycle, 1.2s.

161

Data processing

162

Two levels of identification were performed simultaneously using Tracefinder.

163

Metabolites were first potentially assigned according to endogenous MS database by

164

accurate masses. At the same time, those that can match with the spectra in fragment

165

database were confirmed and potentially assigned metabolites were displayed in the

166

results. 10 ppm and 15 ppm mass tolerance was applied for precursor and fragment

167

matching. Still, 0.25 min retention time shift was allowed for quantitation.

168

Western blot analysis

169

The pTr cells, frozen placenta, endometrium and fetuses were separately homogenized

170

in RIPA lysis buffer (150 mM NaCl, 1 % Triton X-100, 0.5 % sodium deoxycholate,

171

0.1 % SDS, 50 mM Tris–HCl at pH 7.4) supplemented with a protease inhibitor

172

cocktail (Apply gene, Beijing, China). After centrifugation at 1,4000 × g for 15 min at

173

4oC, the protein concentration in the supernatant was determined using the BCA

174

protein assay kit (Pierce, Rockford, IL, USA). Equal amount of proteins was

175

electrophoresed (Bio-Rad, Richmond, CA, USA) on SDS polyacrylamide gels. Then

176

proteins were electrotransferred to polyvinylidene difluoride membrane (Millipore,

177

Bedford, MA, USA) and blocked with 5% Bovine Serum Albumin (BSA) overnight at

178

4 °C or 1 h at room temperature. Prestained protein markers (Fermentas, Waltham,

179

MA, USA) were used in each gel. Samples were incubated with primary antibodies

180

(1:1,000 dilution) against Membrane-associated PGRMC1 (Santa Cruz Biotechnology,

181

Santa Cruz, CA, USA), lamin A/C (Santa Cruz Biotechnology, Santa Cruz, CA, USA), 8

ACS Paragon Plus Environment

Page 8 of 36

Page 9 of 36

Journal of Agricultural and Food Chemistry

182

eNOS (Cell Signaling Technology, Beverly, MA, USA), and anti-β-actin antibody

183

(Santa Cruz Biotechnology, Santa Cruz, CA, USA). After being washed with

184

Tris-Tween 20 buffer (pH 7.4) for three times, membranes were incubated with the

185

HRP-conjugated secondary antibody (Zhongshan Gold Bridge, Beijing, China) for 1 h

186

at room temperature. The membrane was exposed by AlphaImager 2200 (Alpha

187

Innotech, San Leandro, CA, USA) automatically. Band densities were detected with

188

the Western Blot Luminance Reagent (Applygene, Beijing, China) and quantified

189

using AlphaImager 2200 (Alpha Innotech, San Leandro, CA, USA).

190

Statistical analysis

191

For results of reproductive performance, PGRMC1, eNOS, vimentin, and lamin A/C

192

expression of gilts and pTr cells, data were analyzed using MIXED procedures of

193

SAS (version 9.0; SAS Institute Inc., Cary, NC) following a randomized complete

194

block design. Sow was considered the experimental unit. Data for the number of

195

stillborn and mummified, and embryonic mortality were analyzed using the chi-square

196

test of SAS. A value of P < 0.05 was considered statistically significant. Statistical

197

analysis for metabolics results were performed with identified polar metabolites. The

198

student’s t test was evaluated by SAS. Principal component analysis (PCA) and

199

pathway analysis was completed using Metaboanalyst. Compared with the control

200

group, metabolites with a threshold of > 1.50- or < 0.55-fold, values of P < 0.1, and

201

VIP > 1.35 were considered differentially metabolites.

202

RESULTS

203

The reproductive performance of sows and serum concentration of amino acids

204

As shown in Table 2, dietary NCG supplementation during d 1-28 of pregnancy in

205

sows increased the number of total piglets born per litter and total piglets born alive 9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

206

per litter, compared with the control diet and NCG supplementation during d 1-8 or d

207

9-28 of pregnancy (P < 0.05). However, dietary NCG supplementation during three

208

different stages of early pregnancy had no effect on sows’ weight gain, number of

209

stillborn and mummified piglets or birth weight of piglets.

210

The metabolomic profiles in serum and amniotic fluid

211

To identify possible relationships within classes of data, PCA was employed.

212

Performing PCA on the statistics-filtered data set, distinct differences was observed

213

between the NCG group and control group, showing that NCG highly influences the

214

serum and amniotic fluid metabolome. Data were visualized by constructing PC

215

scores, where each point on the plot represented a single sample. In serum, PC1 and

216

PC2 represented 19.4% and 13.1% of the variation, respectively. The first two PCs

217

explained 32.5% of the total variances within the data (Fig. 1A). And in amniotic fluid,

218

PC1 and PC2 represented 8.2% and 8% of variation, respectively (Fig. 1B).

219

Metabolomes in serum from NCG and control groups were differentially

220

expressed (Table 3). To identify which variables account for such a significant

221

separation, we calculated the P value, VIP score (variable in project, the higher the

222

score, the more important it is) and fold change (FC), defined as the ratio of the NCG

223

group to the control group for a given expression profile. Taking the three aspects into

224

account, 19 metabolites are selectively shown in Table 3. Fifteen of the metabolites

225

detected were found to be up-regulated in NCG group, while 4 were down-regulated.

226

The detailed analysis of the most relevant pathways was performed by

227

MetaboAnalysis. The results showed that maternal NCG supplementation

228

up-regulated metabolites mainly involved in fatty acid metabolism, energy

229

metabolism and immunological regulation. 10

ACS Paragon Plus Environment

Page 10 of 36

Page 11 of 36

Journal of Agricultural and Food Chemistry

230

For amniotic fluid, 14 metabolites are selectively shown in Table 4. Ten of the

231

metabolites detected were found to be up-regulated in NCG group, while 4 were

232

down-regulated. The up-regulated metabolites are mainly involved in fatty acid

233

metabolism, energy metabolism and glutathione metabolism while down-regulated

234

metabolites are involved in cysteine metabolism and absorption of vitamins.

235

The protein abundance of PGRMC1, lamin A/C, and eNOS in the endometrium,

236

fetuses, and placentaeplacentae of gilts on d 28 of pregnancy

237

The protein abundance of PGRMC1, eNOS, and lamin A/C in placentae were

238

significantly increased (P < 0.05) in gilts fed the NCG supplemented diet on d 28 of

239

pregnancy, compared with those in gilts fed the control diet (Fig. 2A). Similarly, in

240

fetuses, dietary NCG supplementation significantly increased (P < 0.05) the

241

expression of PGRMC1, eNOS, and lamin A/C on d 28 of pregnancy, compared with

242

the control diet (Fig. 2B). In endometrium, NCG supplementation also dramatically

243

enhanced (P < 0.05) the expression of PGRMC1 and eNOS on d 28 of pregnancy,

244

compared with the control diet (Fig. 2C), while the expression of lamin A/C was not

245

affected.

246

The expression levels of PGRMC1, lamin A/C, eNOS, and vimentin in pTr cells

247

in the presence of arginine, proline, glutamine, or glutamate

248

Western blot analysis revealed that compared to the control treatment, a 24-h arginine

249

treatment markedly increased PGRMC1 expression in a dose dependent manner in

250

pTr cells, which peaked at 0.5 mM. Similarly, the expression levels of eNOS were

251

also dose-dependently increased in pTr cells, with the peak at 0.5 mM (Fig. 3A).

252

However, arginine had no effect on vimentin and lamin A/C expression level in pTr

253

cells (Fig. 3A). 11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

254

Glutamine also dose-dependently stimulated the expression levels of PGRMC1

255

and eNOS in pTr cells, in comparison with the control treatment, with the peak at 0.5

256

mM for PGRMC1, and 1.0 mM for eNOS (Fig. 3B). Glutamine treatment had no

257

obvious effect on the expression of lamin A/C and Vimentin (Fig. 3B).

258

Compared with the control treatment, glutamate stimulated the expression of

259

vimentin and lamin A/C in pTr cells, peaking at 0.25 mM and 0.50 mM, respectively

260

(Fig. 3C). However, glutamate had no effects on the expression of PGRMC1 and

261

eNOS in pTr cells (Fig. 3C).

262

A dose-dependent augmentation of lamin A/C was also observed in pTr cells

263

treated with proline (Fig. 3D). Proline markedly increased the expression levels of

264

lamin A/C at 0.25 mM treatment in pTr cells, compared to the control treatment (Fig.

265

3D). Proline treatment had no effects on the expression of PGRMC1, Vimentin, and

266

eNOS in pTr cells (Fig. 3D).

267

DISCUSSION

268

Our results showed that NCG supply during early pregnancy improved the pregnancy

269

outcome in sows, which indicated that the beneficial effects of NCG on embryonic

270

survival during early pregnancy subsequently improved pregnancy outcome in sows.

271

NCG supplementation had a similar response compared with dietary arginine

272

supplementation during early pregnancy in sows.18 However, NCG has unique

273

advantages over arginine because: (1) there is no impact on intestinal absorption of

274

dietary tryptophan, histidine, or lysine; (2) a low dose is highly effectivein stimulating

275

endogenous arginine synthesis; (3) it has a relatively long half-life in vivo (perhaps

276

8-10 h); and (4) substantially reduced cost.19-21 Therefore, dietary NCG

277

supplementation perhaps is a better choice than arginine to improve pregnancy

278

outcomes in mammals. 12

ACS Paragon Plus Environment

Page 12 of 36

Page 13 of 36

Journal of Agricultural and Food Chemistry

279

To further explore the mechanism of NCG in improving pregnancy outcomes,

280

metabolomic analysis in amniotic fluid and serum was conducted. The results

281

indicated that maternal NCG supplementation increased levels of metabolites in

282

amniotic fluids and serum primarily involved in energy metabolism, lipid metabolism,

283

glutathione metabolism, and immunological regulation. This is consistent with our

284

previous proteome results, which indicates that maternal NCG supplementation

285

modulates endometrial proteins associated with energy metabolism, lipid metabolism,

286

protein metabolism, antioxidative stress, and immune response.15 Energy metabolism

287

is crucially important to embryo growth, differentiation, and viability.22 The

288

reinforcement in energy metabolism probably provides more energy for embryonic

289

and fetal development. During pregnancy, oxidative stress has negative impacts on

290

embryo development, such as a decrease in the percentage of two-cell and blastocyst

291

stage embryos. Reactive oxygen species are generated from embryo metabolism

292

and/or exist in embryo surroundings, which cause embryonic growth retardation and

293

embryonic dysmorphogenesis.23,24 Glutathione status reflects the antioxidant capacity

294

of developing embryos. GSH reduces the formation of free oxygen radical species,

295

ultimately protecting embryos against oxidative stress.24,25 Our data indicated that

296

beta-citryl-L-glutamic acid level in amniotic fluids was increased which indicated that

297

glutathione metabolism was enhanced in NCG supplemented sows. This was

298

beneficial for embryo growth and protecting embryos against damage from free

299

radical oxygen species. Another notable finding of the metabolomics data in amniotic

300

fluids and serum was that immunological regulation pathways were up-regulated in

301

NCG supplemented sows during early pregnancy. During pregnancy, the maternal

302

uterus has tolerance to the semiallogenic fetus, and many immune cells (such as

303

macrophages, dendritic cells, and regulatory T cells) and molecules (such as 13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

304

Leukemia Inhibitory Factor and heme oxygenase-1) are involved in this process.26

305

Platelet-activating factor (PAF) may be used as an indicator of embryo viability and

306

pregnancy outcome prediction. It has been reported that PAF level is highly correlated

307

with pregnancy outcomes in human.27 PAF initiates early pregnancy factor synthesis,

308

which is recognized as an immunosuppressive factor and whose amount is highest on

309

d 30 of pregnancy in gilts.28,29 In this study, the improvement in pregnancy outcome

310

induced by maternal NCG supply could be associated with the increase in serum PAF

311

level. Collectively, alternations in levels of amniotic fluid and serum metabolites

312

involved in energy metabolism, lipid metabolism, glutathione metabolism, and

313

immunological regulation by maternal NCG supply were beneficial to embryonic

314

development, which ultimately enhances pregnancy outcomes in sows.

315

Our previous proteome data indicates that maternal NCG supplementation during

316

early pregnancy up-regulates factors related to embryo implantation and development.

317

15

318

targeted genes to validate the modulation of NCG on the expression of these genes in

319

vivo and in vitro. A recent study demonstrates that PGRMC1 is localized in oocytes,

320

embryos/fetuses, and uteri.30 PGRMC1 is different from classical nuclear PGRs

321

receptors. PGRMC1 and serpine 1 mRNA binding protein (SERBP1) form a complex

322

to bind progesterone, leading to the increase in cAMP levels and activation of protein

323

kinase G,31 to mediate anti-apoptosis and to modulate steroidogenesis and cellular

324

homeostasis.32,33 In bovine endometrium, PGRMC1 is co-expressed with SERBP1

325

and nuclear progesterone receptor (PGR) during the first trimester of pregnancy,

326

indicating PGRMC1 plays an important role in early pregnancy possesses.34

327

PGRMC1 is also expressed in uterus and placenta in mice and humans indicating its

328

potentially important role in cellular differentiation, modulation of apoptosis and

In the present study, PGRMC1, NOS, vimentin, and lamin A/C were chosen as

14

ACS Paragon Plus Environment

Page 14 of 36

Page 15 of 36

Journal of Agricultural and Food Chemistry

329

steroidogenesis during early pregnancy.35 In the current study, the enhanced

330

expression of PGRMC1 observed in endometrium, fetuses, and placentae in NCG

331

supplemented gilts on d 28 of pregnancy indicates that maternal NCG supply might

332

improve the development of endometrium, fetuses, and placentae through modulation

333

of local PGRMC1 expression. The in vitro results showed only arginine and

334

glutamine but not glutamate and proline remarkably induced PGRMC1 expression in

335

pTr cells which indicates that arginine and glutamine are the key metabolites related

336

to PGRMC1 stimulation by maternal NCG supply.

337

Three nitric oxide synthesases (NOS), including neuronal nitric oxide

338

synthesases (nNOS), eNOS, and inducible nitric oxide synthesases (iNOS) are

339

involved in nitro oxide synthesis from arginine in the body.36 NO modulates embryo

340

implantation through remodeling of the extracellular matrix,37 and plays critical roles

341

in placental vascular development.38 NOS expression in the uterus is beneficial for

342

endometrial decidualization and embryo implantation.39 Low expression of eNOS

343

increases embryo loss and decreases embryo implantation sites in the eNOS-knockout

344

mouse model.40 In contrast, high expression of eNOS in fetuses and placentae is

345

benefical for nutrient transfer from mother to fetus and efficient utilization of these

346

nutrients.41 Our results showed that arginine and glutamine supplementation increased

347

eNOS expression in pTr cells. We speculate that dietary NCG supplementation during

348

early pregnancy might improve embryo implantation through up-regulation of eNOS

349

expression induced by arginine and glutamine in pTr cells. NOSs, also highly

350

expressed in placenta, influence the placental vasculogenesis and angiogenesis

351

through modulation of VEGF and angiopoietin signaling pathways.38 We found that

352

eNOS expression was differentially modulated in uterus, fetus, and placenta in

353

response to maternal NCG supply in early pregnancy, with relatively stronger 15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

354

stimulation in the placenta and fetus than that in the uterus. These data indicate that

355

maternal NCG supply exerted a stronger influence on the development of fetuses and

356

placentae, compared with the uterus.

357

Vimentin, one of the intermediate filament proteins, plays pivotal roles in cell

358

migration.42 Trophoblastic giant cells (TGC) exert critical roles in embryo and fetal

359

development through interaction with decidual cells and the extracellular matrix.43-45

360

Vimentin is found to be expressed in mice trophoblastic giant cells, which is benefical

361

for development of the chorioallantoic placenta.46,47 Our data indicated that glutamate

362

enhanced vimentin expression in pTr cells. Serum glutamate concentration was

363

reported to increase by NCG supplementation.15 Therefore, maternal NCG supply

364

appears to benefit embryo and fetal development partially through modulation of

365

vimentin expression in pTr cells. Lamin A/C is expressed in mouse embryonic stem

366

cells and developing blastocyst.48 Lamin A/C mediates cell mechanics, polarization,

367

and migration 49 and is important for determination of cell fate.50 The increased lamin

368

A/C expression in fetuses and placentae could be benefical for the early development

369

of embryos in response to maternal NCG supply.

370

In conclusion, maternal NCG supply during early pregnancy enchanced

371

pregnancy outcomes in sows through differential modulation of factors related to

372

embryo implantation and development in endometrium, fetuses, and placentae, as well

373

as the regulation of metabolites in amniotic fluids and serum, which are primarily

374

involved in energy metabolism, lipid metabolism, glutathione metabolism, and

375

immunological regulation. These findings imply that NCG has great potential to

376

improve pregnancy outcomes in humans and mammalian animals.

16

ACS Paragon Plus Environment

Page 16 of 36

Page 17 of 36

Journal of Agricultural and Food Chemistry

377

AUTHOR INFORMATION

378

Corresponding Authors

379

*(X.Z.)

380

[email protected].

381

Finding

382

This work was supported by National Natural Science Foundation of the P. R of China

383

(NO. 31301980 and NO. 31772614).

384

Notes

385

The authors declare no competing financial interest.

Phone:

+86

10-62733588.

Fax:

+86

10-62733688.

E-mail:

386 387

ABBREVIATIONS USED

388

eNOS, endothelial nitric oxide synthesases; FC, fold change; iNOS, inducible nitric

389

oxide synthesases; ITS, insulin–transferrin–selenium; IVF, in vitro fertilization; NCG,

390

N-carbamylglutamate; nNOS, neuronal nitric oxide synthesases; NOS, nitric oxide

391

synthesases; PAF, Platelet-activating factor; PCA, Principal component analysis; PGR,

392

progesterone receptor; PGRMC1, progesterone receptor membrane component 1; pTr,

393

porcine trophectoderm; SERBP1, serpine 1 mRNA binding protein; TGC,

394

Trophoblastic giant cells; VIP, variable in project

17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

395

Refercences

396

(1) Koot, Y. E. M.; Teklenburg, G.; Salker, M. S.; Brosens, J. J.; Macklon, N. S.

397

Molecular aspects of implantation failure. Biochim. Biophys. Acta. 2010, 1822,

398

1943-1950.

399

(2) Goff, A.K. Embryonic signals and survival. Reprod. Domest. Anim. 2002, 37,

400

133-139.

401

(3) McNamee, K. F.; Dawood, F.; Farquharson, R. G. Thrombophilia and early

402

pregnancy loss. Best. Pract. Res. Clin. Obstet. Gynaecol. 2012, 26, 91-102.

403

(4) Bazer, F.W.; Wu, G.; Johnson, G.A.; Kim, J.; Song, G. Uterine histotroph and

404

conceptus development: select nutrients and secreted phosphoprotein 1 affect

405

mechanistic target of rapamycin cell signaling in ewes. Biol. Reprod. 2011,

406

85,1094-1107.

407

(5) Cross, J.C.; Mickelson, L. Nutritional influences on implantation and placental

408

development. Nutr. Rev. 2006, 64, S12-S18.

409

(6) Ren, W. K.; Luo, W.; Wu, M. M; Liu, G.; Yu, X. L.; Fang, J.; Li, T. J.; Yin, Y. L.;

410

Wu G. Y. Dietary L-glutamine supplementation improves pregnancy outcome in mice

411

infected with type-2 porcine circovirus. Amino. Acids. 2013, 45, 479–488.

412

(7) Wu, X.; Xie, C. Y.; Zhang, Y. Z.; Fan, Z. Y.; Yin, Y. L.; Blachier, F. Glutamate–

413

glutamine cycle and exchange in the placenta–fetus unit during late pregnancy. Amino.

414

Acids. 2015, 47,45-53.

415

(8) Santana, P. D.; Silva, T. V.; Da, C. N.; Da, S. B.; Carter, T. F.; Cordeiro, M. S. Da,

416

S. B.; Santos, S. S.; Herculano, A. M.; Adona, P. R.; Ohashi, O. M.; Miranda, M. S.

417

Supplementation of bovine embryo culture medium with L-arginine improves embryo

418

quality via nitric oxide production. Mol. Reprod. Dev. 2014, 81, 918-927. 18

ACS Paragon Plus Environment

Page 18 of 36

Page 19 of 36

Journal of Agricultural and Food Chemistry

419

(9) Wu, G. Y.; Bazer, F. W.; Satterfield, M. C.; Li, X. L.; Wang, X. Q.; Johnson, G.

420

A.; Burghardt, R. C.; Dai, Z. L.; Wang, J. J.; Wu, Z. L. Impacts of arginine nutrition

421

on embryonic and fetal development in mammals. Amino. Acids. 2013, 45, 241-256.

422

(10) Wu, X.; Yin, Y. L.; Liu, Y. Q.; Liu, X. D.; Liu, Z. D.; Li, T. J.; Huang, R. L.;

423

Ruan, Z.; Deng, Z. Y. Effect of dietary arginine and N-carbamoylglutamate

424

supplementation reproduction and gene expression of eNOS, VEGFA and PlGF1 in s

425

placenta in late pregnancy of sows. Anim. Reprod. Sci. 2012, 132, 187-192.

426

(11) Zeng, X. F.; Mao, X. B.; Huang, Z. M.; Wang, F. L.; Wu, G. Y.; Qiao, S. Y.

427

Arginine enhances embryo implantation in rats through PI3K/PKB/mTOR/NO

428

signaling pathway during early pregnancy. Reproduction. 2013, 145, 1-7.

429

(12) Zeng, X. F.; Wang, F. L.; Fan, X; Yang, W. J.; Zhou, B.; Li, P. F.; Yin, Y. L.;

430

Wu, G. Y.; Wang, J. J. Dietary arginine supplementation during early pregnancy

431

enhances embryonic survival in rats. J. Nutr. 2008, 138, 1421-1425.

432

(13) Zeng, X. F.; Huang, Z. M.; Mao, X. B.; Wang, J. J.; Wu, G. Y.; Qiao, S. Y.

433

N-carbamylglutamate enhances pregnancy outcome in rats through activation of the

434

PI3K/PKB/mTOR signaling pathway. PLos. One. 2012, 7, e41192.

435

(14) Liu, X. D.; Wu, X.; Yin, Y. L.; Liu, Y. Q.; Geng, M. M.; Yang, H. S.; Blachier,

436

F., Wu, G. Y. Effects of dietary L-arginine or N-carbamylglutamate supplementation

437

during late gestation of sows on the miR-15b/16, miR-221/222, VEGFA and Enos

438

expression in umbilical vein. Amino. Acids. 2012, 42, 2111-2119.

439

(15) Zhu, J. L.; Zeng, X. F.; Peng, Q.; Zeng, S. M.; Zhao, H. Y.; Shen, H. X.; Qiao, S.

440

Y. Maternal N-carbamylglutamate supplementation during early pregnancy enhances

441

embryonic survival and development through modulation of the endometrial proteome

442

in gilts. J. Nutr. 2015, 145, 2212-2220. 19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

443

(16) Zhang, B.; Che, L.Q.; Lin, Y.; Zhuo, Y.; Fang, Z. F.; Xu, S. Y.; Song, J.; Wang,

444

Y. S.; Liu, Y.; Wang, P. Wu, D. Effect of dietary N-carbamylglutamate levels on

445

reproductive performance of gilts. Reprod. Domest. Anim. 2014, 49, 740-745.

446

(17) Bazer, F. W.; Song, G.; Kim, J.; Dunlap, K. A.; Satterfield, M. C.; Johnson, G.

447

A.; Burghardt, R. C.; Wu, G. Y. Uterine biology in pigs and sheep. J. Anim. Sci.

448

Biotechnol. 2012, 3, 23.

449

(18) Li, J.; Xia, H.; Yao, W.; Wang, T. T.; Li, J. L.; Piao, X. S.; Thacker, P.; Wu, G.

450

Y.; Wang, F. L. Effects of arginine supplementation during early gestation (day 1 to

451

30) on litter size and plasma metabolites in gilts and sows. J. Anim. Sci. 2015, 93,

452

5291-303.

453

(19) Wu, G. Y.; Knabe, D. A.; Kim, S. W. Arginine nutrition in neonatal pigs. J. Nutr.

454

2004, 134, 2783S-2790S.

455

(20) Geng, M. M.; Li, T. J.; Kong, X. F.; Song, X. Y.; Chu, W. Y.; Huang, R. L.; Wu,

456

G. Y. Reduced expression of intestinal N-acetylglutamate synthase in suckling piglets:

457

a novel molecular mechanism for arginine as a nutritionally essential amino acid for

458

neonates. Amino. Acids. 2011, 40, 1513- 1522.

459

(21) Liu, Z. Q.; Geng, M. M.; Shu, X. G.; Wang, W. C.; Yin, Y. L. Dietary NCG

460

supplementation enhances the expression of N-acetylglutamate synthase in intestine

461

of weaning pig. J. Food. Agric. Enviro. 2012, 10, 408-412.

462

(22) Gardner, D. K.; Pool, T. B.; Lane, M. Embryo nutrition and energy metabolism

463

and its relationship to embryo growth, differentiation, and viability. Semin. Reprod.

464

Med. 2000, 18, 205-18.

465

(23) Guerin, P.; El, M. S.; Menezo, Y. Oxidative stress and protection against reactive

466

oxygen species in the pre-implantation embryo and its surroundings. Hum. Reprod.

467

Update. 2001, 7, 175-189. 20

ACS Paragon Plus Environment

Page 20 of 36

Page 21 of 36

Journal of Agricultural and Food Chemistry

468

(24) Trocino, R. A.; Akazawa, S.; Ishibashi, M.; Matsumoto, K.; Matsuo, H.;

469

Yamamoto, H.; Goto, S.; Urata, Y.; Kondo, T.; Nagataki, S. Significance of

470

glutathione depletion and oxidative stress in early embryogenesis in glucose-induced

471

rat embryo culture. Diabetes. 1995, 44, 992-998.

472

(25) Yin, J.; Ren, W.; Yang, G.; Duan, J.; Huan, X.; Fang, R.; Li, C.; Yin, Y.; Hou, Y.;

473

Kim, S. W.; Wu, G. L-Cysteine metabolism and its nutritional implications. Mol. Nutr.

474

Food. Res. 2016, 60, 134-146.

475

(26) Leber, A.; Zenclussen, M. L.; Teles, A.; Brachwitz, N.; Casalis, P.; El, M. T.;

476

Jensen, F.; Woidacki, K.; Zenclussen, A. C. Pregnancy: tolerance and suppression of

477

immune responses. Methods. Mol. Biol. 2011, 677, 397-417.

478

(27) Roudebush, W. E.; Wininger, J. D.; Jones, A. E.; Wright, G.; Toledo, A. A.; Kort,

479

H. I.; Massey, J. B.; Shapiro, D. B. Embryonic platelet-activating factor: an indicator

480

of embryo viability. Hum. Reprod. 2002, 17, 1306-1310.

481

(28) Grosso, M. C.; Bellingeri, R. V.; Motta, C. E.; Alustiza, F. E.; Picco, N. Y.;

482

Vivas, A. B. Immunohistochemical distribution of early pregnancy factor in ovary,

483

oviduct and placenta of pregnant gilts. Biotech. Histochem. 2015, 90, 14-24.

484

(29) Lash, G. E.; Legge, M. Induction of early pregnancy factor activity in vitro by

485

platelet-activating factor in mice. J. Reprod. Fertil. 1997, 109, 187-191.

486

(30) Luciano, A. M.; Lodde, V.; Franciosi, F.; Ceciliani, F.; Peluso, J. J. Progesterone

487

receptor membrane component 1 expression and putative function in bovine oocyte

488

maturation, fertilization, and early embryonic development. Reproduction. 2010, 140,

489

663-672.

490

(31) Peluso, J. J. Multiplicity of progesterone's actions and receptors in the

491

mammalian ovary. Biol. Reprod. 2006, 75, 2-8.

492

(32) Rohe, H. J.; Ahmed, I. S.; Twist, K. E.; Craven, R. J. PGRMC1 (progesterone 21

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

493

receptor membrane component 1): a targetable protein with multiple functions in

494

steroid signaling, P450 activation and drug binding. Pharmacol. Ther. 2009, 121,

495

14-9.

496

(33) Peluso, J. J.; Romak, J.; Liu, X. Progesterone receptor membrane component-1

497

(PGRMC1) is the mediator of progesterone’s antiapoptotic action in spontaneously

498

immortalized granulosa cells as revealed by PGRMC1 small interfering ribonucleic

499

acid treatment and functional analysis of PGRMC1 mutations. Endocrinology. 2008,

500

149, 534-54.

501

(34) Kowalik, M. K.; Slonina, D.; Rekawiecki, R.; Kotwica, J. Expression of

502

progesterone receptor membrane component (PGRMC) 1 and 2, serpine mRNA

503

binding protein 1 (SERBP1) and nuclear progesterone receptor (PGR) in the bovine

504

endometrium during the estrous cycle and the first trimester of pregnancy. Reprod.

505

Biol. 2013, 13, 5-23.

506

(35) Zhang, L.; Kanda, Y.; Roberts, D. J.; Ecker, J. L.; Losel, R.; Wehling, M.; Peluso,

507

J. J.; Pru, J. K. Expression of progesterone receptor membrane component 1 and its

508

partner serpine 1 mRNA binding protein in uterine and placental tissues of the mouse

509

and human. Mol. Cell. Endocrinol. 2008, 287, 81-89.

510

(36) Rosselli, M.; Keller, R. J.; Dubey, R. K. Role of nitric oxide in the biology,

511

physiology and pathophysiology of reproduction. Hum. Reprod. Update. 1998, 4,

512

3-24.

513

(37) Hansson, S. R.; Nääv, Å.; Erlandsson, L. Oxidative stress in preeclampsia and

514

the role of free fetal hemoglobin. Front. Physiol. 2015, 5, 516.

515

(38) Krause, B. J.; Hanson, M. A.; Casanello, P. Role of nitric oxide in placental

516

vascular development and function. Placenta. 2011, 32, 797-805.

517

(39) Biswas, S.; Kabir, S. N.; Pal, A. K. The role of nitric oxide in the process of 22

ACS Paragon Plus Environment

Page 22 of 36

Page 23 of 36

Journal of Agricultural and Food Chemistry

518

implantation in rats. J. Reprod. fertil. 1998, 114, 157-161.

519

(40) Pallares, P.; Garcia, F. R.; Criado, L. M.; Letelier, C. A.; Esteban, D.; Fernandez,

520

T. J.; Flores, J. M.; Gonzalez, B. A. Disruption of the endothelial nitric oxide synthase

521

gene affects ovulation, fertilization and early embryo survival in a knockout mouse

522

model. Reproduction. 2008, 136, 573-579.

523

(41) Wu, G.; Bazer, F. W.; Wallace, J. M.; Spencer, T. E. Board-invited review:

524

intrauterine growth retardation: implications for the animal sciences. J. Anim. Sci.

525

2006, 84, 2316-2337

526

(42) Chernoivanenko, I. S.; Minin, A. A.; Minin, A. A. Role of vimentin in cell

527

migration. Ontogenez. 2013, 44, 186-202.

528

(43) Katz, S. G. Extracellular breakdown of collagen by mice decidual cells. A

529

cytochemical and ultrastructural study. Biocell. 2005, 29, 261-270.

530

(44) Katz, S.G. Extracellular and intracellular degradation of collagen by trophoblast

531

giant cells in acute fasted mice examined by electron microscopy. Tissue. Cell. 1995,

532

27, 713-721.

533

(45) Welsh, A. O.; Enders, A. C. Chorioallantoic placenta formation in the rat: II.

534

Angiogenesis and maternal blood circulation in the mesometrial region of the

535

implantation chamber prior to placenta formation. Am. J. Anat. 1991, 192, 347-365.

536

(46) Scherholz, P. L.; Souza, D. P.; Spadacci, M. D.; Katz, S. G. Vimentin is

537

synthesized by mouse vascular trophoblast giant cells from embryonic day 7.5

538

onwards and is a characteristic factor of these cells. Placenta. 2013, 34, 518-525.

539

(47) De, S. P.; Katz, S. G. Coexpression of cytokeratin and vimentin in mice

540

trophoblastic giant cells. Tissue. Cell. 2001, 33, 40-45.

541

(48) Eckersley, M. M.; Bergmann, J. H.; Lazar, Z.; Spector, D. L. Lamin A/C is

542

expressed in pluripotent mouse embryonic stem cells. Nucleus. 2013, 4, 53-60. 23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

543

(49) Lee, J. S.; Hale, C. M.; Panorchan, P.; Khatau, S. B.; George, J. P.; Tseng, Y.;

544

Stewart, C. L.; Hodzic, D.; Wirtz, D. Nuclear lamin A/C deficiency induces defects in

545

cell mechanics, polarization, and migration. Biophys. J. 2007, 93, 2542-2552

546

(50) Rober, R. A.; Weber, K.; Osborn, M. Differential timing of nuclear lamin A/C

547

expression in the various organs of the mouse embryo and the young animal: a

548

developmental study. Development. 1989, 105, 365-378.

24

ACS Paragon Plus Environment

Page 24 of 36

Page 25 of 36

Journal of Agricultural and Food Chemistry

549

Table 1 Composition and nutrient composition of the basal diet for gestating sows

550

(as-fed basis) Item

Content

Ingredients, % Corn grain

66.10

Wheat bran

19.50

Soybean meal, 43%

10.00

Soybean oil

0.50

Limestone

0.60

Dicalcium phosphate

2.10

Salt

0.40

L-Lysine hydrochloride

0.10

DL-Methionine

0.05

Choline chloride

0.15

Vitamin mineral premix1

0.50

Nutrient levels, % Dry matter2

89.60

Digestible energy3, Mcal/kg

3.07

Metabolizable energy3, Mcal/kg

2.93

Crude protein2

13.10

Calcium2

0.79

Available phosphorus3

0.49

Total phosphorus3

0.69

Lysine3

0.64

Arginine3

0.66

Methionine + cystine3

0.50

551

1

552

cholecalciferol, 0.04; d-α-tocopheryl acetate, 10.10; menadione sodium bisulfate, 1.60;

553

thiamine, 1.50; riboflavin, 3.00; vitamin B-6, 1.50; vitamin B-12, 0.01; niacin, 22.50;

Provided the following (mg/kg of the basal diet): retinyl acetate, 3.47;

25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

554

calcium pantothenate, 15.00; folic acid, 2.50; biotin, 0.20; manganese, 40.00; iron,

555

85.00; zinc, 75.00; copper, 1.50; iodine, 0.09; and selenium, 0.03.

556

2

Analyzed values.

557

3

Calculated values.

26

ACS Paragon Plus Environment

Page 26 of 36

Page 27 of 36

Journal of Agricultural and Food Chemistry

558

Table 2 Effects of N-Carbamylglutamate (NCG) supplementation on reproductive

559

performance in sows Treatment1

SEM2 P value

Items Weight at breeding (kg)

Control

NCG 1 NCG 2 NCG 3

216.78

215.00

204.72

207.88

10.91

0.85

225.17

222.81

213.11

217.24

10.93

0.87

8.39

7.81

8.39

9.35

0.62

0.40

4.00

3.88

4.00

3.83

0.46

0.99

11.31ab 11.67ab 12.18a

0.46

0.04

10.70a

0.27

0.02

Weight at day 28 of gestation (kg) Weight gain through d 28 of gestaion(kg) Parity Total piglets born per litter Total piglets born alive per

11.11b 9.61b

9.81ab 10.39a

1.50

1.50

1.33

1.53

0.20

0.89

13.32

13.13

11.30

12.43

1.58

0.80

13.73

14.38

15.15

15.43

0.62

0.23

1.44

1.45

1.43

1.40

0.05

0.94

litter Stillborn and mummified (n) Fetal mortality (%)3 Litter birth weight of all piglets born alive (kg) Weight of piglets (kg) 560

Note: 1the numbers of sows for Control group, NCG 1, NCG 2 and NCG 3 were 18,

561

18, 16, and 17, respectively. a, b within a row, significant difference at P < 0.05.

562

2

SEM, Standard error of the mean.

563

3

Fetal mortality = number of stillborn and mummified piglets/number of total

564

piglets born per litter.

27

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 28 of 36

565

Table 3 Serum metabolites that differed between control and N-Carbamylglutamate

566

groups Metabolite

Fold

VIP

P

change1

score

value

3.31

2.90

0.02

Related pathway Cysteine and methionine

Indoleacrylic acid

metabolism Isohyodeoxycholic acid

3.23

2.42

0.00

Platelet activating factor

2.75

1.72

0.05

Lipid metabolism Immunological regulation

Isofucosterol 3-O-6-O-9,12-octadecadi 2.69

2.27

0.01

Lipid metabolism

2.64

1.94

0.01

Hydrolysis of xenobiotic

enoyl -b-d-glucopyranoside Diisobutyl phthalate

Energy metabolism and Retapamulin

2.42

2.77

0.02

immunological regulation

Polysorbate 60

2.25

1.99

0.01

Fatty acid metabolism

Oleamide

2.20

1.65

0.02

Energy metabolism

Benzyl isothiocyanate

2.05

1.74

0.04

Glucosinolate breakdown

Pyridostigmine

2.09

1.70

0.03

Cholinesterase Inhibitors

1.89

1.35

0.04

Energy metabolism

1.88

1.56

0.02

3-cis-hydroxy-b,e-carote n-3'-one 3-hydroxyphenylpyruvic

Tyrosine catabolism

acid

pathway Phenylalanine

Tiropramide

1.84

1.73

0.00 metabolism

Cholesteryl acetate

1.53

1.47

0.04

Steroid biosynthesis

3alpha-acetomethoxy-11

1.60

1.52

0.04

Energy metabolism

28

ACS Paragon Plus Environment

Page 29 of 36

Journal of Agricultural and Food Chemistry

alpha-oxo-12-ursen-24-o ic acid Dilauryl 0.41

1.89

0.04

Redox reaction

0.32

1.76

0.04

Redox reaction

0.43

2.85

0.02

Purine metabolism

3,3'-thiodipropionate O-tyrosine Guanosine hexaphosphate adenosine 567

Note: 1Fold change, which was based on the normalized data, was defined as the fold

568

difference in the observed concentrations between NCG group and control group.

569

2

570

markers for distinguishing the two groups. The higher the score is, the more

571

important the metabolite is.

VIP, variable in project, was used to select distinct variables as potential

29

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 30 of 36

572

Table 4 Metabolites that differed in amniotic fluid between control and

573

N-Carbamylglutamate groups Fold

VIP

P

change1

score

value

12- ketodeoxycholic acid

6.15

3.67

0.00

Fatty acid metabolism

Diisobutyl phthalate

4.30

2.70

0.00

Hydrolysis of xenobiotic

Beta-citryl-l-glutamic acid

3.14

2.57

0.03

Glutathione metabolism

Cladribine

2.92

2.00

0.02

Metabolite name

Related pathway

Immunological suppression Ethyl aconitate

2.41

2.20

0.01

Pristanal

2.15

2.05

0.01

Tricarboxylic acid cycle Oxidation of branched chain fatty acids

Lysopc100

1.94

1.94

0.03

Energy metabolism

9-hexadecenoylcholine

1.77

1.60

0.03

Fatty acid metabolism

1.75

2.76

0.04

1-Pyrroline-4-hydroxy-2-car

Arginine and proline

boxylate

metabolism

Polyoxyethylene 600 1.54

1.50

0.02

Energy metabolism

Benzyl thiocyanate

0.49

1.88

0.02

Cysteine metabolism

Pyroglutamic acid

0.49

2.26

0.02

Glutathione metabolism

Umbelliferone

0.55

1.89

0.03

monoricinoleate

Biosynthesis of phenylpropanoids Metabolism of 2-Nonadecanone

0.45

2.12

0.05 carbohydrates

1

574

Note: Fold change, which was based on the normalized data, was defined as the fold

575

difference in the observed concentrations between NCG group and control group.

576

2

VIP, variable in project, was used to select distinct variables as potential

30

ACS Paragon Plus Environment

Page 31 of 36

Journal of Agricultural and Food Chemistry

577

markers for distinguishing the two groups. The higher the score is, the more

578

important the metabolite is.

31

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

579

Figure legends

580

Figure1. Principal component analysis (PCA). Each point represents an individual

581

sample. (A) PCA of the profiling data from the serum metabolome. (B) PCA of the

582

profiling data from amniotic fluid metabolome. Gilts were fed the control or 0.05%

583

N-Carbamylglutamate supplemented diets during d 1-28 of pregnancy.

584

Figure 2. The protein abundance of PGRMC1, eNOS, and lamin A/C expression in

585

placentae (A), fetuses (B), and endometriums (C) of gilts on d 28 of pregnancy. Gilts

586

were fed the control or 0.05% N-Carbamylglutamate supplemented diets during d

587

1-28 of pregnancy. On day 28 of gestation, the placenta and endometrium were

588

carefully isolated from the uterus of individual fetuses. All tissue samples were

589

immediately snap frozen in liquid nitrogen and stored at -80℃.

590

Figure 3. The protein abundance of PGRMC1, eNOS, lamin A/C, and vimentin in

591

poecine Tr cells treated with arginine (A), glutamine (B), glutamate (C), and proline

592

(D). Cells were serum starved overnight in customized medium, deprived of either

593

arginine, proline, glutamate or glutamine, and then treated with different doses (0,

594

0.25, 0.50, 1.00 mM) of Arg, Pro, Glu or Gln for 24 h. All experiments were repeated

595

at least 2-3 times.

32

ACS Paragon Plus Environment

Page 32 of 36

Page 33 of 36

Journal of Agricultural and Food Chemistry

596

Figure 1

597

33

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

598

Figure 2

34

ACS Paragon Plus Environment

Page 34 of 36

Page 35 of 36

Journal of Agricultural and Food Chemistry

599

Figure 3

35

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

600

TOC Graphic

36

ACS Paragon Plus Environment

Page 36 of 36