Longitudinal Changes of One-Carbon Metabolites and Amino Acid Concentrations during Pregnancy in the Women First Maternal Nutrition Trial

Stephanie P Gilley, Nicholas E Weaver, Evan L Sticca, Purevsuren Jambal, Alexandra Palacios, Mattie E Kerns, Pratibha Anand, Jennifer F Kemp, Jamie E Westcott, Lester Figueroa, Ana Lucía Garcés, Sumera A Ali, Omrana Pasha, Sarah Saleem, K Michael Hambidge, Audrey E Hendricks, Nancy F Krebs, Sarah J Borengasser, Stephanie P Gilley, Nicholas E Weaver, Evan L Sticca, Purevsuren Jambal, Alexandra Palacios, Mattie E Kerns, Pratibha Anand, Jennifer F Kemp, Jamie E Westcott, Lester Figueroa, Ana Lucía Garcés, Sumera A Ali, Omrana Pasha, Sarah Saleem, K Michael Hambidge, Audrey E Hendricks, Nancy F Krebs, Sarah J Borengasser

Abstract

Background: Maternal dietary restriction and supplementation of one-carbon (1C) metabolites can impact offspring growth and DNA methylation. However, longitudinal research of 1C metabolite and amino acid (AA) concentrations over the reproductive cycle of human pregnancy is limited.

Objective: To investigate longitudinal 1C metabolite and AA concentrations prior to and during pregnancy and the effects of a small-quantity lipid-based nutrition supplement (LNS) containing >20 micronutrients and prepregnancy BMI (ppBMI).

Methods: This study was an ancillary study of the Women First Trial (NCT01883193, clinicaltrials.gov) focused on a subset of Guatemalan women (n = 134), 49% of whom entered pregnancy with a BMI ≥25 kg/m2. Ninety-five women received LNS during pregnancy (+LNS group), while the remainder did not (-LNS group). A subset of women from the Pakistan study site (n = 179) were used as a replication cohort, 124 of whom received LNS. Maternal blood was longitudinally collected on dried blood spot (DBS) cards at preconception, and at 12 and 34 wk gestation. A targeted metabolomics assay was performed on DBS samples at each time point using LC-MS/MS. Longitudinal analyses were performed using linear mixed modeling to investigate the influence of time, LNS, and ppBMI.

Results: Concentrations of 23 of 27 metabolites, including betaine, choline, and serine, changed from preconception across gestation after application of a Bonferroni multiple testing correction (P < 0.00185). Sixteen of those metabolites showed similar changes in the replication cohort. Asymmetric and symmetric dimethylarginine were decreased by LNS in the participants from Guatemala. Only tyrosine was statistically associated with ppBMI at both study sites.

Conclusions: Time influenced most 1C metabolite and AA concentrations with a high degree of similarity between the 2 diverse study populations. These patterns were not significantly altered by LNS consumption or ppBMI. Future investigations will focus on 1C metabolite changes associated with infant outcomes, including DNA methylation. This trial was registered at clinicaltrials.gov as NCT01883193.

Keywords: BMI; amino acids; malnutrition; obesity; one-carbon metabolism; preconception; pregnancy; supplementation; triple nutrition burden.

Copyright © The Author(s) 2019.

Figures

FIGURE 1
FIGURE 1
Study and analysis design schematic. At study enrollment women were randomized into 1 of 3 study arms. For the present analysis, blood draws that occurred when the subjects were not consuming LNS (represented by unfilled blood drops) were considered unsupplemented time points; blood draws that occurred when the subjects had taken LNS for at least 12 wk (represented by solid blood drops) were considered supplemented time points. Arms 1 and 2 were combined and represent the +LNS group, while Arm 3 comprised the −LNS group. LNS, small-quantity lipid-based nutrition supplement.

References

    1. Peng W, Berry EM. Global nutrition 1990–2015: a shrinking hungry, and expanding fat world. PLoS One. 2018;13(3):e0194821.
    1. Black RE, Victora CG, Walker SP, Bhutta ZA, Christian P, de Onis M, Ezzati M, Grantham-McGregor S, Katz J, Martorell R et al. .. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet. 2013;382(9890):427–51.
    1. Prentice AM. The double burden of malnutrition in countries passing through the economic transition. Ann Nutr Metab. 2018;72(Suppl 3):47–54.
    1. Corvalan C, Garmendia ML, Jones-Smith J, Lutter CK, Miranda JJ, Pedraza LS, Popkin BM, Ramirez-Zea M, Salvo D, Stein AD. Nutrition status of children in Latin America. Obes Rev. 2017;18(Suppl 2):7–18.
    1. Ramirez-Zea M, Kroker-Lobos MF, Close-Fernandez R, Kanter R. The double burden of malnutrition in indigenous and nonindigenous Guatemalan populations. Am J Clin Nutr. 2014;100(6):1644S–51S.
    1. McGee M, Bainbridge S, Fontaine-Bisson B. A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr Rev. 2018;76(6):469–78.
    1. Painter RC, de Rooij SR, Bossuyt PM, Simmers TA, Osmond C, Barker DJ, Bleker OP, Roseboom TJ. Early onset of coronary artery disease after prenatal exposure to the Dutch famine. Am J Clin Nutr. 2006;84(2):322–7.; quiz 466–7.
    1. Barker DJ, Thornburg KL. The obstetric origins of health for a lifetime. Clin Obstet Gynecol. 2013;56(3):511–9.
    1. Hsu CN, Tain YL. The double-edged sword effects of maternal nutrition in the developmental programming of hypertension. Nutrients. 2018;10(12):E1917.
    1. Voerman E, Santos S, Patro Golab B, Amiano P, Ballester F, Barros H, Bergstrom A, Charles MA, Chatzi L, Chevrier C et al. .. Maternal body mass index, gestational weight gain, and the risk of overweight and obesity across childhood: an individual participant data meta-analysis. PLoS Med. 2019;16(2):e1002744.
    1. Hoffman DJ, Reynolds RM, Hardy DB. Developmental origins of health and disease: current knowledge and potential mechanisms. Nutr Rev. 2017;75(12):951–70.
    1. Dominguez-Salas P, Cox SE, Prentice AM, Hennig BJ, Moore SE. Maternal nutritional status, C(1) metabolism and offspring DNA methylation: a review of current evidence in human subjects. Proc Nutr Soc. 2012;71(1):154–65.
    1. Das JK, Hoodbhoy Z, Salam RA, Bhutta AZ, Valenzuela-Rubio NG, Weise Prinzo Z, Bhutta ZA. Lipid-based nutrient supplements for maternal, birth, and infant developmental outcomes. Cochrane Database Syst Rev. 2018;8:CD012610.
    1. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr. 2007;27:363–88.
    1. James P, Sajjadi S, Tomar AS, Saffari A, Fall CHD, Prentice AM, Shrestha S, Issarapu P, Yadav DK, Kaur L et al. .. Candidate genes linking maternal nutrient exposure to offspring health via DNA methylation: a review of existing evidence in humans with specific focus on one-carbon metabolism. Int J Epidemiol. 2018;47(6):1910–37.
    1. Dominguez-Salas P, Moore SE, Baker MS, Bergen AW, Cox SE, Dyer RA, Fulford AJ, Guan Y, Laritsky E, Silver MJ et al. .. Maternal nutrition at conception modulates DNA methylation of human metastable epialleles. Nat Commun. 2014;5:3746.
    1. Waterland RA, Kellermayer R, Laritsky E, Rayco-Solon P, Harris RA, Travisano M, Zhang W, Torskaya MS, Zhang J, Shen L et al. .. Season of conception in rural Gambia affects DNA methylation at putative human metastable epialleles. PLoS Genet. 2010;6(12):e1001252.
    1. James PT, Dominguez-Salas P, Hennig BJ, Moore SE, Prentice AM, Silver MJ. Maternal one-carbon metabolism and infant DNA methylation between contrasting seasonal environments: a case study from The Gambia. Curr Dev Nutr. 2019;3(1):nzy082.
    1. Steegers-Theunissen RP, Twigt J, Pestinger V, Sinclair KD. The periconceptional period, reproduction and long-term health of offspring: the importance of one-carbon metabolism. Hum Reprod Update. 2013;19(6):640–55.
    1. Kalhan SC. One carbon metabolism in pregnancy: impact on maternal, fetal and neonatal health. Mol Cell Endocrinol. 2016;435:48–60.
    1. Khot V, Kale A, Joshi A, Chavan-Gautam P, Joshi S. Expression of genes encoding enzymes involved in the one carbon cycle in rat placenta is determined by maternal micronutrients (folic acid, vitamin B12) and omega-3 fatty acids. Biomed Res Int. 2014;2014:613078.
    1. Chandler TL, White HM. Choline and methionine differentially alter methyl carbon metabolism in bovine neonatal hepatocytes. PLoS One. 2017;12(2):e0171080.
    1. Yajnik CS, Deshmukh US. Fetal programming: maternal nutrition and role of one-carbon metabolism. Rev Endocr Metab Disord. 2012;13(2):121–7.
    1. Yajnik CS, Chandak GR, Joglekar C, Katre P, Bhat DS, Singh SN, Janipalli CS, Refsum H, Krishnaveni G, Veena S et al. .. Maternal homocysteine in pregnancy and offspring birthweight: epidemiological associations and Mendelian randomization analysis. Int J Epidemiol. 2014;43(5):1487–97.
    1. Rogne T, Tielemans MJ, Chong MF, Yajnik CS, Krishnaveni GV, Poston L, Jaddoe VW, Steegers EA, Joshi S, Chong YS et al. .. Associations of maternal vitamin B12 concentration in pregnancy with the risks of preterm birth and low birth weight: a systematic review and meta-analysis of individual participant data. Am J Epidemiol. 2017;185(3):212–23.
    1. Dominguez-Salas P, Moore SE, Cole D, da Costa KA, Cox SE, Dyer RA, Fulford AJ, Innis SM, Waterland RA, Zeisel SH et al. .. DNA methylation potential: dietary intake and blood concentrations of one-carbon metabolites and cofactors in rural African women. Am J Clin Nutr. 2013;97(6):1217–27.
    1. Rossary A, Farges MC, Lamas B, Miles EA, Noakes PS, Kremmyda LS, Vlachava M, Diaper ND, Robinson SM, Godfrey KM et al. .. Increased consumption of salmon during pregnancy partly prevents the decline of some plasma essential amino acid concentrations in pregnant women. Clin Nutr. 2014;33(2):267–73.
    1. Yan J, Jiang X, West AA, Perry CA, Malysheva OV, Devapatla S, Pressman E, Vermeylen F, Stabler SP, Allen RH et al. .. Maternal choline intake modulates maternal and fetal biomarkers of choline metabolism in humans. Am J Clin Nutr. 2012;95(5):1060–71.
    1. Diaz SO, Pinto J, Graca G, Duarte IF, Barros AS, Galhano E, Pita C, Almeida Mdo C, Goodfellow BJ, Carreira IM et al. .. Metabolic biomarkers of prenatal disorders: an exploratory NMR metabolomics study of second trimester maternal urine and blood plasma. J Proteome Res. 2011;10(8):3732–42.
    1. Maruta E, Wang J, Kotani T, Tsuda H, Nakano T, Imai K, Sumigama S, Niwa Y, Mitsui T, Yoshida S et al. .. Association of serum asymmetric dimethylarginine, homocysteine, and l-arginine concentrations during early pregnancy with hypertensive disorders of pregnancy. Clin Chim Acta. 2017;475:70–7.
    1. Lindsay KL, Hellmuth C, Uhl O, Buss C, Wadhwa PD, Koletzko B, Entringer S. Longitudinal metabolomic profiling of amino acids and lipids across healthy pregnancy. PLoS One. 2015;10(12):e0145794.
    1. Viskova H, Vesela K, Janosikova B, Krijt J, Visek JA, Calda P. Plasma cysteine concentrations in uncomplicated pregnancies. Fetal Diagn Ther. 2007;22(4):254–8.
    1. Pinto J, Barros AS, Domingues MR, Goodfellow BJ, Galhano E, Pita C, Almeida Mdo C, Carreira IM, Gil AM. Following healthy pregnancy by NMR metabolomics of plasma and correlation to urine. J Proteome Res. 2015;14(2):1263–74.
    1. Cikot RJ, Steegers-Theunissen RP, Thomas CM, de Boo TM, Merkus HM, Steegers EA. Longitudinal vitamin and homocysteine levels in normal pregnancy. Br J Nutr. 2001;85(1):49–58.
    1. Hambidge KM, Krebs NF, Westcott JE, Garces A, Goudar SS, Kodkany BS, Pasha O, Tshefu A, Bose CL, Figueroa L et al. .. Preconception maternal nutrition: a multi-site randomized controlled trial. BMC Pregnancy Childbirth. 2014;14:111.
    1. Hambidge KM, Westcott JE, Garces A, Figueroa L, Goudar SS, Dhaded SM, Pasha O, Ali SA, Tshefu A, Lokangaka A et al. .. A multicountry randomized controlled trial of comprehensive maternal nutrition supplementation initiated before conception: the Women First Trial. Am J Clin Nutr. 2019;109(2):457–69.
    1. da Silva VR, Rios-Avila L, Lamers Y, Ralat MA, Midttun O, Quinlivan EP, Garrett TJ, Coats B, Shankar MN, Percival SS et al. .. Metabolite profile analysis reveals functional effects of 28-day vitamin B-6 restriction on one-carbon metabolism and tryptophan catabolic pathways in healthy men and women. J Nutr. 2013;143(11):1719–27.
    1. Loughlin AF, Skiles GL, Alberts DW, Schaefer WH. An ion exchange liquid chromatography/mass spectrometry method for the determination of reduced and oxidized glutathione and glutathione conjugates in hepatocytes. J Pharm Biomed Anal. 2001;26(1):131–42.
    1. Lander RL, Hambidge KM, Westcott JE, Tejeda G, Diba TS, Mastiholi SC, Khan US, Garces A, Figueroa L, Tshefu A et al. .. Pregnant women in four low-middle income countries have a high prevalence of inadequate dietary intakes that are improved by dietary diversity. Nutrients. 2019;11(7):E1560.
    1. Velzing-Aarts FV, Holm PI, Fokkema MR, van der Dijs FP, Ueland PM, Muskiet FA. Plasma choline and betaine and their relation to plasma homocysteine in normal pregnancy. Am J Clin Nutr. 2005;81(6):1383–9.
    1. Di Giulio AM, Carelli S, Castoldi RE, Gorio A, Taricco E, Cetin I. Plasma amino acid concentrations throughout normal pregnancy and early stages of intrauterine growth restricted pregnancy. J Matern Fetal Neonatal Med. 2004;15(6):356–62.
    1. Friesen RW, Novak EM, Hasman D, Innis SM. Relationship of dimethylglycine, choline, and betaine with oxoproline in plasma of pregnant women and their newborn infants. J Nutr. 2007;137(12):2641–6.
    1. Thame MM, Hsu JW, Gibson R, Baker TM, Tang GJ, Badaloo AV, Fletcher HM, Jackson AA, Jahoor F. Adaptation of in vivo amino acid kinetics facilitates increased amino acid availability for fetal growth in adolescent and adult pregnancies alike. Br J Nutr. 2014;112(11):1779–86.
    1. Dickinson H, Davies-Tuck M, Ellery SJ, Grieger JA, Wallace EM, Snow RJ, Walker DW, Clifton VL. Maternal creatine in pregnancy: a retrospective cohort study. BJOG. 2016;123(11):1830–8.
    1. Valtonen P, Laitinen T, Lyyra-Laitinen T, Raitakari OT, Juonala M, Viikari JS, Heiskanen N, Vanninen E, Punnonen K, Heinonen S. Serum L-homoarginine concentration is elevated during normal pregnancy and is related to flow-mediated vasodilatation. Circ J. 2008;72(11):1879–84.
    1. Dasarathy J, Gruca LL, Bennett C, Parimi PS, Duenas C, Marczewski S, Fierro JL, Kalhan SC. Methionine metabolism in human pregnancy. Am J Clin Nutr. 2010;91(2):357–65.
    1. Jacob S, Nodzenski M, Reisetter AC, Bain JR, Muehlbauer MJ, Stevens RD, Ilkayeva OR, Lowe LP, Metzger BE, Newgard CB et al. .. Targeted metabolomics demonstrates distinct and overlapping maternal metabolites associated with BMI, glucose, and insulin sensitivity during pregnancy across four ancestry groups. Diabetes Care. 2017;40(7):911–9.
    1. Luan H, Meng N, Liu P, Feng Q, Lin S, Fu J, Davidson R, Chen X, Rao W, Chen F et al. .. Pregnancy-induced metabolic phenotype variations in maternal plasma. J Proteome Res. 2014;13(3):1527–36.
    1. Desforges M, Parsons L, Westwood M, Sibley CP, Greenwood SL. Taurine transport in human placental trophoblast is important for regulation of cell differentiation and survival. Cell Death Dis. 2013;4:e559.
    1. Rees WD, Hay SM, Buchan V, Antipatis C, Palmer RM. The effects of maternal protein restriction on the growth of the rat fetus and its amino acid supply. Br J Nutr. 1999;81(3):243–50.
    1. Gumus E, Atalay MA, Cetinkaya Demir B, Sahin Gunes E. Possible role of asymmetric dimethylarginine (ADMA) in prediction of perinatal outcome in preeclampsia and fetal growth retardation related to preeclampsia. J Matern Fetal Neonatal Med. 2016;29(23):3806–11.
    1. Tamas P, Bodis J, Sulyok E, Kovacs GL, Hantosi E, Molnar G, Martens-Lobenhoffer J, Bode-Boger SM. L-arginine metabolism in early-onset and late-onset pre-eclamptic pregnancies. Scand J Clin Lab Invest. 2013;73(5):436–43.
    1. Bian Z, Shixia C, Duan T. First-trimester maternal serum levels of sFLT1, PGF and ADMA predict preeclampsia. PLoS One. 2015;10(4):e0124684.
    1. Nemeth B, Muranyi E, Hegyi P, Matrai P, Szakacs Z, Varju P, Hamvas S, Tinusz B, Budan F, Czimmer J et al. .. Asymmetric dimethylarginine levels in preeclampsia—systematic review and meta-analysis. Placenta. 2018;69:57–63.
    1. Tsikas D, Bollenbach A, Savvidou MD. Inverse correlation between maternal plasma asymmetric dimethylarginine (ADMA) and birthweight percentile in women with impaired placental perfusion: circulating ADMA as an NO-independent indicator of fetal growth restriction?. Amino Acids. 2018;50(2):341–51.
    1. Du YF, Wei Y, Yang J, Cheng ZY, Zuo XF, Wu TC, Shi HF, Wang XL. Maternal betaine status, but not that of choline or methionine, is inversely associated with infant birth weight. Br J Nutr. 2019;121(11):1279–86.
    1. Hogeveen M, den Heijer M, Semmekrot BA, Sporken JM, Ueland PM, Blom HJ. Umbilical choline and related methylamines betaine and dimethylglycine in relation to birth weight. Pediatr Res. 2013;73(6):783–7.
    1. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA et al. .. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9(4):311–26.
    1. Turner E, Brewster JA, Simpson NA, Walker JJ, Fisher J. Aromatic amino acid biomarkers of preeclampsia–a nuclear magnetic resonance investigation. Hypertens Pregnancy. 2008;27(3):225–35.
    1. Han YS, Ha EH, Park HS, Kim YJ, Lee SS. Relationships between pregnancy outcomes, biochemical markers and pre-pregnancy body mass index. Int J Obes (Lond). 2011;35(4):570–7.
    1. Choi R, Choi S, Lim Y, Cho YY, Kim HJ, Kim SW, Chung JH, Oh SY, Lee SY. A prospective study on serum methylmalonic acid and homocysteine in pregnant women. Nutrients. 2016;8(12):797.
    1. Wang J, Kotani T, Tsuda H, Mano Y, Sumigama S, Li H, Komatsu K, Miki R, Maruta E, Niwa Y et al. .. Is the serum l-arginine level during early pregnancy a predictor of pregnancy-induced hypertension?. J Clin Biochem Nutr. 2015;57(1):74–81.
    1. Hellmuth C, Lindsay KL, Uhl O, Buss C, Wadhwa PD, Koletzko B, Entringer S. Association of maternal prepregnancy BMI with metabolomic profile across gestation. Int J Obes (Lond). 2017;41(1):159–69.
    1. Headen I, Cohen AK, Mujahid M, Abrams B. The accuracy of self-reported pregnancy-related weight: a systematic review. Obes Rev. 2017;18(3):350–69.
    1. Chace DH, Adam BW, Smith SJ, Alexander JR, Hillman SL, Hannon WH. Validation of accuracy-based amino acid reference materials in dried-blood spots by tandem mass spectrometry for newborn screening assays. Clin Chem. 1999;45(8 Pt 1):1269–77.
    1. Wagner M, Tonoli D, Varesio E, Hopfgartner G. The use of mass spectrometry to analyze dried blood spots. Mass Spectrom Rev. 2016;35(3):361–438.
    1. Pearce EN, Caldwell KL. Urinary iodine, thyroid function, and thyroglobulin as biomarkers of iodine status. Am J Clin Nutr. 2016;104(Suppl 3):898S–901S.
    1. Bruno C, Dufour-Rainfray D, Patin F, Vourc'h P, Guilloteau D, Maillot F, Labarthe F, Tardieu M, Andres CR, Emond P et al. .. Validation of amino-acids measurement in dried blood spot by FIA-MS/MS for PKU management. Clin Biochem. 2016;49(13–14):1047–50.

Source: PubMed

3
Tilaa