Longitudinal Metabolomic Profiling of Amino Acids and Lipids across Healthy Pregnancy

Karen L Lindsay, Christian Hellmuth, Olaf Uhl, Claudia Buss, Pathik D Wadhwa, Berthold Koletzko, Sonja Entringer, Karen L Lindsay, Christian Hellmuth, Olaf Uhl, Claudia Buss, Pathik D Wadhwa, Berthold Koletzko, Sonja Entringer

Abstract

Pregnancy is characterized by a complexity of metabolic processes that may impact fetal development and ultimately, infant health outcomes. However, our understanding of whole body maternal and fetal metabolism during this critical life stage remains incomplete. The objective of this study is to utilize metabolomics to profile longitudinal patterns of fasting maternal metabolites among a cohort of non-diabetic, healthy pregnant women in order to advance our understanding of changes in protein and lipid concentrations across gestation, the biochemical pathways by which they are metabolized and to describe variation in maternal metabolites between ethnic groups. Among 160 pregnant women, amino acids, tricarboxylic acid (TCA) cycle intermediates, keto-bodies and non-esterified fatty acids were detected by liquid chromatography coupled with mass spectrometry, while polar lipids were detected through flow-injected mass spectrometry. The maternal plasma concentration of several essential and non-essential amino acids, long-chain polyunsaturated fatty acids, free carnitine, acetylcarnitine, phosphatidylcholines and sphingomyelins significantly decreased across pregnancy. Concentrations of several TCA intermediates increase as pregnancy progresses, as well as the keto-body β-hydroxybutyrate. Ratios of specific acylcarnitines used as indicators of metabolic pathways suggest a decreased beta-oxidation rate and increased carnitine palmitoyltransferase-1 enzyme activity with advancing gestation. Decreasing amino acid concentrations likely reflects placental uptake and tissue biosynthesis. The absence of any increase in plasma non-esterified fatty acids is unexpected in the catabolic phase of later pregnancy and may reflect enhanced placental fatty acid uptake and utilization for fetal tissue growth. While it appears that energy production through the TCA cycle increases as pregnancy progresses, decreasing patterns of free carnitine and acetylcarnitine as well as increased carnitine palmitoyltransferase-1 rate and β-hydroxybutyrate levels suggest a concomitant upregulation of ketogenesis to ensure sufficient energy supply in the fasting state. Several differences in metabolomic profiles between Hispanic and non-Hispanic women demonstrate phenotypic variations in prenatal metabolism which should be considered in future studies.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Barplot comparing plasma amino acid…
Fig 1. Barplot comparing plasma amino acid concentrations across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted. * p-value

Fig 2. Barplot comparing plasma non-esterified fatty…

Fig 2. Barplot comparing plasma non-esterified fatty acid concentrations across trimesters among pregnant women.

Median…

Fig 2. Barplot comparing plasma non-esterified fatty acid concentrations across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value

Fig 3. Barplot comparing metabolic ratios as…

Fig 3. Barplot comparing metabolic ratios as indicators of β-oxidation (right) and carnitine palmitoyl transferase-1…

Fig 3. Barplot comparing metabolic ratios as indicators of β-oxidation (right) and carnitine palmitoyl transferase-1 activity (left) across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value

Fig 4. Barplot comparing plasma concentrations of…

Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across…

Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value
Similar articles
Cited by
References
    1. Herrera A. Metabolic alterations in pregnancy and their implications for the availability of substrates for the fetus. Eur J Clin Nutr. 2000; 54: S47–S51. - PubMed
    1. Hadden DR, McLaughlin C. Normal and abnormal maternal metabolism during pregnancy. Sem Neonat Fet Med. 2009; 14: 66–71. - PubMed
    1. Butte NF. Carbohydrate and lipid metabolism in pregnancy: normal compared with gestational diabetes mellitus. Am J Clin Nutr. 2000; 71: 1256S–61S. - PubMed
    1. Herrera E, Amusquivar E, Lopez-Soldado L, Ortego H. Maternal lipid metabolism and placental lipid transfer. Horm Res. 2006; 65(suppl 3): 59–64. - PubMed
    1. Iva SK. Metabolic adaptations in pregnancy in lean and obese women—a literature review. Res Obstet Gynecol. 2013; 2: 37–47.
Show all 50 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 2. Barplot comparing plasma non-esterified fatty…
Fig 2. Barplot comparing plasma non-esterified fatty acid concentrations across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value

Fig 3. Barplot comparing metabolic ratios as…

Fig 3. Barplot comparing metabolic ratios as indicators of β-oxidation (right) and carnitine palmitoyl transferase-1…

Fig 3. Barplot comparing metabolic ratios as indicators of β-oxidation (right) and carnitine palmitoyl transferase-1 activity (left) across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value

Fig 4. Barplot comparing plasma concentrations of…

Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across…

Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value
Similar articles
Cited by
References
    1. Herrera A. Metabolic alterations in pregnancy and their implications for the availability of substrates for the fetus. Eur J Clin Nutr. 2000; 54: S47–S51. - PubMed
    1. Hadden DR, McLaughlin C. Normal and abnormal maternal metabolism during pregnancy. Sem Neonat Fet Med. 2009; 14: 66–71. - PubMed
    1. Butte NF. Carbohydrate and lipid metabolism in pregnancy: normal compared with gestational diabetes mellitus. Am J Clin Nutr. 2000; 71: 1256S–61S. - PubMed
    1. Herrera E, Amusquivar E, Lopez-Soldado L, Ortego H. Maternal lipid metabolism and placental lipid transfer. Horm Res. 2006; 65(suppl 3): 59–64. - PubMed
    1. Iva SK. Metabolic adaptations in pregnancy in lean and obese women—a literature review. Res Obstet Gynecol. 2013; 2: 37–47.
Show all 50 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 3. Barplot comparing metabolic ratios as…
Fig 3. Barplot comparing metabolic ratios as indicators of β-oxidation (right) and carnitine palmitoyl transferase-1 activity (left) across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value

Fig 4. Barplot comparing plasma concentrations of…

Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across…

Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value
Similar articles
Cited by
References
    1. Herrera A. Metabolic alterations in pregnancy and their implications for the availability of substrates for the fetus. Eur J Clin Nutr. 2000; 54: S47–S51. - PubMed
    1. Hadden DR, McLaughlin C. Normal and abnormal maternal metabolism during pregnancy. Sem Neonat Fet Med. 2009; 14: 66–71. - PubMed
    1. Butte NF. Carbohydrate and lipid metabolism in pregnancy: normal compared with gestational diabetes mellitus. Am J Clin Nutr. 2000; 71: 1256S–61S. - PubMed
    1. Herrera E, Amusquivar E, Lopez-Soldado L, Ortego H. Maternal lipid metabolism and placental lipid transfer. Horm Res. 2006; 65(suppl 3): 59–64. - PubMed
    1. Iva SK. Metabolic adaptations in pregnancy in lean and obese women—a literature review. Res Obstet Gynecol. 2013; 2: 37–47.
Show all 50 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 4. Barplot comparing plasma concentrations of…
Fig 4. Barplot comparing plasma concentrations of tricarboxylic acid cycle intermediates and keto acids across trimesters among pregnant women.
Median (+ Interquartile range/2) was plotted * p-value

References

    1. Herrera A. Metabolic alterations in pregnancy and their implications for the availability of substrates for the fetus. Eur J Clin Nutr. 2000; 54: S47–S51.
    1. Hadden DR, McLaughlin C. Normal and abnormal maternal metabolism during pregnancy. Sem Neonat Fet Med. 2009; 14: 66–71.
    1. Butte NF. Carbohydrate and lipid metabolism in pregnancy: normal compared with gestational diabetes mellitus. Am J Clin Nutr. 2000; 71: 1256S–61S.
    1. Herrera E, Amusquivar E, Lopez-Soldado L, Ortego H. Maternal lipid metabolism and placental lipid transfer. Horm Res. 2006; 65(suppl 3): 59–64.
    1. Iva SK. Metabolic adaptations in pregnancy in lean and obese women—a literature review. Res Obstet Gynecol. 2013; 2: 37–47.
    1. Pinto J, Barros AS, Domingues MRM, Goodfellow BJ, Galhano E, Pita C, et al. Following healthy pregnancy by NMR metabolomics of plasma and correlation to urine. J Proteome Res. 2015; 14: 1263–74. 10.1021/pr5011982
    1. Luan H, Meng N, Liu P, Feng Q, Lin S, Fu J, et al. Pregnancy-induced metabolic phenotype variations in maternal plasma. J Proteome Res. 2014; 13: 1527–36. 10.1021/pr401068k
    1. Diaz SO, Barros AS, Goodfellow BJ, Duarte IF, Carreira IM, Galhano E, et al. Following healthy pregnancy by nuclear magnetic resonance (NMR) metabolic profiling of human urine. J Proteome Res. 2013; 12: 969–79. 10.1021/pr301022e
    1. Gibbons H, O’Gorman A, Brennan L. Metabolomics as a tool in nutrition research. Curr Opin Lipidol. 2015; 26: 30–4.
    1. Dumas M-E, Maibaum EC, Teague C, Ueshima H, Zhou B, Lindon JC, et al. Assessment of analytical reproducibility of 1H NMR spectroscopy based metabolomics for large-scale epidemiological research: the INTERMAP study. Anal Chem. 2006; 78: 2199–208.
    1. Harder U, Koletzko B, Peissner W. Quantification of 22 plasma amino acids combining derivatization and ion pair LC-MS/MS. J Chromatography B. 2011; 879: 495–504.
    1. Birkler RI, Stottrup NB, Hermannson S, Nielsen TT, Gregersen N, Bothker HE, et al. A UPLC-MS/MS application for profiling of intermediate energy metabolites in microdialysis samples—a method for high throughput. J Pharm Biomed Anal. 2010; 53: 983–90.
    1. Luo B, Groenke K, Takors R, Wandrey C, Oldiges M. Simultaneous determination of multiple intracellular metabolites in glycolysis, pentose phosphate pathway and tricarboxylic acid cycle by liquid chromatography-mass spectrometry. J Chromatogr A. 2007; 1147: 153–64.
    1. Hellmuth C, Weber M, Koletzko B, Peissner W. Nonesterified fatty acid determination for functional lipidomics: Comprehensive ultrahigh performance liquid chromatography—tandem mass spectrometry quantitation, qualification and parameter prediction. Anal Chem. 2012; 84: 1483–90.
    1. Cetin I, Nobile de Santos MS, Taricco E, Radaelli T, Teng C, Ronzoni S, et al. Maternal and fetal amino acid concentrations in normal pregnancies and in pregnancies with gestational diabetes mellitus. Am J Obstet Gynecol. 2005; 192: 610–7.
    1. Pappa KI, Vlachos G, Theodora M, Roubelaki M, Angelidou K, Antsaklis A. Intermediate metabolism in association with the amino acid profile during the third trimester of normal pregnancy and diet controlled diabetes. Am J Obstet Gynecol. 2007; 196: 65e1–65e5.
    1. Scholtens DM, Muelhbauer MJ, Daya NR, Stevens RD, Dyer AR, Lowe LP, et al. Metabolomics reveals broad-scale metabolic perturbations in hyperglycemic mothers during pregnancy. Diabetes Care. 2014; 37: 158–66. 10.2337/dc13-0989
    1. Duggleby SL, Jackson AA. Higher weight at birth is related to decreased maternal amino acid oxidation during pregnancy. Am J Clin Nutr. 2002; 76: 852–7.
    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 Mat Fet Neonat Med. 2004; 15: 356–62.
    1. Van den Akker CH, Schierbeek H, Dorst KY, Schoonderwaldt EM, Vermes A, Duvekot JJ, et al. Human fetal amino acid metabolism at term gestation. Am J Clin Nutr. 2009; 89: 153–60. 10.3945/ajcn.2008.26553
    1. Kalhan SC. Protein metabolism in pregnancy. Am J Clin Nutr. 2000; 71: 1249S–55S.
    1. Felig P, Kim YJ, Lynch V, Hendler R. Amino acid metabolism during starvation in human pregnancy. J Clin Invest. 1972; 51: 1195–202.
    1. Metzger BE, Ravnikar V, Vileisis RA, Freinkel N. ‘Accelerated starvation’ and the sipped breakfast in late normal pregnancy. Lancet. 1982; i: 588–92.
    1. Kalhan SC, Parimi PS. Transamination of leucine and nitrogen accretion in human pregnancy and the newborn infant. J Nutr. 2006; 136: 281S–7S.
    1. Whittaker PG, Lee CH, Cooper BG, Taylor R. Evaluation of phenylalanine and tyrosine metabolism in late human pregnancy. Metabolism. 1999; 48: 849–52.
    1. Rees WD, Hay SM, Antipatis C. The effect of dietary protein on the amino acid supply and threonine metabolism in the pregnant rat. Reprod Nutr Dev. 2006; 46: 227–39.
    1. Darmaun D, Decholette P. Role of leucine as a precursor of glutamine alpha-amino nitrogen in vivo in humans. Am J Physiol. 1991; 260: E326–9.
    1. Battaglia FC, Regnault TRH. Placental transport and metabolism of amino acids. Placenta. 2001; 22: 145–61.
    1. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched-chain amino acid metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009; 9: 311–26.
    1. McCormack SE, Shaham O, McCarthy MA, Deik AA, Wang TJ, Gerzsten RE, et al. Circulating branched-chain amino acid concentrations are associated with obesity and future insulin resistance in children and adolescents. Pediatr Obes. 2013; 8: 52–61. 10.1111/j.2047-6310.2012.00087.x
    1. Rauschert S, Uhl O, Koletzko B, Hellmuth C. Metabolomic biomarkers for obesity in humans: a short review. Ann Nutr Metab. 2014; 63: 314–24.
    1. Ditchfield AM, Desforges M, Mills TA, Glazier JD, Wareing M, Mynett K, et al. Maternal obesity is associated with a reduction in placental taurine transporter activity. Int J Obes. 2015; 39: 557–64.
    1. Naismith DJ, Rana SK, Emery PW. Metabolism of taurine during reproduction in women. Hum Nutr Clin Nutr. 1987; 41: 37–45.
    1. Elliott JA. The effect of pregnancy on the control of lipolysis in fat cells isolated from human adipose tissue. Eur J Clin Invest. 1975; 5: 1659–63.
    1. Larqué E, Demmelmair H, Gil-Sánchez A, Prieto-Sánchez MT, Blanco JE, Pagán A, et al. Placental transfer of fatty acids and fetal implications. Am J Clin Nutr. 2011; 94: 1908S–13S. 10.3945/ajcn.110.001230
    1. Campbell FM, Taffesse S, Gordon MJ, Dutta-Roy AK. Plasma membrane fatty-acid binding protein in human placenta: identification and characterization. Biochem Biophys Res Commun. 1995; 209: 1011–7.
    1. Herrera A. Lipid metabolism in pregnancy and its consequences in the fetus and newborn. Endocrine. 2002; 19: 43–55.
    1. Nochera CL, Goossen LH, Brutus AR, Cristales M, Eastman B. Nutr Clin Prat. 2011; 26: 445–50.
    1. Cetin I, Koletzko B. Long-chain omega-3 fatty acid supply in pregnancy and lactation. Curr Opin Clin Nutr Metab Care. 2008; 11: 297–302. 10.1097/MCO.0b013e3282f795e6
    1. Marzo A, Cardace G, Corbelletta C, Pace S, D’Iddio S, Verrotti C, et al. Plasma concentration, urinary excretion and renal clearance of L-carnitine during pregnancy: a reversible secondary L-carnitine deficiency. Gynecol Endocrinol. 1994; 8: 115–20.
    1. Cho S-W, Cha Y-S. Pregnancy increases urinary loss of carnitine and reduces plasma carnitine in Korean women. Br J Nutr. 2005; 93: 685–91.
    1. Reuter SE, Evans AM. Carnitine and acylcarnitines. Pharmacokinetic, pharmacological and clinical aspects. Clin Pharmacokinet. 2012; 51: 553–72. 10.2165/11633940-000000000-00000
    1. Houten SM, Wanders RJA. A general introduction to the biochemistry of mitochondrial β-oxidation. J Inherit Metab Dis. 2010; 33: 469–77.
    1. Violante S, Iljst L, Ruiter J, Koster J, van Lenthe H, Duran M, et al. Substrate specificity of human carnitine acyltransferases: Implications for fatty acid and branched-chain amino acid metabolism. Biochimicaet Biophysica Acta. 2013; 1832: 773–9.
    1. Longo VD, Matson MP. Fasting: Molecular mechanisms and clinical applications. Cell Metab. 2014; 19: 181–92. 10.1016/j.cmet.2013.12.008
    1. Arenas J, Rubio JC, Martin MA, Campos Y. Biological roles of L-carnitine in perinatal metabolism. Early Hum Dev. 1998; 53: S43–50.
    1. Grube M, Meyer Z, Schweibedissen H, Draber K, Präger D, Möritz KU, et al. Expression, localization and function of the carnitine transporter octn2 (slc22a5) in human placenta. Drug Metab Dispos. 2005; 33: 31–7.
    1. Oey NA, Van Vlies N, Wijburg FA, Wanders RJA, Attie-Bitach T, Vaz FM. L-Carnitine is synthesized in the human fetal-placental unit: Potential roles in placental and fetal metabolism. Placenta. 2006; 27: 841–46.
    1. Krajcovicová-Kudlácková M, Šimončič R, Béderová A, Babinská K, Béder I. Correlation of carnitine levels to methionine and lysine intake. Physiol Res. 2000; 49: 399–402.
    1. Cederblad G, Fahraeus L, Lindgren K. Plasma carnitine and renal carnitine clearance during pregnancy. Am J Clin Nutr. 1986; 44: 379–83.

Source: PubMed

Подписаться