Horm Metab Res 2021; 53(02): 112-123
DOI: 10.1055/a-1300-2294
Endocrine Care

Investigating the Metabolic Model in Preterm Neonates by Tandem Mass Spectrometry: A Cohort Study

Benjing Wang
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Qin Zhang
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Qi Wang
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Jun Ma
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Xiaoju Cao
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Yaping Chen
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Yuhong Pan
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Hong Li
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Jingjing Xiang
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
,
Ting Wang
1   Center for Reproduction and Genetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
› Author Affiliations
Funding: This study was supported by grants from Suzhou Key Medical Center (SZZX201505), Jiangsu Maternal and Children health care research project (F201715), Jiangsu Provincial Medical Innovation Team (CXTDB2017013), Suzhou Clinical Medical Expert Team (SZYJTD201708), Jiangsu Maternal and Children health care key discipline (FXK201748), Suzhou Science and Technology Support Program(SS2019066).

Abstract

The changes of metabolite profiles in preterm birth have been demonstrated using newborn screening data. However, little is known about the holistic metabolic model in preterm neonates. The aim was to investigate the holistic metabolic model in preterm neonates. All metabolite values were obtained from a cohort data of routine newborn screening. A total of 261 758 newborns were recruited and randomly divided into a training subset and a testing subset. Using the training subset, 949 variates were considered to establish a logistic regression model for identifying preterm birth (<37 weeks) from term birth (≥37 weeks). Sventy-two variates (age at collection, TSH, 17α-OHP, proline, tyrosine, C16:1-OH, C18:2, and 65 ratios) entered into the final metabolic model for identifying preterm birth from term birth. Among the variates entering into the final model of PTB [Leucine+Isoleucine+Proline-OH)/Valine (OR=38.36], (C3DC+C4-OH)/C12 (OR=15.58), Valine/C5 (OR=6.32), [Leucine+isoleucine+Proline-OH)/Ornithine (OR=2.509)], and Proline/C18:1 (OR=2.465) have the top five OR values, and [Leucine+Isoleucine+Proline-OH)/C5 (OR=0.05)], [Leucine+Isoleucine+Proline-OH)/Phenylalanine (OR=0.214)], proline/valine (OR=0.230), C16/C18 (OR=0.259), and Alanine/free carnitine (OR=0.279) have the five lowest OR values. The final metabolic model had a capacity of identifying preterm infants with >80% accuracy in both the training and testing subsets. When identifying neonates ≤32 weeks from those >32 weeks, it had a robust performance with nearly 95% accuracy in both subsets. In summary, we have established an excellent metabolic model in preterm neonates. These findings could provide new insights for more efficient nutrient supplements and etiology of preterm birth.

Supplementary Material



Publication History

Received: 14 July 2020

Accepted: 20 October 2020

Article published online:
27 November 2020

© 2020. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Luu TM, Rehman Mian MO. et al. Long-term impact of preterm birth: neurodevelopmental and physical health outcomes. Clin Perinatol 2017; 44: 305-314
  • 2 Lapillonne A, O'Connor DL, Wang D. et al. Nutritional recommendations for the late-preterm infant and the preterm infant after hospital discharge. J Pediatr 2013; 162: S90-S100
  • 3 Ombrone D, Giocaliere E, Forni G. et al. Expanded newborn screening by mass spectrometry: New tests, future perspectives. Mass Spectrom Rev 2016; 35: 71-84
  • 4 Therrell BL, Padilla CD, Loeber JG. et al. Current status of newborn screening worldwide: 2015. Semin Perinatol 2015; 39: 171-187
  • 5 Roelfsema F, Veldhuis JD. Thyrotropin secretion patterns in health and disease. Endocr Rev 2013; 34: 619-657
  • 6 Shields BM, Knight BA, Hill A. et al. Fetal thyroid hormone level at birth is associated with fetal growth. J Clin Endocrinol Metab 2011; 96: E934-E938
  • 7 Manuck TA, Watkins WS, Esplin MS. et al. Pharmacogenomics of 17-alpha hydroxyprogesterone caproate for recurrent preterm birth: a case-control study. BJOG 2018; 125: 343-350
  • 8 Saccone G, Khalifeh A, Elimian A. et al. Vaginal progesterone vs intramuscular 17α-hydroxyprogesterone caproate for prevention of recurrent spontaneous preterm birth in singleton gestations: systematic review and meta-analysis of randomized controlled trials. Ultrasound Obstet Gynecol 2017; 49: 315-321
  • 9 Manuck TA. 17-alpha hydroxyprogesterone caproate for preterm birth prevention: Where have we been, how did we get here, and where are we going?. Semin Perinatol 2017; 41: 461-467
  • 10 Ning A, Vladutiu CJ, Dotters-Katz SK. et al. Gestational age at initiation of 17-alpha hydroxyprogesterone caproate and recurrent preterm birth. Am J Obstet Gynecol 2017; 217: 371.e1-371.e7
  • 11 Wang B, Zhang Q, Gao A. et al. New ratios for performance improvement for identifying Acyl-CoA dehydrogenase deficiencies in expanded newborn screening: A retrospective study. Front Genet 2019; 10: 811
  • 12 Wang T, Ma J, Zhang Q. et al. Expanded newborn screening for inborn errors of metabolism by tandem mass spectrometry in Suzhou, China: Disease spectrum, prevalence, genetic characteristics in a Chinese Population. Front Genet 2019; 10: 1052
  • 13 Wilson K, Hawken S, Murphy MSQ. et al. Postnatal Prediction of gestational age using newborn fetal hemoglobin levels. EBioMedicine 2017; 15: 203-209
  • 14 Hawken S, Ducharme R, Murphy MSQ. et al. Performance of a postnatal metabolic gestational age algorithm: a retrospective validation study among ethnic subgroups in Canada. BMJ Open 2017; 7: e015615
  • 15 Wilson K, Hawken S, Potter BK. et al. Accurate prediction of gestational age using newborn screening analyte data. Am J Obstet Gynecol 2015; 214: 513.e1-513.e9
  • 16 Jelliffe-Pawlowski LL, Norton ME, Baer RJ. et al. Gestational dating by metabolic profile at birth: a California cohort study. Am J Obstet Gynecol 2016; 214: 511.e1-511
  • 17 Ryckman KK, Berberich SL, Dagle JM. Predicting gestational age using neonatal metabolic markers. Am J Obstet Gynecol 2016; 214: 515.e1-515
  • 18 Yang Y, Yu B, Long W. et al. Investigating the changes in amino acid values in premature infants: a pilot study. J Pediatr Endocrinol Metab 2018; 31: 435-441
  • 19 Mandour I, El Gayar D, Amin M. et al. Amino acid and acylcarnitine profiles in premature neonates: a pilot study. Indian J Pediatr 2013; 80: 736-744
  • 20 Koster MP, Vreeken RJ, Harms AC. et al. First-trimester serum acylcarnitine levels to predict preeclampsia: A metabolomics approach. Dis Markers 2015; 2015: 857108
  • 21 Kuc S, Koster MP, Pennings JL. et al. Metabolomics profiling for identification of novel potential markers in early prediction of preeclampsia. PLoS One 2014; 9: e98540
  • 22 Ryckman KK, Shchelochkov OA, Cook DE. et al. The influence of maternal disease on metabolites measured as part of newborn screening. J Matern Fetal Neonatal Med 2013; 26: 1380-1383
  • 23 Thiele IG, Niezen-Koning KE, van Gennip AH. et al. Increased plasma carnitine concentrations in preeclampsia. Obstet Gynecol 2004; 103: 876-880
  • 24 Chace DH, Pons R, Chiriboga CA. et al. Neonatal blood carnitine concentrations: normative data by electrospray tandem mass spectometry. Pediatr Res 2003; 53: 823-829
  • 25 Sánchez-Pintos P, Pérez-Muñuzuri A, Cocho JÁ. et al. Evaluation of carnitine deficit in very low birth weight preterm newborns small for their gestational age. J Matern Fetal Neonatal Med 2015; 29: 933-937
  • 26 Clark RH, Kelleher AS, Chace DH. et al. Gestational age and age at sampling influence metabolic profiles in premature infants. Pediatrics 2015; 134: e37-e46
  • 27 Honzík T, Chrastina R, Hansíková H. et al. Carnitine concentrations in term and preterm newborns at birth and during the first days of life. Prague Med Rep 2005; 106: 297-306
  • 28 Gucciardi A, Zaramella P, Costa I. et al. Analysis and interpretation of acylcarnitine profiles in dried blood spot and plasma of preterm and full-term newborns. Pediatr Res 2015; 77: 36-47
  • 29 Antonucci R, Atzori L, Barberini L. et al. Metabolomics: the "new clinical chemistry" for personalized neonatal medicine. Minerva Pediatr 2010; 62: 145-148
  • 30 Chou PY, Fasman GD. Structural and functional role of leucine residues in proteins. J Mol Biol 1973; 74: 263-281
  • 31 Markus KW, Michael WP, Franz XR. The effects of branched-chain amino acid interactions on growth performance, blood metabolites, enzyme kinetics and transcriptomics in weaned pigs. Br. J. Nutr 2010; 103: 964-976
  • 32 Cota D, Proulx K, Smith KA. et al. Hypothalamic mTOR signaling regulates food intake. Science 2006; 312: 927-930
  • 33 Chace DH, Lim T, Hansen CR. et al. Quantification of malonylcarnitine in dried blood spots by use of MS/MS varies by stable isotope internal standard composition. Clin Chim Acta 2009; 402: 14-18
  • 34 Wolf A, Weir P, Segar P. et al. Impaired fatty acid oxidation in propofol infusion syndrome. Lancet 2001; 357: 606-607
  • 35 Santer R, Fingerhut R, Lassker U. et al. Tandem mass spectrometric determination of malonylcarnitine: Diagnosis and neonatal screening of malonyl-CoA decarboxylase deficiency. Clin Chem 2003; 49: 660-662
  • 36 Abo Alrob O, Lopaschuk GD. Role of CoA and acetyl-CoA in regulating cardiac fatty acid and glucose oxidation. Biochem Soc Trans 2014; 42: 1043-1051
  • 37 McGarry JD, Takabayashi Y, Foster DW. The role of malonyl-CoA in the coordination of fatty acid synthesis and oxidation in isolated rat hepatocytes. J Biol Chem 1978; 253: 8294-8300
  • 38 McGarry JD, Foster DW. Regulation of hepatic fatty acid oxidation and ketone body production. Annu Rev Biochem 1980; 49: 395-420
  • 39 Yamada K, Taketani T. Management and diagnosis of mitochondrial fatty acid oxidation disorders: Focus on very-long-chain acyl-CoA dehydrogenase deficiency. J Hum Genet 2019; 64: 73-85
  • 40 Kwon WB, Touchette KJ, Simongiovanni A. et al. Excess dietary leucine in diets for growing pigs reduces growth performance, biological value of protein, protein retention, and serotonin synthesis1. J Anim Sci 2019; 97: 4282-4292
  • 41 Couce ML, Aldamiz-Echevarría L, Bueno MA. et al. Genotype and phenotype characterization in a Spanish cohort with isovaleric acidemia. J Hum Genet 2017; 62: 355-360
  • 42 Wieland LS, Santesso N. A Summary of a Cochrane Review: Supplementation with long chain polyunsaturated fatty acids (LCPUFA) to breastfeeding mothers for improving child growth and development. Eur J Integr Med 2016; 8: 113-114
  • 43 Nakamura K, Kido J, Mitsubuchi H. et al. Diagnosis and Treatment of Urea Cycle Disorder in Japan. Pediatr Int 2014; 56: 506-509
  • 44 Bijarnia-Mahay S, Häberle J, Jalan AB. et al. Urea Cycle Disorders in India: Clinical Course, Biochemical and Genetic Investigations, and Prenatal Testing. Orphanet J Rare Dis 2018; 13: 174
  • 45 Molema F, Gleich F, Burgard P. et al. Decreased Plasma l-Arginine Levels in Organic Acidurias (MMA and PA) and Decreased Plasma Branched-Chain Amino Acid Levels in Urea Cycle Disorders as a Potential Cause of Growth Retardation: Options for Treatment. Mol Genet Metab 2019; 126: 397-405
  • 46 Minkler PE, Stoll MSK, Ingalls ST. et al. Selective and accurate C5 acylcarnitine quantitation by UHPLC-MS/MS: Distinguishing true isovaleric acidemia from pivalate derived interference. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1061–1062: 128-133
  • 47 Alfardan J, Mohsen AW, Copeland S. et al. Characterization of new ACADSB gene sequence mutations and clinical implications in patients with 2-methylbutyrylglycinuria identified by newborn screening. Mol Genet Metab 2010; 100: 333-338
  • 48 van Calcar SC, Gleason LA, Lindh H. et al. 2-methylbutyryl-CoA dehydrogenase deficiency in Hmong infants identified by expanded newborn screen. WMJ 2007; 106: 12-15
  • 49 Lin Y, Gao H, Lin C. et al. Biochemical, Clinical, and Genetic Characteristics of Short/Branched Chain Acyl-CoA Dehydrogenase Deficiency in Chinese Patients by Newborn Screening. Front Genet 2019; 10: 802
  • 50 Ibarra-González I, Fernández-Lainez C, Guillén-López S. et al. Molecular analysis using targeted next generation DNA sequencing and clinical spectrum of Mexican patients with isovaleric acidemia. Clin Chim Acta 2020; 501: 216-221
  • 51 Porta F, Chiesa N, Martinelli D. et al. Clinical, biochemical, and molecular spectrum of short/branched-chain acyl-CoA dehydrogenase deficiency: two new cases and review of literature. J Pediatr Endocrinol Metab 2019; 32: 101-108
  • 52 Chinen Y, Nakamura S, Tamashiro K. et al. Isovaleric acidemia: Therapeutic response to supplementation with glycine, l-carnitine, or both in combination and a 10-year follow-up case study. Mol Genet Metab Rep 2017; 11: 2-5
  • 53 Pinto A, Daly A, Evans S. et al. Dietary practices in isovaleric acidemia: A European survey. Mol Genet Metab Rep 2017; 12: 16-22
  • 54 Korman SH. Inborn errors of isoleucine degradation: A review. Mol Genet Metab 2006; 89: 289-299
  • 55 Carpentier AC. Branched chain amino acid catabolism by brown adipose tissue. Endocrinology 2020; 161: bqaa060
  • 56 Teruya KI, Remor E, Schwartz IVD. Development of an inventory to assess perceived barriers related to PKU treatment. J Patient Rep Outcomes 2020; 4: 29
  • 57 Harper AE, Miller RH, Block KP. Branched-chain amino acid metabolism. Annu Rev Nutr 1984; 4: 409-454
  • 58 To CY, Freeman M, Van Winkle LJ.. Consumption of a Branched-chain amino acid (BCAA) during days 2-10 of pregnancy causes abnormal Fetal and Placental Growth: Implications for BCAA Supplementation in Humans. Int J Environ Res Public Health 2020; 17: 2445
  • 59 Christensen EM, Patel SM, Korasick DA. et al. Resolving the cofactor-binding site in the proline biosynthetic enzyme human pyrroline-5-carboxylate reductase 1. J Biol Chem 2017; 292: 7233-7243
  • 60 Hu CA, Khalil S, Zhaorigetu S. et al. Human Delta1-pyrroline -5-carboxylate synthase: function and regulation. Amino Acids 2008; 35: 665-672
  • 61 McHugh D, Cameron CA, Abdenur JE. et al. Clinical validation of cutoff target ranges in newborn screening of metabolic disorders by tandem mass spectrometry: A worldwide collaborative project. Genet Med 2011; 13: 230-254
  • 62 Quinonez SC, Seeley AH, Seeterlin M. et al. Newborn screening for dihydrolipoamide dehydrogenase deficiency: Citrulline as a useful analyte. Mol Genet Metab Rep 2014; 1: 345-349
  • 63 Quinonez SC, Thoene JG. Dihydrolipoamide Dehydrogenase Deficiency. In Adam MP, Ardinger HH, Pagon RA. Free Books & Documents. Source GeneReviews [Internet]. Seattle (WA): University of Washington, Seattle; 2014: 1993-2020
  • 64 Sim KG, Wiley V, Carpenter K. et al. Carnitine palmitoyltransferase I deficiency in neonate identified by dried blood spot free carnitine and acylcarnitine profile. J Inherit Metab Dis 2001; 24: 51-59
  • 65 Korman SH, Waterham HR, Gutman A. et al. Novel metabolic and molecular findings in hepatic carnitine palmitoyltransferase I deficiency. Mol Genet Metab 2005; 86: 337-343
  • 66 Rovelli V, Manzoni F, Viau K. et al. Clinical and biochemical outcome of patients with very long-chain acyl-CoA dehydrogenase deficiency. Mol Genet Metab 2019; 127: 64-73