Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter May 24, 2020

Genetic characteristics and follow-up of patients with fatty acid β-oxidation disorders through expanded newborn screening in a Northern Chinese population

  • Shuting Wang , Junhong Leng , Chengming Diao , Yuan Wang and Rongxiu Zheng EMAIL logo

Abstract

Background

Fatty acid β-oxidation disorders (FAODs) include more than 15 distinct disorders and have a wide variety of symptoms, usually not evident between episodes of acute decompensation. After the introduction of newborn screening (NBS) using tandem mass spectrometry (MS/MS), early identification of FAODs has become feasible. We analyzed the MS/MS results in Tianjin, China during a six-year period to evaluate the incidence, disease spectrum, and genetic characteristics of FAODs.

Methods

We analyzed the MS/MS results for screening FAODs from May 2013 to December 2018 in Tianjin, China. Infants with positive screening results were confirmed through next-generation sequencing and validated by Sanger sequencing.

Results

A total of 220,443 infants were screened and 25 FAODs patients were identified (1:8,817). Primary carnitine deficiency (PCD) with an incidence rate up to 1:20,040 was the most common disorder among all FAODs. Recurrent mutations of relatively common diseases, like PCD and short-chain acyl-CoA dehydrogenase deficiency (SCADD), were identified. During the follow-up, two patients suffered from sudden death due to carnitine palmitoyl transferase-Ⅱ deficiency (CPT Ⅱ) and very-long-chain acyl-CoA dehydrogenase deficiency (VLCAD).

Conclusion

Our data indicated that FAODs are relatively common in Tianjin and may even cause infant death in certain cases. The elucidated disease spectrum and genetic backgrounds elucidated in this study may contribute to the treatment and prenatal genetic counseling of FAODs.


Corresponding author: Dr. Rongxiu Zheng, Pediatric Department, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, 300052, Tianjin, PR China, Phone: +86-22-60361673, E-mail:

Funding source: Tianjin Women and Children's Health Center

Funding source: Beijing Genomics Institute

Acknowledgments

This study was supported by the Tianjin Women and Children's Health Center and Beijing Genomics Institute (BGI).

  1. Research funding: None declared.

  2. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The authors declare that they have no competing interests.

  6. Ethical statement: The study was approved by the Tianjin Women and Children's Health Center Ethics Committee.

References

1. Tucci S, Herebian D, Sturm M, Seibt A, Spiekerkoetter U. Tissue-specific strategies of the very-long chain acyl-CoA dehydrogenase-deficient (VLCAD-/-) mouse to compensate a defective fatty acid beta-oxidation. PloS One 2012;7:e45429. https://doi.org/10.1371/journal.pone.0045429.Search in Google Scholar

2. Vockley J, Whiteman DA. Defects of mitochondrial beta-oxidation: a growing group of disorders. Neuromuscul Disord 2002;12:235–46. https://doi.org/10.1016/s0960-8966(01)00308-x.Search in Google Scholar

3. Garg U, Dasouki M. Expanded newborn screening of inherited metabolic disorders by tandem mass spectrometry: clinical and laboratory aspects. Clin Biochem 2006;39:315–32. https://doi.org/10.1016/j.clinbiochem.2005.12.009.Search in Google Scholar

4. Wilcken B, Wiley V, Hammond J, Carpenter K. Screening newborns for inborn errors of metabolism by tandem mass spectrometry. N Engl J Med 2003;348:2304–12. https://doi.org/10.1056/nejmoa025225.10.1056/NEJMoa025225Search in Google Scholar PubMed

5. Moczulski D, Majak I, Mamczur D. An overview of beta-oxidation disorders. Postepy Hig Med Dosw (Online) 2009;63:266–77.Search in Google Scholar

6. Gregersen N, Andresen BS, Pedersen CB, Olsen RK, Corydon TJ, Bross P. Mitochondrial fatty acid oxidation defects—remaining challenges. J Inherit Metab Dis 2008;31:643–57. https://doi.org/10.1007/s10545-008-0990-y.Search in Google Scholar

7. Gregersen N, Andresen BS, Corydon MJ, Corydon TJ, Olsen RK, Bolund L, et al. Mutation analysis in mitochondrial fatty acid oxidation defects: Exemplified by acyl-CoA dehydrogenase deficiencies, with special focus on genotype-phenotype relationship. Hum Mutat 2001;18:169–89. https://doi.org/10.1002/humu.1174.Search in Google Scholar

8. Spiekerkoetter U. Mitochondrial fatty acid oxidation disorders: clinical presentation of long-chain fatty acid oxidation defects before and after newborn screening. J Inherit Metab Dis 2010;33:527–32. https://doi.org/10.1007/s10545-010-9090-x.Search in Google Scholar

9. Andresen BS, Lund AM, Hougaard DM, Christensen E, Gahrn B, Christensen M, et al. MCAD deficiency in Denmark. Mol Genet Metabol 2012;106:175–88. https://doi.org/10.1016/j.ymgme.2012.03.018.Search in Google Scholar

10. Champion MP. An approach to the diagnosis of inherited metabolic disease. Arch Dis Child Educ Pract 2010;95:40–6. https://doi.org/10.1136/adc.2008.151183.Search in Google Scholar

11. Niu DM, Chien YH, Chiang CC, Ho HC, Hwu WL, Kao SM, et al. Nationwide survey of extended newborn screening by tandem mass spectrometry in Taiwan. J Inherit Metab Dis 2010;33:S295–305. https://doi.org/10.1007/s10545-010-9129-z.Search in Google Scholar

12. Guo K, Zhou X, Chen X, Wu Y, Liu C, Kong Q. Expanded newborn screening for inborn errors of metabolism and genetic characteristics in a Chinese population. Front Genet 2018;9:122. https://doi.org/10.3389/fgene.2018.00122.Search in Google Scholar

13. Huang XW, Yang JB, Tong F, Yang RL, Mao HQ, Zhou XL, et al. [Screening for neonatal inborn errors of metabolism by electrospray ionization-tandem mass spectrometry and follow-up]. Zhonghua er ke za zhi = Chin J Pediatr 2011;49:765–70.Search in Google Scholar

14. Lin Y, Zheng Q, Zheng T, Zheng Z, Lin W, Fu Q. Expanded newborn screening for inherited metabolic disorders and genetic characteristics in a southern Chinese population. Clin Chim Acta; Int J Clin Chem 2019;494:106–11. https://doi.org/10.1016/j.cca.2019.03.1622.Search in Google Scholar

15. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics (Oxford, England) 2010;26:589–95. https://doi.org/10.1093/bioinformatics/btp698.Search in Google Scholar

16. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet 2011;43:491–8. https://doi.org/10.1038/ng.806.Search in Google Scholar

17. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinf 2013;43:11.10.1–.33. https://doi.org/10.1002/0471250953.bi1110s43.Search in Google Scholar

18. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297–303. https://doi.org/10.1101/gr.107524.110.Search in Google Scholar

19. Shibata N, Hasegawa Y, Yamada K, Kobayashi H, Purevsuren J, Yang Y, et al. Diversity in the incidence and spectrum of organic acidemias, fatty acid oxidation disorders, and amino acid disorders in Asian countries: Selective screening vs. expanded newborn screening. Mol Genet Metab Rep 2018;16:5–10. https://doi.org/10.1016/j.ymgmr.2018.05.003.Search in Google Scholar

20. Lindner M, Hoffmann GF, Matern D. Newborn screening for disorders of fatty-acid oxidation: experience and recommendations from an expert meeting. J Inherit Metab Dis 2010;33:521–6. https://doi.org/10.1007/s10545-010-9076-8.Search in Google Scholar

21. Han L, Han F, Ye J, Qiu W, Zhang H, Gao X, et al. Spectrum analysis of common inherited metabolic diseases in Chinese patients screened and diagnosed by tandem mass spectrometry. J Clin Lab Anal 2015;29:162–8. https://doi.org/10.1002/jcla.21745.Search in Google Scholar

22. Sun Y, Wang YY, Jiang T. Clinical features and genotyping of patients with primary carnitine deficiency identified by newborn screening. JPEM (J Pediatr Endocrinol Metab) 2017;30:879–83. https://doi.org/10.1515/jpem-2017-0002.10.1515/jpem-2017-0002Search in Google Scholar PubMed

23. Feuchtbaum L, Carter J, Dowray S, Currier RJ, Lorey F. Birth prevalence of disorders detectable through newborn screening by race/ethnicity. Genet Med Off J Am Coll Med Genet 2012;14:937–45. https://doi.org/10.1038/gim.2012.76.Search in Google Scholar

24. Kasper DC, Ratschmann R, Metz TF, Mechtler TP, Moslinger D, Konstantopoulou V, et al. The national Austrian newborn screening program - eight years experience with mass spectrometry. past, present, and future goals. Wien Klin Wochenschr 2010;122:607–13. https://doi.org/10.1007/s00508-010-1457-3.Search in Google Scholar

25. Vilarinho L, Rocha H, Sousa C, Marcao A, Fonseca H, Bogas M, et al. Four years of expanded newborn screening in Portugal with tandem mass spectrometry. J Inherit Metab Dis 2010;33:S133–8. https://doi.org/10.1007/s10545-010-9048-z.Search in Google Scholar

26. Oerton J, Khalid JM, Besley G, Dalton RN, Downing M, Green A, et al. Newborn screening for medium chain acyl-CoA dehydrogenase deficiency in England: prevalence, predictive value and test validity based on 1.5 million screened babies. J Med Screen 2011;18:173–81. https://doi.org/10.1258/jms.2011.011086.Search in Google Scholar

27. Li FY, El-Hattab AW, Bawle EV, Boles RG, Schmitt ES, Scaglia F, et al. Molecular spectrum of SLC22A5 (OCTN2) gene mutations detected in 143 subjects evaluated for systemic carnitine deficiency. Hum Mutat 2010;31:E1632–51. https://doi.org/10.1002/humu.21311.Search in Google Scholar

28. Yoon YA, Lee DH, Ki CS, Lee SY, Kim JW, Lee YW, et al. SLC22A5 mutations in a patient with systemic primary carnitine deficiency: the first Korean case confirmed by biochemical and molecular investigation. Ann Clin Lab Sci 2012;42:424–8.Search in Google Scholar

29. Koizumi A, Nozaki J, Ohura T, Kayo T, Wada Y, Nezu J, et al. Genetic epidemiology of the carnitine transporter OCTN2 gene in a Japanese population and phenotypic characterization in Japanese pedigrees with primary systemic carnitine deficiency. Hum Mol Genet 1999;8:2247–54. https://doi.org/10.1093/hmg/8.12.2247.Search in Google Scholar

30. Schimmenti LA, Crombez EA, Schwahn BC, Heese BA, Wood TC, Schroer RJ, et al. Expanded newborn screening identifies maternal primary carnitine deficiency. Mol Genet Metabol 2007;90:441–5. https://doi.org/10.1016/j.ymgme.2006.10.003.Search in Google Scholar

31. Kim SH, Park HD, Sohn YB, Park SW, Cho SY, Ji S, et al. Mutations of ACADS gene associated with short-chain acyl-coenzyme A dehydrogenase deficiency. Ann Clin Lab Sci 2011;41:84–8.Search in Google Scholar

32. Shirao K, Okada S, Tajima G, Tsumura M, Hara K, Yasunaga S, et al. Molecular pathogenesis of a novel mutation, G108D, in short-chain acyl-CoA dehydrogenase identified in subjects with short-chain acyl-CoA dehydrogenase deficiency. Hum Genet 2010;127:619–28. https://doi.org/10.1007/s00439-010-0822-7.Search in Google Scholar

33. Gallant NM, Leydiker K, Tang H, Feuchtbaum L, Lorey F, Puckett R, et al. Biochemical, molecular, and clinical characteristics of children with short chain acyl-CoA dehydrogenase deficiency detected by newborn screening in California. Mol Genet Metabol 2012;106:55–61. https://doi.org/10.1016/j.ymgme.2012.02.007.Search in Google Scholar

34. van Maldegem BT, Kloosterman SF, Janssen WJ, Augustijn PB, van der Lee JH, Ijlst L, et al. High prevalence of short-chain acyl-CoA dehydrogenase deficiency in the Netherlands, but no association with epilepsy of unknown origin in childhood. Neuropediatrics 2011;42:13–7. https://doi.org/10.1055/s-0031-1275342.Search in Google Scholar

35. Dessein AF, Fontaine M, Joncquel-Chevalier Curt M, Briand G, Sechter C, Mention-Mulliez K, et al. Fluxomic evidence for impaired contribution of short-chain acyl-CoA dehydrogenase to mitochondrial palmitate beta-oxidation in symptomatic patients with ACADS gene susceptibility variants. Clin Chim Acta; Int J Clin Chem 2017;471:101–6. https://doi.org/10.1016/j.cca.2017.05.026.Search in Google Scholar

36. Tajima G, Hara K, Tsumura M, Kagawa R, Okada S, Sakura N, et al. Screening of MCAD deficiency in Japan: 16years' experience of enzymatic and genetic evaluation. Mol Genet Metabol 2016;119:322–8. https://doi.org/10.1016/j.ymgme.2016.10.007.Search in Google Scholar

37. Woo HI, Park HD, Lee YW, Lee DH, Ki CS, Lee SY, et al. Clinical, biochemical and genetic analyses in two Korean patients with medium-chain acyl-CoA dehydrogenase deficiency. J Korean Lab Med 2011;31:54–60. https://doi.org/10.3343/kjlm.2011.31.1.54.Search in Google Scholar

38. Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, et al. An integrated map of genetic variation from 1,092 human genomes. Nature 2012;491:56–65. https://doi.org/10.1038/nature11632.Search in Google Scholar

39. Nochi Z, Olsen RKJ, Gregersen N. Short-chain acyl-CoA dehydrogenase deficiency: from gene to cell pathology and possible disease mechanisms. J Inherit Metab Dis 2017;40:641–55. https://doi.org/10.1007/s10545-017-0047-1.Search in Google Scholar

40. Baruteau J, Sachs P, Broue P, Brivet M, Abdoul H, Vianey-Saban C, et al. Clinical and biological features at diagnosis in mitochondrial fatty acid beta-oxidation defects: a French pediatric study of 187 patients. J Inherit Metab Dis 2013;36:795–803. https://doi.org/10.1007/s10545-012-9542-6.Search in Google Scholar

41. Iafolla AK, Thompson RJJr., Roe CR. Medium-chain acyl-coenzyme A dehydrogenase deficiency: clinical course in 120 affected children. J Pediatr 1994;124:409–15. https://doi.org/10.1016/s0022-3476(94)70363-9.Search in Google Scholar

42. Chace DH, DiPerna JC, Mitchell BL, Sgroi B, Hofman LF, Naylor EW. Electrospray tandem mass spectrometry for analysis of acylcarnitines in dried postmortem blood specimens collected at autopsy from infants with unexplained cause of death. Clin Chem 2001;47:1166–82. https://doi.org/10.1093/clinchem/47.7.1166.Search in Google Scholar

43. Qian J, Wang X, Liu J, Zhong J, Le Y, Melchior Tellier LCA, et al. Applying targeted next generation sequencing to dried blood spot specimens from suspicious cases identified by tandem mass spectrometry-based newborn screening. JPEM (J Pediatr Endocrinol Metab) 2017;30:979–88. https://doi.org/10.1515/jpem-2017-0003.Search in Google Scholar

Received: 2019-11-24
Accepted: 2020-03-16
Published Online: 2020-05-24

© 2020 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 17.5.2024 from https://www.degruyter.com/document/doi/10.1515/jpem-2019-0551/html
Scroll to top button