Skip to main content
Log in

Mitochondrial FAD shortage in SLC25A32 deficiency affects folate-mediated one-carbon metabolism

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The SLC25A32 dysfunction is associated with neural tube defects (NTDs) and exercise intolerance, but very little is known about disease-specific mechanisms due to a paucity of animal models. Here, we generated homozygous (Slc25a32Y174C/Y174C and Slc25a32K235R/K235R) and compound heterozygous (Slc25a32Y174C/K235R) knock-in mice by mimicking the missense mutations identified from our patient. A homozygous knock-out (Slc25a32−/−) mouse was also generated. The Slc25a32K235R/K235R and Slc25a32Y174C/K235R mice presented with mild motor impairment and recapitulated the biochemical disturbances of the patient. While Slc25a32−/− mice die in utero with NTDs. None of the Slc25a32 mutations hindered the mitochondrial uptake of folate. Instead, the mitochondrial uptake of flavin adenine dinucleotide (FAD) was specifically blocked by Slc25a32Y174C/K235R, Slc25a32K235R/K235R, and Slc25a32−/− mutations. A positive correlation between SLC25A32 dysfunction and flavoenzyme deficiency was observed. Besides the flavoenzymes involved in fatty acid β-oxidation and amino acid metabolism being impaired, Slc25a32−/− embryos also had a subunit of glycine cleavage system—dihydrolipoamide dehydrogenase damaged, resulting in glycine accumulation and glycine derived-formate reduction, which further disturbed folate-mediated one-carbon metabolism, leading to 5-methyltetrahydrofolate shortage and other folate intermediates accumulation. Maternal formate supplementation increased the 5-methyltetrahydrofolate levels and ameliorated the NTDs in Slc25a32−/− embryos. The Slc25a32K235R/K235R and Slc25a32Y174C/K235R mice had no glycine accumulation, but had another formate donor—dimethylglycine accumulated and formate deficiency. Meanwhile, they suffered from the absence of all folate intermediates in mitochondria. Formate supplementation increased the folate amounts, but this effect was not restricted to the Slc25a32 mutant mice only. In summary, we established novel animal models, which enabled us to understand the function of SLC25A32 better and to elucidate the role of SLC25A32 dysfunction in human disease development and progression.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Data availability

The datasets generated during or analyzed during the current study are all available in the manuscript.

References

  1. Schiff M, Veauville-Merllié A, Su CH et al (2016) SLC25A32 mutations and riboflavin-responsive exercise intolerance. N Engl J Med. https://doi.org/10.1056/NEJMc1513610

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hellebrekers DMEI, Sallevelt SCEH, Theunissen TEJ et al (2017) Novel SLC25A32 mutation in a patient with a severe neuromuscular phenotype. Eur J Hum Genet. https://doi.org/10.1038/ejhg.2017.62

    Article  PubMed  PubMed Central  Google Scholar 

  3. Al Shamsi B, Al Murshedi F, Al Habsi A, Al-Thihli K (2021) Hypoketotic hypoglycemia without neuromuscular complications in patients with slc25a32 deficiency. Eur J Hum Genet. https://doi.org/10.1038/s41431-021-00995-7

    Article  PubMed  Google Scholar 

  4. Titus SA, Moran RG (2000) Retrovirally mediated complementation of the glyb phenotype: cloning of a human gene encoding the carrier for entry of folates into mitochondria. J Biol Chem. https://doi.org/10.1074/jbc.M005163200

    Article  PubMed  Google Scholar 

  5. Haitina T, Lindblom J, Renström T, Fredriksson R (2006) Fourteen novel human members of mitochondrial solute carrier family 25 (SLC25) widely expressed in the central nervous system. Genomics. https://doi.org/10.1016/j.ygeno.2006.06.016

    Article  PubMed  Google Scholar 

  6. Spaan AN, Ijlst L, van Roermund CWT et al (2005) Identification of the human mitochondrial FAD transporter and its potential role in multiple acyl-coa dehydrogenase deficiency. Mol Genet Metab. https://doi.org/10.1016/j.ymgme.2005.07.014;86:441-7

    Article  PubMed  Google Scholar 

  7. McCarthy EA, Titus SA, Taylor SM et al (2004) A mutation inactivating the mitochondrial inner membrane folate transporter creates a glycine requirement for survival of Chinese hamster cells. J Biol Chem. https://doi.org/10.1074/jbc.M403677200

    Article  PubMed  Google Scholar 

  8. Becker ML, van Haandel L, Gaedigk R et al (2012) Red blood cell folate concentrations and polyglutamate distribution in juvenile arthritis: predictors of folate variability. Pharmacogenet Genomics. https://doi.org/10.1097/FPC.0b013e3283500202

    Article  PubMed  Google Scholar 

  9. Kim J, Lei Y, Guo J et al (2018) Formate rescues neural tube defects caused by mutations in slc25a32. Proc Natl Acad Sci USA. https://doi.org/10.1073/pnas.1800138115

    Article  PubMed  PubMed Central  Google Scholar 

  10. Steele JW, Kim SE, Finnell RH (2020) One-carbon metabolism and folate transporter genes: do they factor prominently in the genetic etiology of neural tube defects? Biochimie. https://doi.org/10.1016/j.biochi.2020.02.005

    Article  PubMed  PubMed Central  Google Scholar 

  11. Ducker GS, Rabinowitz JD (2017) One-carbon metabolism in health and disease. Cell Metab. https://doi.org/10.1016/j.cmet.2016.08.009

    Article  PubMed  Google Scholar 

  12. Pavone V, Chisari E, Vescio A et al (2018) The etiology of idiopathic congenital talipes equinovarus: a systematic review. J Orthop Surg Res. https://doi.org/10.1186/s13018-018-0913-z

    Article  PubMed  PubMed Central  Google Scholar 

  13. Frerman FE, Goodman SI (2019) Defects of electron transfer flavoprotein and electron transfer flavoprotein-ubiquinone oxidoreductase: glutaric acidemia type II. In: Valle DL, Antonarakis S, Ballabio A et al (eds) The online metabolic and molecular bases of inherited disease. McGraw Hill, New York

    Google Scholar 

  14. Frezza C, Cipolat S, Scorrano L (2007) Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat Protoc. https://doi.org/10.1038/nprot.2006.478

    Article  PubMed  Google Scholar 

  15. Chaiyarit S, Thongboonkerd V (2009) Comparative analyses of cell disruption methods for mitochondrial isolation in high-throughput proteomics study. Anal Biochem. https://doi.org/10.1016/j.ab.2009.07.026

    Article  PubMed  Google Scholar 

  16. Luongo TS, Eller JM, Lu MJ et al (2020) SLC25A51 is a mammalian mitochondrial NAD+ transporter. Nature. https://doi.org/10.1038/s41586-020-2741-7

    Article  PubMed  PubMed Central  Google Scholar 

  17. Leung K-Y, De Castro SCP, Cabreiro F et al (2013) Folate metabolite profiling of different cell types and embryos suggests variation in folate one-carbon metabolism, including developmental changes in human embryonic brain. Mol Cell Biochem. https://doi.org/10.1007/s11010-013-1613-y

    Article  PubMed  PubMed Central  Google Scholar 

  18. Liu H, Xu F, Gao Y et al (2020) An integrated LC-MS/MS strategy for quantifying the oxidative-redox metabolome in multiple biological samples. Anal Chem. https://doi.org/10.1021/acs.analchem.0c00242

    Article  PubMed  PubMed Central  Google Scholar 

  19. Peng M, Fang X, Huang Y et al (2013) Separation and identification of underivatized plasma acylcarnitine isomers using liquid chromatography–tandem mass spectrometry for the differential diagnosis of organic acidemias and fatty acid oxidation defects. J Chromatogr A. https://doi.org/10.1016/j.chroma.2013.10.036

    Article  PubMed  Google Scholar 

  20. Peng M-Z, Cai Y-N, Shao Y-X et al (2019) Simultaneous quantification of 48 plasma amino acids by liquid chromatography-tandem mass spectrometry to investigate urea cycle disorders. Clin Chim Acta. https://doi.org/10.1016/j.cca.2019.05.011

    Article  PubMed  Google Scholar 

  21. Tan B, Lu Z, Dong S et al (2014) Derivatization of the tricarboxylic acid intermediates with o-benzylhydroxylamine for liquid chromatography–tandem mass spectrometry detection. Anal Biochem. https://doi.org/10.1016/j.ab.2014.07.027

    Article  PubMed  Google Scholar 

  22. Kirsch SH, Herrmann W, Rabagny Y, Obeid R (2010) Quantification of acetylcholine, choline, betaine, and dimethylglycine in human plasma and urine using stable-isotope dilution ultra performance liquid chromatography–tandem mass spectrometry. J Chromatogr B. https://doi.org/10.1016/j.jchromb.2010.10.016

    Article  Google Scholar 

  23. Lamarre SG, MacMillan L, Morrow GP et al (2014) An isotope-dilution, GC–MS assay for formate and its application to human and animal metabolism. Amino Acids. https://doi.org/10.1007/s00726-014-1738-7

    Article  PubMed  Google Scholar 

  24. Tajima G, Sakura N, Yofune H et al (2005) Establishment of a practical enzymatic assay method for determination of isovaleryl-CoA dehydrogenase activity using high-performance liquid chromatography. Clin Chim Acta. https://doi.org/10.1016/j.cccn.2004.11.007

    Article  PubMed  Google Scholar 

  25. Bouvier D, Vianey-Saban C, Ruet S et al (2017) Development of a tandem mass spectrometry method for rapid measurement of medium- and very-long-chain acyl-CoA dehydrogenase activity in fibroblasts. In: Morava E, Baumgartner M, Patterson M, Rahman S, Zschocke J, Peters V (eds) JIMD Reports. Springer, Berlin, pp 71–78

    Google Scholar 

  26. Kerr D, Grahame G, Nakouzi G (2012) Assays of pyruvate dehydrogenase complex and pyruvate carboxylase activity. Methods Mol Biol. https://doi.org/10.1007/978-1-61779-504-6_7

    Article  PubMed  Google Scholar 

  27. Morrow GP, MacMillan L, Lamarre SG et al (2015) In vivo kinetics of formate metabolism in folate-deficient and folate-replete rats. J Biol Chem. https://doi.org/10.1074/jbc.M114.600718

    Article  PubMed  Google Scholar 

  28. Aartsma-Rus A, van Putten M (2014) Assessing functional performance in the mdx mouse model. J Vis Exp. https://doi.org/10.3791/51303

    Article  PubMed  PubMed Central  Google Scholar 

  29. Brooks SP, Trueman RC, Dunnett SB (2012) Assessment of motor coordination and balance in mice using the rotarod, elevated bridge, and footprint tests. Curr Protoc Mouse Biol. https://doi.org/10.1002/9780470942390.mo110165

    Article  PubMed  Google Scholar 

  30. Lin C-Y, Jhang Y-S, Lai S-C et al (2017) Antifatigue properties of tanshinone iia in mice subjected to the forced swimming test. Pharm Biol. https://doi.org/10.1080/13880209.2017.1401648

    Article  PubMed  PubMed Central  Google Scholar 

  31. Brautigam CA, Chuang JL, Tomchick DR et al (2005) Crystal structure of human dihydrolipoamide dehydrogenase: NAD+/NADH binding and the structural basis of disease-causing mutations. J Mol Biol. https://doi.org/10.1016/j.jmb.2005.05.014

    Article  PubMed  Google Scholar 

  32. Lienhart W-D, Gudipati V, Macheroux P (2013) The human flavoproteome. Arch Biochem Biophys. https://doi.org/10.1016/j.abb.2013.02.015

    Article  PubMed  PubMed Central  Google Scholar 

  33. Barile M, Passarella S, Bertoldi A et al (1993) Flavin adenine dinucleotide synthesis in isolated rat liver mitochondria caused by imported flavin mononucleotide. Arch Biochem Biophys. https://doi.org/10.1006/abbi.1993.1444

    Article  PubMed  Google Scholar 

  34. Saijo T, Tanaka K (1995) Isoalloxazine ring of FAD is required for the formation of the core in the Hsp60-assisted folding of medium chain acyl-CoA dehydrogenase subunit into the assembly competent conformation in mitochondria. J Biol Chem. https://doi.org/10.1074/jbc.270.4.1899

    Article  PubMed  Google Scholar 

  35. Nagao M, Tanaka K (1992) FAD-dependent regulation of transcription, translation, post-translational processing, and post-processing stability of various mitochondrial acyl-CoA dehydrogenases and of electron transfer flavoprotein and the site of holoenzyme formation. J Biol Chem 267(25):17925–17932

    Article  CAS  Google Scholar 

  36. Leung K-Y, Pai YJ, Chen Q et al (2017) Partitioning of one-carbon units in folate and methionine metabolism is essential for neural tube closure. Cell Rep. https://doi.org/10.1016/j.celrep.2017.10.072

    Article  PubMed  PubMed Central  Google Scholar 

  37. Narisawa A, Komatsuzaki S, Kikuchi A et al (2012) Mutations in genes encoding the glycine cleavage system predispose to neural tube defects in mice and humans. Hum Mol Genet. https://doi.org/10.1093/hmg/ddr585

    Article  PubMed  Google Scholar 

  38. Pai YJ, Leung K-Y, Savery D et al (2015) Glycine decarboxylase deficiency causes neural tube defects and features of non-ketotic hyperglycinemia in mice. Nat Commun. https://doi.org/10.1038/ncomms7388

    Article  PubMed  Google Scholar 

  39. Tzagoloff A, Jang J, Glerum DM, Wu M (1996) FLX1 codes for a carrier protein involved in maintaining a proper balance of flavin nucleotides in yeast mitochondria. J Biol Chem. https://doi.org/10.1074/jbc.271.13.7392

    Article  PubMed  Google Scholar 

  40. Zhang X, Li M, Xu Y et al (2019) Quantitative study of H protein lipoylation of the glycine cleavage system and a strategy to increase its activity by co-expression of LplA. J Biol Eng. https://doi.org/10.1186/s13036-019-0164-5

    Article  PubMed  PubMed Central  Google Scholar 

  41. Tibbetts AS, Appling DR (2010) Compartmentalization of mammalian folate-mediated one-carbon metabolism. Annu Rev Nutr. https://doi.org/10.1146/annurev.nutr.012809.104810

    Article  PubMed  Google Scholar 

  42. Bryant JD, Sweeney SR, Sentandreu E et al (2018) Deletion of the neural tube defect-associated gene Mthfd1l disrupts one-carbon and central energy metabolism in mouse embryos. J Biol Chem. https://doi.org/10.1074/jbc.RA118.002180

    Article  PubMed  PubMed Central  Google Scholar 

  43. Kao F, Chasin L, Puck TT (1969) Genetics of somatic mammalian cells X Complementation analysis of glycine-requiring mutants. Proc Natl Acad Sci USA. https://doi.org/10.1073/pnas.64.4.1284

    Article  PubMed  PubMed Central  Google Scholar 

  44. Zheng Y, Lin TY, Lee G et al (2018) Mitochondrial one-carbon pathway supports cytosolic folate integrity in cancer cells. Cell. https://doi.org/10.1016/j.cell.2018.09.041

    Article  PubMed  PubMed Central  Google Scholar 

  45. Ulrich M, Kristoffersen K, Rolschau J et al (1999) The influence of folic acid supplement on the outcome of pregnancies in the county of Funen in Denmark. Part ii. Congenital anomalies. A randomised study. Eur J Obstet Gynecol Reprod Biol. https://doi.org/10.1016/S0301-2115(99)00085-8

    Article  PubMed  Google Scholar 

  46. Sharp L, Miedzybrodzka Z, Cardy AH et al (2006) The C677T polymorphism in the methylenetetrahydrofolate reductase gene (MTHFR), maternal use of folic acid supplements, and risk of isolated clubfoot: a case-parent-triad analysis. Am J Epidemiol. https://doi.org/10.1093/aje/kwj285

    Article  PubMed  Google Scholar 

  47. Overholser MD, Whitley JR, O’Dell BL, Hogan AG (1954) The ventricular system in hydrocephalic rat brains produced by a deficiency of vitamin B12 or of folic acid in the maternal diet. Anat Rec. https://doi.org/10.1002/ar.1091200407

    Article  PubMed  Google Scholar 

  48. Woodard JC, Newberne PM (1966) Relation of vitamin B12 and one-carbon metabolism to hydrocephalus in the rat. J Nutr. https://doi.org/10.1093/jn/88.4.375

    Article  PubMed  Google Scholar 

  49. Santos C, Pai YJ, Mahmood MR et al (2020) Impaired folate 1-carbon metabolism causes formate-preventable hydrocephalus in glycine decarboxylase-deficient mice. J Clin Investig. https://doi.org/10.1172/jci132360

    Article  PubMed  PubMed Central  Google Scholar 

  50. Ippolito E, Gorgolini G (2021) Clubfoot pathology in fetus and pathogenesis. A new pathogenetic theory based on pathology, imaging findings and biomechanics-a narrative review. Ann Transl Med. 9(13):1095–1095

    Article  Google Scholar 

  51. Hwang SY, Sung B, Kim ND (2019) Roles of folate in skeletal muscle cell development and functions. Arch Pharm Res. https://doi.org/10.1007/s12272-018-1100-9

    Article  PubMed  Google Scholar 

  52. Olsen RKJ, Olpin SE, Andresen BS et al (2007) ETFDH mutations as a major cause of riboflavin-responsive multiple acyl-CoA dehydrogenation deficiency. Brain. https://doi.org/10.1093/brain/awm135

    Article  PubMed  Google Scholar 

  53. O’Callaghan B, Bosch AM, Houlden H (2019) An update on the genetics, clinical presentation, and pathomechanisms of human riboflavin transporter deficiency. J Inherit Metab Dis. https://doi.org/10.1002/jimd.12053

    Article  PubMed  PubMed Central  Google Scholar 

  54. Olsen RKJ, Koňaříková E, Giancaspero TA et al (2016) Riboflavin-responsive and -non-responsive mutations in fad synthase cause multiple acyl-CoA dehydrogenase and combined respiratory-chain deficiency. Am J Hum Genet. https://doi.org/10.1016/j.ajhg.2016.04.006

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We warmly thank the patient and her parents for participating in this study. We also thank Zhu-Yuan Xu (Account Manager, ProteinSimple) and Zu-Sen Weng (FAS, ProteinSimple) for their help in the Western blot assays, Zhe Li (Ph.D, School of Pharmaceutical Sciences, Sun Yat-sen University) for his help in the homology modeling analysis, and Prof. Ya-Ping Tang (Guangzhou Women and Children’s Medical Center) and Prof. Guo-Jun Shi (The third affiliated hospital, Sun-Yat Sen University) for their valuable suggestions in preparing the manuscript.

Funding

This study was funded by the National Natural Science Foundation of China (81802125 to M-Z P., 81700755 to Y-X S., and 81873661 to L L.), and Guangzhou Women and Children’s Medical Center/Guangzhou Institute of Pediatrics (IP-2018-024 to M-Z P.). The funding sources are not involved with the preparation of the article.

Author information

Authors and Affiliations

Authors

Contributions

Study concept and design were undertaken by M-Z P, Y-X S, and L L. Data acquisition and analysis were undertaken by M-Z P, Y-X S, X-Z L, K-D Z, Y-N C, Y-T L, M-Y J, Z-C L, X-Y S, W Z, and X-L J. The manuscript was written by M-Z P. and revised by L L, Y-X S, and X-L J.

Corresponding authors

Correspondence to Wen Zhang, Xiao-Ling Jiang or Li Liu.

Ethics declarations

Conflict of interest

The authors have no conflict of interest to declare.

Ethical approval and consent to participate

This study was performed in line with the principles of the Declaration of Helsinki. The ethics committee of Guangzhou Women and Children’s Medical Center authorized this study, and the Institutional Animal Care and Use Committee of Guangzhou Medical University approved the animal studies. Informed consent has been obtained from the patient and her guardian.

Consent for publication

The authors affirm that human research participants provided informed consent for publication of the images in Fig. 1b, 1c and 1d.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Peng, MZ., Shao, YX., Li, XZ. et al. Mitochondrial FAD shortage in SLC25A32 deficiency affects folate-mediated one-carbon metabolism. Cell. Mol. Life Sci. 79, 375 (2022). https://doi.org/10.1007/s00018-022-04404-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00018-022-04404-0

Keywords

Navigation