Skip to main content

Disorders of Pyruvate Metabolism and the Tricarboxylic Acid Cycle

  • Chapter
Inborn Metabolic Diseases

Zusammenfassung

Owing to the role of pyruvate and the TCA cycle in energy metabolism, as well as in gluconeogenesis, lipogenesis and amino acid synthesis, defects in pyruvate metabolism and in the TCA cycle almost invariably affect the central nervous system. The severity and the different clinical phenotypes vary widely among patients and are not always specific, the range of manifestations extending from overwhelming neonatal lactic acidosis and early death to relatively normal adult life and variable effects on systemic functions. The same clinical manifestations may be caused by other defects of energy metabolism, especially defects of the respiratory chain. Diagnosis depends primarily on biochemical analyses of metabolites in body fluids, followed by definitive enzymatic assays in cells or tissues, and DNA analysis. PC deficiency constitutes a defect both in the Krebs cycle and in gluconeogenesis, but generally presents with severe neurological dysfunction and lactic acidosis rather than with fasting hypoglycaemia. Deficiency of PEPCK is now considered to be a secondary phenomenon. Deficiency of PDHC impedes glucose oxidation and aerobic energy production, and ingestion of carbohydrate aggravates lactic acidosis. Treatment of disorders of pyruvate metabolism comprises avoidance of fasting (PC) or minimising dietary carbohydrate intake (PDHC) and enhancing anaplerosis (restoration of pools of intermediate metabolites). Dihydrolipoamide dehydrogenase (E3) deficiency affects PDHC and also the 2-ketoglutarate dehydrogenase complex (KDHC) and the branched-chain 2-ketoacid dehydrogenase (BCKD) complex, with biochemical manifestations of all three disorders. The deficiencies of the TCA cycle enzymes, KDHC and fumarase, interrupt the cycle, resulting in accumulation of the corresponding substrates. Succinate dehydrogenase deficiency represents a unique disorder affecting both the TCA cycle and the respiratory chain. Defects of mitochondrial transport of pyruvate and ketoglutarate have also been identified. Treatment strategies for the TCA cycle defects are limited.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 149.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Marin-Valencia I, Roe CR, Pascual JM (2010) Pyruvate carboxylase deficiency: mechanisms, mimics and anaplerosis. Mol Genet Metab 101:9–17

    Google Scholar 

  2. Garcia-Cazorla A, Rabier D, Touati G et al. (2006) Pyruvate carboxylase deficiency: metabolic characteristics and new neurological aspects. Ann Neurol 59:121–127

    Google Scholar 

  3. Ostergaard E, Duno M, Møller LB et al. (2013) Novel Mutations in the PC Gene in Patients with Type B Pyruvate Carboxylase Deficiency. JIMD Rep 9:1–5

    Google Scholar 

  4. Murin R, Cesar M, Kowtharapu BS et al. (2009) Expression of pyruvate carboxylase in cultured oligodendroglial, microglial and ependymal cells. Neurochem Res 34:480–489

    Google Scholar 

  5. Park LC, Zhang H, Sheu KF et al. (1999) Metabolic impairment induces oxidative stress, compromises inflammatory responses, and inactivates a key mitochondrial enzyme in microglia. J Neurochem 72:1948–1958

    Google Scholar 

  6. Unal O, Orhan D, Ostergaard E et al. (2013) A patient with pyruvate carboxylase deficiency and nemaline rods on muscle biopsy. J Child Neurol 28:1505–1508

    Google Scholar 

  7. Carbone MA, Applegarth DA, Robinson BH (2002) Intron retention and frameshift mutations result in severe pyruvate carboxylase deficiency in two male siblings. Hum Mutat 20:48–56

    Google Scholar 

  8. Wang D, Yang H, de Braganca KC et al. (2008) The molecular basis of pyruvate carboxylase deficiency: mosaicism correlates with progrolonged survival. Mol Genet Metab 95:31–38

    Google Scholar 

  9. Monnot S, Serre V, Chadefaux-Vekemans B et al. (2009) Structural insights on pathogenic effects of novel mutations causing pyruvate carboxylase deficiency. Hum Mutat 30:734–740

    Google Scholar 

  10. Ahmad A, Kahler SG, Kishani PS et al. (1999) Treatment with pyruvate carboxylase deficiency with high doses of citrate and aspartate. Am J Med Genet 87:331–338

    Google Scholar 

  11. Ortez C, Jou C, Cortès-Saladelafont E (2013) Infantile parkinsonism and GABAergic hypotransmission in a patient with pyruvate carboxylase deficiency. Gene 532:302–306

    Google Scholar 

  12. Nyhan WL, Khanna A, Barshop BA et al. (2002) Pyruvate carboxylase deficiency – insights from liver transplantation. Mol Genet Metab 77:143–149

    Google Scholar 

  13. Mochel F, DeLonlay P, Touati G, et al. (2005) Pyruvate carboxylase deficiency: clinical and biochemical response to anaplerotic diet therapy. Mol Genet Metab 84:305–312

    Google Scholar 

  14. Breen C, White FJ, Scott CA et al. (2014) Unsuccessful treatment of severe pyruvate carboxylase deficiency with triheptanoin. Eur J Pediatr 173:361–366

    Google Scholar 

  15. Clayton PT, Hyland K, Brand M, Leonard JV (1986) Mitochondrial phosphoenolpyruvate carboxykinase deficiency Eur J Pediatr 145:46–50

    Google Scholar 

  16. Leonard JV, Hyland K, Furukawa N, Clayton PT (1991) Mitochondrial phosphoenolpyruvate carboxykinase deficiency. Eur J Pediatr 150:198–199

    Google Scholar 

  17. Bodnar AG, Cooper JM, Holt LJ et al. (1993) Nuclear complementation restores mtDNA levels in cultured cells from a patient with mtDNA depletion. Am J Hum Genet 53:663–669

    Google Scholar 

  18. Bodnar AG, Cooper JM, Leonard JV, Schapira AH (1995) Respiratory-deficient human fibroblasts exhibiting defective mitochondrial DNA replication. Biochem J 305:817–822

    Google Scholar 

  19. Barnerias C, Saudubray JM, Touati G et al. (2010) Pyruvate dehydrogenase complex deficiency: four neurological phenotypes with differing pathogenesis. Dev Med Child Neurol 52:2:e1–9

    Google Scholar 

  20. Sperl W, Fleuren L, Freisinger P et al. (2015) The spectrum of pyruvate oxidation defects in the diagnosis of mitochondrial disorders. J Inherit Metab Dis 38:391–403

    Google Scholar 

  21. Strassburg HM, Koch J, Mayr J et al. (2006) Acute flaccid paralysis as initial symptom in 4 patients with novel E1-alpha mutations of the pyruvate dehydrogenase complex. Neuropediatrics 37:137–141

    Google Scholar 

  22. Debray FG, Lambert M, Gagne R et al. (2008) Pyruvate dehydrogenase deficiency presenting as intermittent isolated acute ataxia. Neuropediatrics 39:20–23

    Google Scholar 

  23. Sedel F, Challe G, Layer JM et al. (2008) Thiamine responsive pyruvate dehydrogenase deficiency in an adult with peripheral neuropathy and optic neuropathy. J Neurol Neurosurg Psychiatry 79:846–847

    Google Scholar 

  24. De Meirleir L, Specola N, Seneca S, Lissens W (1998) Pyruvate dehydrogenase E1-alpha deficiency in a family: different clinical presentation in two siblings. J Inherit Metab Dis 21:224–226

    Google Scholar 

  25. De Meirleir L (2002) Defects of pyruvate metabolism and the Krebs cycle. J Child Neurol 17:S26–33

    Google Scholar 

  26. Miné M, Zater M, Ogier H et al. (2004) A puzzling case of mosaicism with mutation leading to nonsense mRNA decay (NMD) in a male patient with pyruvate dehydrogenase (PDH) deficiency. J Inherit Metab Dis 27:S123

    Google Scholar 

  27. Okajima K, Worman ML, Byrne LC et al. (2006) Somatic mosaicism in a male with an exon skipping mutation in PDHA1 of the pyruvate dehydrogenase complex results in a milder phenotype. Mol Genet Metab 87:162–168

    Google Scholar 

  28. Coughlin C, Krantz I, Schmitt E et al. (2010) Somatic mosaicism for PDHA1 mutation in a male with pyruvate dehydrogenase deficiency. Mol Genet Metab 100:296–269

    Google Scholar 

  29. Ridout C, Brown R, Walter J et al. (2008) Somatic mosaicism for a PDHA1 mutation in a female with pyruvate dehydrogenase deficiency. Hum Genet 124:187–193

    Google Scholar 

  30. Castiglioni C, Verrigni D, Okuma C, Diaz A, et al. (2015) Pyruvate dehydrogenase deficiency presenting as isolated exercise induced dystonia successfully reversed with thiamine supplemenation. Eur J Ped Neurology 19:497–503

    Google Scholar 

  31. Michotte A, De Meirleir L, Lissens W et al. (1993) Neuropathological findings of a patient with pyruvate dehydrogenase E1 alpha deficiency presenting as a cerebral lactic acidosis. Acta Neuropathol (Berl) 85:674–678

    Google Scholar 

  32. Han Z, Srivastava A, Stacpoole P (2008) Pyruvate dehydrogenase complex deficiency caused by ubiquitination and proteasome-mediated degradation of the E1 subunit. J Biol Chem 283:237–243

    Google Scholar 

  33. Okajima K, Korotchkina L, Rupar T et al. (2008) Mutations of the E1 beta subunit gene (PDHB) in four families with pyruvate dehydrogenase deficiency. Mol Genet Metab 93:371–380

    Google Scholar 

  34. Quintana E, Mayr J, Garcia Silva M et al. (2009) PDHE(1) beta deficiency with novel mutations in two patients with Leigh syndrome. J Inherit Metab Dis 32 (Suppl 1):S330–S343

    Google Scholar 

  35. Imbard A, Boutron A, Vequaud C et al. (2011) Molecular characterization of 82 patients with pyruvate dehydrogenase complex deficiency Structural implications of novel amino acid substitutions in E1 protein. Mol Genet Metabol 104:507–516

    Google Scholar 

  36. Ito M, Kobashi H, Naito E et al. (1992) Decrease of pyruvate dehydrogenase phosphatase activity in patients with congenital lactic acidemia. Clin Chim Acta 209:1–7

    Google Scholar 

  37. Maj M, Mackay N, Levandovskyi V et al. (2005) Pyruvate dehydrogenase phosphatase deficiency: identification of the first mutation in two brothers and restoration of activity by protein complementation. J Clin Endocrinol Metab 90:4101–4107

    Google Scholar 

  38. Cameron J, Maj M, Levandovskyi V et al. (2009) Pyruvate dehydrogenase phosphatase (PDP1) null mutation produces a lethal infantile phenotype. Hum Genet 125:319–326

    Google Scholar 

  39. Head R, Brown R, Zolkipli Z et al. (2005) Clinical and genetic spectrum of pyruvate dehydrogenase deficiency: dihydrolipoamide acetyltransferase (E2) deficiency. Ann Neurol 58:234–241

    Google Scholar 

  40. McWilliam C, Ridout C, Brown R et al. (2010) Pyruvate dehydrogenase E2 deficiency: a potentially treatable cause of episodic dystonia. Eur J Paediatr Neurol 14:349–353

    Google Scholar 

  41. Brown RM, Head RA, Brown GK (2002) Pyruvate dehydrogenase E3 binding protein deficiency. Hum Genet 110:187–191

    Google Scholar 

  42. Brown RM, Head R, Morris AA et al. (2006) Pyruvate dehydrogenase E3 binding protein (protein X) deficiency. Dev Med Child Neurol 48:756–760

    Google Scholar 

  43. Schiff M, Miné M, Brivet M et al. (2006) Leigh’s disease due to a new mutation in the PDHX gene. Ann Neurol 59:709–714

    Google Scholar 

  44. Ivanov IS, Azmanov DN, Ivanova MB et al. (2014) Founder p.Arg446* mutation in PDHX gene explains over half of cases with congenital lactic acidosis in Roma children. Mol Genet Metabol 113:76–83

    Google Scholar 

  45. Kennerson ML, Yiu EM, Chuang DT, et al (2013) A new locus for X-linked dominant Charcot–Marie–Tooth disease (CMTX6) is caused by mutations in the pyruvate dehydrogenase kinase isoenzyme3 (PDK3) gene Human Molecular Genetics 22:1404–1416

    Google Scholar 

  46. Lissens W, De Meirleir L, Seneca S et al. (2000) Mutations in the X-linked pyruvate dehydrogenase (E1) α sububit gene (PDHA1) in patients with a pyruvate dehydrogenase complex deficiency. Hum Mutat 15:209–219

    Google Scholar 

  47. Quintana E, Gort L, Busquets C et al. (2009) Mutational study in the PDHA1 gene of 40 patients suspected of pyruvate dehydrogenase complex deficiency. Clin Gen 77:274–282

    Google Scholar 

  48. Ridout CK, Keighley P, Krywawych S et al. (2008) A putative exonic solicing enhancer in exon 7 of the PDHA1 gene affects splicing of adjacent exons. Hum Mutat 29:451

    Google Scholar 

  49. Boichard A, Venet L, Naas T et al. (2008) Two silent substitutions in the PDHA1 gene cause exon 5 skipping by disruption of a putative exonic splicing enhancer. Mol Genet Metab 93:323–330

    Google Scholar 

  50. Miné M, Brivet M, Touati G et al. (2003) Splicing error in E1 alpha pyruvate dehydrogenase mRNA caused by novel intronic mutation responsible for lactic acidosis and mental retardation. J Biol Chem 278:11768–11772

    Google Scholar 

  51. Brivet M, Moutard ML, Zater M et al. (2005) First characterization of a large deletion of the PDHA1 gene. Mol Genet Metab 86:456–461

    Google Scholar 

  52. Miné M, Brivet M, Schiff et al. (2006) A novel gross deletion caused by non-homologous recombination of the PDHX gene in a patient with pyruvate dehydrogenase deficiency. Mol Genet Metab 89:106–110

    Google Scholar 

  53. Miné M, Chen JM, Brivet M et al. (2007) A large genomic deletion in the PDHX gene caused by the retrotranspositional insertion of a full length LINE-1 element. Hum Mutat 28:137–142

    Google Scholar 

  54. De Meirleir L, Lissens W, Denis R et al. (1993) Pyruvate dehydrogenase deficiency: clinical and biochemical diagnosis. Pediatr Neurol 9:216–220

    Google Scholar 

  55. Willemsen M, Rodenburg RJT, Teszas A et al. (2006) Females with PDHA1 gene mutations: a diagnostic challenge. Mitochondrion 6:155–159

    Google Scholar 

  56. Naito E, Ito M, Yokota I et al. (2002) Diagnosis and molecular analysis of three male patients with thiamine-responsive pyruvate dehydrogenase complex deficiency. J Neurol Sci 201:33–37

    Google Scholar 

  57. Falk RE, Cederbaum SD, Blass JP et al. (1976) Ketogenic diet in the management of pyruvate dehydrogenase deficiency. Pediatrics 58:713–721

    Google Scholar 

  58. Wexler ID, Hemalatha SG, McConnell J et al. (1997) Outcome of pyruvate dehydrogenase deficiency treated with ketogenic diets. Studies in patients with identical mutations. Neurology 49:1655–1661

    Google Scholar 

  59. Naito E, Ito M, Yokota I et al. (2002) Thiamine-responsive pyruvate dehydrogenase deficiency in two patients caused by a point mutation (F2005L and L216F) within the thiamine pyrophosphate binding site. Biochim Biophys Acta 1588:79–84

    Google Scholar 

  60. Stacpoole PW, Barnes CL, Hurbanis MD et al. (1997) Treatment of congenital lactic acidosis with dichloroacetate: a review. Arch Pediatr Adolesc Med 77:535–541

    Google Scholar 

  61. Fouque F, Brivet M, Boutron A et al. (2003) Differential effect of DCA treatment on the pyruvate dehydrogenase complex in patients with severe PDHC deficiency. Pediatr Res 53:793–799

    Google Scholar 

  62. Ferriero R, Iannuzzi C, Manco G et al. (2015) Differential inhibition of PDKs by phenylbutyrate and enhancement of pyruvate dehydrogenase complex activity by combination with dichloroacetate. J Inher Metab Dis 38:895–904

    Google Scholar 

  63. Ferriero R, Boutron A, Brivet M et al. (2014) Phenylbutyrate increases pyruvate dehydrogenase complex activity in cells harboring a variety of defects. Ann Clin Translational Neurology 1:462–470

    Google Scholar 

  64. Elpeleg ON, Ruitenbeek W, Jakobs C et al. (1995) Congenital lactic acidemia caused by lipoamide dehydrogenase deficiency with favorable outcome. J Pediatr 126:72–74

    Google Scholar 

  65. Elpeleg ON, Shaag A, Glustein JZ et al. (1997) Lipoamide dehydrogenase deficiency in Ashkenazi Jews: an insertion mutation in the mitochondrial leader sequence. Hum Mutat 10:256–257

    Google Scholar 

  66. Grafakou O, Oexle K, van den Heuvel L et al. (2003) Leigh syndrome due to compound heterozygosity of dihydrolipoamide dehydrogenase gene mutations. Description of the first E3 splice site mutation. Eur J Pediatr 162:714–718

    Google Scholar 

  67. Cameron JM, Levandovskyi V, McKay N et al. (2006) Novel mutations in dihydrolipoamide dehydrogenase deficiency in two cousins with borderline-normal PDH complex activity. Am J Med Genet 140A:1542–1552

    Google Scholar 

  68. Shaag A, Saada A, Berger I et al. (1999) Molecular basis of lipoamide dehydrogenase deficiency in Ashkenazi Jews. Am J Med Genet 82:177–182

    Google Scholar 

  69. Hong YS, Korman SH, Lee J et al. (2003) Identification of a common mutation (Gly194Cys) in both Arab Moslem and Ashkenazi Jewish patients with dihydrolipoamide dehydrogenase (E3) deficiency: possible beneficial effect of vitamin therapy. J Inherit Metab Dis 26:816–818

    Google Scholar 

  70. Odièvre M, Chrétien D, Munnich A et al. (2005) A novel mutation in the dihydrolipoamide dehydrogenase E3 subunit gene (DLD) resulting in an atypical form of a-ketoglutarate dehydrogenase deficiency Hum Mutat 25:323–324

    Google Scholar 

  71. Carozzo R, Torraco A, Fiermonte G et al. (2014) Riboflavin responsive mitochondrial myopathy is a new phenotype of dihydrolipoamide dehydrogenase deficiency. The chaperone-like effect of vitamine B2. Mitochondrion 14:49–57

    Google Scholar 

  72. Rustin P, Bourgeron T, Parfait B et al. (1997) Inborn errors of the Krebs cycle: a group of unusual mitochondrial diseases in human. Biochim Biophys Acta 1361:185–197

    Google Scholar 

  73. Dunckelman RJ, Ebinger F, Schulze A et al. (2000) 2-Ketoglutarate dehydrogenase deficiency with intermittent 2-ketoglutaric aciduria. Neuropediatrics 31:35–38

    Google Scholar 

  74. Al Aqeel A, Rashed M, Ozand PT et al. (1994) A new patient with alpha-ketoglutaric aciduria and progressive extrapyramidal tract disease. Brain Dev 16:S33–37

    Google Scholar 

  75. Surendran S, Michals-Matalon K, Krywawych S et al. (2002) DOOR syndrome: deficiency of E1 component of the 2-oxoglutarate dehydrogenase complex. Am J Med Genet 113:371–374

    Google Scholar 

  76. Kerrigan JF, Aleck KA, Tarby TJ et al. (2000) Fumaric aciduria: clinical and imaging features. Ann Neurol 47:583–588

    Google Scholar 

  77. Coughlin EM, Chalmers RA, Slaugenhaupt SA et al. (1993) Identification of a molecular defect in a fumarase deficient patient and mapping of the fumarase gene. Am J Hum Genet 53:86–89

    Google Scholar 

  78. Remes AM, Filppula SA, Rantala H et al. (2004) A novel mutation of the fumarase gene in a family with autosomal recessive fumarase deficiency J Mol Med 82:550–554

    Google Scholar 

  79. Gross KL, Panhuysen CI, Kleinman MS et al. (2004) Involvement of fumarate hydratase in nonsyndromic uterine leiomyomas: genetic linkage analysis and FISH studies. Genes Chromosomes Cancer 41:183–190

    Google Scholar 

  80. Bourgeron T, Rustin P, Chretien D et al. (1995) Mutation of a nuclear succinate dehydrogenase gene results in mitochondrial respiratory chain deficiency. Nat Genet 11:144–149

    Google Scholar 

  81. Haller RG, Henriksson KG, Jorfeldt L et al. (1991) Deficiency of skeletal muscle succinate dehydrogenase and aconitase. Pathophysiology of exercise in a novel human muscle oxidative defect. J Clin Invest 88:1197–1206

    Google Scholar 

  82. Hall RE, Henriksson KG, Lewis SF et al. (1993) Mitochondrial myopathy with succinate dehydrogenase and aconitase deficiency. Abnormalities of several iron-sulfur proteins. J Clin Invest 92:2660–2666

    Google Scholar 

  83. Van Coster R, Seneca S, Smet J et al. (2003) Homozygous Gly555Glu mutation in the nuclear-encoded 70kDa flavoprotein gene causes instability of the respiratory chain complex II. Am J Med Genet 120A:13–18

    Google Scholar 

  84. Parfait B, Chretien D, Rotig A et al. (2000) Compound heterozygous mutations in the flavoprotein gene of the respiratory chain complex II in a patient with Leigh syndrome. Hum Genet 106:236–243

    Google Scholar 

  85. Kollberg G, Tulinius M, Melberg A et al. (2009) Clinical manifestation and a new ISCU mutation in iron-sulphur cluster deficiency myopathy. Brain 132:2170–2179

    Google Scholar 

  86. Birch-Machin MA, Taylor RW, Cochran B et al. (2000) Late-onset optic atrophy, ataxia, and myopathy associated with a mutation of a complex II gene. Ann Neurol 48:330–335

    Google Scholar 

  87. Rustin P, Munnich A, Rotig A (2002) Succinate dehydrogenase and human diseases: new insights into a well-known enzyme. Eur J Hum Genet 10:289–291

    Google Scholar 

  88. Hartong DT, Dange M, McGee EL et al. (2008) Novel insights into the contributions of isocitrate dehydrogenases to the Krebs cycle from patients with retinitis pigmentosa. Nat Genet 40:1230–1234

    Google Scholar 

  89. Brivet M, Garcia-Cazorla A, Lyonnet S et al. (2003) Impaired mitochondrial pyruvate importation in a patient and a fetus at risk. Mol Gen Metab 78:186–192

    Google Scholar 

  90. Bricker DK, Taylor EB, Schell JC et al. (2012) A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila and humans. Science 337:96–100

    Google Scholar 

  91. Santra S, Cameron J, Shyr C et al. (2016) Cytosolic phosphoenolpyruvate carboxykinase deficiency presenting with acute liver failure following gastroenteritis. J Inherit Metab Dis 39 (Suppl1) S47

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Linda de Meirleir , Angels Garcia-Cazorla or Michèle Brivet .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

de Meirleir, L., Garcia-Cazorla, A., Brivet, M. (2016). Disorders of Pyruvate Metabolism and the Tricarboxylic Acid Cycle. In: Saudubray, JM., Baumgartner, M., Walter, J. (eds) Inborn Metabolic Diseases. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-662-49771-5_11

Download citation

  • DOI: https://doi.org/10.1007/978-3-662-49771-5_11

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-662-49769-2

  • Online ISBN: 978-3-662-49771-5

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics