Skip to main content
Log in

Isoforms of the DHTKD1-Encoded 2-Oxoadipate Dehydrogenase, Identified in Animal Tissues, Are not Observed upon the Human DHTKD1 Expression in Bacterial or Yeast Systems

  • ACCELERATED PUBLICATION
  • Published:
Biochemistry (Moscow) Aims and scope Submit manuscript

Abstract

Unlike the OGDH-encoded 2-oxoglutarate dehydrogenase (OGDH), which is an essential enzyme present in all animal tissues, expression of the DHTKD1-encoded isoenzyme, 2-oxoadipate dehydrogenase (OADH), depends on a number of factors, and mutant DHTKD1 phenotypes are rarely manifested. Physiological significance of OADH is also obscured by the fact that both isoenzymes transform 2-oxoglutarate and 2-oxoadipate. By analogy with other members of the 2-oxo acid dehydrogenases family, OADH is assumed to be a component of the multienzyme complex that catalyzes oxidative decarboxylation of 2-oxoadipate. This study aims at molecular characterization of OADH from animal tissues. Phylogenetic analysis of 2-oxo acid dehydrogenases reveals OADH only in animals and Dictyostelium discoideum slime mold, within a common branch with bacterial OGDH. Examination of partially purified animal OADH by immunoblotting and mass spectrometry identifies two OADH isoforms with molecular weights of about 130 and 70 kDa. These isoforms are not observed upon the expression of human DHTKD1 protein in either bacterial or yeast system, where the synthesized OADH is of expected molecular weight (about 100 kDa). Thus, the OADH isoforms present in animal tissues, may result from the animal-specific regulation of the DHTKD1 expression and/or posttranslational modifications of the encoded protein. Mapping of the peptides identified in the OADH preparations, onto the protein structure suggests that the 70-kDa isoform is truncated at the N-terminus, but retains the active site. Since the N-terminal domain of OGDH is required for the formation of the multienzyme complex, it is possible that the 70-kDa isoform catalyzes non-oxidative transformation of dicarboxylic 2-oxo acids that does not require the multienzyme structure. In this case, the ratio of the OADH isoforms in animal tissues may correspond to the ratio between the oxidative and non-oxidative decarboxylation of 2-oxoadipate.

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.

(1)
Fig. 1.
Fig. 2.
Fig. 3.
Fig. 4.
Fig. 5.
Fig. 6.

Similar content being viewed by others

Abbreviations

DHTKD1 :

2-oxoadipate dehydrogenase (dehydrogenase E1 and transketolase domain-containing 1) gene

OADH:

2-oxoadipate dehydrogenase

OGDH:

2-oxoglutarate dehydrogenase

REFERENCES

  1. Bunik, V. I., and Degtyarev, D. (2008) Structure-function relationships in the 2-oxo acid dehydrogenase family: substrate-specific signatures and functional predictions for the 2-oxoglutarate dehydrogenase-like proteins, Proteins, 71, 874-890, doi: https://doi.org/10.1002/prot.21766.

    Article  CAS  PubMed  Google Scholar 

  2. Tsepkova, P. M., Artiukhov, A. V., Boyko, A. I., Aleshin, V. A., Mkrtchyan, G. V., Zvyagintseva, M. A., Ryabov, S. I., Ksenofontov, A. L., Baratova, L. A., Graf, A. V., and Bunik, V. I. (2017) Thiamine induces long-term changes in amino acid profiles and activities of 2-oxoglutarate and 2-oxoadipate dehydrogenases in rat brain, Biochemistry (Moscow), 82, 723-736, doi: https://doi.org/10.1134/S0006297917060098.

    Article  CAS  Google Scholar 

  3. Artiukhov, A. V., Grabarska, A., Gumbarewicz, E., Aleshin, V. A., Kahne, T., Obata, T., Kazantsev, A. V., Lukashev, N. V., Stepulak, A., Fernie, A. R., and Bunik, V. I. (2020) Synthetic analogues of 2-oxo acids discriminate metabolic contribution of the 2-oxoglutarate and 2-oxoadipate dehydrogenases in mammalian cells and tissues, Sci. Rep., 10, 1886, doi: https://doi.org/10.1038/s41598-020-58701-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Nemeria, N. S., Gerfen, G., Yang, L., Zhang, X., and Jordan, F. (2018) Evidence for functional and regulatory cross-talk between the tricarboxylic acid cycle 2-oxoglutarate dehydrogenase complex and 2-oxoadipate dehydrogenase on the l-lysine, l-hydroxylysine and l-tryptophan degradation pathways from studies in vitro, Biochim. Biophys. Acta Bioenerg., 1859, 932-939, doi: https://doi.org/10.1016/j.bbabio.2018.05.001.

    Article  CAS  PubMed  Google Scholar 

  5. Nemeria, N. S., Gerfen, G., Nareddy, P. R., Yang, L., Zhang, X., Szostak, M., and Jordan, F. (2018) The mitochondrial 2-oxoadipate and 2-oxoglutarate dehydrogenase complexes share their E2 and E3 components for their function and both generate reactive oxygen species, Free Radic. Biol. Med., 115, 136-145, doi: https://doi.org/10.1016/j.freeradbiomed.2017.11.018.

    Article  CAS  PubMed  Google Scholar 

  6. Danhauser, K., Sauer, S. W., Haack, T. B., Wieland, T., Staufner, C., Graf, E., Zschocke, J., Strom, T. M., Traub, T., Okun, J. G., Meitinger, T., Hoffmann, G. F., Prokisch, H., and Kolker, S. (2012) DHTKD1 mutations cause 2-aminoadipic and 2-oxoadipic aciduria, Am. J. Hum. Genet., 91, 1082-1087, doi: https://doi.org/10.1016/j.ajhg.2012.10.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Stiles, A. R., Venturoni, L., Mucci, G., Elbalalesy, N., Woontner, M., Goodman, S., and Abdenur, J. E. (2016) New cases of DHTKD1 mutations in patients with 2-ketoadipic aciduria, JIMD Rep., 25, 15-19, doi: https://doi.org/10.1007/8904_2015_462.

    Article  PubMed  Google Scholar 

  8. Amsterdam, A., Nissen, R. M., Sun, Z., Swindell, E. C., Farrington, S., and Hopkins, N. (2004) Identification of 315 genes essential for early zebrafish development, Proc. Natl. Acad. Sci. USA, 101, 12792-12797, doi: https://doi.org/10.1073/pnas.0403929101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yap, Z. Y., Strucinska, K., Matsuzaki, S., Lee, S., Si, Y., Humphries, K., Tarnopolsky, M. A., and Yoon, W. H. (2020) A biallelic pathogenic variant in the OGDH gene results in a neurological disorder with features of a mitochondrial disease, J. Inherit. Metab. Dis., doi: https://doi.org/10.1002/jimd.12248.

  10. Xu, W., Zhu, H., Gu, M., Luo, Q., Ding, J., Yao, Y., Chen, F., and Wang, Z. (2013) DHTKD1 is essential for mitochondrial biogenesis and function maintenance, FEBS Lett., 587, 3587-3592, doi: https://doi.org/10.1016/j.febslet.2013.08.047.

    Article  CAS  PubMed  Google Scholar 

  11. Plubell, D. L., Fenton, A. M., Wilmarth, P. A., Bergstrom, P., Zhao, Y., Minnier, J., Heinecke, J. W., Yang, X., and Pamir, N. (2018) GM-CSF driven myeloid cells in adipose tissue link weight gain and insulin resistance via formation of 2-aminoadipate, Sci. Rep., 8, 11485, doi: https://doi.org/10.1038/s41598-018-29250-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Timmons, J. A., Atherton, P. J., Larsson, O., Sood, S., Blokhin, I. O., Brogan, R. J., Volmar, C. H., Josse, A. R., Slentz, C., Wahlestedt, C., Phillips, S. M., Phillips, B. E., Gallagher, I. J., and Kraus, W. E. (2018) A coding and non-coding transcriptomic perspective on the genomics of human metabolic disease, Nucleic Acids Res., 46, 7772-7792, doi: https://doi.org/10.1093/nar/gky570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lim, J., Liu, Z., Apontes, P., Feng, D., Pessin, J. E., Sauve, A. A., Angeletti, R. H., and Chi, Y. (2014) Dual mode action of mangiferin in mouse liver under high fat diet, PLoS One, 9, e90137, doi: https://doi.org/10.1371/journal.pone.0090137.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Xu, W. Y., Zhu, H., Shen, Y., Wan, Y. H., Tu, X. D., Wu, W. T., Tang, L., Zhang, H. X., Lu, S. Y., Jin, X. L., Fei, J., and Wang, Z. G. (2018) DHTKD1 deficiency causes Charcot–Marie–Tooth disease in mice, Mol. Cell. Biol., 38, doi: https://doi.org/10.1128/MCB.00085-18.

    Google Scholar 

  15. O’Neill, E., Chiara Goisis, R., Haverty, R., and Harkin, A. (2019) L-alpha-aminoadipic acid restricts dopaminergic neurodegeneration and motor deficits in an inflammatory model of Parkinson’s disease in male rats, J. Neurosci. Res., 97, 804-816, doi: https://doi.org/10.1002/jnr.24420.

    Article  CAS  PubMed  Google Scholar 

  16. Graf, A., Kabysheva, M. S., Klimuk, E. I., Trofimova, L., Dunaeva, T., Zundorf, G., Kahlert, S., Reiser, G., Storozhevykh, T. P., Pinelis, V. G., Sokolova, N., and Bunik, V. (2009) Role of 2-oxoglutarate dehydrogenase in brain pathologies involving glutamate neurotoxicity, J. Mol. Catal. B Enzym., 61, 80-87, doi: https://doi.org/10.1016/j.molcatb.2009.02.016.

    Article  CAS  Google Scholar 

  17. Musayev, F. N., Di Salvo, M. L., Ko, T. P., Schirch, V., and Safo, M. K. (2003) Structure and properties of recombinant human pyridoxine 5′-phosphate oxidase, Protein Sci., 12, 1455-1463, doi: https://doi.org/10.1110/ps.0356203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wu, S., and Letchworth, G. J. (2004) High efficiency transformation by electroporation of Pichia pastoris pretreated with lithium acetate and dithiothreitol, Biotechniques, 36, 152-154, doi: https://doi.org/10.2144/04361DD02.

    Article  CAS  PubMed  Google Scholar 

  19. Aleshin, V. A., Mkrtchyan, G. V., Kaehne, T., Graf, A. V., Maslova, M. V., and Bunik, V. I. (2020) Diurnal regulation of the function of the rat brain glutamate dehydrogenase by acetylation and its dependence on thiamine administration, J. Neurochem., 153, 80-102, doi: https://doi.org/10.1111/jnc.14951.

    Article  CAS  PubMed  Google Scholar 

  20. Altschul, S. F., Gish, W., Miller, W., Myers, E. W., and Lipman, D. J. (1990) Basic local alignment search tool, J. Mol. Biol., 215, 403-410, doi: https://doi.org/10.1016/S0022-2836(05)80360-2.

    Article  CAS  PubMed  Google Scholar 

  21. Kumar, S., Stecher, G., Li, M., Knyaz, C., and Tamura, K. (2018) MEGA X: molecular evolutionary genetics analysis across computing platforms, Mol. Biol. Evol., 35, 1547-1549, doi: https://doi.org/10.1093/molbev/msy096.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Roy, A., Kucukural, A., and Zhang, Y. (2010) I-TASSER: a unified platform for automated protein structure and function prediction, Nat. Protoc., 5, 725-738, doi: https://doi.org/10.1038/nprot.2010.5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Payne, S. H., and Loomis, W. F. (2006) Retention and loss of amino acid biosynthetic pathways based on analysis of whole-genome sequences, Eukaryot. Cell, 5, 272-276, doi: https://doi.org/10.1128/EC.5.2.272-276.2006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bunik, V. (2017) Vitamin-Dependent Multienzyme Complexes of 2-Oxo Acid Dehydrogenases: Structure, Function, Regulation and Medical Implications, Hauppaughe, NY, Nova Science Publishers Inc.

  25. Bunik, V., Shoubnikova, A., Bisswanger, H., and Follmann, H. (1997) Characterization of thioredoxins by sodium dodecyl sulfate-slab gel electrophoresis and high performance capillary electrophoresis, Electrophoresis, 18, 762-766, doi: https://doi.org/10.1002/elps.1150180517.

    Article  CAS  PubMed  Google Scholar 

  26. Bonacci, T., Audebert, S., Camoin, L., Baudelet, E., Bidaut, G., Garcia, M., Witzel, II, Perkins, N. D., Borg, J. P., Iovanna, J. L., and Soubeyran, P. (2014) Identification of new mechanisms of cellular response to chemotherapy by tracking changes in post-translational modifications by ubiquitin and ubiquitin-like proteins, J. Proteome Res., 13, 2478-2494, doi: https://doi.org/10.1021/pr401258d.

    Article  CAS  PubMed  Google Scholar 

  27. Akimov, V., Barrio-Hernandez, I., Hansen, S. V. F., Hallenborg, P., Pedersen, A. K., Bekker-Jensen, D. B., Puglia, M., Christensen, S. D. K., Vanselow, J. T., Nielsen, M. M., Kratchmarova, I., Kelstrup, C. D., Olsen, J. V., and Blagoev, B. (2018) UbiSite approach for comprehensive mapping of lysine and N-terminal ubiquitination sites, Nat. Struct. Mol. Biol., 25, 631-640, doi: https://doi.org/10.1038/s41594-018-0084-y.

    Article  CAS  PubMed  Google Scholar 

  28. Hornbeck, P. V., Zhang, B., Murray, B., Kornhauser, J. M., Latham, V., and Skrzypek, E. (2015) PhosphoSitePlus, 2014: mutations, PTMs and recalibrations, Nucleic Acids Res., 43, D512-520, doi: https://doi.org/10.1093/nar/gku1267.

    Article  CAS  PubMed  Google Scholar 

  29. Udeshi, N. D., Mertins, P., Svinkina, T., and Carr, S. A. (2013) Large-scale identification of ubiquitination sites by mass spectrometry, Nat. Protoc., 8, 1950-1960, doi: https://doi.org/10.1038/nprot.2013.120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. McCartney, R. G., Rice, J. E., Sanderson, S. J., Bunik, V., Lindsay, H., and Lindsay, J. G. (1998) Subunit interactions in the mammalian alpha-ketoglutarate dehydrogenase complex. Evidence for direct association of the alpha-ketoglutarate dehydrogenase and dihydrolipoamide dehydrogenase components, J. Biol. Chem., 273, 24158-24164.

    Article  CAS  PubMed  Google Scholar 

  31. Arndt, C., Koristka, S., Feldmann, A., Bartsch, H., and Bachmann, M. (2012) Coomassie-Brilliant Blue staining of polyacrylamide gels, Methods Mol. Biol., 869, 465-469, doi: https://doi.org/10.1007/978-1-61779-821-4_40.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are thankful to Dr. A. V. Graf and Dr. M. V. Maslova (Lomonosov Moscow State University) for providing rat tissue samples.

Funding

This work is supported by the Russian Foundation for Basic Research (project no. 18-54-7812) and the Council of National Research, Italy (grant SAC.AD002.020.017).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to A. I. Boyko or V. I. Bunik.

Ethics declarations

The authors declare no conflict of interest in financial or any other sphere. All applicable international, national, and/or institutional guidelines for the care and use of animals were followed.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boyko, A.I., Artiukhov, A.V., Kaehne, T. et al. Isoforms of the DHTKD1-Encoded 2-Oxoadipate Dehydrogenase, Identified in Animal Tissues, Are not Observed upon the Human DHTKD1 Expression in Bacterial or Yeast Systems. Biochemistry Moscow 85, 920–929 (2020). https://doi.org/10.1134/S0006297920080076

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1134/S0006297920080076

Keywords

Navigation