Biallelic variants in HPDL, encoding 4-hydroxyphenylpyruvate dioxygenase-like protein, lead to an infantile neurodegenerative condition

Shereen G Ghosh, Sangmoon Lee, Rudy Fabunan, Guoliang Chai, Maha S Zaki, Ghada Abdel-Salam, Tipu Sultan, Tawfeg Ben-Omran, Javeria Raza Alvi, Jennifer McEvoy-Venneri, Valentina Stanley, Aakash Patel, Danica Ross, Jeffrey Ding, Mohit Jain, Daqiang Pan, Philipp Lübbert, Bernd Kammerer, Nils Wiedemann, Nanda M Verhoeven-Duif, Judith J Jans, David Murphy, Mehran Beiraghi Toosi, Farah Ashrafzadeh, Shima Imannezhad, Ehsan Ghayoor Karimiani, Khalid Ibrahim, Elizabeth R Waters, Reza Maroofian, Joseph G Gleeson, Shereen G Ghosh, Sangmoon Lee, Rudy Fabunan, Guoliang Chai, Maha S Zaki, Ghada Abdel-Salam, Tipu Sultan, Tawfeg Ben-Omran, Javeria Raza Alvi, Jennifer McEvoy-Venneri, Valentina Stanley, Aakash Patel, Danica Ross, Jeffrey Ding, Mohit Jain, Daqiang Pan, Philipp Lübbert, Bernd Kammerer, Nils Wiedemann, Nanda M Verhoeven-Duif, Judith J Jans, David Murphy, Mehran Beiraghi Toosi, Farah Ashrafzadeh, Shima Imannezhad, Ehsan Ghayoor Karimiani, Khalid Ibrahim, Elizabeth R Waters, Reza Maroofian, Joseph G Gleeson

Abstract

Purpose: Dioxygenases are oxidoreductase enzymes with roles in metabolic pathways necessary for aerobic life. 4-hydroxyphenylpyruvate dioxygenase-like protein (HPDL), encoded by HPDL, is an orphan paralogue of 4-hydroxyphenylpyruvate dioxygenase (HPD), an iron-dependent dioxygenase involved in tyrosine catabolism. The function and association of HPDL with human diseases remain unknown.

Methods: We applied exome sequencing in a cohort of over 10,000 individuals with neurodevelopmental diseases. Effects of HPDL loss were investigated in vitro and in vivo, and through mass spectrometry analysis. Evolutionary analysis was performed to investigate the potential functional separation of HPDL from HPD.

Results: We identified biallelic variants in HPDL in eight families displaying recessive inheritance. Knockout mice closely phenocopied humans and showed evidence of apoptosis in multiple cellular lineages within the cerebral cortex. HPDL is a single-exonic gene that likely arose from a retrotransposition event at the base of the tetrapod lineage, and unlike HPD, HPDL is mitochondria-localized. Metabolic profiling of HPDL mutant cells and mice showed no evidence of altered tyrosine metabolites, but rather notable accumulations in other metabolic pathways.

Conclusion: The mitochondrial localization, along with its disrupted metabolic profile, suggests HPDL loss in humans links to a unique neurometabolic mitochondrial infantile neurodegenerative condition.

Keywords: 4-hydroxyphenylpyruvate dioxygenase-like protein; HPD; HPDL; neurodegenerative disease; oxidoreductase.

References

    1. Brassier A, Ottolenghi C, Boddaert N, et al. Prenatal symptoms and diagnosis of inherited metabolic diseases. Arch Pediatr. 2012;19:959–969.
    1. Tarailo-Graovac M, Shyr C, Ross CJ, et al. Exome sequencing and the management of neurometabolic disorders. N Engl J Med. 2016;374:2246–2255.
    1. BoAli AY, Alfadhel M, Tabarki B. Neurometabolic disorders and congenital malformations of the central nervous system. Neurosciences (Riyadh). 2018;23:97–103.
    1. Willemsen MA, Harting I, Wevers RA. Neurometabolic disorders: five new things. Neurol Clin Pract. 2016;6:348–357.
    1. Abu-Omar MM, Loaiza A, Hontzeas N. Reaction mechanisms of mononuclear non-heme iron oxygenases. Chem Rev. 2005;105:2227–2252.
    1. Burlina A, Zacchello F, Dionisi-Vici C, et al. New clinical phenotype of branched-chain acyl-CoA oxidation defect. Lancet. 1991;338:1522–1523.
    1. Boissel S, Reish O, Proulx K, et al. Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. Am J Hum Genet. 2009;85:106–111.
    1. Fernández-Cañón JM, Granadino B, Beltrán-Valero de Bernabé D, et al. The molecular basis of alkaptonuria. Nat Genet. 1996;14:19–24.
    1. Ferreira P, Shin I, Sosova I, et al. Hypertryptophanemia due to tryptophan 2,3-dioxygenase deficiency. Mol Genet Metab. 2017;120:317–324.
    1. Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science. 2001;294:1337–1340.
    1. Moran GR. 4-Hydroxyphenylpyruvate dioxygenase. Arch Biochem Biophys. 2005;433:117–128.
    1. Rüetschi U, Cerone R, Pérez-Cerda C, et al. Mutations in the 4-hydroxyphenylpyruvate dioxygenase gene (HPD) in patients with tyrosinemia type III. Hum Genet. 2000;106:654–662.
    1. Kubo S, Kiwaki K, Awata H, et al. In vivo correction with recombinant adenovirus of 4-hydroxyphenylpyruvic acid dioxygenase deficiencies in strain III mice. Hum Gene Ther. 1997;8:65–71.
    1. Szymanska E, Sredzinska M, Ciara E, et al. Tyrosinemia type III in an asymptomatic girl. Mol Genet Metab Rep. 2015;5:48–50.
    1. Tomoeda K, Awata H, Matsuura T, et al. Mutations in the 4-hydroxyphenylpyruvic acid dioxygenase gene are responsible for tyrosinemia type III and hawkinsinuria. Mol Genet Metab. 2000;71:506–510.
    1. Item CB, Mihalek I, Lichtarge O, et al. Manifestation of hawkinsinuria in a patient compound heterozygous for hawkinsinuria and tyrosinemia III. Mol Genet Metab. 2007;91:379–383.
    1. Barroso F, Correia J, Bandeira A, et al. Tyrosinemia type III: a case report of siblings and literature review. Rev Paul Pediatr. 2020;38:e2018158.
    1. Haijes HA, Willemsen M, Van der Ham M, et al. Direct infusion based metabolomics identifies metabolic disease in patients’ dried blood spots and plasma. Metabolites. 2019;9:12.
    1. Pei J, Grishin NV. PROMALS3D: multiple protein sequence alignment enhanced with evolutionary and three-dimensional structural information. Methods Mol Biol. 2014;1079:263–271.
    1. Stamatakis A. RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics. 2014;30:1312–1313.
    1. Letunic I, Bork P. Interactive tree of life (iTOL) v3: an online tool for the display and annotation of phylogenetic and other trees. Nucleic Acids Res. 2016;44:W242–245.
    1. Yang Z. PAML 4: phylogenetic analysis by maximum likelihood. Mol Biol Evol. 2007;24:1586–1591.
    1. Abascal F, Zardoya R, Telford MJ. TranslatorX: multiple alignment of nucleotide sequences guided by amino acid translations. Nucleic Acids Res. 2010;38:W7–13.
    1. Charif D, Thioulouse J, Lobry JR, Perriere G. Online synonymous codon usage analyses with the ade4 and seqinR packages. Bioinformatics. 2005;21:545–547.
    1. Karczewski KJ, Francioli LC, Tiao G, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434–443.
    1. GTEx Consortium. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–660.
    1. Zhang Y, Sloan SA, Clarke LE, et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron. 2016;89:37–53.
    1. Saunders A, Macosko EZ, Wysoker A, et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell. 2018;174:1015–.e1016.
    1. Vanlandewijck M, He L, Mäe MA, et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature. 2018;554:475–480.
    1. Thul PJ, Akesson L, Wiking M, et al. A subcellular map of the human proteome. Science. 2017;356:eaal3321.
    1. Fukasawa Y, Tsuji J, Fu SC, Tomii K, Horton P, Imai K. MitoFates: improved prediction of mitochondrial targeting sequences and their cleavage sites. Mol Cell Proteomics. 2015;14:1113–1126.
    1. Roise D, Theiler F, Horvath SJ, et al. Amphiphilicity is essential for mitochondrial presequence function. Embo j. 1988;7:649–653.
    1. Uhlén M, Fagerberg L, Hallström BM, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347:1260419.
    1. Natt E, Kida K, Odievre M, Di Rocco M, Scherer G. Point mutations in the tyrosine aminotransferase gene in tyrosinemia type II. Proc Natl Acad Sci U S A. 1992;89:9297–9301.
    1. Phaneuf D, Lambert M, Laframboise R, Mitchell G, Lettre F, Tanguay RM. Type 1 hereditary tyrosinemia. Evidence for molecular heterogeneity and identification of a causal mutation in a French Canadian patient. J Clin Invest. 1992;90:1185–1192.
    1. Tiranti V, Viscomi C, Hildebrandt T, et al. Loss of ETHE1, a mitochondrial dioxygenase, causes fatal sulfide toxicity in ethylmalonic encephalopathy. Nat Med. 2009;15:200–205.

Source: PubMed

3
Abonner