Novel variants in TUBA1A cause congenital fibrosis of the extraocular muscles with or without malformations of cortical brain development

Julie A Jurgens, Brenda J Barry, Gabrielle Lemire, Wai-Man Chan, Mary C Whitman, Sherin Shaaban, Caroline D Robson, Sarah MacKinnon, Eleina M England, Hugh J McMillan, Christopher Kelly, Brandon M Pratt, Care4Rare Canada Consortium, Anne O'Donnell-Luria, Daniel G MacArthur, Kym M Boycott, David G Hunter, Elizabeth C Engle, Julie A Jurgens, Brenda J Barry, Gabrielle Lemire, Wai-Man Chan, Mary C Whitman, Sherin Shaaban, Caroline D Robson, Sarah MacKinnon, Eleina M England, Hugh J McMillan, Christopher Kelly, Brandon M Pratt, Care4Rare Canada Consortium, Anne O'Donnell-Luria, Daniel G MacArthur, Kym M Boycott, David G Hunter, Elizabeth C Engle

Abstract

Variants in multiple tubulin genes have been implicated in neurodevelopmental disorders, including malformations of cortical development (MCD) and congenital fibrosis of the extraocular muscles (CFEOM). Distinct missense variants in the beta-tubulin encoding genes TUBB3 and TUBB2B cause MCD, CFEOM, or both, suggesting substitution-specific mechanisms. Variants in the alpha tubulin-encoding gene TUBA1A have been associated with MCD, but not with CFEOM. Using exome sequencing (ES) and genome sequencing (GS), we identified 3 unrelated probands with CFEOM who harbored novel heterozygous TUBA1A missense variants c.1216C>G, p.(His406Asp); c.467G>A, p.(Arg156His); and c.1193T>G, p.(Met398Arg). MRI revealed small oculomotor-innervated muscles and asymmetrical caudate heads and lateral ventricles with or without corpus callosal thinning. Two of the three probands had MCD. Mutated amino acid residues localize either to the longitudinal interface at which α and β tubulins heterodimerize (Met398, His406) or to the lateral interface at which tubulin protofilaments interact (Arg156), and His406 interacts with the motor domain of kinesin-1. This series of individuals supports TUBA1A variants as a cause of CFEOM and expands our knowledge of tubulinopathies.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1. TUBA1A variants are associated with…
Fig. 1. TUBA1A variants are associated with CFEOM.
AI Five positions of gaze in Individual 1 prior to her first strabismus surgery at 2.5 years of age, demonstrating ophthalmic features consistent with CFEOM. The individual’s eye movements were directed as follows: (A) up; (B) right; (C) forward; (D) left; (E) down. Notable ophthalmic features include: (A) bilateral limitations of upgaze and convergence on attempted upgaze; (C) Bilateral ptosis and primary eye position in downgaze; (B, D): full horizontal eye movements; (E): mild bilateral limitations of downgaze and preferred chin-up head positioning. F Axial T2 weighted MR image (2 mm/0 mm gap thickness) at the level of the interpeduncular and suprasellar cisterns faintly demonstrates the oculomotor nerves (arrows). Although the slice thickness was not optimized for cranial nerve imaging, these nerves appear hypoplastic. G Coronal T1 MPRAGE image (0.9 mm/0 mm gap thickness) through the orbits demonstrates small superior and medial rectus muscles bilaterally (arrows).
Fig. 2. Brain MRI demonstrates features of…
Fig. 2. Brain MRI demonstrates features of classic tubulinopathies with or without MCD.
AF MR images from Individual 1 at 20 months of age. A Midline sagittal T1 FLAIR MR image reveals deficiency of the rostrum of the corpus callosum. B Axial T2 and (C) coronal T1 MPRAGE MR images demonstrate an asymmetrically smaller, foreshortened body of the left caudate nucleus (white arrows) with mild prominence and dysmorphism of the ipsilateral lateral ventricle. D Axial T1 FLAIR MR image (5 mm/2.25 mm gap thickness) demonstrates thickening and irregularity of the posterior perisylvian cortex (white arrows), most pronounced on the right. E, F Coronal T1 MPRAGE MR images (0.9 mm/0 mm gap thickness) confirm bilateral perisylvian polymicrogyria (white arrows). GI MR images from Individual 2 at 13 months of age. G 1.5T Siemens Symphony midline sagittal T1 MR image (4 mm/0.8 mm gap thickness) reveals mild thinning of the body and splenium (arrow) of the corpus callosum. H Axial (4 mm/0.8 mm gap) and (I) coronal (2 mm/ 0 mm gap) turbo spin echo T2 weighted MR images show asymmetric lateral ventricles and caudate nuclei, with the body of the right caudate nucleus appearing slightly smaller than the left (arrow). J Axial spin echo 1.5 T Siemens Symphony T1 MR image (4 mm/ 0.8 mm gap) demonstrating the perisylvian cortex. The cortical architecture is not well assessed for MCD due to a combination of technical parameters and age-appropriate incompletely myelinated subcortical white matter. K Anterior coronal (2 mm/ 0 mm gap) turbo spin echo T2 weighted MR image shows interdigitating frontal lobes (arrows) attributed to deficiency of the falx cerebri. LP MR images from Individual 3 at 7 years of age. L Midline reconstructed sagittal T1 MPRAGE MR image (0.8 mm/ 0 mm interslice gap) reveals accentuation of the usual thinning of the posterior body of the corpus callosum (arrow). M Axial T2 (4 mm/1.2 mm gap) and N coronal T1 MPRAGE (1.2 mm/0 mm gap) MR images show asymmetric lateral ventricles and caudate nuclei, with the body of the left caudate nucleus appearing smaller than the right (arrow). O Reformatted axial (0.8 mm thickness) and (P) direct coronal (1.2 mm/ 0 mm gap) T1 3D MPRAGE MR images demonstrate irregular, thickened perisylvian cortex, more pronounced on the right, consistent with polymicrogyria (arrows).
Fig. 3. Structural modeling of tubulinopathy variants.
Fig. 3. Structural modeling of tubulinopathy variants.
Structures were obtained from PDB ID: 3JAK (AC, E) and PDB ID: 4HNA (D). A α and β tubulin monomers within a portion of a microtubule. Representative lateral and longitudinal interfaces are indicated. B α tubulin monomer with TUBA1A tubulinopathy-associated residues colored. C α and β tubulin monomers at the heterodimerization interface. Colored beta tubulin residues are associated with substitutions in TUBB3 and/or TUBB2B. For simplicity, residues associated with isolated MCD or MCD with involvement of cranial nerves other than CN3 are omitted in this view. D α and β tubulin complexed with kinesin-1. Colored beta tubulin residues are associated with substitutions in TUBB3 and/or TUBB2B. For simplicity, residues associated with isolated MCD or MCD with involvement of cranial nerves other than CN3 are omitted in this view. Polar interactions between kinesin and alpha or beta tubulin residues are shown with dashed lines. Distance between kinesin-Lys256 and α-His406 is shown. E Lateral interface between α tubulin residues in adjacent microtubule protofilaments. Key: α tubulin (gold); β tubulin (silver); kinesin-1 (pink); residues associated with CFEOM but not MCD (red); residues associated with CFEOM+MCD (purple); residues associated with MCD in the absence of CFEOM (cyan); residues associated with CFEOM ± MCD (orange); residues associated with CFEOM in the absence of MCD, or associated with MCD in the absence of CFEOM depending on substitution (brown); residues associated with MCD ± CFEOM (yellow); previously reported residues associated with putative CFEOM in the absence of MCD (black; [50]); reported residues associated with MCD and anomalies of cranial nerves other than CN III (green; [25]). *residues reported for the first time in this work (Arg156, Met398, and His406). ●-Previously reported residues associated with putative CN3 phenotypes [50].

References

    1. Lasser M, Tiber J, Lowery LA. The role of the microtubule cytoskeleton in neurodevelopmental disorders. Front Cell Neurosci. 2018;12:165.
    1. Lopata MA, Cleveland DW. In vivo microtubules are copolymers of available beta-tubulin isotypes: localization of each of six vertebrate beta-tubulin isotypes using polyclonal antibodies elicited by synthetic peptide antigens. J Cell Biol. 1987;105:1707–20.
    1. Tilney LG, Bryan J, Bush DJ, Fujiwara K, Mooseker MS, Murphy DB, et al. Microtubules: evidence for 13 protofilaments. J Cell Biol. 1973;59:267–75.
    1. Mitchison T, Kirschner M. Dynamic instability of microtubule growth. Nature. 1984;312:237–42.
    1. Fulton C, Simpson P. Selective synthesis and utilization of flagellar tubulin. The multi-tubulin hypothesis. Cell Motil. 1976;3:987–1005.
    1. Gonçalves FG, de AL Freddi T, Taranath A, Lakshmanan R, Goetti R, Feltrin FS, et al. Tubulinopathies. Topics Magn Reson Imaging. 2018;27:395–408.
    1. Köhler S, Carmody L, Vasilevsky N, Jacobsen J, Danis D, Gourdine J, et al. Expansion of the Human Phenotype Ontology (HPO) knowledge base and resources. Nucleic Acids Res. 2019;47:D1018–27.
    1. Engle EC, Goumnerov BC, McKeown CA, Schatz M, Johns DR, Porter JD, et al. Oculomotor nerve and muscle abnormalities in congenital fibrosis of the extraocular muscles. Ann Neurol. 1997;41:314–25.
    1. Demer JL, Clark RA, Engle EC. Magnetic Resonance Imaging Evidence For Widespread Orbital Dysinnervation in Congenital Fibrosis of Extraocular Muscles Due to Mutations in KIF21A. Invest Ophth Vis Sci. 2005;46:530–9.
    1. Lim K, Engle E, Demer J. Abnormalities of the oculomotor nerve in congenital fibrosis of the extraocular muscles and congenital oculomotor palsy. Invest Ophth Vis Sci. 2007;48:1601–6.
    1. Online Mendelian Inheritance in Man, OMIM. Mc-Kusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore, MD), accessed December 10, 2020.
    1. Yamada K, Andrews C, Chan W-M, McKeown CA, Magli A, Berardinis Tde, et al. Heterozygous mutations of the kinesin KIF21A in congenital fibrosis of the extraocular muscles type 1 (CFEOM1) Nat Genet. 2003;35:318–21.
    1. Tischfield M, Baris H, Wu C, Rudolph G, Maldergem L, He W, et al. Human TUBB3 mutations perturb microtubule dynamics, kinesin interactions, and axon guidance. Cell. 2010;140:74–87.
    1. Chew S, Balasubramanian R, Chan W, Kang P, Andrews C, Webb B, et al. A novel syndrome caused by the E410K amino acid substitution in the neuronal β-tubulin isotype 3. Brain. 2013;136:522–35.
    1. Cederquist GY, Luchniak A, Tischfield MA, Peeva M, Song Y, Menezes MP, et al. An inherited TUBB2B mutation alters a kinesin-binding site and causes polymicrogyria, CFEOM and axon dysinnervation. Hum Mol Genet. 2012;21:5484–99.
    1. Whitman MC, Andrews C, Chan W-M, Tischfield MA, Stasheff SF, Brancati F, et al. Two unique TUBB3 mutations cause both CFEOM3 and malformations of cortical development. Am J Med Genet A. 2016;170A:297–305.
    1. Poirier K, Keays D, Francis F, Saillour Y, Bahi N, Manouvrier S, et al. Large spectrum of lissencephaly and pachygyria phenotypes resulting from de novo missense mutations in tubulin alpha1A (TUBA1A) Hum Mutat. 2007;28:1055–64.
    1. Jaglin X, Poirier K, Saillour Y, Buhler E, Tian G, Bahi-Buisson N, et al. Mutations in the beta-tubulin gene TUBB2B result in asymmetrical polymicrogyria. Nat Genet. 2009;41:746–52.
    1. Poirier K, Saillour Y, Bahi-Buisson N, Jaglin X, Fallet-Bianco C, Nabbout R, et al. Mutations in the neuronal ß-tubulin subunit TUBB3 result in malformation of cortical development and neuronal migration defects. Hum Mol Genet. 2010;19:4462–73.
    1. Latremoliere A, Cheng L, DeLisle M, Wu C, Chew S, Hutchinson EB, et al. Neuronal-specific TUBB3 is not required for normal neuronal function but is essential for timely axon regeneration. Cell Rep. 2018;24:1865–79.e9.
    1. Bittermann E, Abdelhamed Z, Liegel R, Menke C, Timms A, Beier D, et al. Differential requirements of tubulin genes in mammalian forebrain development. Plos Genet. 2019;15:e1008243–e1008243.
    1. Keays D, Tian G, Poirier K, Huang G-J, Siebold C, Cleak J, et al. Mutations in alpha-tubulin cause abnormal neuronal migration in mice and lissencephaly in humans. Cell. 2007;128:45–57.
    1. Furuse T, Yamada I, Kushida T, Masuya H, Miura I, Kaneda H, et al. Behavioral and neuromorphological characterization of a novel Tuba1 mutant mouse. Behavioural Brain Res. 2012;227:167–74.
    1. Hanson MG, Aiken J, Sietsema DV, Sept D, Bates EA, Niswander L, et al. Novel α-tubulin mutation disrupts neural development and tubulin proteostasis. Dev Biol. 2016;409:406–19.
    1. Hebebrand M, Hüffmeier U, Trollmann R, Hehr U, Uebe S, Ekici A, et al. The mutational and phenotypic spectrum of TUBA1A-associated tubulinopathy. Orphanet J Rare Dis. 2019;14:38–38.
    1. Nellhaus G. Head circumference from birth to eighteen years. Pediatrics. 1968;41:106–14.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–60.
    1. 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.
    1. McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GRS, Thormann A, et al. The ensembl variant effect predictor. Genome Biol. 2016;17:122.
    1. Beaulieu CL, Majewski J, Schwartzentruber J, Samuels ME, Fernandez BA, Bernier FP, et al. FORGE Canada Consortium: outcomes of a 2-year national rare-disease gene-discovery project. Am J Hum Genetics. 2014;94:809–17.
    1. Paila U, Chapman BA, Kirchner R, Quinlan AR. GEMINI: integrative exploration of genetic variation and genome annotations. Plos Comput Biol. 2013;9:e1003153.
    1. Srour M, Schwartzentruber J, Hamdan F, Ospina L, Patry L, Labuda D, et al. Mutations in C5ORF42 cause Joubert syndrome in the French Canadian population. Am J Hum Genetics. 2012;90:693–700.
    1. Lek M, Karczewski K, Minikel E, Samocha K, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–91.
    1. Karczewski K, Francioli L, Tiao G, Cummings B, Alföldi J, Wang Q, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434–43.
    1. Auton A, Abecasis GR, Altshuler DM, Durbin RM, Abecasis GR, Bentley DR, et al. A global reference for human genetic variation. Nature. 2015;526:68–74.
    1. Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S, et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018;46:D1062–7.
    1. Zhang R, Alushin GM, Brown A, Nogales E. Mechanistic origin of microtubule dynamic instability and its modulation by EB proteins. Cell. 2015;162:849–59.
    1. Gigant B, Wang W, Dreier B, Jiang Q, Pecqueur L, Plückthun A, et al. Structure of a kinesin–tubulin complex and implications for kinesin motility. Nat Struct Mol Biol. 2013;20:1001–7.
    1. Sievers F, Wilm A, Dineen D, Gibson TJ, Karplus K, Li W, et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol. 2011;7:539–539.
    1. Waterhouse AM, Procter JB, Martin D, Clamp M, Barton GJ. Jalview Version 2—a multiple sequence alignment editor and analysis workbench. Bioinformatics. 2009;25:1189–91.
    1. Shoshany T, Robson C, Hunter D. Anomalous superior oblique muscles and tendons in congenital fibrosis of the extraocular muscles. J Am Assoc Pediatr Ophthalmol Strabismus. 2019;23:325.e1–325.e6.
    1. Pollard KS, Hubisz MJ, Rosenbloom KR, Siepel A. Detection of nonneutral substitution rates on mammalian phylogenies. Genome Res. 2010;20:110–21.
    1. Rentzsch P, Witten D, Cooper G, Shendure J, Kircher M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 2019;47:D886–94.
    1. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–23.
    1. Samocha K, Robinson E, Sanders S, Stevens C, Sabo A, McGrath L, et al. A framework for the interpretation of de novo mutation in human disease. Nat Genet. 2014;46:944–50.
    1. Nogales E, Whittaker M, Milligan RA, Downing KH. High-resolution model of the microtubule. Cell. 1999;96:79–88.
    1. Löwe J, Li H, Downing KH, Nogales E. Refined structure of αβ-tubulin at 3.5 Å resolution. J Mol Biol. 2001;313:1045–57.
    1. Romaniello R, Zucca C, Arrigoni F, Bonanni P, Panzeri E, Bassi MT, et al. Epilepsy in tubulinopathy: personal series and literature review. Cells. 2019;8:669.
    1. Aiken J, Buscaglia G, Bates E, Moore J. The α-tubulin gene TUBA1A in brain development: a key ingredient in the neuronal isotype blend. J Dev Biol. 2017;5:8.
    1. Romaniello R, Arrigoni F, Panzeri E, Poretti A, Micalizzi A, Citterio A, et al. Tubulin-related cerebellar dysplasia: definition of a distinct pattern of cerebellar malformation. Eur Radio. 2017;27:5080–92.

Source: PubMed

3
Abonnere