Combined genomics and proteomics unveils elusive variants and vast aetiologic heterogeneity in dystonia
Jazyk angličtina Země Velká Británie, Anglie Médium print
Typ dokumentu časopisecké články
Grantová podpora
#458949627
DFG Research Infrastructure NGS_CC
423957469
DFG Research Infrastructure NGS_CC
ZE 1213/2-1
DFG Research Infrastructure NGS_CC
SCHO 1644/4-1
DFG Research Infrastructure NGS_CC
WI 1820/14-1
DFG Research Infrastructure NGS_CC
#458949627
DFG Research Infrastructure NGS_CC
01GM2302
Federal Ministry of Education and Research
2022_EKSE.185
Federal Ministry of Education and Research
LX22NPO5107
National Institute for Neurological Research
European Union
NW24-04-0006
Next Generation EU
APVV-22-0279
Next Generation EU
09I03-03-V03-00007
Next Generation EU
1/0712/22
Next Generation EU
GR-2009-1594645
Next Generation EU
825575
Fondazione Regionale per la Ricerca Biomedica
01GM1906A
Fondazione Regionale per la Ricerca Biomedica
PubMed
39937650
PubMed Central
PMC12316014
DOI
10.1093/brain/awaf059
PII: 8010552
Knihovny.cz E-zdroje
- Klíčová slova
- dystonia, genomics, multi-omics, proteomics, transcriptomics, whole-genome sequencing,
- MeSH
- dospělí MeSH
- dystonické poruchy * genetika MeSH
- dystonie * genetika MeSH
- genetická heterogenita MeSH
- genomika * metody MeSH
- lidé středního věku MeSH
- lidé MeSH
- proteomika * metody MeSH
- sekvenování celého genomu MeSH
- sekvenování exomu MeSH
- Check Tag
- dospělí MeSH
- lidé středního věku MeSH
- lidé MeSH
- mužské pohlaví MeSH
- ženské pohlaví MeSH
- Publikační typ
- časopisecké články MeSH
Dystonia is a rare disease trait for which large-scale genomic investigations are still underrepresented. Genetic heterogeneity among patients with unexplained dystonia warrants interrogation of entire genome sequences, but this has not yet been systematically evaluated. To significantly enhance our understanding of the genetic contribution to dystonia, we (re)analysed 2874 whole-exome sequencing (WES), 564 whole-genome sequencing (WGS), as well as 80 fibroblast-derived proteomics datasets, representing the output of high-throughput analyses in 1990 patients and 973 unaffected relatives from 1877 families. Recruitment and precision-phenotyping procedures were driven by long-term collaborations of international experts with access to overlooked populations. By exploring WES data, we found that continuous scaling of sample sizes resulted in steady gains in the number of associated disease genes without plateauing. On average, every second diagnosis involved a gene not previously implicated in our cohort. Second-line WGS focused on a subcohort of undiagnosed individuals with high likelihood of having monogenic forms of dystonia, comprising large proportions of patients with early onset (81.3%), generalized symptom distribution (50.8%) and/or coexisting features (68.9%). We undertook extensive searches for variants in nuclear and mitochondrial genomes to uncover 38 (ultra)rare diagnostic-grade findings in 37 of 305 index patients (12.1%), many of which had remained undetected due to methodological inferiority of WES or pipeline limitations. WGS-identified elusive variations included alterations in exons poorly covered by WES, RNA-gene variants, mitochondrial-DNA mutations, small copy-number variants, complex rearranged genome structure and short tandem repeats. For improved variant interpretation in WGS-inconclusive cases, we employed systematic integration of quantitative proteomics. This aided in verifying diagnoses related to technically challenging variants and in upgrading a variant of uncertain significance (3 of 70 WGS-inconclusive index patients, 4.3%). Further, unsupervised proteomic outlier analysis supplemented with transcriptome sequencing revealed pathological gene underexpression induced by transcript disruptions in three more index patients with underlying (deep) intronic variants (3/70, 4.3%), highlighting the potential for targeted antisense-oligonucleotide therapy development. Finally, trio-WGS prioritized a de novo missense change in the candidate PRMT1, encoding a histone methyltransferase. Data-sharing strategies supported the discovery of three distinct PRMT1 de novo variants in four phenotypically similar patients, associated with loss-of-function effects in in vitro assays. This work underscores the importance of continually expanding sequencing cohorts to characterize the extensive spectrum of gene aberrations in dystonia. We show that a pool of unresolved cases is amenable to WGS and complementary multi-omic studies, directing advanced aetiopathological concepts and future diagnostic-practice workflows for dystonia.
Academic Center for Education Culture and Research Khorasan Razavi Mashhad 9177948564 Iran
Bavarian Genomes Network for Rare Disorders Munich 81675 Germany
Brain and Mind Research Program CEITEC Masaryk University Brno 62500 Czech Republic
Center for Advanced Studies Research and Development in Sardinia NGS Bioinformatics Pula 09010 Italy
Center for Rare Diseases University Hospital of Würzburg Wurzburg 97080 Germany
Chair of Social Pediatrics Technical University of Munich Munich 80333 Germany
Department of Dermatovenerology University Hospital of L Pasteur Kosice 4001 Slovakia
Department of Developmental Neurology Medical University of Gdansk Gdansk 80210 Poland
Department of Genetics Pitié Salpêtrière Hospital APHP Sorbonne University Paris 75013 France
Department of Neurology Asklepios Fachklinikum Stadtroda Stadtroda 07646 Germany
Department of Neurology Medical University of Vienna Vienna 1090 Austria
Department of Neurology P J Safarik University Kosice 4001 Slovakia
Department of Neurology University Hospital of L Pasteur Kosice 4001 Slovakia
Department of Neurology University Hospital of Würzburg Wurzburg 97080 Germany
Department of Neurology University of Leipzig Leipzig 04103 Germany
Department of Neurology Zvolen Hospital Zvolen 96001 Slovakia
Department of Paediatric Neurology Thomayer University Hospital Prague 12108 Czech Republic
Division of Child Neurology Department of Pediatrics Children's Hospital St Gallen 9000 Switzerland
DZKJ Deutsches Zentrum Für Kinder und Jugendgesundheit Munich 80333 Germany
DZPG Deutsches Zentrum Für Psychische Gesundheit Munich 80333 Germany
Faculty of Medicine Department of Dermatovenerology P J Safarik University Kosice 4001 Slovakia
Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Neurology Unit Milan 20122 Italy
Institute for Advanced Study Technical University of Munich Garching 85748 Germany
Institute of Medical Genetics University of Zurich Zurich 8952 Switzerland
Institute of Neurogenomics Helmholtz Zentrum München Munich 85764 Germany
Institute of Structural Biology Helmholtz Center Munich Munich 85764 Germany
kbo Kinderzentrum München Munich 81377 Germany
Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad 9177948564 Iran
Munich Cluster for Systems Neurology SyNergy Munich 81377 Germany
Munich Data Science Institute Technical University of Munich Garching 85748 Germany
Nantes Université CHU de Nantes CNRS INSERM l'institut du thorax Nantes 44000 France
Nantes Université CHU de Nantes Service de Génétique médicale Nantes 44000 France
Parkinsonism and Movement Disorders Treatment Center Zvolen Hospital Zvolen 96001 Slovakia
Zobrazit více v PubMed
Albanese A, Bhatia K, Bressman SB, et al. Phenomenology and classification of dystonia: A consensus update. Mov Disord. 2013;28:863–873. PubMed PMC
Grutz K, Klein C. Dystonia updates: Definition, nomenclature, clinical classification, and etiology. J Neural Transm (Vienna). 2021;128:395–404. PubMed PMC
van Egmond ME, Kuiper A, Eggink H, et al. Dystonia in children and adolescents: A systematic review and a new diagnostic algorithm. J Neurol Neurosurg Psychiatry. 2015;86:774–781. PubMed
van Egmond ME, Lagrand TJ, Lizaitiene G, Smit M, Tijssen MAJ. A novel diagnostic approach for patients with adult-onset dystonia. J Neurol Neurosurg Psychiatry. 2022;93:1039–1048. PubMed
Pozojevic J, Beetz C, Westenberger A. The importance of genetic testing for dystonia patients and translational research. J Neural Transm (Vienna). 2021;128:473–481. PubMed PMC
Sarva H, Rodriguez-Porcel F, Rivera F, et al. The role of genetics in the treatment of dystonia with deep brain stimulation: Systematic review and meta-analysis. J Neurol Sci. 2024;459:122970. PubMed
Zech M, Jech R, Boesch S, et al. Monogenic variants in dystonia: An exome-wide sequencing study. Lancet Neurol. 2020;19:908–918. PubMed PMC
Dzinovic I, Boesch S, Skorvanek M, et al. Genetic overlap between dystonia and other neurologic disorders: A study of 1,100 exomes. Parkinsonism Relat Disord. 2022;102:1–6. PubMed
Ahn JH, Kim AR, Park WY, Cho JW, Park J, Youn J. Whole exome sequencing and clinical investigation of young onset dystonia: What can we learn? Parkinsonism Relat Disord. 2023;115:105814. PubMed
Dhar D, Holla VV, Kumari R, et al. Clinical and genetic profile of patients with dystonia: An experience from a tertiary neurology center from India. Parkinsonism Relat Disord. 2024;120:105986. PubMed
Atasu B, Simon-Sanchez J, Hanagasi H, et al. Dissecting genetic architecture of rare dystonia: Genetic, molecular and clinical insights. J Med Genet. 2024;61:443–451. PubMed PMC
Thomsen M, Marth K, Loens S, et al. Large-scale screening: Phenotypic and mutational spectrum in isolated and combined dystonia genes. Mov Disord. 2024;39:526–538. PubMed
Hamosh A, Scott AF, Amberger JS, Bocchini CA, McKusick VA. Online Mendelian inheritance in man (OMIM), a knowledgebase of human genes and genetic disorders. Nucleic Acids Res. 2005;33(Database issue):D514–D517. PubMed PMC
Stark Z, Foulger RE, Williams E, et al. Scaling national and international improvement in virtual gene panel curation via a collaborative approach to discordance resolution. Am J Hum Genet. 2021;108:1551–1557. PubMed PMC
Thomsen M, Lange LM, Zech M, Lohmann K. Genetics and pathogenesis of dystonia. Annu Rev Pathol. 2024;19:99–131. PubMed
Wright CF, Campbell P, Eberhardt RY, et al. Genomic diagnosis of rare pediatric disease in the United Kingdom and Ireland. N Engl J Med. 2023;388:1559–1571. PubMed PMC
Zech M, Jech R, Boesch S, et al. Scoring algorithm-based genomic testing in dystonia: A prospective validation study. Mov Disord. 2021;36:1959–1964. PubMed
100,000 Genomes Project Pilot Investigators; Smedley D, Smith KR, et al. 100,000 genomes pilot on rare-disease diagnosis in health care—Preliminary report. N Engl J Med. 2021;385:1868–1880. PubMed PMC
Wojcik MH, Reuter CM, Marwaha S, et al. Beyond the exome: What’s next in diagnostic testing for Mendelian conditions. Am J Hum Genet. 2023;110:1229–1248. PubMed PMC
Zech M, Winkelmann J. Next-generation sequencing and bioinformatics in rare movement disorders. Nat Rev Neurol. 2024;20:114–126. PubMed
Kumar KR, Davis RL, Tchan MC, et al. Whole genome sequencing for the genetic diagnosis of heterogenous dystonia phenotypes. Parkinsonism Relat Disord. 2019;69:111–118. PubMed
Bertoli-Avella AM, Beetz C, Ameziane N, et al. Successful application of genome sequencing in a diagnostic setting: 1007 index cases from a clinically heterogeneous cohort. Eur J Hum Genet. 2021;29:141–153. PubMed PMC
Kopajtich R, Smirnov D, Stenton SL, et al. Integration of proteomics with genomics and transcriptomics increases the diagnostic rate of Mendelian disorders. bioRxiv. [Preprint] 10.1101/2021.03.09.21253187 DOI
Invernizzi F, Legati A, Nasca A, et al. Myopathic mitochondrial DNA depletion syndrome associated with biallelic variants in LIG3. Brain. 2021;144:e74. PubMed PMC
Lunke S, Bouffler SE, Patel CV, et al. Integrated multi-omics for rapid rare disease diagnosis on a national scale. Nat Med. 2023;29:1681–1691. PubMed PMC
Di Fonzo A, Jinnah HA, Zech M. Dystonia genes and their biological pathways. Int Rev Neurobiol. 2023;169:61–103. PubMed
Braconi D, Bernardini G, Spiga O, Santucci A. Leveraging proteomics in orphan disease research: Pitfalls and potential. Expert Rev Proteomics. 2021;18:315–327. PubMed
Smirnov D, Konstantinovskiy N, Prokisch H. Integrative omics approaches to advance rare disease diagnostics. J Inherit Metab Dis. 2023;46:824–838. PubMed
Prokisch H, Zech M. Proteomic profiling in dystonia: The next frontier for pathophysiology research and biomarker exploration. Mov Disord. 2024;39:1478–1479. PubMed
Harrer P, Skorvanek M, Kittke V, et al. Dystonia linked to EIF4A2 haploinsufficiency: A disorder of protein translation dysfunction. Mov Disord. 2023;38:1914–1924. PubMed
Dolzhenko E, Deshpande V, Schlesinger F, et al. ExpansionHunter: A sequence-graph-based tool to analyze variation in short tandem repeat regions. Bioinformatics. 2019;35:4754–4756. PubMed PMC
Lim SY, Tan AH, Ahmad-Annuar A, et al. Uncovering the genetic basis of Parkinson’s disease globally: From discoveries to the clinic. Lancet Neurol. 2024;23:1267–1280. PubMed
Stranneheim H, Lagerstedt-Robinson K, Magnusson M, et al. Integration of whole genome sequencing into a healthcare setting: High diagnostic rates across multiple clinical entities in 3219 rare disease patients. Genome Med. 2021;13:40. PubMed PMC
Richards S, Aziz N, Bale S, 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;17:405–424. PubMed PMC
Riggs ER, Andersen EF, Cherry AM, et al. Technical standards for the interpretation and reporting of constitutional copy-number variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics (ACMG) and the Clinical Genome Resource (ClinGen). Genet Med. 2020;22:245–257. PubMed PMC
Steel D, Zech M, Zhao C, et al. Loss-of-function variants in HOPS complex genes VPS16 and VPS41 cause early onset dystonia associated with lysosomal abnormalities. Ann Neurol. 2020;88:867–877. PubMed
Guo MH, Plummer L, Chan YM, Hirschhorn JN, Lippincott MF. Burden testing of rare variants identified through exome sequencing via publicly available control data. Am J Hum Genet. 2018;103:522–534. PubMed PMC
Coutelier M, Holtgrewe M, Jager M, et al. Combining callers improves the detection of copy number variants from whole-genome sequencing. Eur J Hum Genet. 2022;30:178–186. PubMed PMC
Ibanez K, Polke J, Hagelstrom RT, et al. Whole genome sequencing for the diagnosis of neurological repeat expansion disorders in the UK: A retrospective diagnostic accuracy and prospective clinical validation study. Lancet Neurol. 2022;21:234–245. PubMed PMC
Karczewski KJ, Francioli LC, Tiao G, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434–443. PubMed PMC
Indelicato E, Eberl A, Boesch S, et al. Genome aggregation database version 4-allele frequency changes and impact on variant interpretation in dystonia. Mov Disord. 2025;40:357–362. PubMed PMC
Lappalainen I, Lopez J, Skipper L, et al. Dbvar and DGVa: Public archives for genomic structural variation. Nucleic Acids Res. 2013;41(Database issue):D936–D941. PubMed PMC
MacDonald JR, Ziman R, Yuen RK, Feuk L, Scherer SW. The database of genomic variants: A curated collection of structural variation in the human genome. Nucleic Acids Res. 2014;42(Database issue):D986–D992. PubMed PMC
Brandon MC, Lott MT, Nguyen KC, et al. MITOMAP: A human mitochondrial genome database–2004 update. Nucleic Acids Res. 2005;33(Database issue):D611–D613. PubMed PMC
Landrum MJ, Lee JM, Benson M, et al. ClinVar: Public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 2016;44(D1):D862–D868. PubMed PMC
Stenson PD, Mort M, Ball EV, et al. The human gene mutation database: Towards a comprehensive repository of inherited mutation data for medical research, genetic diagnosis and next-generation sequencing studies. Hum Genet. 2017;136:665–677. PubMed PMC
Chintalaphani SR, Pineda SS, Deveson IW, Kumar KR. An update on the neurological short tandem repeat expansion disorders and the emergence of long-read sequencing diagnostics. Acta Neuropathol Commun. 2021;9:98. PubMed PMC
Ellingford JM, Ahn JW, Bagnall RD, et al. Recommendations for clinical interpretation of variants found in non-coding regions of the genome. Genome Med. 2022;14:73. PubMed PMC
Bamshad MJ, Nickerson DA, Chong JX. Mendelian gene discovery: Fast and furious with no end in sight. Am J Hum Genet. 2019;105:448–455. PubMed PMC
Robinson JT, Thorvaldsdottir H, Winckler W, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29:24–26. PubMed PMC
Waldmann TA, McIntire KR. Serum-alpha-fetoprotein levels in patients with ataxia-telangiectasia. Lancet. 1972;2:1112–1115. PubMed
van Kuilenburg ABP, Tarailo-Graovac M, Richmond PA, et al. Glutaminase deficiency caused by short tandem repeat expansion in GLS. N Engl J Med. 2019;380:1433–1441. PubMed PMC
Remerand G, Boespflug-Tanguy O, Tonduti D, et al. Expanding the phenotypic spectrum of Allan-Herndon-Dudley syndrome in patients with SLC16A2 mutations. Dev Med Child Neurol. 2019;61:1439–1447. PubMed
Zech M, Kopajtich R, Steinbrucker K, et al. Variants in mitochondrial ATP synthase cause variable neurologic phenotypes. Ann Neurol. 2022;91:225–237. PubMed PMC
Yepez VA, Gusic M, Kopajtich R, et al. Clinical implementation of RNA sequencing for Mendelian disease diagnostics. Genome Med. 2022;14:38. PubMed PMC
Brechtmann F, Mertes C, Matuseviciute A, et al. OUTRIDER: A statistical method for detecting aberrantly expressed genes in RNA sequencing data. Am J Hum Genet. 2018;103:907–917. PubMed PMC
Brugger M, Lauri A, Zhen Y, et al. Bi-allelic variants in SNF8 cause a disease spectrum ranging from severe developmental and epileptic encephalopathy to syndromic optic atrophy. Am J Hum Genet. 2024;111:594–613. PubMed PMC
Pascual-Alonso A, Xiol C, Smirnov D, Kopajtich R, Prokisch H, Armstrong J. Identification of molecular signatures and pathways involved in Rett syndrome using a multi-omics approach. Hum Genomics. 2023;17:85. PubMed PMC
Tyanova S, Temu T, Cox J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat Protoc. 2016;11:2301–2319. PubMed
Kaplanis J, Samocha KE, Wiel L, et al. Evidence for 28 genetic disorders discovered by combining healthcare and research data. Nature. 2020;586:757–762. PubMed PMC
Dummler A, Lawrence AM, de Marco A. Simplified screening for the detection of soluble fusion constructs expressed in PubMed PMC
Zanuttigh E, Derderian K, Gura MA, et al. Identification of autophagy as a functional target suitable for the pharmacological treatment of mitochondrial membrane protein-associated neurodegeneration (MPAN) in vitro. Pharmaceutics. 2023;15:267. PubMed PMC
Coe BP, Stessman HAF, Sulovari A, et al. Neurodevelopmental disease genes implicated by de novo mutation and copy number variation morbidity. Nat Genet. 2019;51:106–116. PubMed PMC
Teunissen MWA, Lewerissa E, van Hugte EJH, et al. ANK2 loss-of-function variants are associated with epilepsy, and lead to impaired axon initial segment plasticity and hyperactive network activity in hiPSC-derived neuronal networks. Hum Mol Genet. 2023;32:2373–2385. PubMed PMC
Sobreira N, Schiettecatte F, Valle D, Hamosh A. GeneMatcher: A matching tool for connecting investigators with an interest in the same gene. Hum Mutat. 2015;36:928–930. PubMed PMC
Kochinke K, Zweier C, Nijhof B, et al. Systematic phenomics analysis deconvolutes genes mutated in intellectual disability into biologically coherent modules. Am J Hum Genet. 2016;98:149–164. PubMed PMC
Indelicato E, Boesch S, Zech M. Reply to: “early onset nonprogressive generalized dystonia is caused by Biallelic SHQ1 variants”. Mov Disord. 2023;38:1119–1120. PubMed
van der Spek J, den Hoed J, Snijders Blok L, et al. Inherited variants in CHD3 show variable expressivity in Snijders Blok-Campeau syndrome. Genet Med. 2022;24:1283–1296. PubMed
Harrer P, Leppmeier V, Berger A, et al. A de novo BCL11B variant case manifesting with dystonic movement disorder regarding the article “BCL11B-related disorder in two Canadian children: Expanding the clinical phenotype (Prasad et al. 2020)”. Eur J Med Genet. 2022;65:104635. PubMed
Garone G, Capuano A, Amodio D, et al. BCL11B-related dystonia: Further evidence of an emerging cause of childhood-onset generalized dystonia. Mov Disord Clin Pract. 2024;11:897–901. PubMed PMC
Yang Y, Muzny DM, Reid JG, et al. Clinical whole-exome sequencing for the diagnosis of Mendelian disorders. N Engl J Med. 2013;369:1502–1511. PubMed PMC
Guo F, Liu R, Pan Y, et al. Evidence from 2100 index cases supports genome sequencing as a first-tier genetic test. Genet Med. 2024;26:100995. PubMed
Sanghvi RV, Buhay CJ, Powell BC, et al. Characterizing reduced coverage regions through comparison of exome and genome sequencing data across 10 centers. Genet Med. 2018;20:855–866. PubMed PMC
Quinodoz M, Kaminska K, Cancellieri F, et al. Detection of elusive DNA copy-number variations in hereditary disease and cancer through the use of noncoding and off-target sequencing reads. Am J Hum Genet. 2024;111:701–713. PubMed PMC
Belkadi A, Bolze A, Itan Y, et al. Whole-genome sequencing is more powerful than whole-exome sequencing for detecting exome variants. Proc Natl Acad Sci USA. 2015;112:5473–5478. PubMed PMC
Turro E, Astle WJ, Megy K, et al. Whole-genome sequencing of patients with rare diseases in a national health system. Nature. 2020;583:96–102. PubMed PMC
Yuan B, Wang L, Liu P, et al. CNVs cause autosomal recessive genetic diseases with or without involvement of SNV/indels. Genet Med. 2020;22:1633–1641. PubMed PMC
Pfundt R, Del Rosario M, Vissers L, et al. Detection of clinically relevant copy-number variants by exome sequencing in a large cohort of genetic disorders. Genet Med. 2017;19:667–675. PubMed PMC
Balasubramanian M, Willoughby J, Fry AE, et al. Delineating the phenotypic spectrum of Bainbridge-Ropers syndrome: 12 new patients with de novo, heterozygous, loss-of-function mutations in ASXL3 and review of published literature. J Med Genet. 2017;54:537–543. PubMed
Svantnerova J, Minar M, Radova S, Kolnikova M, Vlkovic P, Zech M. ASXL3 de novo variant-related neurodevelopmental disorder presenting as dystonic cerebral palsy. Neuropediatrics. 2022;53:361–365. PubMed
Jouan L, Rocheford D, Szuto A, et al. An 18 alanine repeat in a severe form of oculopharyngeal muscular dystrophy. Can J Neurol Sci. 2014;41:508–511. PubMed
Marcogliese PC, Shashi V, Spillmann RC, et al. IRF2BPL is associated with neurological phenotypes. Am J Hum Genet. 2018;103:245–260. PubMed PMC
Frints SG, Lenzner S, Bauters M, et al. MCT8 mutation analysis and identification of the first female with Allan-Herndon-Dudley syndrome due to loss of MCT8 expression. Eur J Hum Genet. 2008;16:1029–1037. PubMed
Olivati C, Favilla BP, Freitas EL, et al. Allan-Herndon-Dudley syndrome in a female patient and related mechanisms. Mol Genet Metab Rep. 2022;31:100879. PubMed PMC
Masnada S, Sarret C, Antonello CE, et al. Movement disorders in MCT8 deficiency/Allan-Herndon-Dudley syndrome. Mol Genet Metab. 2022;135:109–113. PubMed
Chong JX, Berger SI, Baxter S, et al. Considerations for reporting variants in novel candidate genes identified during clinical genomic testing. Genet Med. 2024;26:101199. PubMed PMC
The UniProt C . UniProt: The universal protein knowledgebase. Nucleic Acids Res. 2017;45(D1):D158–D169. PubMed PMC
Wiel L, Baakman C, Gilissen D, Veltman JA, Vriend G, Gilissen C. MetaDome: Pathogenicity analysis of genetic variants through aggregation of homologous human protein domains. Hum Mutat. 2019;40:1030–1038. PubMed PMC
Shen S, Zhou H, Xiao Z, et al. PRMT1 in human neoplasm: Cancer biology and potential therapeutic target. Cell Commun Signal. 2024;22:102. PubMed PMC
Fedoriw A, Rajapurkar SR, O'Brien S, et al. Anti-tumor activity of the type I PRMT inhibitor, GSK3368715, synergizes with PRMT5 inhibition through MTAP loss. Cancer Cell. 2019;36:100–114.e25. PubMed
Meyer E, Carss KJ, Rankin J, et al. Mutations in the histone methyltransferase gene KMT2B cause complex early-onset dystonia. Nat Genet. 2017;49:223–237. PubMed
Hashimoto M, Kumabe A, Kim JD, et al. Loss of PRMT1 in the central nervous system (CNS) induces reactive astrocytes and microglia during postnatal brain development. J Neurochem. 2021;156:834–847. PubMed
Centen LM, Pinter D, van Egmond ME, et al. Dystonia management across Europe within ERN-RND: Current state and future challenges. J Neurol. 2023;270:797–809. PubMed PMC
Dzinovic I, Winkelmann J, Zech M. Genetic intersection between dystonia and neurodevelopmental disorders: Insights from genomic sequencing. Parkinsonism Relat Disord. 2022;102:131–140. PubMed
Peall KJ, Owen MJ, Hall J. Rare genetic brain disorders with overlapping neurological and psychiatric phenotypes. Nat Rev Neurol. 2024;20:7–21. PubMed
Indelicato E, Schlieben LD, Stenton SL, et al. Dystonia and mitochondrial disease: The movement disorder connection revisited in 900 genetically diagnosed patients. J Neurol. 2024;271:4685–4692. PubMed PMC
Shambetova C, Klein C. Genetic testing for non-Parkinsonian movement disorders: Navigating the diagnostic maze. Parkinsonism Relat Disord. 2024;121:106033. PubMed
Baide-Mairena H, Coget A, Leboucq N, et al. Infantile-onset Parkinsonism, dyskinesia, and developmental delay: Do not forget polyglutamine defects!. Ann Clin Transl Neurol. 2023;10:1937–1943. PubMed PMC
Herzog R, Weissbach A, Baumer T, Munchau A. Complex dystonias: An update on diagnosis and care. J Neural Transm (Vienna). 2021;128:431–445. PubMed PMC
Aartsma-Rus A, van Roon-Mom W, Lauffer M, et al. Development of tailored splice-switching oligonucleotides for progressive brain disorders in Europe: Development, regulation, and implementation considerations. RNA. 2023;29:446–454. PubMed PMC
Kim J, Woo S, de Gusmao CM, et al. A framework for individualized splice-switching oligonucleotide therapy. Nature. 2023;619:828–836. PubMed PMC
Hindorff LA, Bonham VL, Brody LC, et al. Prioritizing diversity in human genomics research. Nat Rev Genet. 2018;19:175–185. PubMed PMC
Li S, Zhao S, Sinson JC, et al. The clinical utility and diagnostic implementation of human subject cell transdifferentiation followed by RNA sequencing. Am J Hum Genet. 2024;111:841–862. PubMed PMC
Terkelsen T, Mikkelsen NS, Bak EN, et al. CRISPR activation to characterize splice-altering variants in easily accessible cells. Am J Hum Genet. 2024;111:309–322. PubMed PMC
Zhao B, Nguyen MA, Woo S, Kim J, Yu TW, Lee EA. Contribution and therapeutic implications of retroelement insertions in ataxia telangiectasia. Am J Hum Genet. 2023;110:1976–1982. PubMed PMC