Skip to main content

Advertisement

Log in

Genetic testing in adults with neurologic disorders: indications, approach, and clinical impacts

  • Review
  • Published:
Journal of Neurology Aims and scope Submit manuscript

Abstract

The role of genetic testing in neurologic clinical practice has increased dramatically in recent years, driven by research on genetic causes of neurologic disease and increased availability of genetic sequencing technology. Genetic testing is now indicated for adults with a wide range of common neurologic conditions. The potential clinical impacts of a genetic diagnosis are also rapidly expanding, with a growing list of gene-specific treatments and clinical trials, in addition to important implications for prognosis, surveillance, family planning, and diagnostic closure. The goals of this review are to provide practical guidance for clinicians about the role of genetics in their practice and to provide the neuroscience research community with a broad survey of current progress in this field. We aim to answer three questions for the neurologist in practice: Which of my patients need genetic testing? What testing should I order? And how will genetic testing help my patient? We focus on common neurologic disorders and presentations to the neurology clinic. For each condition, we review the most current guidelines and evidence regarding indications for genetic testing, expected diagnostic yield, and recommended testing approach. We also focus on clinical impacts of genetic diagnoses, highlighting a number of gene-specific therapies recently approved for clinical use, and a rapidly expanding landscape of gene-specific clinical trials, many using novel nucleotide-based therapeutic modalities like antisense oligonucleotides and gene transfer. We anticipate that more widespread use of genetic testing will help advance therapeutic development and improve the care, and outcomes, of patients with neurologic conditions.

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

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Abbreviations

ES:

Exome sequencing

GS:

Genome sequencing

VUS:

Variant of uncertain significance

References

  1. Hadjivassiliou M, Martindale J, Shanmugarajah P, Grünewald RA, Sarrigiannis PG, Beauchamp N et al (2017) Causes of progressive cerebellar ataxia: prospective evaluation of 1500 patients. J Neurol Neurosurg Psychiatry 88:301–309

    Article  CAS  PubMed  Google Scholar 

  2. Koriath CAM, Kenny J, Ryan NS, Rohrer JD, Schott JM, Houlden H et al (2021) Genetic testing in dementia—utility and clinical strategies. Nat Rev Neurol 17:23–36

    Article  PubMed  Google Scholar 

  3. Wood EM, Falcone D, Suh E, Irwin DJ, Chen-Plotkin AS, Lee EB et al (2013) Development and validation of pedigree classification criteria for frontotemporal lobar degeneration. JAMA Neurol 70:1411–1417

    Article  PubMed  PubMed Central  Google Scholar 

  4. Krey I, Platzer K, Esterhuizen A, Berkovic SF, Helbig I, Hildebrand MS et al (2022) Current practice in diagnostic genetic testing of the epilepsies. Epileptic Disord 24:765–786

    Article  PubMed  PubMed Central  Google Scholar 

  5. Smith L, Malinowski J, Ceulemans S, Peck K, Walton N, Sheidley BR et al (2023) Genetic testing and counseling for the unexplained epilepsies: an evidence-based practice guideline of the National Society of Genetic Counselors. J Genet Couns 32:266–280

    Article  PubMed  Google Scholar 

  6. Lynch DS, Wade C, de Paiva ARB, John N, Kinsella JA, Merwick Á et al (2019) Practical approach to the diagnosis of adult-onset leukodystrophies: an updated guide in the genomic era. J Neurol Neurosurg Psychiatry 90:543–554

    Article  PubMed  Google Scholar 

  7. Roggenbuck J, Eubank BHF, Wright J, Harms MB, Kolb SJ, ALS Genetic Testing and Counseling Guidelines Expert Panel (2023) Evidence-based consensus guidelines for ALS genetic testing and counseling. Ann Clin Transl Neurol 2023:1

    Google Scholar 

  8. Salmon K, Kiernan MC, Kim SH, Andersen PM, Chio A, van den Berg LH et al (2022) The importance of offering early genetic testing in everyone with amyotrophic lateral sclerosis. Brain 145:1207–1210

    Article  PubMed  PubMed Central  Google Scholar 

  9. Nicolau S, Milone M, Liewluck T (2021) Guidelines for genetic testing of muscle and neuromuscular junction disorders. Muscle Nerve 64:255–269

    Article  PubMed  Google Scholar 

  10. Wang W, Wang C, Dawson DB, Thorland EC, Lundquist PA, Eckloff BW et al (2016) Target-enrichment sequencing and copy number evaluation in inherited polyneuropathy. Neurology 86:1762–1771

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cook L, Schulze J, Naito A, Alcalay RN (2021) The role of genetic testing for Parkinson’s disease. Curr Neurol Neurosci Rep 21:17

    Article  PubMed  Google Scholar 

  12. Zech M, Jech R, Boesch S, Škorvánek M, Weber S, Wagner M et al (2020) Monogenic variants in dystonia: an exome-wide sequencing study. Lancet Neurol 19:908–918

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Beaudin M, Matilla-Dueñas A, Soong B-W, Pedroso JL, Barsottini OG, Mitoma H et al (2019) The classification of autosomal recessive cerebellar ataxias: a consensus statement from the society for research on the cerebellum and ataxias task force. Cerebellum 18:1098–1125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sun M, Johnson AK, Nelakuditi V, Guidugli L, Fischer D, Arndt K et al (2019) Targeted exome analysis identifies the genetic basis of disease in over 50% of patients with a wide range of ataxia-related phenotypes. Genet Med 21:195–206

    Article  CAS  PubMed  Google Scholar 

  15. Fogel BL, Lee H, Deignan JL, Strom SP, Kantarci S, Wang X et al (2014) Exome sequencing in the clinical diagnosis of sporadic or familial cerebellar ataxia. JAMA Neurol 71:1237–1246

    Article  PubMed  PubMed Central  Google Scholar 

  16. Pellerin D, Danzi MC, Wilke C, Renaud M, Fazal S, Dicaire M-J et al (2023) Deep intronic FGF14 GAA repeat expansion in late-onset cerebellar ataxia. N Engl J Med 388:128–141

    Article  CAS  PubMed  Google Scholar 

  17. van de Warrenburg BPC, van Gaalen J, Boesch S, Burgunder J-M, Dürr A, Giunti P et al (2014) EFNS/ENS consensus on the diagnosis and management of chronic ataxias in adulthood. Eur J Neurol 21:552–562

    Article  PubMed  Google Scholar 

  18. Zesiewicz TA, Wilmot G, Kuo S-H, Perlman S, Greenstein PE, Ying SH et al (2018) Comprehensive systematic review summary: treatment of cerebellar motor dysfunction and ataxia. Neurology 90:464–471

    Article  PubMed  PubMed Central  Google Scholar 

  19. Mariani L-L, Tesson C, Charles P, Cazeneuve C, Hahn V, Youssov K et al (2016) Expanding the spectrum of genes involved in Huntington disease using a combined clinical and genetic approach. JAMA Neurol 73:1105–1114

    Article  PubMed  Google Scholar 

  20. Cubo E, Ramos-Arroyo MA, Martinez-Horta S, Martínez-Descalls A, Calvo S, Gil-Polo C et al (2016) Clinical manifestations of intermediate allele carriers in Huntington disease. Neurology 87:571–578

    Article  CAS  PubMed  Google Scholar 

  21. Hensman Moss DJ, Poulter M, Beck J, Hehir J, Polke JM, Campbell T et al (2014) C9orf72 expansions are the most common genetic cause of Huntington disease phenocopies. Neurology 82:292–299

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Huq AJ, Sexton A, Lacaze P, Masters CL, Storey E, Velakoulis D et al (2021) Genetic testing in dementia—a medical genetics perspective. Int J Geriatr Psychiatry 36:1158–1170

    Article  PubMed  Google Scholar 

  23. Goldman JS, Hahn SE, Catania JW, LaRusse-Eckert S, Butson MB, Rumbaugh M et al (2011) Genetic counseling and testing for Alzheimer disease: joint practice guidelines of the American College of Medical Genetics and the National Society of Genetic Counselors. Genet Med 13:597–605

    Article  PubMed  PubMed Central  Google Scholar 

  24. Salloway S, Chalkias S, Barkhof F, Burkett P, Barakos J, Purcell D et al (2022) Amyloid-related imaging abnormalities in 2 phase 3 studies evaluating Aducanumab in patients with early Alzheimer disease. JAMA Neurol 79:13–21

    Article  PubMed  Google Scholar 

  25. Orme T, Guerreiro R, Bras J (2018) The genetics of dementia with Lewy bodies: current understanding and future directions. Curr Neurol Neurosci Rep 18:67

    Article  PubMed  PubMed Central  Google Scholar 

  26. Roggenbuck J, Fong JC (2020) Genetic testing for amyotrophic lateral sclerosis and frontotemporal dementia: impact on clinical management. Clin Lab Med 40:271–287

    Article  PubMed  Google Scholar 

  27. Koriath C, Kenny J, Adamson G, Druyeh R, Taylor W, Beck J et al (2020) Predictors for a dementia gene mutation based on gene-panel next-generation sequencing of a large dementia referral series. Mol Psychiatry 25:3399–3412

    Article  CAS  PubMed  Google Scholar 

  28. McKnight D, Bristow SL, Truty RM, Morales A, Stetler M, Westbrook MJ et al (2022) Multigene panel testing in a large cohort of adults with epilepsy: diagnostic yield and clinically actionable genetic findings. Neurol Genet 8:e650

    Article  PubMed  Google Scholar 

  29. Truty R, Patil N, Sankar R, Sullivan J, Millichap J, Carvill G et al (2019) Possible precision medicine implications from genetic testing using combined detection of sequence and intragenic copy number variants in a large cohort with childhood epilepsy. Epilepsia Open 4:397–408

    Article  PubMed  PubMed Central  Google Scholar 

  30. Bayat A, Fenger CD, Techlo TR, Højte AF, Nørgaard I, Hansen TF et al (2022) Impact of genetic testing on therapeutic decision-making in childhood-onset epilepsies—a study in a tertiary epilepsy center. Neurotherapeutics 19:1353–1367

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Perucca P, Perucca E (2019) Identifying mutations in epilepsy genes: impact on treatment selection. Epilepsy Res 152:18–30

    Article  CAS  PubMed  Google Scholar 

  32. Ayrignac X, Carra-Dalliere C, Menjot de Champfleur N, Denier C, Aubourg P, Bellesme C et al (2015) Adult-onset genetic leukoencephalopathies: a MRI pattern-based approach in a comprehensive study of 154 patients. Brain 138:284–292

    Article  PubMed  Google Scholar 

  33. Helman G, Lajoie BR, Crawford J, Takanohashi A, Walkiewicz M, Dolzhenko E et al (2020) Genome sequencing in persistently unsolved white matter disorders. Ann Clin Transl Neurol 7:144–152

    Article  PubMed  PubMed Central  Google Scholar 

  34. Eichler F, Duncan C, Musolino PL, Orchard PJ, De Oliveira S, Thrasher AJ et al (2017) Hematopoietic stem-cell gene therapy for cerebral adrenoleukodystrophy. N Engl J Med 377:1630–1638

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Köhler W, Engelen M, Eichler F, Lachmann R, Fatemi A, Sampson J et al (2023) Safety and efficacy of leriglitazone for preventing disease progression in men with adrenomyeloneuropathy (ADVANCE): a randomised, double-blind, multi-centre, placebo-controlled phase 2–3 trial. Lancet Neurol 22:127–136

    Article  PubMed  Google Scholar 

  36. Amado DA, Davidson BL (2021) Gene therapy for ALS: a review. Mol Ther 29:3345–3358

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Majounie E, Renton AE, Mok K, Dopper EGP, Waite A, Rollinson S et al (2012) Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol 11:323–330

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chia R, Chiò A, Traynor BJ (2018) Novel genes associated with amyotrophic lateral sclerosis: diagnostic and clinical implications. Lancet Neurol 17:94–102

    Article  CAS  PubMed  Google Scholar 

  39. Shepheard SR, Parker MD, Cooper-Knock J, Verber NS, Tuddenham L, Heath P et al (2021) Value of systematic genetic screening of patients with amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 92:510–518

    Article  PubMed  Google Scholar 

  40. Miller TM, Cudkowicz ME, Genge A, Shaw PJ, Sobue G, Bucelli RC et al (2022) Trial of antisense oligonucleotide tofersen for SOD1 ALS. N Engl J Med 387:1099–1110

    Article  CAS  PubMed  Google Scholar 

  41. Walters J, Baborie A (2020) Muscle biopsy: What and why and when? Pract Neurol 20:385–395

    Article  PubMed  Google Scholar 

  42. Cirak S, Arechavala-Gomeza V, Guglieri M, Feng L, Torelli S, Anthony K et al (2011) Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet 378:595–605

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Clemens PR, Rao VK, Connolly AM, Harper AD, Mah JK, Smith EC et al (2020) Safety, tolerability, and efficacy of viltolarsen in boys with duchenne muscular dystrophy amenable to exon 53 skipping: a phase 2 randomized clinical trial. JAMA Neurol 77:982–991

    Article  PubMed  Google Scholar 

  44. Diaz-Manera J, Kishnani PS, Kushlaf H, Ladha S, Mozaffar T, Straub V et al (2021) Safety and efficacy of avalglucosidase alfa versus alglucosidase alfa in patients with late-onset Pompe disease (COMET): a phase 3, randomised, multicentre trial. Lancet Neurol 20:1012–1026

    Article  CAS  PubMed  Google Scholar 

  45. Fridman V, Bundy B, Reilly MM, Pareyson D, Bacon C, Burns J et al (2015) CMT subtypes and disease burden in patients enrolled in the Inherited Neuropathies Consortium natural history study: a cross-sectional analysis. J Neurol Neurosurg Psychiatry 86:873–878

    Article  CAS  PubMed  Google Scholar 

  46. Murphy SM, Laura M, Fawcett K, Pandraud A, Liu Y-T, Davidson GL et al (2012) Charcot–Marie–Tooth disease: frequency of genetic subtypes and guidelines for genetic testing. J Neurol Neurosurg Psychiatry 83:706–710

    Article  PubMed  Google Scholar 

  47. Lee DC, Dankwa L, Edmundson C, Cornblath DR, Scherer SS (2019) Yield of next-generation neuropathy gene panels in axonal neuropathies. J Peripher Nerv Syst 24:324–329

    Article  PubMed  PubMed Central  Google Scholar 

  48. Di Stefano V, Prinzi F, Luigetti M, Russo M, Tozza S, Alonge P et al (2023) Machine learning for early diagnosis of ATTRv amyloidosis in non-endemic areas: a multicenter study from Italy. Brain Sci 13:805

    Article  PubMed  PubMed Central  Google Scholar 

  49. Berk JL, Suhr OB, Obici L, Sekijima Y, Zeldenrust SR, Yamashita T et al (2013) Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA 310:2658–2667

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, Waddington-Cruz M et al (2018) Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med 379:1007–1016

    Article  CAS  PubMed  Google Scholar 

  51. Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang C-C, Ueda M, Kristen AV et al (2018) Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med 379:11–21

    Article  CAS  PubMed  Google Scholar 

  52. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK et al (2018) Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med 379:22–31

    Article  CAS  PubMed  Google Scholar 

  53. Adams D, Tournev IL, Taylor MS, Coelho T, Planté-Bordeneuve V, Berk JL et al (2023) Efficacy and safety of vutrisiran for patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy: a randomized clinical trial. Amyloid 30:1–9

    Article  PubMed  Google Scholar 

  54. Gillmore JD, Gane E, Taubel J, Kao J, Fontana M, Maitland ML et al (2021) CRISPR-Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med 385:493–502

    Article  CAS  PubMed  Google Scholar 

  55. Iacobucci I, Hay Mele B, Cozzolino F, Monaco V, Cimmaruta C, Monti M et al (2023) Enzyme replacement therapy for FABRY disease: possible strategies to improve its efficacy. Int J Mol Sci 24:4548

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Balwani M, Sardh E, Ventura P, Peiró PA, Rees DC, Stölzel U et al (2020) Phase 3 trial of RNAi therapeutic Givosiran for acute intermittent porphyria. N Engl J Med 382:2289–2301

    Article  CAS  PubMed  Google Scholar 

  57. McCray BA, Scherer SS (2021) Axonal Charcot–Marie–Tooth disease: from common pathogenic mechanisms to emerging treatment opportunities. Neurotherapeutics 18:2269–2285

    Article  PubMed  PubMed Central  Google Scholar 

  58. Hill EJ, Robak LA, Al-Ouran R, Deger J, Fong JC, Vandeventer PJ et al (2022) Genome sequencing in the parkinson disease clinic. Neurol Genet 8:e200002

    Article  PubMed  PubMed Central  Google Scholar 

  59. Członkowska A, Litwin T, Dusek P, Ferenci P, Lutsenko S, Medici V et al (2018) Wilson disease. Nat Rev Dis Primers 4:1–20

    Article  Google Scholar 

  60. Schneider SA, Hizli B, Alcalay RN (2020) Emerging targeted therapeutics for genetic subtypes of parkinsonism. Neurotherapeutics 17:1378–1392

    Article  PubMed  PubMed Central  Google Scholar 

  61. Mullin S, Smith L, Lee K, D’Souza G, Woodgate P, Elflein J et al (2020) Ambroxol for the treatment of patients with parkinson disease with and without glucocerebrosidase gene mutations: a nonrandomized. Noncontrolled Trial JAMA Neurol 77:427–434

    Article  PubMed  Google Scholar 

  62. Powis Z, Towne MC, Hagman KDF, Blanco K, Palmaer E, Castro A et al (2020) Clinical diagnostic exome sequencing in dystonia: genetic testing challenges for complex conditions. Clin Genet 97:305–311

    Article  CAS  PubMed  Google Scholar 

  63. Wirth T, Tranchant C, Drouot N, Keren B, Mignot C, Cif L et al (2020) Increased diagnostic yield in complex dystonia through exome sequencing. Parkinsonism Relat Disord 74:50–56

    Article  PubMed  Google Scholar 

  64. Osmanovic A, Rangnau I, Kosfeld A, Abdulla S, Janssen C, Auber B et al (2017) FIG4 variants in central European patients with amyotrophic lateral sclerosis: a whole-exome and targeted sequencing study. Eur J Hum Genet 25:324–331

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Ellis CA, Petrovski S, Berkovic SF (2020) Epilepsy genetics: clinical impacts and biological insights. Lancet Neurol 19:93–100

    Article  CAS  PubMed  Google Scholar 

  66. Mersch J, Brown N, Pirzadeh-Miller S, Mundt E, Cox HC, Brown K et al (2018) Prevalence of variant reclassification following hereditary cancer genetic testing. JAMA 320:1266–1274

    Article  PubMed  PubMed Central  Google Scholar 

  67. Russo M, Gentile L, Di Stefano V, Di Bella G, Minutoli F, Toscano A et al (2021) Use of drugs for ATTRv amyloidosis in the real world: how therapy is changing survival in a non-endemic area. Brain Sci 11:545

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Hayeems RZ, Luca S, Assamad D, Bhatt A, Ungar WJ (2021) Utility of genetic testing from the perspective of parents/caregivers: a scoping review. Children 8:259

    Article  PubMed  PubMed Central  Google Scholar 

  69. Park H-K, Lee K-J, Park J-M, Kang K, Lee SJ, Kim JG et al (2023) Prevalence of mutations in Mendelian stroke genes in early onset stroke patients. Ann Neurol 93:768–782

    Article  CAS  PubMed  Google Scholar 

  70. Harder AV, Terwindt GM, Nyholt DR, van den Maagdenberg AM (2023) Migraine genetics: status and road forward. Cephalalgia 43:3331024221145962

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Colin A. Ellis.

Ethics declarations

Conflicts of interest

L. Dratch has consulted for Passage Bio, Biogen, and Sano Genetics. None of these represent a conflict for this manuscript. M. Azage has no disclosures. A. Baldwin has no disclosures. K. Johnson has no disclosures. R.A. Paul is a site genetic counselor for the Parkinson Foundation PDGENEration study. T.M. Bardakjian is an employee and equity holder of Sarepta Therapeutics, Inc. S. Michon has no disclosures. D.A. Amado has no disclosures. M. Baer has no disclosures. A.F. Deik has served on the Advisory Board for Supernus Therapeutics and Acorda therapeutics. He has received royalties from UpToDate and Elsevier. He also has received clinical trial funding from Addex therapeutics, Cerevel therapeutics, Annovis Bio, Lundbeck pharmaceuticals, Eli Lilly, and Teva pharmaceuticals. L.B. Elman has had financial relationships with Biogen, Genentech, PTC Therapeutics, Edgewise Therapeutics, and Apellis Pharmaceuticals. P. Gonzalez-Alegre is an employee and equity holder of Spark Therapeutics, a Roche company. M.H. Guo has no disclosures. A.G. Hamedani has received grants from the National Eye Institute and Michael J. Fox Foundation. He has been part of clinical trials sponsored by Biogen and Biohaven. None are conflicts of interest with respect to this manuscript. D.J. Irwin has no disclosures. A. Lasker has no disclosures. J. Orthmann-Murphy is a site PI for Vigil Neurosciences. She has received grants from National MS Society and GLIA-CTN, and honorarium from Novoglia. C. Quinn has served on Advisory Boards for Amylyx Pharmaceuticals and Biogen. He has Consulted for Guidepoint, GLG and Pinnacle Research Group. T.F. Tropea has received grants from the NIH/NINDS (K23-NS114167), and research support from the Parkinson Foundation and The Michael J Fox Foundation. He has received consulting fees and honoraria from Sanofi Genzyme, Bial, and the Parkinson Foundation. S.S. Scherer has served on the Scientific Advisory Board for Mitochondria in Motion and Disarm Therapeutics, and consulted for Applied Therapeutics, DTx Therapeutics, Passage Bio, Pfizer, and Toray Industries. C.A. Ellis is supported by NIH Award Number K23NS121520; and by the American Academy of Neurology Susan S. Spencer Clinical Research Training Scholarship. He also receives consulting fees from an EEG company called EpiMinder (no relationship to this manuscript).

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dratch, L., Azage, M., Baldwin, A. et al. Genetic testing in adults with neurologic disorders: indications, approach, and clinical impacts. J Neurol 271, 733–747 (2024). https://doi.org/10.1007/s00415-023-12058-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00415-023-12058-6

Keywords

Navigation