open access

Vol 58, No 1 (2024)
Invited Review Article
Submitted: 2023-10-11
Accepted: 2023-11-28
Published online: 2024-01-04
Get Citation

Genetics of Parkinson’s Disease: state-of-the-art and role in clinical settings

Jarosław Dulski1234, Owen A. Ross45, Zbigniew K. Wszolek1
·
Pubmed: 38175148
·
Neurol Neurochir Pol 2024;58(1):38-46.
Affiliations
  1. Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
  2. Division of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
  3. Neurology Department, St Adalbert Hospital, Copernicus PL Ltd., Gdansk, Poland
  4. Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
  5. Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida, USA

open access

Vol 58, No 1 (2024)
Invited review articles
Submitted: 2023-10-11
Accepted: 2023-11-28
Published online: 2024-01-04

Abstract

Introduction. Advances in sequencing technologies have enabled extensive genetic testing on an individual basis. Genome-wide association studies (GWAS) have provided insight into the pathophysiology of PD. Additionally, direct-to-consumer genetic testing has enabled the identification of genetic diseases and risk factors without genetic counselling. As genetics increasingly permeates clinical practice, this paper aims to summarise the most important information on genetics in PD forclinical practitioners.

State-of-the-art. LRRK2 mutations may be found in c.1% of all PD patients with an indistinguishable phenotype from sporadic PD. LRRK2-PD is more prevalent in patients with a positive family history (5-6%) and among certain populations (e.g. up to 41% in North Africans and Ashkenazi Jews). Other familial forms include PRKN (patients with early onset, EOPD), VPS35 (Western European ancestry), PINK1 (EOPD), DJ-1 (EOPD), and SNCA. GBA mutations are found in a large number of PD patients and are associated with faster progression and a poorer prognosis. GWAS have identified 90 genetic risk variants for developing PD and several genetic modifiers for the age at onset, disease progression, and response to treatment.


Clinical implications. Multigene panels using next-generation sequencing (NGS) are the first choice for genetic testing in clinical settings. Whole exome sequencing is increasingly being used, particularly as the second-tier testing in patients with negative results of multigene panels. NGS may not detect accurately copy number variants (CNV), meaning that additional analysis is warranted. In a case of a variant of unknown significance (VUS), we suggest firstly searching the up-to-date literature. Segregation studies and in silico predictions may shed more light on the character of the VUS; however, functional studies remain the gold standard. Several interventional clinical trials are active for carriers of LRRK2 and/or GBA mutations.

Future directions. Application of artificial intelligence and machine learning will enable high-throughput analysis of large
sets of multimodal data. We speculate that, in the future, the treatment landscape for PD will be similar to that in oncological conditions, in which the presence of certain gene mutations or gene overexpression determines the prognosis and treatment decision-making.

Abstract

Introduction. Advances in sequencing technologies have enabled extensive genetic testing on an individual basis. Genome-wide association studies (GWAS) have provided insight into the pathophysiology of PD. Additionally, direct-to-consumer genetic testing has enabled the identification of genetic diseases and risk factors without genetic counselling. As genetics increasingly permeates clinical practice, this paper aims to summarise the most important information on genetics in PD forclinical practitioners.

State-of-the-art. LRRK2 mutations may be found in c.1% of all PD patients with an indistinguishable phenotype from sporadic PD. LRRK2-PD is more prevalent in patients with a positive family history (5-6%) and among certain populations (e.g. up to 41% in North Africans and Ashkenazi Jews). Other familial forms include PRKN (patients with early onset, EOPD), VPS35 (Western European ancestry), PINK1 (EOPD), DJ-1 (EOPD), and SNCA. GBA mutations are found in a large number of PD patients and are associated with faster progression and a poorer prognosis. GWAS have identified 90 genetic risk variants for developing PD and several genetic modifiers for the age at onset, disease progression, and response to treatment.


Clinical implications. Multigene panels using next-generation sequencing (NGS) are the first choice for genetic testing in clinical settings. Whole exome sequencing is increasingly being used, particularly as the second-tier testing in patients with negative results of multigene panels. NGS may not detect accurately copy number variants (CNV), meaning that additional analysis is warranted. In a case of a variant of unknown significance (VUS), we suggest firstly searching the up-to-date literature. Segregation studies and in silico predictions may shed more light on the character of the VUS; however, functional studies remain the gold standard. Several interventional clinical trials are active for carriers of LRRK2 and/or GBA mutations.

Future directions. Application of artificial intelligence and machine learning will enable high-throughput analysis of large
sets of multimodal data. We speculate that, in the future, the treatment landscape for PD will be similar to that in oncological conditions, in which the presence of certain gene mutations or gene overexpression determines the prognosis and treatment decision-making.

Get Citation

Keywords

hereditary, monogenic, familial, sequencing, GWAS

About this article
Title

Genetics of Parkinson’s Disease: state-of-the-art and role in clinical settings

Journal

Neurologia i Neurochirurgia Polska

Issue

Vol 58, No 1 (2024)

Article type

Invited Review Article

Pages

38-46

Published online

2024-01-04

Page views

682

Article views/downloads

615

DOI

10.5603/pjnns.97806

Pubmed

38175148

Bibliographic record

Neurol Neurochir Pol 2024;58(1):38-46.

Keywords

hereditary
monogenic
familial
sequencing
GWAS

Authors

Jarosław Dulski
Owen A. Ross
Zbigniew K. Wszolek

References (62)
  1. Polymeropoulos MH, Higgins JJ, Golbe LI, et al. Mapping of a gene for Parkinson's disease to chromosome 4q21-q23. Science. 1996; 274(5290): 1197–1199.
  2. Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997; 276(5321): 2045–2047.
  3. Dulski J, Uitti RJ, Ross OA, et al. Genetic architecture of Parkinson's disease subtypes - Review of the literature. Front Aging Neurosci. 2022; 14: 1023574.
  4. Cook L, Schulze J, Verbrugge J, et al. ClinGen Parkinson's Disease Gene Curation Expert Panel and the MDS Task Force for Recommendations for Genetic Testing in Parkinson's Disease, Clinical Genome Resource (ClinGen) Parkinson's Disease Gene Curation Expert Panel Authors, Movement Society Disorder (MDS) Task Force on Recommendations for Clinical Genetic Testing in Parkinson's Disease Authors. The commercial genetic testing landscape for Parkinson's disease. Parkinsonism Relat Disord. 2021; 92: 107–111.
  5. Balestrino R, Schapira AHV. Parkinson disease. Eur J Neurol. 2020; 27(1): 27–42.
  6. Hall A, Bandres-Ciga S, Diez-Fairen M, et al. Genetic Risk Profiling in Parkinson's Disease and Utilizing Genetics to Gain Insight into Disease-Related Biological Pathways. Int J Mol Sci. 2020; 21(19).
  7. Dulski J, Fujioka S, Wszolek ZK. Genetic parkinsonisms. In: Wolters E, Baumann C, Truong D. ed. IAPRD Textbook of Movement Disorders. Cambridge University Press, Cambridge forthcoming.
  8. Ali S, Wszolek ZK. LRRK2 R1441C mutation causing Parkinson's Disease in an Egyptian family. Neurol Neurochir Pol. 2022; 56(2): 191–192.
  9. Saunders-Pullman R, Raymond D, Elango S. LRRK2 Parkinson Disease. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJ, Gripp KW, Amemiya A. ed. GeneReviews(®). University of Washington, Seattle 1993-2023.
  10. 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(6): 665–677.
  11. Milanowski ŁM, Ross OA, Friedman A, et al. Genetics of Parkinson's disease in the Polish population. Neurol Neurochir Pol. 2021; 55(3): 241–252.
  12. Brüggemann N, Klein C. Parkin Type of Early-Onset Parkinson Diseas. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJ, Gripp KW, Amemiya A. ed. GeneReviews(®). University of Washington, Seattle 1993-2023.
  13. Simon DK, Tanner CM, Brundin P. Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. Clin Geriatr Med. 2020; 36(1): 1–12.
  14. Periquet M, Latouche M, Lohmann E, et al. French Parkinson's Disease Genetics Study Group, European Consortium on Genetic Susceptibility in Parkinson's Disease. Parkin mutations are frequent in patients with isolated early-onset parkinsonism. Brain. 2003; 126(Pt 6): 1271–1278.
  15. Yoshino H, Li Y, Nishioka K, et al. investigators of Japan Parkinson disease genetic study. Genotype-phenotype correlation of Parkinson's disease with PRKN variants. Neurobiol Aging. 2022; 114: 117–128.
  16. Dulski J, Ross OA, Wszolek ZK. VPS35-Related Parkinson Disease. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LH, Gripp KW, Amemiya A. ed. GeneReviews(®). University of Washington, Seattle 1993-2023.
  17. Schneider SA, Klein C. PINK1 Type of Young-Onset Parkinson. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJ, Gripp KW, Amemiya A. ed. DiseaseGeneReviews(®). University of Washington, Seattle 1993-2023.
  18. Book A, Guella I, Candido T, et al. SNCA Multiplication Investigators of the GEoPD Consortium. A Meta-Analysis of α-Synuclein Multiplication in Familial Parkinsonism. Front Neurol. 2018; 9: 1021.
  19. Edvardson S, Cinnamon Y, Ta-Shma A, et al. A deleterious mutation in DNAJC6 encoding the neuronal-specific clathrin-uncoating co-chaperone auxilin, is associated with juvenile parkinsonism. PLoS One. 2012; 7(5): e36458.
  20. Kurian MA, Abela L. DNAJC6 Parkinson Disease. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJ, Gripp KW, Amemiya A. ed. GeneReviews(®). University of Washington, Seattle 1993-2023.
  21. Fernández-Santiago R, Sharma M. What have we learned from genome-wide association studies (GWAS) in Parkinson's disease? Ageing Res Rev. 2022; 79: 101648.
  22. Senkevich K, Rudakou U, Gan-Or Z. Genetic mechanism vs genetic subtypes: The example of GBA. Handb Clin Neurol. 2023; 193: 155–170.
  23. Vieira SRL, Schapira AHV. Glucocerebrosidase mutations and Parkinson disease. J Neural Transm. 2022; 129: 1105–1117.
  24. Höglinger G, Schulte C, Jost WH, et al. GBA-associated PD: chances and obstacles for targeted treatment strategies. J Neural Transm (Vienna). 2022; 129(9): 1219–1233.
  25. Nalls MA, Blauwendraat C, Vallerga CL, et al. 23andMe Research Team, System Genomics of Parkinson's Disease Consortium, International Parkinson's Disease Genomics Consortium. Identification of novel risk loci, causal insights, and heritable risk for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 2019; 18(12): 1091–1102.
  26. Blauwendraat C, Heilbron K, Vallerga CL, et al. 23andMe Research Team, International Parkinson's Disease Genomics Consortium (IPDGC). Parkinson's disease age at onset genome-wide association study: Defining heritability, genetic loci, and α-synuclein mechanisms. Mov Disord. 2019; 34(6): 866–875.
  27. Grover S, Kumar Sreelatha AA, Pihlstrom L, et al. and the Comprehensive Unbiased Risk Factor Assessment for Genetics and Environment in Parkinson's Disease (COURAGE-PD) Consortium. Genome-wide Association and Meta-analysis of Age at Onset in Parkinson Disease: Evidence From the COURAGE-PD Consortium. Neurology. 2022; 99(7): e698–e710.
  28. Iwaki H, Blauwendraat C, Leonard HL, et al. International Parkinson's Disease Genomics Consortium. Genomewide association study of Parkinson's disease clinical biomarkers in 12 longitudinal patients' cohorts. Mov Disord. 2019; 34(12): 1839–1850.
  29. Iwaki H, Blauwendraat C, Leonard HL, et al. Genetic risk of Parkinson disease and progression:: An analysis of 13 longitudinal cohorts. Neurol Genet. 2019; 5(4): e348.
  30. Tan MMX, Lawton MA, Jabbari E, et al. Genome-Wide Association Studies of Cognitive and Motor Progression in Parkinson's Disease. Mov Disord. 2021; 36(2): 424–433.
  31. König E, Nicoletti A, Pattaro C, et al. Exome-wide association study of levodopa-induced dyskinesia in Parkinson's disease. Sci Rep. 2021; 11(1): 19582.
  32. Liu G, Peng J, Liao Z, et al. International Genetics of Parkinson Disease Progression (IGPP) Consortium. Genome-wide survival study identifies a novel synaptic locus and polygenic score for cognitive progression in Parkinson's disease. Nat Genet. 2021; 53(6): 787–793.
  33. Weintraub D, Posavi M, Fontanillas P, et al. 23andMe Research Team. Genetic prediction of impulse control disorders in Parkinson's disease. Ann Clin Transl Neurol. 2022; 9(7): 936–949.
  34. Alfradique-Dunham I, Al-Ouran R, von Coelln R, et al. Genome-Wide Association Study Meta-Analysis for Parkinson Disease Motor Subtypes. Neurol Genet. 2021; 7(2): e557.
  35. Visanji NP, Ghani M, Yu E, et al. Axial Impairment Following Deep Brain Stimulation in Parkinson's Disease: A Surgicogenomic Approach. J Parkinsons Dis. 2022; 12(1): 117–128.
  36. Tam V, Patel N, Turcotte M, et al. Benefits and limitations of genome-wide association studies. Nat Rev Genet. 2019; 20(8): 467–484.
  37. Bialecka M, Hui S, Klodowska-Duda G, et al. Analysis of LRRK 2 G 2019 S and I 2020 T mutations in Parkinson's disease. Neurosci Lett. 2005; 390(1): 1–3.
  38. Kachergus J, Mata IF, Hulihan M, et al. Identification of a novel LRRK2 mutation linked to autosomal dominant parkinsonism: evidence of a common founder across European populations. Am J Hum Genet. 2005; 76(4): 672–680.
  39. Gaweda-Walerych K, Safranow K, Jasinska-Myga B, et al. PARK2 variability in Polish Parkinson's disease patients--interaction with mitochondrial haplogroups. Parkinsonism Relat Disord. 2012; 18(5): 520–524.
  40. Koziorowski D, Hoffman-Zacharska D, Sławek J, et al. Incidence of mutations in the PARK2, PINK1, PARK7 genes in Polish early-onset Parkinson disease patients. Neurol Neurochir Pol. 2013; 47(4): 319–324.
  41. Puschmann A, Fiesel FC, Caulfield TR, et al. Heterozygous PINK1 p.G411S increases risk of Parkinson's disease via a dominant-negative mechanism. Brain. 2017; 140(1): 98–117.
  42. Hoffman-Zacharska D, Koziorowski D, Ross OA, et al. Novel A18T and pA29S substitutions in α-synuclein may be associated with sporadic Parkinson's disease. Parkinsonism Relat Disord. 2013; 19(11): 1057–1060.
  43. Malec-Litwinowicz M, Plewka A, Plewka D, et al. The relation between plasma α-synuclein level and clinical symptoms or signs of Parkinson's disease. Neurol Neurochir Pol. 2018; 52(2): 243–251.
  44. Jamrozik Z, Lugowska A, Kosiorowski D. Beta-Glucocerebrosidase Gene Mutations P.Asn409Ser and P.Leu483Pro in Polish Patients with Parkinson's Disease. Journal of Neurology and Neuroscience. 2015; 06(04).
  45. Quirini-Popławska D. Polska azylem europejskich emigrantów na przełomie wieków średnich i nowożytnych. Odrodzenie i Reformacja w Polsce. 1993; 37: 34–46.
  46. Turski P, Chaberska I, Szukało P, et al. Review of the epidemiology and variability of LRRK2 non-p.Gly2019Ser pathogenic mutations in Parkinson's disease. Front Neurosci. 2022; 16: 971270.
  47. Kaja E, Lejman A, Sielski D, et al. The Thousand Polish Genomes-A Database of Polish Variant Allele Frequencies. Int J Mol Sci. 2022; 23(9).
  48. Tipton PW, Bülbül N, Crook J, et al. Effects of sex and APOE on Parkinson's Disease-related cognitive decline. Neurol Neurochir Pol. 2021; 55(6): 559–566.
  49. Pal G, Mangone G, Hill EJ, et al. Parkinson Disease and Subthalamic Nucleus Deep Brain Stimulation: Cognitive Effects in GBA Mutation Carriers. Ann Neurol. 2022; 91(3): 424–435.
  50. Avenali M, Zangaglia R, Cuconato G, et al. PARKNET Study Group. Are patients with GBA-Parkinson disease good candidates for deep brain stimulation? A longitudinal multicentric study on a large Italian cohort. J Neurol Neurosurg Psychiatry. 2023 [Epub ahead of print].
  51. Cook L, Schulze J, Naito A, et al. The Role of Genetic Testing for Parkinson's Disease. Curr Neurol Neurosci Rep. 2021; 21(4): 17.
  52. Radziwonik W, Elert-Dobkowska E, Tomczuk F, et al. C9orf72 hexanucleotide repeat expansion found in suspected spinobulbar muscular atrophy (SBMA). Neurol Neurochir Pol. 2022; 56(3): 276–280.
  53. Wszolek Z. When is it useful to genotype Parkinson's disease patients? Henry Stewart Talks 2023.
  54. Kovanda A, Rački V, Bergant G, et al. A multicenter study of genetic testing for Parkinson's disease in the clinical setting. NPJ Parkinsons Dis. 2022; 8(1): 149.
  55. Alcalay RN, Kehoe C, Shorr E, et al. Genetic testing for Parkinson disease: current practice, knowledge, and attitudes among US and Canadian movement disorders specialists. Genet Med. 2020; 22(3): 574–580.
  56. Wetterstrand KA. The Cost of Sequencing a Human Genome. National Human Genome Research Institute. https://www.genome.gov/about-genomics/fact-sheets/Sequencing-Human-Genome-cost (23.22.2023).
  57. Pennisi E. Upstart DNA sequencers could be a 'game changer'. Science. 2022; 376(6599): 1257–1258.
  58. PPD GENEration: Mapping the Future of Parkinson's Disease. Parkinson's Foundation. https://www.parkinson.org/advancing-research/our-research/pdgeneration (23.11.2023).
  59. Parkinson's Progression Markers Initiative. The Michael J. Fox Foundation. https://www.michaeljfox.org/news/parkinsons-progression-markers-initiative (23.11.2023).
  60. Cardoso F, Kyriakides S, Ohno S, et al. ESMO Guidelines Committee. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2019; 30(10): 1674.
  61. Gennari A, André F, Barrios CH, et al. ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. ESMO Clinical Practice Guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol. 2021; 32(12): 1475–1495.
  62. Makarious MB, Leonard HL, Vitale D, et al. Multi-modality machine learning predicting Parkinson's disease. NPJ Parkinsons Dis. 2022; 8(1): 35.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk, Poland
tel.:+48 58 320 94 94, fax:+48 58 320 94 60, e-mail: viamedica@viamedica.pl