Online first
Invited Review Article
Published online: 2025-03-03

open access

Page views 268
Article views/downloads 181
Get Citation

Connect on Social Media

Connect on Social Media

Do Parkinson’s Disease clinical subtypes really exist?

Marta Filidei1, Luca Marsili2, Carlo Colosimo1

Abstract

Introduction. Parkinson’s Disease (PD) is a highly heterogeneous entity in terms of clinical manifestations, progression, and treatment response. This variability has given rise to the hypothesis that different clinical subtypes of the disease exist.

State of the art.
To date, several clinical subtypes have been described, mostly based on different clinical features, and sometimes with the support of biomarkers, either fluid, neuroimaging, or neurophysiological. The most homogeneous subtypes detected are a ‘benign subtype’, characterised by younger age at onset, mild non-motor symptoms, and a slower rate of disease progression, and a ‘malignant subtype’, which features an older age at onset, a higher burden of non-motor symptoms, and faster disease progression.

Clinical implications. Despite extensive research, none of the subtypes identified so far seem to be biologically supported, so clinical subtyping does not elucidate PD aetiology and does not allow for the prediction of prognosis or treatment response. This study was aimed to review the literature on this topic and to examine the studies on PD subtyping. We also reviewed the proposed biomarkers for a biological classification of PD, and outlined the role of genetics and pathology within this context.

Future directions.
In light of the recent proposal of a biological classification of PD, which might overcome the limits of the clinical diagnosis, PD subtyping should hopefully shepherd researchers towards a biological approach, also aided by recent advances in the field of biomarkers.

Article available in PDF format

View PDF Download PDF file

References

  1. Postuma RB, et al. MDS clinical diagnostic criteria for Parkinson's disease. Movement Disorders. 2015; 30(12): 1591–1601.
  2. Virameteekul S, Revesz T, Jaunmuktane Z, et al. Clinical Diagnostic Accuracy of Parkinson's Disease: Where Do We Stand? Mov Disord. 2023; 38(4): 558–566.
  3. Beach TG, Adler CH, Beach TG, et al. Importance of low diagnostic Accuracy for early Parkinson's disease. Mov Disord. 2018; 33(10): 1551–1554.
  4. Mestre T, Fereshtehnejad SM, Berg D, et al. Parkinson’s Disease Subtypes: Critical Appraisal and Recommendations. Journal of Parkinson's Disease. 2021; 11(2): 395–404.
  5. Puschmann A, Puschmann A. Monogenic Parkinson's disease and parkinsonism: clinical phenotypes and frequencies of known mutations. Parkinsonism Relat Disord. 2013; 19(4): 407–415.
  6. Sieber, B.-A. Prioritized research recommendations from the National Institute of ogical Disorders and Stroke Parkinson’s Disease 2014 conference. Ann Neurol. 2014; 76(4): 469–472.
  7. Merola A, Romagnolo A, Dwivedi AK, et al. Benign versus malignant Parkinson disease: the unexpected silver lining of motor complications. J Neurol. 2020; 267(10): 2949–2960.
  8. Jankovic J, McDermott M, Carter J, et al. Variable expression of Parkinson's disease: a base-line analysis of the DATATOP cohort. The Parkinson Study Group. Neurology. 1990; 40(10): 1529–1534.
  9. Marsili L, Mahajan A, Marsili L, et al. Clinical milestones in Parkinson's disease: Past, present, and future. J Neurol Sci. 2022; 432: 120082.
  10. Marras C, Fereshtehnejad SM, Berg D, et al. Transitioning from Subtyping to Precision Medicine in Parkinson's Disease: A Purpose-Driven Approach. Mov Disord. 2024; 39(3): 462–471.
  11. Höglinger GU, Adler CH, Berg D, et al. A biological classification of Parkinson's disease: the SynNeurGe research diagnostic criteria. Lancet Neurol. 2024; 23(2): 191–204.
  12. Dulski J, Ross OA, Wszolek ZK, et al. Genetics of Parkinson's Disease: state-of-the-art and role in clinical settings. Neurol Neurochir Pol. 2024; 58(1): 38–46.
  13. Dulski J, Uitti RJ, Beasley A, et al. Genetics of Parkinson's disease heterogeneity: A genome-wide association study of clinical subtypes. Parkinsonism Relat Disord. 2024; 119: 105935.
  14. Brooks DJ, Playford ED, Ibanez V, et al. Isolated tremor and disruption of the nigrostriatal dopaminergic system: an 18F-dopa PET study. Neurology. 1992; 42(8): 1554–1560.
  15. Marshall V, Grosset DG, Marshall V, et al. Role of dopamine transporter imaging in the diagnosis of atypical tremor disorders. Mov Disord. 2003; 18 Suppl 7: S22–S27.
  16. Josephs KA, Matsumoto JY, Ahlskog JE, et al. Benign tremulous parkinsonism. Arch Neurol. 2006; 63(3): 354–357.
  17. Emre M, et al. Clinical diagnostic criteria for dementia associated with Parkinson’s disease. 2007; 22(15): quiz.
  18. Surendranathan A, Kane JPM, Bentley A, et al. Clinical prevalence of Lewy body dementia. Alzheimers Res Ther. 2018; 10(1): 19.
  19. McKeith IG, McKeith IG, Boeve BF, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017; 89(1): 88–100.
  20. Jellinger KA, Korczyn AD, Jellinger KA, et al. Are dementia with Lewy bodies and Parkinson's disease dementia the same disease? BMC Med. 2018; 16(1): 34.
  21. Jellinger KA, Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna). 2018; 125(4): 615–650.
  22. Jellinger KA, Korczyn AD, Attems J, et al. Are there morphological differences between Parkinson’s disease-dementia and dementia with Lewy bodies? Parkinsonism Relat. Disord. 2022; 100: 24–32.
  23. Dickson DW, Braak H, Duda JE, et al. Neuropathological assessment of Parkinson's disease: refining the diagnostic criteria. Lancet Neurol. 2009; 8(12): 1150–1157.
  24. Marsili L, Rizzo G, Colosimo C, et al. Diagnostic Criteria for Parkinson's Disease: From James Parkinson to the Concept of Prodromal Disease. Front Neurol. 2018; 9: 156.
  25. Geroin C, Artusi CA, Nonnekes J, et al. International Parkinson and Movement Disorders Society Task Force on Postural Abnormalities, International Parkinson and Movement Disorders Society Task Force on Postural Abnormalities. Axial Postural Abnormalities in Parkinsonism: Gaps in Predictors, Pathophysiology, and Management. Mov Disord. 2023; 38(5): 732–739.
  26. Doherty KM, Davagnanam I, Molloy S, et al. Postural deformities in Parkinson's disease. Lancet Neurol. 2011; 10(6): 538–549.
  27. Köllensperger M, Geser F, Seppi K, et al. European MSA Study Group, European MSA Study Group. Red flags for multiple system atrophy. Mov Disord. 2008; 23(8): 1093–1099.
  28. Tiple D, Fabbrini G, Colosimo C, et al. Camptocormia in Parkinson disease: an epidemiological and clinical study. J Neurol Neurosurg Psychiatry. 2009; 80(2): 145–148.
  29. Schäbitz WR, Glatz K, Schuhan C, et al. Severe forward flexion of the trunk in Parkinson's disease: focal myopathy of the paraspinal muscles mimicking camptocormia. Mov Disord. 2003; 18(4): 408–414.
  30. Chaudhuri KR, Healy DG, Schapira AHV, et al. National Institute for Clinical Excellence, National Institute for Clinical Excellence. Non-motor symptoms of Parkinson's disease: diagnosis and management. Lancet Neurol. 2006; 5(3): 235–245.
  31. Kim HS, Cheon SM, Seo JW, et al. Nonmotor symptoms more closely related to Parkinson's disease: comparison with normal elderly. J Neurol Sci. 2013; 324(1-2): 70–73.
  32. Krishnan S, Sarma G, Sarma S, et al. Do nonmotor symptoms in Parkinson's disease differ from normal aging? Mov Disord. 2011; 26(11): 2110–2113.
  33. O'Sullivan SS, Williams DR, Gallagher DA, et al. Nonmotor symptoms as presenting complaints in Parkinson's disease: a clinicopathological study. Mov Disord. 2008; 23(1): 101–106.
  34. Heinzel S, Berg D, Gasser T, et al. MDS Task Force on the Definition of Parkinson's Disease, MDS Task Force on the Definition of Parkinson's Disease. MDS research criteria for prodromal Parkinson's disease. Mov Disord. 2015; 30(12): 1600–1611.
  35. Sauerbier A, Rosa-Grilo M, Qamar MA, et al. Nonmotor Subtyping in Parkinson's Disease. Int Rev Neurobiol. 2017; 133: 447–478.
  36. Dulski J, Sławek J, et al. Incidence and characteristics of post-COVID-19 parkinsonism and dyskinesia related to COVID-19 vaccines. Neurol Neurochir Pol. 2023; 57(1): 53–62.
  37. Fereshtehnejad SM, Postuma RB, Fereshtehnejad SM, et al. Subtypes of Parkinson's Disease: What Do They Tell Us About Disease Progression? Curr Neurol Neurosci Rep. 2017; 17(4): 34.
  38. Berg D, Borghammer P, Fereshtehnejad SM, et al. Prodromal Parkinson disease subtypes - key to understanding heterogeneity. Nat Rev Neurol. 2021; 17(6): 349–361.
  39. DATATOP: a multicenter controlled clinical trial in early Parkinson's disease. Parkinson Study Group. Arch Neurol. 1989; 46(10): 1052–1060.
  40. Graham J, Sagar H, Graham J, et al. A data-driven approach to the study of heterogeneity in idiopathic Parkinson's disease: identification of three distinct subtypes. Movement Disorders. 1999; 14(1): 10–20, doi: 10.1002/1531-8257(199901)14:1<10::aid-mds1005>3.0.co;2-4.
  41. van Rooden SM, et al. Clinical subtypes of Parkinson's disease. Mov Disord. 2011; 26(1): 51–58.
  42. Profiling Parkinson’s disease | Research with human participants. https://onderzoekmetmensen.nl/en/trial/54513 (12.11.2024).
  43. Grupo ELEP. [A longitudinal study of patients with Parkinson's disease (ELEP): aims and methodology]. Rev Neurol. 2006; 42(6): 360–365.
  44. Erro R, Vitale C, Amboni M, et al. The heterogeneity of early Parkinson's disease: a cluster analysis on newly diagnosed untreated patients. PLoS One. 2013; 8(8): e70244.
  45. Parkinson ression Marker Initiative. The Parkinson ression Marker Initiative (PPMI). Prog Neurobiol. 2011; 95: 629–635.
  46. Fereshtehnejad SM, Zeighami Y, Dagher A, et al. Clinical criteria for subtyping Parkinson's disease: biomarkers and longitudinal progression. Brain. 2017; 140(7): 1959–1976.
  47. Shakya S, Prevett J, Hu X, et al. Characterization of Parkinson's Disease Subtypes and Related Attributes. Front Neurol. 2022; 13: 810038.
  48. Franzén E, Johansson H, Freidle M, et al. The EXPANd trial: effects of exercise and exploring neuroplastic changes in people with Parkinson's disease: a study protocol for a double-blinded randomized controlled trial. BMC Neurol. 2019; 19(1): 280.
  49. Albrecht F, Poulakis K, Freidle M, et al. Unraveling Parkinson's disease heterogeneity using subtypes based on multimodal data. Parkinsonism Relat Disord. 2022; 102: 19–29.
  50. De Pablo-Fernández E, Lees AJ, Holton JL, et al. Prognosis and Neuropathologic Correlation of Clinical Subtypes of Parkinson Disease. JAMA Neurol. 2019; 76(4): 470–479.
  51. Fereshtehnejad SM, Romenets SR, Anang JBM, et al. New Clinical Subtypes of Parkinson Disease and Their Longitudinal Progression: A Prospective Cohort Comparison With Other Phenotypes. JAMA Neurol. 2015; 72(8): 863–873.
  52. Erro R, Picillo M, Vitale C, et al. Clinical clusters and dopaminergic dysfunction in de-novo Parkinson disease. Parkinsonism Relat Disord. 2016; 28: 137–140.
  53. Parnetti L, Gaetani L, Eusebi P, et al. CSF and blood biomarkers for Parkinson's disease. Lancet Neurol. 2019; 18(6): 573–586.
  54. Kang JH, Mollenhauer B, Coffey CS, et al. Parkinson’s Progression Marker Initiative, Parkinson’s Progression Marker Initiative. CSF biomarkers associated with disease heterogeneity in early Parkinson's disease: the Parkinson's Progression Markers Initiative study. Acta Neuropathol. 2016; 131(6): 935–949.
  55. Hansson O, Hall S, Ohrfelt A, et al. Levels of cerebrospinal fluid α-synuclein oligomers are increased in Parkinson's disease with dementia and dementia with Lewy bodies compared to Alzheimer's disease. Alzheimers Res Ther. 2014; 6(3): 25.
  56. Park MJ, Cheon SM, Bae HR, et al. Elevated levels of α-synuclein oligomer in the cerebrospinal fluid of drug-naïve patients with Parkinson's disease. J Clin Neurol. 2011; 7(4): 215–222.
  57. Chi X, Yin S, Sun Y, et al. Phosphorylated α-synuclein in Parkinson's disease. Sci Transl Med. 2012; 4(121): 121ra20–1929.
  58. Stewart T, Sossi V, Aasly JO, et al. Phosphorylated α-synuclein in Parkinson's disease: correlation depends on disease severity. Acta Neuropathol Commun. 2015; 3: 7.
  59. Barbour R, Kling K, Anderson JP, et al. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008; 5(2): 55–59.
  60. Concha-Marambio L, Farris CM, Holguin B, et al. Seed Amplification Assay to Diagnose Early Parkinson's and Predict Dopaminergic Deficit Progression. Mov Disord. 2021; 36(10): 2444–2446.
  61. Russo MJ, Orru CD, Concha-Marambio L, et al. High diagnostic performance of independent alpha-synuclein seed amplification assays for detection of early Parkinson's disease. Acta Neuropathol Commun. 2021; 9(1): 179.
  62. Rossi M, Baiardi S, Teunissen CE, et al. Diagnostic Value of the CSF α-Synuclein Real-Time Quaking-Induced Conversion Assay at the Prodromal MCI Stage of Dementia With Lewy Bodies. Neurology. 2021; 97(9): e930–e940.
  63. Singer W, Schmeichel AM, Shahnawaz M, et al. Alpha-Synuclein Oligomers and Neurofilament Light Chain Predict Phenoconversion of Pure Autonomic Failure. Ann Neurol. 2021; 89(6): 1212–1220.
  64. Concha‐Marambio L, Weber S, Farris C, et al. Accurate Detection of α‐Synuclein Seeds in Cerebrospinal Fluid from Isolated Rapid Eye Movement Sleep Behavior Disorder and Patients with Parkinson's Disease in the DeNovo Parkinson (DeNoPa) Cohort. Movement Disorders. 2023; 38(4): 567–578.
  65. Siderowf A, Concha-Marambio L, Lafontant DE, et al. Parkinson's Progression Markers Initiative, Parkinson's Progression Markers Initiative. Assessment of heterogeneity among participants in the Parkinson's Progression Markers Initiative cohort using α-synuclein seed amplification: a cross-sectional study. Lancet Neurol. 2023; 22(5): 407–417.
  66. Manne S, Kondru N, Jin H, et al. Blinded RT-QuIC Analysis of α-Synuclein Biomarker in Skin Tissue From Parkinson's Disease Patients. Mov Disord. 2020; 35(12): 2230–2239.
  67. Mammana A, Baiardi S, Quadalti C, et al. RT-QuIC Detection of Pathological α-Synuclein in Skin Punches of Patients with Lewy Body Disease. Mov Disord. 2021; 36(9): 2173–2177.
  68. De Luca CM, Elia AE, Portaleone SM, et al. Efficient RT-QuIC seeding activity for α-synuclein in olfactory mucosa samples of patients with Parkinson's disease and multiple system atrophy. Transl Neurodegener. 2019; 8: 24.
  69. Manne S, Kondru N, Jin H, et al. α-Synuclein real-time quaking-induced conversion in the submandibular glands of Parkinson's disease patients. Mov Disord. 2020; 35(2): 268–278.
  70. Luan M, Sun Y, Chen J, et al. Diagnostic Value of Salivary Real-Time Quaking-Induced Conversion in Parkinson's Disease and Multiple System Atrophy. Mov Disord. 2022; 37(5): 1059–1063.
  71. Mammana A, Baiardi S, Rossi M, et al. Improving protocols for α-synuclein seed amplification assays: analysis of preanalytical and analytical variables and identification of candidate parameters for seed quantification. Clin Chem Lab Med. 2024; 62(10): 2001–2010.
  72. Bellomo G, De Luca CM, Paoletti FP, et al. α-Synuclein Seed Amplification Assays for Diagnosing Synucleinopathies: The Way Forward. Neurology. 2022; 99(5): 195–205.
  73. Vaughan DP, Fumi R, Theilmann Jensen M, et al. Evaluation of Cerebrospinal Fluid α-Synuclein Seed Amplification Assay in Progressive Supranuclear Palsy and Corticobasal Syndrome. Mov Disord. 2024; 39(12): 2285–2291.
  74. Jack CR, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018; 14(4): 535–562.
  75. Jack CR, Graf A, Burnham SC, et al. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement. 2024; 20(8): 5143–5169.
  76. Petrou M, Dwamena BA, Foerster BR, et al. Amyloid deposition in Parkinson's disease and cognitive impairment: a systematic review. Mov Disord. 2015; 30(7): 928–935.
  77. Zhang X, Gao F, Wang D, et al. Tau Pathology in Parkinson's Disease. Front Neurol. 2018; 9: 809.
  78. Parnetti L, Farotti L, Eusebi P, et al. Differential role of CSF alpha-synuclein species, tau, and Aβ42 in Parkinson's Disease. Front Aging Neurosci. 2014; 6: 53.
  79. Terrelonge M, Marder KS, Weintraub D, et al. CSF β-Amyloid 1-42 Predicts Progression to Cognitive Impairment in Newly Diagnosed Parkinson Disease. J Mol Neurosci. 2016; 58(1): 88–92.
  80. McMillan CT, Wolk DA, McMillan CT, et al. Presence of cerebral amyloid modulates phenotype and pattern of neurodegeneration in early Parkinson's disease. J Neurol Neurosurg Psychiatry. 2016; 87(10): 1112–1122.
  81. Cousins KAQ, Irwin DJ, Tropea TF, et al. Parkinson's Progression Markers Initiative, Parkinson’s Progression Markers Initiative (PPMI), Parkinson's Progression Markers Initiative, Parkinson’s Progression Markers Initiative (PPMI). CSF amyloid {beta} 1-42 predicts cognitive decline in Parkinson disease. Neurology. 2010; 75(12): 1055–1061.
  82. Hall S, Surova Y, Öhrfelt A, et al. Swedish BioFINDER Study, Swedish BioFINDER Study. Longitudinal Measurements of Cerebrospinal Fluid Biomarkers in Parkinson's Disease. Mov Disord. 2016; 31(6): 898–905.
  83. Liu C, Cholerton B, Shi M, et al. Parkinson Study Group DATATOP Investigators, Parkinson Study Group DATATOP Investigators. CSF tau and tau/Aβ42 predict cognitive decline in Parkinson's disease. Parkinsonism Relat Disord. 2015; 21(3): 271–276.
  84. Magdalinou NK, Paterson RW, Schott JM, et al. A panel of nine cerebrospinal fluid biomarkers may identify patients with atypical parkinsonian syndromes. J Neurol Neurosurg Psychiatry. 2015; 86(11): 1240–1247.
  85. Herbert MK, Aerts MB, Beenes M, et al. CSF Neurofilament Light Chain but not FLT3 Ligand Discriminates Parkinsonian Disorders. Front Neurol. 2015; 6: 91.
  86. Holmberg B, Rosengren L, Karlsson JE, et al. Increased cerebrospinal fluid levels of neurofilament protein in progressive supranuclear palsy and multiple-system atrophy compared with Parkinson's disease. Mov Disord. 1998; 13(1): 70–77.
  87. Mollenhauer B, et al. Validation of Serum Neurofilament Light Chain as a Biomarker of Parkinson's Disease Progression. Mov Disord. 2020; 35(11): 1999–2008.
  88. Aamodt W, Waligorska T, Shen J, et al. Neurofilament Light Chain as a Biomarker for Cognitive Decline in Parkinson Disease. Movement Disorders. 2021; 36(12): 2945–2950.
  89. Olsson B, Portelius E, Cullen NC, et al. Association of Cerebrospinal Fluid Neurofilament Light Protein Levels With Cognition in Patients With Dementia, Motor Neuron Disease, and Movement Disorders. JAMA Neurol. 2019; 76(3): 318–325.
  90. Kasuga K, Tokutake T, Ishikawa A, et al. Differential levels of alpha-synuclein, beta-amyloid42 and tau in CSF between patients with dementia with Lewy bodies and Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2010; 81(6): 608–610.
  91. Buhmann C, Magnus T, Choe CU, et al. Blood neurofilament light chain in Parkinson's disease. J Neural Transm (Vienna). 2023; 130(6): 755–762.
  92. Gaetani L, Blennow K, Calabresi P, et al. Neurofilament light chain as a biomarker in neurological disorders. J Neurol Neurosurg Psychiatry. 2019; 90(8): 870–881.
  93. Bartl M, Xylaki M, Bähr M, et al. Evidence for immune system alterations in peripheral biological fluids in Parkinson's disease. Neurobiol Dis. 2022; 170: 105744.
  94. Gerhard A, Pavese N, Hotton G, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson's disease. Neurobiol Dis. 2006; 21(2): 404–412.
  95. Ouchi Y, Yagi S, Yokokura M, et al. Neuroinflammation in the living brain of Parkinson's disease. Parkinsonism Relat Disord. 2009; 15 Suppl 3: S200–S204.
  96. Bonneh-Barkay D, Bissel SJ, Kofler J, et al. Astrocyte and macrophage regulation of YKL-40 expression and cellular response in neuroinflammation. Brain Pathol. 2012; 22(4): 530–546.
  97. Hepp DH, van Wageningen TA, Kuiper KL, et al. Inflammatory Blood Biomarkers Are Associated with Long-Term Clinical Disease Severity in Parkinson's Disease. Int J Mol Sci. 2023; 24(19).
  98. Williams-Gray CH, et al. Serum immune markers and disease progression in an incident Parkinson's disease cohort (ICICLE-PD). Movement Disorders. 2016; 31(7): 995–1003.
  99. Lawton M, Baig F, Toulson G, et al. Blood biomarkers with Parkinson's disease clusters and prognosis: The oxford discovery cohort. Mov Disord. 2020; 35(2): 279–287.
  100. Deng X, Mehta A, Xiao B, et al. Parkinson's disease subtypes: Approaches and clinical implications. Parkinsonism Relat Disord. 2025; 130: 107208.
  101. Belvisi D, Fabbrini A, De Bartolo MI, et al. The Pathophysiological Correlates of Parkinson's Disease Clinical Subtypes. Mov Disord. 2021; 36(2): 370–379.
  102. Filipović SR, Kačar A, Milanović S, et al. Neurophysiological Predictors of Response to Medication in Parkinson's Disease. Front Neurol. 2021; 12: 763911.
  103. Yassine S, Gschwandtner U, Auffret M, et al. Identification of Parkinson's Disease Subtypes from Resting-State Electroencephalography. Mov Disord. 2023; 38(8): 1451–1460.
  104. Braak H, Del Tredici K, Del Tredici K, et al. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. 2003; 24(2): 197–211.
  105. Radziwon J, Sławek J, Radziwon J, et al. Ultrasonographically measured atrophy of vagus nerve in Parkinson's Disease: clinical and pathogenetic insights plus systematic review and meta-analysis. Neurol Neurochir Pol. 2024; 58(5): 471–483.
  106. Liu B, Wanders A, Wirdefeldt K, et al. Vagotomy and Parkinson disease: A Swedish register-based matched-cohort study. Neurology. 2017; 88(21): 1996–2002.
  107. Verstraeten A, Theuns J, Van Broeckhoven C, et al. Progress in unraveling the genetic etiology of Parkinson disease in a genomic era. Trends Genet. 2015; 31(3): 140–149.
  108. Chang D, Nalls MA, Hallgrímsdóttir IB, et al. International Parkinson's Disease Genomics Consortium, 23andMe Research Team, International Parkinson's Disease Genomics Consortium, 23andMe Research Team. A meta-analysis of genome-wide association studies identifies 17 new Parkinson's disease risk loci. Nat Genet. 2017; 49(10): 1511–1516.
  109. Chen-Plotkin A, Zetterberg H, Chen-Plotkin A, et al. Updating Our Definitions of Parkinson’s Disease for a Molecular Age. Journal of Parkinson’s Disease. 2018; 8(s1): S53–S57.
  110. Espay AJ, Brundin P, Lang AE, et al. Precision medicine for disease modification in Parkinson disease. Nat Rev Neurol. 2017; 13(2): 119–126.
  111. Zimmermann M, Gaenslen A, Prahl K, et al. Patient's perception: shorter and more severe prodromal phase in GBA-associated PD. Eur J Neurol. 2019; 26(4): 694–698.
  112. Krohn L, Ruskey JA, Rudakou U, et al. variants in REM sleep behavior disorder: A multicenter study. Neurology. 2020; 95(8): e1008–e1016.
  113. Cilia R, Tunesi S, Marotta G, et al. Survival and dementia in GBA-associated Parkinson's disease: The mutation matters. Ann Neurol. 2016; 80(5): 662–673.
  114. Davis MY, Johnson CO, Leverenz JB, et al. Association of GBA Mutations and the E326K Polymorphism With Motor and Cognitive Progression in Parkinson Disease. JAMA Neurol. 2016; 73(10): 1217–1224.
  115. Caminiti S, Avenali M, Galli A, et al. Combined detrimental effect of male sex and GBA1 variants on cognitive decline in Parkinson’s Disease. .
  116. Pont-Sunyer C, Tolosa E, Caspell-Garcia C, et al. LRRK2 Cohort Consortium, LRRK2 Cohort Consortium. The prodromal phase of leucine-rich repeat kinase 2-associated Parkinson disease: Clinical and imaging Studies. Mov Disord. 2017; 32(5): 726–738.
  117. Mirelman A, Heman T, Yasinovsky K, et al. LRRK2 Ashkenazi Jewish Consortium, LRRK2 Ashkenazi Jewish Consortium. Fall risk and gait in Parkinson's disease: the role of the LRRK2 G2019S mutation. Mov Disord. 2013; 28(12): 1683–1690.
  118. Alcalay RN, Mejia-Santana H, Tang MX, et al. Motor phenotype of LRRK2 G2019S carriers in early-onset Parkinson disease. Arch Neurol. 2009; 66(12): 1517–1522.
  119. Lücking CB, Dürr A, Bonifati V, et al. French Parkinson's Disease Genetics Study Group, European Consortium on Genetic Susceptibility in Parkinson's Disease, French Parkinson's Disease Genetics Study Group, European Consortium on Genetic Susceptibility in Parkinson's Disease. Association between early-onset Parkinson's disease and mutations in the parkin gene. N Engl J Med. 2000; 342(21): 1560–1567.
  120. Nybø CJ, Gustavsson EK, Farrer MJ, et al. Neuropathological findings in PINK1-associated Parkinson's disease. Parkinsonism Relat Disord. 2020; 78: 105–108.
  121. Kalia LV, Lang AE, Hazrati LN, et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol. 2015; 72(1): 100–105.
  122. Beyer K, Domingo-Sàbat M, Ariza A, et al. Molecular pathology of Lewy body diseases. Int J Mol Sci. 2009; 10(3): 724–745.
  123. Ibáñez P, Lesage S, Janin S, et al. French Parkinson's Disease Genetics Study Group, French Parkinson's Disease Genetics Study Group. Alpha-synuclein gene rearrangements in dominantly inherited parkinsonism: frequency, phenotype, and mechanisms. Arch Neurol. 2009; 66(1): 102–108.
  124. Lesage S, Anheim M, Letournel F, et al. French Parkinson's Disease Genetics Study Group, French Parkinson's Disease Genetics Study Group. G51D α-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol. 2013; 73(4): 459–471.
  125. 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.
  126. Cooper CA, Jain N, Gallagher MD, et al. Common variant rs356182 near SNCA defines a Parkinson's disease endophenotype. Ann Clin Transl Neurol. 2017; 4(1): 15–25.
  127. Marsili L, Vizcarra JA, Sturchio A, et al. When does postural instability appear in monogenic parkinsonisms? An individual-patient meta-analysis. J Neurol. 2021; 268(9): 3203–3211.
  128. McCann H, Stevens CH, Cartwright H, et al. α-Synucleinopathy phenotypes. Parkinsonism Relat. Disord. 2014; 20(Suppl 1): S62–S67.
  129. Compta Y, Parkkinen L, O'Sullivan SS, et al. Lewy- and Alzheimer-type pathologies in Parkinson's disease dementia: which is more important? Brain. 2011; 134(Pt 5): 1493–1505.
  130. Bassil F, Brown HJ, Pattabhiraman S, et al. Amyloid-Beta (Aβ) Plaques Promote Seeding and Spreading of Alpha-Synuclein and Tau in a Mouse Model of Lewy Body Disorders with Aβ Pathology. Neuron. 2020; 105(2): 260–275.e6.
  131. Menšíková K, Matěj R, Colosimo C, et al. Lewy body disease or diseases with Lewy bodies? npj Parkinson's Disease. 2022; 8(1).
  132. Wardlaw JM, Smith EE, Biessels GJ, et al. STandards for ReportIng Vascular changes on nEuroimaging (STRIVE v1). Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013; 12(8): 822–838.
  133. Yu P, Venkat P, Chopp M, et al. Deficiency of tPA Exacerbates White Matter Damage, Neuroinflammation, Glymphatic Dysfunction and Cognitive Dysfunction in Aging Mice. Aging Dis. 2019; 10(4): 770–783.
  134. Zhu S, Wei X, Yang X, et al. Plasma Lipoprotein-associated Phospholipase A2 and Superoxide Dismutase are Independent Predicators of Cognitive Impairment in Cerebral Small Vessel Disease Patients: Diagnosis and Assessment. Aging Dis. 2019; 10(4): 834–846.
  135. Sunwoo MK, Jeon S, Ham JH, et al. The burden of white matter hyperintensities is a predictor of progressive mild cognitive impairment in patients with Parkinson's disease. Eur J Neurol. 2014; 21(6): 922–e50.
  136. Jones JD, Tanner JJ, Okun M, et al. Are Parkinson's Patients More Vulnerable to the Effects of Cardiovascular Risk: A Neuroimaging and Neuropsychological Study. J Int Neuropsychol Soc. 2017; 23(4): 322–331.
  137. Hanning U, Teuber A, Lang E, et al. White matter hyperintensities are not associated with cognitive decline in early Parkinson's disease - The DeNoPa cohort. Parkinsonism Relat Disord. 2019; 69: 61–67.
  138. Liu H, Deng B, Xie F, et al. The influence of white matter hyperintensity on cognitive impairment in Parkinson's disease. Ann Clin Transl Neurol. 2021; 8(9): 1917–1934.
  139. Carvalho de Abreu DC, Pieruccini-Faria F, Son S, et al. Is white matter hyperintensity burden associated with cognitive and motor impairment in patients with parkinson's disease? A systematic review and meta-analysis. Neurosci Biobehav Rev. 2024; 161: 105677.
  140. Amor S, Woodroofe MN, Amor S, et al. Innate and adaptive immune responses in neurodegeneration and repair. Immunology. 2014; 141(3): 287–291.
  141. Heneka M, Kummer M, Latz E, et al. Innate immune activation in neurodegenerative disease. Nature Reviews Immunology. 2014; 14(7): 463–477.
  142. Caggiu E, Arru G, Hosseini S, et al. Inflammation, Infectious Triggers, and Parkinson's Disease. Front Neurol. 2019; 10: 122.
  143. Adler CH, Beach TG, Adler CH, et al. Neuropathological basis of nonmotor manifestations of Parkinson's disease. Mov Disord. 2016; 31(8): 1114–1119.
  144. Borghammer P, Horsager J, Andersen K, et al. Neuropathological evidence of body-first vs. brain-first Lewy body disease. Neurobiol Dis. 2021; 161: 105557.
  145. Borghammer P, Berge NV, Borghammer P, et al. Brain-First versus Gut-First Parkinson’s Disease: A Hypothesis. Journal of Parkinson’s Disease. 2019; 9(s2): S281–S295.
  146. Dong S, Shen Bo, Jiang Xu, et al. Comparison of vagus nerve cross-sectional area between brain-first and body-first Parkinson's disease. NPJ Parkinsons Dis. 2024; 10(1): 231.
  147. Burbulla LF, Song P, Mazzulli JR, et al. Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson's disease. Science. 2017; 357(6357): 1255–1261.
  148. Espay AJ, Vizcarra JA, Marsili L, et al. Revisiting protein aggregation as pathogenic in sporadic Parkinson and Alzheimer diseases. Neurology. 2019; 92(7): 329–337.
  149. Espay AJ, Lees AJ, Espay AJ, et al. Loss of monomeric alpha-synuclein (synucleinopenia) and the origin of Parkinson's disease. Parkinsonism Relat Disord. 2024; 122: 106077.
  150. Markopoulou K, Biernacka JM, Armasu SM, et al. Does α-synuclein have a dual and opposing effect in preclinical vs. clinical Parkinson's disease? Parkinsonism Relat Disord. 2014; 20(6): 584–9; discussion 584.
  151. Gorbatyuk O, Li S, Nash K, et al. In Vivo RNAi-Mediated α-Synuclein Silencing Induces Nigrostriatal Degeneration. Molecular Therapy. 2010; 18(8): 1450–1457.
  152. Collier TJ, Redmond DE, Steece-Collier K, et al. Is Alpha-Synuclein Loss-of-Function a Contributor to Parkinsonian Pathology? Evidence from Non-human Primates. Front Neurosci. 2016; 10: 12.
  153. Galkin M, Topcheva O, Priss A, et al. Dopamine-Induced Oligomers of α-Synuclein Inhibit Amyloid Fibril Growth and Show No Toxicity. ACS Chem Neurosci. 2023; 14(11): 2027–2034.
  154. Abanto J, Dwivedi AK, Imbimbo BP, et al. Increases in amyloid-β42 slow cognitive and clinical decline in Alzheimer's disease trials. Brain. 2024; 147(10): 3513–3521.
  155. Greffard S, Verny M, Bonnet AM, et al. Motor score of the Unified Parkinson Disease Rating Scale as a good predictor of Lewy body-associated neuronal loss in the substantia nigra. Arch Neurol. 2006; 63(4): 584–588.