• Aucun résultat trouvé

The sleep-wake abnormalities in DLB challenge the study of sleep in this disease

6.6. Future directions

Here I propose the questions that go beyond this thesis and that will need future research. To validate the three clinical subtypes of DLB a study of the

158

ferences that may relate to the different phenotypes. Most of the neuropatho-logical studies addressing the clinical variety in DLB centred in the presence of AD copathology (65,93). To properly understand the contribution of AD copathology and other copathologies, as TDP-43 aggregates, in these clusters, neuropathological studies aimed to quantify these copathologies in the differ-ent cortical and subcortical brain areas in DLB patidiffer-ents with extensive and de-tailed clinical reports from the stage of MCI are needed. On the other hand, a neuropathological study of DLB patients subdivided according to the proposed clinical subtypes would also allow the study of the cellular and molecular bi-ology beyond the Lewy body related pathbi-ology and AD copathbi-ology that may underlie the clinical heterogeneity in DLB. New techniques assessing the dif-ferent pointed α-synuclein strains and specific seeding capacity as well as their relationship with tau aggregation should be applied in the research of the clin-ical heterogeneity, not merely along synucleinopathies, but within DLB itself.

The development and application of biochemical and neuroimaging bio-markers is highly relevant to disentangle the biological basis supporting the clinical heterogeneity in DLB. These markers would be essential in the detection of the prodromal phase of DLB. The recognition of prodromal DLB is highly relevant considering future disease-modifying drugs that may be more effective in this phase of the disease. The study of DLB patients from their earlier stages would allow longitudinal studies to investigate the evolution of the biological markers together with the different clinical features. Although iRBD patients are amenable to longitudinal follow-up (107), to avoid biases in extrapolating features typical from those patients to others without RBD and that may present very different characteristics (107), such studies should not be limited to this group.

Furthermore, studies aimed at exploring the relationship between the brain structure and function by MRI and the three clinical subtypes presented in

Chapter III are needed. This study could be complemented with functional neuroimaging based on PET scan, which would also be informative of the underlying damage to neural networks underlying the proposed clinical sub-types. New neuroimaging technics as amyloid- and tau- PET could also be useful to study copathology and pathological synergies in DLB. To obtain this objective, larger cohorts with appropriate and quantified neuroimaging examinations and a complete and prospective clinical assessment from the stage of MCI, are required. Since DLB has a lower prevalence and incidence than AD, and is frequently underdiagnosed, the creation of large, sufficiently powered cohorts to perform different studies is challenging. To solve this problem, it is crucial to create and enlarge common and public repositories based on multicentric and international recruitment following the example of the Alzheimer Disease Neuroimaging Initiative (ADNI). With this aim, sev-eral initiatives such as the European DLB consortium in Europe or the De-mentia with Lewy bodies Consortium in United States have started recently.

From a clinical point of view, the importance of the V-PSG in the study of the sleep disturbances in DLB has been highlighted in Chapter IV. Larger studies with a systematic V-PSG analysis aimed to establish new descriptive criteria for the assessment of sleep in demented patients, specifically in DLB, are needed. These criteria would help in the harmonization of sleep stud-ies focused on better understanding the sleep pathology in DLB. Improved systematic and specific studies of sleep disturbances in DLB in specialized clinical centres improves the clinical management of these patients. Given the limited access and the difficulties associated with performing a V-PSG on demented patients, the search for other early markers that can identi-fy candidates who would benefit from V-PSG would be of help in clinical practice. How the different sleep pathologies in DLB correlate with specific phenotypes, biomarker profile and underlying neuropathology should also

160

Finally, neuropathological and experimental studies using cellular and ani-mal models and aimed at understanding the role of glial activation in DLB should be undertaken. Specifically, in vitro and in vivo studies of how glial activity influences tau pathology in synucleinopathies should be performed.

Besides basic experimental studies, we should consider the importance of neuropathological studies specifically aimed to better understand the role of glial cells in the human DLB brain. Specific studies aimed at unravelling the role of glial cells in the beginning and the evolution of the DLB are needed.

7. CONCLUSIONS

Three subtypes of DLB that differ in their clini-cal manifestations and progression can be distin-guished based on the predominant symptoms dur-ing the prodromal phase of the disease: cognitive-, psychotic- and parkinsonism-predominant.

Sleep-wake disturbances are common and com-plex in patients with DLB. V-PSG is essential to di-agnose RBD and rule out mimics.

DLB and AD show different patterns of glial

acti-vation markers in CSF. sTREM2, PRGN and YKL-40

levels in CSF are normal in prodromal DLB and DLB

in the absence of AD co-pathology. YKL40 levels are

only elevated in those DLB patients with abnormal

core AD biomarkers.

8. REFERENCES

1. Holdorff B. Friedrich Heinrich Lewy (1885?1950) and His Work. J Hist Neurosci. 2002;

2. Woodard JS. Concentric Hyaline Inclusion Body Formation in Men-tal Disease Analysis of Twenty-Seven Cases†. J Neuropathol Exp Neurol [Internet]. 1962;21(3):442–9. Available from: https://doi.

org/10.1097/00005072-196207000-00012

3. McKeith IG, Perry RH, Fairbairn AF, Jabeen S, Perry EK. Operational criteria for senile dementia of lewy body type (SDLT). Psychol Med.

1992;22(4):911–22.

4. Kosaka K, Oyanagi S, Matsushita M, Hori A, Iwase S. Presenile de-mentia with Alzheimer-, Pick- and Lewy-body changes. Acta Neuro-pathol. 1976;36(3):221–33.

5. Kosaka K. Lewy bodies in cerebral cortex. Report of three cases. Acta Neuropathol. 1978;42(2):127–34.

6. Forno LS, Barbour PJ, Norville RL. Presenile Dementia with Lewy Bod-ies and Neurofibrillary Tangles. Arch Neurol. 1978;35(12):818–22.

7. Gibb WR, Luthert PJ, Janota I, Lantos PL. Cortical Lewy body demen-tia: clinical features and classification. J Neurol Neurosurg Psychiatry [Internet]. 1989 Feb 1;52(2):185–92. Available from: http://jnnp.

bmj.com/cgi/doi/10.1136/jnnp.52.2.185

8. McKeith IG, Galasko D, Kosaka K, Perry EK, Dickson DW, Hansen LA, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): Report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113–24.

9. Mckeith I, Mintzer J, Aarsland D, Burn D, Chiu H, Cohen-mansfield J, et al. McKeith et al 2004. 44(0):19–28.

10. Spillantini GM, Schmidt ML, Lee VM-Y, Trojanowski JQ, Jakes R, Goedert M. Ȋ-Synuclein in Lewy bodies Endogenous proviruses as “ mementos ”? Nature. 1997;

11. Mckeith IG, Dickson DW, Lowe J, Emre M, Brien JTO, Feldman H, et al.

Diagnosis and management of dementia with Lewy bodies. Third report of the DLB consortium. Neurology. 2005;65(September 2004):1863–72.

12. Sachdev PS, Blacker D, Blazer DG, Ganguli M, Jeste D V., Paulsen JS,

166

Rev Neurol. 2014;10(11):634–42.

13. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor J-P, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies. Neu-rology [Internet]. 2017 Jul 4;89(1):88–100. Available from: http://

www.neurology.org/lookup/doi/10.1212/WNL.0000000000004058 14. Ferman TJ, Smith GE, Boeve BF, Graff-Radford NR, Lucas JA,

Knop-man DS, et al. Neuropsychological Differentiation of Dementia with Lewy Bodies from Normal Aging and Alzheimer’s Disease. Clin Neu-ropsychol [Internet]. 2006 Dec 1;20(4):623–36. Available from:

https://doi.org/10.1080/13854040500376831

15. Walker Z, Possin KL, Boeve BF, Aarsland D. Lewy body dementias. Lan-cet (London, England) [Internet]. 2015 Oct 24;386(10004):1683–97.

Available from: https://www.ncbi.nlm.nih.gov/pubmed/26595642 16. Hamilton JM, Landy KM, Salmon DP, Hansen LA, Masliah E, Galasko

D. Early Visuospatial Deficits Predict the Occurrence of Visual Halluci-nations in Autopsy-Confirmed Dementia With Lewy Bodies. Am J Ger-iatr PsychGer-iatry [Internet]. 2012 Sep;20(9):773–81. Available from:

https://linkinghub.elsevier.com/retrieve/pii/S1064748112619940 17. Yokoi K, Nishio Y, Uchiyama M, Shimomura T, Iizuka O, Mori E.

Hal-lucinators find meaning in noises: Pareidolic illusions in dementia with Lewy bodies. Neuropsychologia. 2014;

18. Fereshtehnejad S-M, Yao C, Pelletier A, Montplaisir JY, Gagnon J-F, Pos-tuma RB. Evolution of prodromal Parkinson’s disease and dementia with Lewy bodies: a prospective study. Brain. 2019;142(7):2051–67.

19. Walker MP, Ayre GA, Cummings JL, Wesnes K, McKeith IG, O’Brien JT, et al. Quantifying fluctuation in dementia with Lewy bodies, Alzheim-er’s disease, and vascular dementia. Neurology [Internet]. 2000 Apr 25;54(8):1616–25. Available from: http://www.neurology.org/cgi/

doi/10.1212/WNL.54.8.1616

20. Walker MP, Ayre GA, Cummings JL, Wesnes K, McKeith IG, O’Brien JT, et al. The Clinician Assessment of Fluctuation and the One Day Fluctuation Assessment Scale. Br J Psychiatry [Internet].

2018/01/02. 2000 Sep 2;177(3):252–6. Available from: https://

www.cambridge.org/core/article/clinician-assessment-of-fluc- tuation-and-the-one-day-fluctuation-assessment-scale/3551BC-D9A7C1039BF7170CC63ADE5F5D

21. Ferman TJ, Boeve BF, Smith GE, Lin S-C, Silber MH, Pedraza O, et al.

Inclusion of RBD improves the diagnostic classification of dementia

with Lewy bodies. Neurology [Internet]. 2011 Aug 30;77(9):875–

82. Available from: http://www.neurology.org/cgi/doi/10.1212/

WNL.0b013e31822c9148

22. Postuma RB, Gagnon JF, Vendette M, Fantini ML, Mas-sicotte-Marquez J, Montplaisir J., et al. Quantifying the risk of neu-rodegenerative disease in idiopathic REM. Neurology [Internet].

2009;72(15):1296–300. Available from: http://www.scopus.com/

inward/record.url?eid=2-s2.0-65349168853&partnerID=tZOtx- 3y1%5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?ar-tid=2828948&tool=pmcentrez&rendertype=abstract

23. Iranzo A, Fernández-Arcos A, Tolosa E, Serradell M, Molinuevo JL, Valldeoriola F, et al. Neurodegenerative Disorder Risk in Idiopathic REM Sleep Behavior Disorder: Study in 174 Patients. Toft M, edi-tor. PLoS One [Internet]. 2014 Feb 26;9(2):e89741. Available from:

https://dx.plos.org/10.1371/journal.pone.0089741

24. McKeith I, O’Brien J, Walker Z, Tatsch K, Booij J, Darcourt J, et al.

Sensitivity and specificity of dopamine transporter imaging with 123I-FP-CIT SPECT in dementia with Lewy bodies: a phase III, multicentre study. Lancet Neurol [Internet]. 2007 Apr;6(4):305–

13. Available from: https://linkinghub.elsevier.com/retrieve/pii/

S1474442207700571

25. Thomas AJ, Donaghy P, Roberts G, Colloby SJ, Barnett NA, Petrides G, et al. Diagnostic accuracy of dopaminergic imaging in prodro-mal dementia with Lewy bodies. Psychol Med [Internet]. 2019 Feb 25;49(3):396–402. Available from: https://www.cambridge.org/

core/product/identifier/S0033291718000995/type/journal_article 26. Kiferle L, Ceravolo R, Giuntini M, Linsalata G, Puccini G, Volterrani

D, et al. Parkinsonism and Related Disorders Caudate dopaminergic denervation and visual hallucinations : Evidence from a 123 I-FP-CIT SPECT study. Park Relat Disord [Internet]. 2014;4–8. Available from:

http://dx.doi.org/10.1016/j.parkreldis.2014.04.006

27. Colloby SJ, McParland S, O’Brien JT, Attems J. Neuropathological correlates of dopaminergic imaging in Alzheimer’s disease and Lewy body dementias. Brain [Internet]. 2012 Sep 1;135(9):2798–808.

Available from: https://academic.oup.com/brain/article-lookup/

doi/10.1093/brain/aws211

28. Tiraboschi P, Corso A, Guerra UP, Nobili F, Piccardo A, Calcagni ML, et al. 123 I-2β-carbomethoxy-3β-(4-iodophenyl)- N -(3-fluoropropyl)

168

I-metaiodobenzylguanidine myocardial scintigraphy in differentiat-ing dementia with lewy bodies from other dementias: A comparative s. Ann Neurol [Internet]. 2016 Sep;80(3):368–78. Available from:

http://doi.wiley.com/10.1002/ana.24717

29. Minoshima S, Foster NL, Sima AAF, Frey KA, Albin RL, Kuhl DE.

Alzheimer’s disease versus dementia with Lewy bodies: Cere-bral metabolic distinction with autopsy confirmation. Ann Neurol.

2001;50(3):358–65.

30. Lobotesis K, Fenwick JD, Phipps A, Ryman A, Swann A, Ballard C, et al. Occipital hypoperfusion on SPECT in dementia with Lewy bodies but not AD. Neurology [Internet]. 2001 Mar 13;56(5):643 LP – 649.

Available from: http://n.neurology.org/content/56/5/643.abstract 31. O’Brien JT, Firbank MJ, Davison C, Barnett N, Bamford C, Donaldson C,

et al. 18F-FDG PET and Perfusion SPECT in the Diagnosis of Alzheim-er and Lewy Body Dementias. J Nucl Med. 2014;55(12):1959–65.

32. Lim SM, Katsifis A, Villemagne VL, Best R, Jones G, Saling M, et al.

The 18F-FDG PET Cingulate Island Sign and Comparison to 123I- -CIT SPECT for Diagnosis of Dementia with Lewy Bodies. J Nucl Med.

2009;50(10):1638–45.

33. Graff-Radford J, Murray ME, Lowe VJ, Boeve BF, Ferman TJ, Przybel-ski Timothy G Lesnick Matthew L Senjem Jeffrey L Gunter Glenn E Smith SA, et al. Dementia with Lewy bodies Basis of cingulate island sign From the Departments of Neurology (. 2014;801–9.

34. Harper L, Fumagalli GG, Barkhof F, Scheltens P, O’Brien JT, Bouwman F, et al. MRI visual rating scales in the diagnosis of dementia: Evaluation in 184 post-mortem confirmed cases. Brain. 2016;139(4):1211–25.

35. Llorens F, Schmitz M, Gloeckner SF, Kaerst L, Hermann P, Schmidt C, et al. Increased albumin CSF/serum ratio in dementia with Lewy bodies. J Neurol Sci [Internet]. 2015 Nov;358(1–2):398–403. Avail-able from: http://dx.doi.org/10.1016/j.jns.2015.10.011

36. Sohma H, Imai S, Takei N, Honda H, Matsumoto K, Utsumi K, et al.

Evaluation of annexin A5 as a biomarker for Alzheimer’s disease and dementia with lewy bodies [Internet]. Vol. 5, Frontiers in Aging Neuroscience . 2013. p. 15. Available from: https://www.frontiersin.

org/article/10.3389/fnagi.2013.00015

37. Maetzler W, Tian Y, Baur SM, Gauger T, Odoj B, Schmid B, et al. Se-rum and Cerebrospinal Fluid Levels of Transthyretin in Lewy Body Disorders with and without Dementia. PLoS One. 2012;7(10):1–6.

38. Nielsen HM, Londos E, Minthon L, Janciauskiene SM. Soluble ad-hesion molecules and angiotensin-converting enzyme in dementia.

Neurobiol Dis. 2007;26(1):27–35.

39. Parnetti L, Paciotti S, Farotti L, Bellomo G, Sepe FN, Eusebi P. Par-kinson’s and Lewy body dementia CSF biomarkers. Clin Chim Acta [Internet]. 2019;495(April):318–25. Available from: https://doi.

org/10.1016/j.cca.2019.04.078

40. Ottolini D, Calí T, Szabò I, Brini M. Alpha-synuclein at the intracellu-lar and the extracelluintracellu-lar side: functional and dysfunctional implica-tions. Biol Chem [Internet]. 2017 Jan 1;398(1):77. Available from:

https://www.degruyter.com/view/j/bchm.2017.398.issue-1/hsz-2016-0201/hsz-2016-0201.xml

41. Wennström M, Surova Y, Hall S, Nilsson C, Minthon L, Boström F, et al. Low CSF Levels of Both α-Synuclein and the α-Synuclein Cleaving Enzyme Neurosin in Patients with Synucleinopathy. Ginsberg SD, ed-itor. PLoS One [Internet]. 2013 Jan 8;8(1):e53250. Available from:

https://dx.plos.org/10.1371/journal.pone.0053250

42. Schmitz M, Villar-Piqué A, Llorens F, Gmitterová K, Hermann P, Varg-es D, et al. Cerebrospinal Fluid Total and Phosphorylated α-Synuclein in Patients with Creutzfeldt–Jakob Disease and Synucleinopathy. Mol Neurobiol [Internet]. 2019;56(5):3476–83. Available from: https://

doi.org/10.1007/s12035-018-1313-4

43. Gao L, Tang H, Nie K, Wang L, Zhao J, Gan R, et al. Cerebrospinal flu-id alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: a systematic review and meta-analysis. Int J Neurosci [Internet]. 2015 Sep 2;125(9):645–54. Available from: https://doi.org/10.3109/002 07454.2014.961454

44. Førland MG, Tysnes O, Aarsland D, Maple-Grødem J, Pedersen KF, Alves G, et al. The value of cerebrospinal fluid α-synuclein and the tau/α-synuclein ratio for diagnosis of neurodegenerative disorders with Lewy pathology. Eur J Neurol [Internet]. 2019;ene.14032.

Available from: https://onlinelibrary.wiley.com/doi/abs/10.1111/

ene.14032

45. Hansson O, Hall S, Öhrfelt A, Zetterberg H, Blennow K, Minthon L, 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. Alzheimer’s Res Ther. 2014;6(1):4–9.

170

as potential CSF biomarkers for Parkinson’s disease. Mol Neurode-gener [Internet]. 2016;11(1):1–15. Available from: http://dx.doi.

org/10.1186/s13024-016-0072-9

47. Chiasserini D, Biscetti L, Eusebi P, Salvadori N, Frattini G, Simoni S, et al. Differential role of CSF fatty acid binding protein 3, α-synuclein, and Alzheimer’s disease core biomarkers in Lewy body disorders and Alzheimer’s dementia. Alzheimer’s Res Ther. 2017;9(1):1–12.

48. Lemstra AW, De Beer MH, Teunissen CE, Schreuder C, Scheltens P, Van Der Flier WM, et al. Concomitant AD pathology affects clinical manifestation and survival in dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2017;88(2):113–8.

49. J. Irwin D, I. Hurtig H. The Contribution of Tau, Amyloid-Beta and Alpha-Synuclein Pathology to Dementia in Lewy Body Disorders. J Alzheimer’s Dis Park [Internet]. 2018;08(04). Available from: http://

www.ncbi.nlm.nih.gov/pubmed/30473927%0Ahttp://www.pub-medcentral.nih.gov/articlerender.fcgi?artid=PMC6248323

50. Donaghy PC, McKeith IG. The clinical characteristics of dementia with Lewy bodies and a consideration of prodromal diagnosis. Alzheimers Res Ther [Internet]. 2014;6(4):46. Available from: https://doi.

org/10.1186/alzrt274

51. Desplats P, Lee H-J, Bae E-J, Patrick C, Rockenstein E, Crews L, et al.

Inclusion formation and neuronal cell death through neuron-to-neuron transmission of -synuclein. Proc Natl Acad Sci. 2009;106(31):13010–5.

52. Colom-Cadena M, Pegueroles J, Herrmann AG, Henstridge CM, Muñoz L, Querol-Vilaseca M, et al. Synaptic phosphorylated α-syn-uclein in dementia with Lewy bodies. Brain [Internet]. 2017 Dec 1;140(12):3204–14. Available from: https://www.ncbi.nlm.nih.gov/

pubmed/29177427

53. Hepp DH, Vergoossen DLE, Huisman E, Lemstra AW, Bank NB, Ber-endse HW, et al. Distribution and Load of Amyloid-β Pathology in Parkinson Disease and Dementia with Lewy Bodies. J Neuropathol Exp Neurol [Internet]. 2016 Aug 11;75(10):936–45. Available from:

https://doi.org/10.1093/jnen/nlw070

54. Irwin DJ, Xie SX, Coughlin D, Nevler N, Akhtar RS, McMillan CT, et al. CSF tau and β-amyloid predict cerebral synucleinopathy in autop-sied Lewy body disorders. Neurology [Internet]. 2018/02/21. 2018 Mar 20;90(12):e1038–46. Available from: https://www.ncbi.nlm.

nih.gov/pubmed/29467305

55. Dugger BN, Adler CH, Shill HA, Caviness J, Jacobson S, Driver-Dunck-ley E, et al. Concomitant pathologies among a spectrum of parkinso-nian disorders. Parkinsonism Relat Disord [Internet]. 2014/02/22.

2014 May;20(5):525–9. Available from: https://www.ncbi.nlm.nih.

gov/pubmed/24637124

56. Ossenkoppele R, Jansen WJ, Rabinovici GD, Knol DL, van der Flier WM, van Berckel BNM, et al. Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. JAMA [Internet]. 2015 May 19;313(19):1939–49. Available from: https://www.ncbi.nlm.nih.

gov/pubmed/25988463

57. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science (80- ) [Internet]. 1993 Aug 13;261(5123):921 LP – 923. Available from:

http://science.sciencemag.org/content/261/5123/921.abstract

58. Saeed U, Mirza SS, MacIntosh BJ, Herrmann N, Keith J, Ramirez J, et al. APOE-ε4 associates with hippocampal volume, learning, and memory across the spectrum of Alzheimer’s disease and dementia with Lewy bodies. Alzheimer’s Dement [Internet]. 2018;14(9):1137–

47. Available from: https://doi.org/10.1016/j.jalz.2018.04.005 59. Dickson DW, Heckman MG, Murray ME, Soto AI, Walton RL, Diehl

NN, et al. <em>APOE</em> ε4 is associated with severity of Lewy body pathology independent of Alzheimer pathology. Neu-rology [Internet]. 2018 Sep 18;91(12):e1182 LP-e1195. Available from: http://n.neurology.org/content/91/12/e1182.abstract

60. Halliday GM, Holton JL, Revesz T, Dickson DW. Neuropathology un-derlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011;187–204.

61. Tiraboschi P, Attems J, Thomas A, Brown A, Jaros E, Lett DJ, et al.

Clinicians’ ability to diagnose dementia with Lewy bodies is not af-fected by -amyloid load. Neurology [Internet]. 2014/12/31. 2015 Feb 3;84(5):496–9. Available from: https://www.ncbi.nlm.nih.gov/

pubmed/25552579

62. Selvackadunco S, Langford K, Shah Z, Hurley S, Bodi I, King A, et al. Comparison of clinical and neuropathological diagnoses of neu-rodegenerative diseases in two centres from the Brains for Demen-tia Research (BDR) cohort. J Neural Transm [Internet]. 2019 Mar 7;126(3):327–37. Available from: http://dx.doi.org/10.1007/

172

63. Nelson PT, Abner EL, Patel E, Anderson S, Wilcock DM, Kryscio RJ, et al. The Amygdala as a Locus of Pathologic Misfolding in Neuro-degenerative Diseases. J Neuropathol Exp Neurol [Internet]. 2018 Jan 1;77(1):2–20. Available from: https://www.ncbi.nlm.nih.gov/

pubmed/29186501

64. Colom-Cadena M, Gelpi E, Charif S, Belbin O, Blesa R, Martí MJ, et al. Confluence of α-Synuclein, Tau, and β-Amyloid Pathologies in De-mentia With Lewy Bodies. J Neuropathol Exp Neurol [Internet]. 2013 Dec 1;72(12):1203–12. Available from: https://doi.org/10.1097/

NEN.0000000000000018

65. Coughlin D, Xie SX, Liang M, Williams A, Peterson C, Weintraub D, et al. Cognitive and Pathological Influences of Tau Pathology in Lewy Body Disorders. Ann Neurol [Internet]. 2019/01/07. 2019 Feb;85(2):259–71. Available from: https://www.ncbi.nlm.nih.gov/

pubmed/30549331

66. Badiola N, de Oliveira RM, Herrera F, Guardia-Laguarta C, Gonçalves SA, Pera M, et al. Tau enhances α-synuclein aggregation and toxicity in cellular models of synucleinopathy. PLoS One. 2011;6(10):1–9.

67. Ono K, Takahashi R, Ikeda T, Yamada M. Cross-seeding effects of am-yloid β-protein and α-synuclein. J Neurochem [Internet]. 2012 Sep 1;122(5):883–90. Available from: https://doi.org/10.1111/j.1471-4159.2012.07847.x

68. Arnaoutoglou NA, O’Brien JT, Underwood BR. Dementia with Lewy bodies — from scientific knowledge to clinical insights. Nat Rev Neu-rol [Internet]. 2019 Feb 17;15(2):103–12. Available from: http://

www.nature.com/articles/s41582-018-0107-7

69. Orme T, Guerreiro R, Bras J. The Genetics of Dementia with Lewy Bodies: Current Understanding and Future Directions. Curr Neurol Neurosci Rep. 2018;18(10).

70. Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schnei-der JA, et al. APOE ε4 increases risk for dementia in pure synuclein-opathies. JAMA Neurol [Internet]. 2013 Feb;70(2):223–8. Available from: https://www.ncbi.nlm.nih.gov/pubmed/23407718

71. Keogh MJ, Kurzawa-Akanbi M, Griffin H, Douroudis K, Ayers KL, Hussein RI, et al. Exome sequencing in dementia with Lewy bodies.

Transl Psychiatry [Internet]. 2016 Feb 2;6(2):e728–e728. Available from: https://www.ncbi.nlm.nih.gov/pubmed/26836416

72. Nielsen AS, Ravid R, Kamphorst W, Jørgensen OS. Apolipoprotein E

ε4 in an autopsy series of various dementing disorders. J Alzheimer’s Dis. 2003;5(2):119–25.

73. Ibáñez P, Bonnet AM, Débarges B, Lohmann E, Tison F, Pollak P, et al.

Causal relation between α-synuclein gene duplication and familial Parkinson’s disease. Lancet. 2004;364(9440):1169–71.

74. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kacher-gus J, et al. α-Synuclein Locus Triplication Causes Parkinson's Disease. Science (80- ) [Internet]. 2003 Oct 31;302(5646):841 LP – 841. Available from: http://science.sciencemag.org/con-tent/302/5646/841.abstract

75. Bras J, Guerreiro R, Darwent L, Parkkinen L, Ansorge O, Escott-Price V, et al. Genetic analysis implicates APOE, SNCA and suggests lysoso-mal dysfunction in the etiology of dementia with Lewy bodies. Hum Mol Genet [Internet]. 2014/06/27. 2014 Dec 1;23(23):6139–46.

Available from: https://www.ncbi.nlm.nih.gov/pubmed/24973356 76. Funahashi Y, Yoshino Y, Yamazaki K, Mori Y, Mori T, Ozaki Y, et al.

DNA methylation changes at SNCA intron 1 in patients with demen-tia with Lewy bodies. Psychiatry Clin Neurosci [Internet]. 2017 Jan 1;71(1):28–35. Available from: https://doi.org/10.1111/pcn.12462 77. Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G,

DNA methylation changes at SNCA intron 1 in patients with demen-tia with Lewy bodies. Psychiatry Clin Neurosci [Internet]. 2017 Jan 1;71(1):28–35. Available from: https://doi.org/10.1111/pcn.12462 77. Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G,