AccScience Publishing / AN / Volume 2 / Issue 1 / DOI: 10.36922/an.326
Cite this article
91
Download
1300
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
ORIGINAL RESEARCH ARTICLE

Alpha-synuclein at the interface between depression and neurodegeneration: Evidence from epidemiological and genetic data

Ravi Philip Rajkumar*
Show Less
1 Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
Advanced Neurology 2023, 2(1), 326 https://doi.org/10.36922/an.326
Submitted: 16 January 2023 | Accepted: 21 February 2023 | Published: 6 March 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Parkinson’s disease (PD) and Alzheimer’s disease (AlzD) are the two most common neurodegenerative disorders. Although these two disorders differ in terms of their underlying pathophysiology, clinical features, and course, there is a certain degree of overlap between them. This overlap may be partly related to alpha-synuclein (α-synuclein)-mediated neuropathological changes. Recent evidence has found that depression is associated with increased subsequent risk of both these neurological disorders and α-synuclein may also play a pathogenic role in depression. In the current study, epidemiological, population genetic, and environmental exposure data were examined in relation to the estimated prevalence of depressive disorders, PD, and AlzD using a cross-sectional, country-level analysis. The results showed a significant relationship between depressive disorders and neurodegenerative disorders, a possible shared genetic vulnerability related to the functional polymorphisms of SNCA gene, and potential gene-environment interactions involving fine particulate matter pollution. The significance of these results is discussed in light of existing translational, clinical, and epidemiological research on the links between these disorders.

Keywords
Alpha-synuclein
SNCA
Major depression
Dysthymia
Parkinson’s disease
Epidemiology
Funding
None.
References
[1]

Ou Z, Pan J, Tang S, et al., 2021, Global trends in the incidence, prevalence and years lived with disability in Parkinson’s disease in 204 countries/territories from 1990 to 2019. Front Public Health, 9: 776847. https://doi.org/10.3389/fpubh.2021.776847

[2]

GBD 2019 Dementia Forecasting Collaborators, 2022, Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: An analysis for the Global Burden of Disease Study 2019. Lancet Public Health, 7: e105–e125.https://doi.org/10.1016/S2468-2667(21)00249-8 

[3]

Farina N, Ibnidris A, Alladi S, et al., 2020, A systematic review and meta-analysis of dementia prevalence in seven developing countries: A STRiDE project. Glob Public Health, 15: 1878–1893. https://doi.org/10.1080/17441692.2020.1792527

[4]

Singhal BS, Khadilkar SV, 2014, Neurology in the developing world. Handb Clin Neurol, 121: 1773–1782. https://doi.org/10.1016/b978-0-7020-4088-7.00114-0

[5]

Borumandnia N, Majd HA, Doosti H, et al., 2022, The trend analysis of neurological disorders as major causes of death and disability according to human development, 1990-2019. Environ Sci Pollut Res Int, 29: 14348–14354. https://doi.org/10.1007/s11356-021-16604-5 

[6]

van den Kieboom R, Snaphaan L, Mark R, et al., 2020, The trajectory of caregiver burden and risk factors in dementia progression: A systematic review. J Alzheimers Dis, 77: 1107–1115. https://doi.org/10.3233/JAD-200647

[7]

Boland DF, Stacy M, 2012, The economic and quality of life burden associated with Parkinson’s disease: A focus on symptoms. Am J Manag Care, 18: S168–S175.

[8]

Mattap SM, Mohan D, McGrattan AM, et al., 2022, The economic burden of dementia in low-and middle-income countries (LMICs): A systematic review. BMJ Glob Health, 7: e007409. https://doi.org/10.1136/bmjgh-2021-007409

[9]

Roheger M, Brenning J, Riemann S, et al., 2022, Progression of socio-cognitive impairment from healthy aging to Alzheimer’s dementia: A systematic review and meta-analysis. Neurosci Biobehav Rev, 140: 104796. https://doi.org/10.1016/j.neubiorev.2022.104796 

[10]

Martinez-Martin P, Chaudhuri KR, 2018, Comprehensive grading of Parkinson’s disease using motor and non-motor assessments: Addressing a key unmet need. Expert Rev Neurother, 18: 41–50. https://doi.org/10.1080/14737175.2018.1400383

[11]

Saredakis D, Collins-Praino L, Gutteridge DS, et al., 2019, Conversion to MCI and dementia in Parkinson’s disease: A systematic review and meta-analysis. Parkinsonism Relat Disord, 65: 20–31. https://doi.org/10.1016/j.parkreldis.2019.04.020 

[12]

Merello M, Sabe L, Teson A, et al., 1994, Extrapyramidalism in Alzheimer’s disease: Prevalence, psychiatric, and neuropsychological correlates. J Neurol Neurosurg Psychiatry, 57: 1503–1509. https://doi.org/10.1136/jnnp.57.12.1503

[13]

Petrou M, Dwamena BA, Foerster BR, et al., 2015, Amyloid deposition in Parkinson disease and cognitive impairment: A systematic review. Mov Disord, 30: 928–935. https://doi.org/10.1002/mds.26191

[14]

Kotagal V, Spino C, Bohnen NI, et al., 2018, Serotonin, beta-amyloid, and cognition in Parkinson disease. Ann Neurol, 83: 994–1002. https://doi.org/10.1002/ana.25236

[15]

Twohig D, Nielsen HM, 2019, α-synuclein in the pathophysiology of Alzheimer’s disease. Mol Neurodegener, 14: 23. https://doi.org/10.1186/s13024-019-0320-x

[16]

Roberts HL, Schneider BL, Brown DR, 2017, α-synuclein increases β-amyloid secretion by promoting β-/γ-secretase processing of APP. PLoS One, 12: e0171295. https://doi.org/10.1371/journal.pone.0171925

[17]

Cong S, Xiang C, Zhang S, et al., 2022, Prevalence and clinical aspects of depression in Parkinson’s disease: A systematic review and meta-analysis of 129 studies. Neurosci Biobehav Rev, 141: 104749. https://doi.org/10.1016/j.neubiorev.2022.104749

[18]

Broen MP, Narayen NE, Kuijf ML, et al., 2016, Prevalence of anxiety in Parkinson’s disease: Systematic review and meta-analysis. Mov Disord, 31: 1125–1133. https://doi.org/10.1002/mds.26643

[19]

Leung DK, Chan WC, Spector A, et al., 2021, Prevalence of depression, anxiety, and apathy symptoms across dementia stages: A systematic review and meta-analysis. Int J Geriatr Psychiatry, 36: 1330–1344. https://doi.org/10.1002/gps.5556

[20]

Lee Y, Oh JS, Chung SJ, et al., 2018, The presence of depression in de novo Parkinson’s disease reflects poor motor compensation. PLoS One, 13: e0203303. https://doi.org/10.1371/journal.pone.0203303

[21]

Jones JD, Kurniadi NE, Kuhn TP, et al., 2019, Depressive symptoms precede cognitive impairment in de novo Parkinson’s disease patients: Analysis of the PPMI cohort. Neuropsychology, 33: 1111–1120. https://doi.org/10.1037/neu0000583

[22]

van der Velden RM, Broen MP, Kuijf ML, et al., 2018, Frequency of mood and anxiety fluctuations in Parkinson’s disease patients with motor fluctuations: A systematic review. Mov Disord, 33: 1521–1527. https://doi.org/10.1002/mds.27465

[23]

Botto R, Callai N, Cermelli A, et al., 2022, Anxiety and depression in Alzheimer’s disease: A systematic review of pathogenetic mechanisms and relation to cognitive decline. Neurol Sci, 43: 4107–4124. https://doi.org/10.1007/s10072-022-06068-x

[24]

Fan DY, Wang YJ, 2020, Early intervention in Alzheimer’s disease: How early is early enough? Neurosci Bull, 36: 195–197. https://doi.org/10.1007/s12264-019-00429-x

[25]

Mahlknecht P, Marini K, Werkmann M, et al., 2022, Prodromal Parkinson’s disease: Hype or hope for disease-modification trials? Transl Neurodegener, 11: 11. https://doi.org/10.1186/s40035-022-00286-1 

[26]

Alvarez-Sanchez L, Pena-Bautista C, Baquero M, et al., 2022, Novel ultrasensitive detection technologies for the identification of early and minimally invasive Alzheimer’s disease blood biomarkers. J Alzheimers Dis, 86: 1337–1369. https://doi.org/10.3233/jad-215093 

[27]

Chelliah SS, Bhuvanendran S, Magalingam KB, et al., 2022, Identification of blood-based biomarkers for diagnosis and prognosis of Parkinson’s disease: A systematic review of proteomics studies. Ageing Res Rev, 73: 101514. https://doi.org/10.1016/j.arr.2021.101514 

[28]

Aisen PS, Jimenez-Maggiora GA, Rafii MS, et al., 2022, Early-stage Alzheimer disease: Getting trial-ready. Nat Rev Neurol, 18: 389–399. https://doi.org/10.1038/s41582-022-00645-6

[29]

Kuring JK, Mathias JL, Ward L, 2020, Risk of dementia in persons who have previously experienced clinically-significant depression, anxiety, or PTSD: A systematic review and meta-analysis. J Affect Disord, 274: 247–261. https://doi.org/10.1016/j.jad.2020.05.020

[30]

Bareeqa SB, Samar SS, Kamal S, et al., 2022, Prodromal depression and subsequent risk of developing Parkinson’s disease: A systematic review with meta-analysis. Neurodegener Dis Manag, 12: 155–164. https://doi.org/10.2217/nmt-2022-0001

[31]

Zhao Q, Xiang H, Cai Y, et al., 2022, Systematic evaluation of the associations between mental disorders and dementia: An umbrella review of systematic reviews and meta-analyses. J Affect Disord, 307: 301–309. https://doi.org/10.1016/j.jad.2022.03.010

[32]

Walter U, Heilmann R, Kaulitz L, et al., 2015, Prediction of Parkinson’s disease subsequent to severe depression: A ten-year follow-up study. J Neural Transm, 122: 789–797. https://doi.org/10.1007/s00702-014-1313-0

[33]

Wang S, Mao S, Xiang D, et al., 2018, Association between depression and the subsequent risk of Parkinson’s disease: A meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry, 86: 186–192. https://doi.org/10.1016/j.pnpbp.2018.05.025

[34]

Stafford J, Chung WT, Sommerlad A, et al., 2022, Psychiatric disorders and risk of subsequent dementia: Systematic review and meta-analysis of longitudinal studies. Int J Geriatr Psychiatry, 37: 5711. https://doi.org/10.1002/gps.5711

[35]

Saiz-Vazquez O, Gracia-Garcia P, Ubillos-Landa P, et al., 2021, Depression as a risk factor for Alzheimer’s disease: A systematic review of longitudinal meta-analyses. J Clin Med, 10: 1809. https://doi.org/10.3390/jcm10091809

[36]

Tran AA, De Smet M, Grant GD, et al., 2020, Investigating the convergent mechanisms between major depressive disorder and Parkinson’s disease. Complex Psychiatry, 6: 47–61. https://doi.org/10.1159/000512657

[37]

Novellino F, Sacca V, Donato A, et al., 2020, Innate immunity: A common denominator between neurodegenerative and neuropsychiatric diseases. Int J Mol Sci, 21: 1115. https://doi.org/10.3390/ijms21031115

[38]

Hussain M, Kumar P, Khan S, et al., 2020, Similarities between depression and neurodegenerative diseases: Pathophysiology, challenges in diagnosis and treatment options. Cureus, 12: e11613. https://doi.org/10.7759/cureus.11613 

[39]

Luo J, Beam CR, Gatz M, 2022, Is stress an overlooked risk factor for dementia? A systematic review from a lifespan perspective. Prev Sci. https://doi.org/10.1007/s11121-022-01385-1

[40]

Hahad O, Lelieveld J, Birklein F, et al., 2020, Ambient air pollution increases the risk of cerebrovascular and neuropsychiatric disorders through induction of inflammation and oxidative stress. Int J Mol Sci, 21: 4306. https://doi.org/10.3390/ijms21124306

[41]

Chiavegatto S, Izidio GS, Mendes-Lana A, et al., 2009, Expression of α-synuclein is increased in the hippocampus of rats with high levels of innate anxiety. Mol Psychiatry, 14: 894–905. https://doi.org/10.1038/mp.2008.43

[42]

Miquel-Rio L, Alarcon-Aris D, Torres-Lopez M, et al., 2022, Human α-synuclein overexpression in mouse serotonin neurons triggers a depressive-like phenotype. Rescue by oligonucleotide therapy. Transl Psychiatry, 12: 79. https://doi.org/10.1038/s41398-022-01842-z

[43]

Zhao X, Kong D, Zhou Q, et al., 2021, Baicalein alleviates depression-like behavior in rotenone-induced Parkinson’s disease model in mice through activating the BDNF/TrkB/ CREB pathway. Biomed Pharmacother, 140: 111556. https://doi.org/10.1016/j.biopha.2021.111556

[44]

Burtscher J, Copin JC, Rodrigues J, et al., 2019, Chronic corticosterone aggravates behavioral and neuronal  symptomatology in a mouse model of alpha-synuclein pathology. Neurobiol Aging, 83: 11–20. https://doi.org/10.1016/j.neurobiolaging.2019.08.007

[45]

Ishiguro M, Baba H, Maeshima H, et al., 2019, Increased serum levels of α-synuclein in patients with major depressive disorder. Am J Geriatr Psychiatry, 27: 280–286. https://doi.org/10.1016/j.jagp.2018.10.015

[46]

Rotter A, Lenz B, Pitsch R, et al., 2019, Alpha-synuclein RNA expression is increased in major depression. Int J Mol Sci, 20: 2029. https://doi.org/10.3390/ijms20082029

[47]

Fieling H, Gozner A, Romer KD, et al., 2008, Alpha-synuclein mRNA levels correspond to Beck Depression Inventory scores in females with eating disorders. Neuropsychobiology, 58: 48–52. https://doi.org/10.1159/000155991

[48]

Bruno D, Plaska CR, Clark DP, et al., 2021, CSF α-synuclein correlates with CSF neurogranin in late-life depression. Int J Neurosci, 131: 357–361. https://doi.org/10.1080/00207454.2020.1744596

[49]

Eyre HA, Eskin A, Nelson SF, et al., 2016, Genomic predictors of remission to antidepressant treatment in geriatric depression using genome-wide expression analyses: A pilot study. Int J Geriatr Psychiatry, 31: 510–517. https://doi.org/10.1002/gps.4356 

[50]

Murray CJ, 2022, The Global Burden of Disease Study at 30 years. Nat Med, 28: 2019–2026. https://doi.org/10.1038/s41591-022-01990-1

[51]

Global Burden of Disease Study, 2019, (GBD 2019) Data Resources. Available from: https://ghdx.healthdata.org/gbd- 2019 [Last accessed on 2022 Dec 31]. 

[52]

Akiskal HS, Bolis CL, Cazzullo C, et al., 1996, Dysthymia in neurological disorders. Mol Psychiatry, 1: 478–491.

[53]

Han W, Liu Y, Mi Y, et al., 2015, Alpha-synuclein (SNCA) polymorphisms and susceptibility to Parkinson’s disease: A meta-analysis. Am J Med Genet B Neuropsychiatr Genet, 168: 123–134. https://doi.org/10.1002/ajmg.b.32288

[54]

Li C, Ou R, Chen Y, et al., 2021, Genetic modifiers of age at onset for Parkinson’s disease in Asians: A genome-wide association study. Mov Disord, 36: 2077–2084. https://doi.org/10.1002/mds.28621

[55]

Wang Q, Tian Q, Song X, et al., 2016, SNCA gene polymorphism may contribute to an increased risk of Alzheimer’s disease. J Clin Lab Anal, 30: 1092–1099. https://doi.org/10.1002/jcla.21986 

[56]

Han Z, Tian R, Ren P, et al., 2018, Parkinson’s disease and Alzheimer’s disease: A Mendelian randomization study. BMC Med Genet, 19: 215. https://doi.org/10.1186/s12881-018-0721-7 

[57]

Talwar P, Gupta R, Kushwaha S, et al., 2019, Viral induced oxidative and inflammatory response in Alzheimer’s disease pathogenesis with identification of potential drug candidates: A systematic review using systems biology approach. Curr Neuropharmacol, 17: 352–365. https://doi.org/10.2174/1570159x16666180419124508

[58]

Fransquet PD, Lacaze P, Saffery R, et al., 2020, DNA methylation analysis of candidate genes associated with dementia in peripheral blood. Epigenomics, 12: 2109–2123. https://doi.org/10.2217/epi-2020-0236

[59]

Gibbons A, McPherson K, Gogos A, Dean B, 2021, An investigation into nicotinic receptor involvement in mood disorders uncovers novel depression candidate genes. J Affect Disord, 288: 154–160. https://doi.org/10.1016/j.jad.2021.04.007

[60]

Rajeevan H, Osier MV, Cheung KH, et al., 2003, ALFRED: The ALlele FREquency database. Update. Nucleic Acids Res, 31: 270–271. https://doi.org/10.1093/nar/gkg043

[61]

ALFRED: The Allele Frequency Database. Available from: https://alfred.med.yale.edu/alfred/index.asp [Last accessed on 2023 Jan 02]. 

[62]

Bi M, Kang S, Du X, et al., 2020, Association between SNCA rs356220 polymorphism and Parkinson’s disease: A meta-analysis. Neurosci Lett, 717: 134703. https://doi.org/10.1016/j.neulet.2019.134703

[63]

Magistrelli L, Contaldi E, Comi C, 2021, The impact of SNCA variations and its product alpha-synuclein on non-motor features of Parkinson’s disease. Life, 11: 804. https://doi.org/10.3390/life11080804

[64]

Zhang Y, Shu L, Pan H, et al., 2018, A comprehensive analysis of the association between SNCA polymorphisms and the risk of Parkinson’s disease. Front Mol Neurosci, 11: 391. https://doi.org/10.3389/fnmol.2018.00391

[65]

Heckman MG, Soto-Ortolaza AI, Diehl NN, et al., 2012, Evaluation of the role of SNCA variants in survival without neurological disease. PLoS One, 7: e42877. https://doi.org/10.1371/journal.pone.0042877

[66]

Biernacka JM, Armasu SM, Cunningham JM, et al., 2011, Do interactions between SNCA, MAPT, and LRRK2 genes contribute to Parkinson’s disease susceptibility? Parkinsonism Relat Disord, 17: 730–736. https://doi.org/10.1016/j.parkreldis.2011.07.001

[67]

Cristaldi A, Fiore M, Conti GO, et al., 2022, Possible association between PM2.5 and neurodegenerative diseases: A systematic review. Environ Res, 208: 112581. https://doi.org/10.1016/j.envres.2021.112581

[68]

Yan D, Zhang Y, Liu L, et al., 2018, Pesticide exposure and the risk of Parkinson’s disease: Dose-response meta-analysis of observational studies. Reg Toxicol Pharmacol, 96: 57–63. https://doi.org/10.1016/j.yrtph.2018.05.005

[69]

Yan D, Zhang Y, Liu L, et al., 2016, Pesticide exposure and risk of Alzheimer’s disease: A systematic review and meta-analysis. Sci Rep, 6: 32222. https://doi.org/10.1038/srep32222 

[70]

Fu P, Guo X, Cheung FM, et al., 2019, The association between PM2.5 exposure and neurological disorders: A systematic review and meta-analysis. Sci Total Environ, 655: 1240–1248. https://doi.org/10.1016/j.scitotenv.2018.11.218

[71]

Braithwaite I, Zhang S, Kirkbride JB, et al., 2019, Air pollution (particulate matter) exposure and associations with depression, anxiety, bipolar, psychosis and suicide risk: A systematic review and meta-analysis. Environ Health Perspect, 127: 126002. https://doi.org/10.1289/ehp4595

[72]

Gunnarsson LG, Bodin L, 2019, Occupational exposures and neurodegenerative diseases-a systematic literature review and meta-analyses. Int J Environ Res Public Health, 16: 337. http://doi.org/10.3390/ijerph16030337

[73]

Freire C, Koifman S, 2013, Pesticides, depression and suicide: A systematic review of the epidemiological evidence. Int J Hyg Environ Health, 216: 445–460. https://doi.org/10.1016/j.ijheh.2012.12.003

[74]

Ahmed H, Abushouk AI, Gabr M, et al., 2017, Parkinson’s disease and pesticides: A meta-analysis of disease connection and genetic alterations. Biomed Pharmacother, 90: 638–649. https://doi.org/10.1016/j.biopha.2017.03.100

[75]

Global Health Observatory. Available from: https://www. who.int/data/gho [Last accessed on 2023 Jan 05].

[76]

FAOSTAT. Available from: https://www.fao.org/faostat/ en/#search/pesticide [Last accessed on 2023 Jan 02].

[77]

Roumeliotis S, ElHafeez SA, Jager KJ, et al., 2021, Be careful with ecological associations. Nephrology, 26: 501–505. https://doi.org/10.1111/nep.13861

[78]

Dormann C, Griffin MA, 2015, Optimal time lags in panel studies. Psychol Methods, 20: 489–505. https://doi.org/10.1037/met0000041

[79]

Akoglu H, 2019, User’s guide to correlation coefficients. Turk J Emerg Med, 18, 91–93. https://doi.org/10.1016/j.tjem.2018.08.001

[80]

Wolfe N, Katz DI, Albert ML, et al., 1990, Neuropsychological profile linked to low dopamine: In Alzheimer’s disease, major depression, and Parkinson’s disease. J Neurol Neurosurg Psychiatry, 53: 915–917. https://doi.org/10.1136/jnnp.53.10.915

[81]

Shen Y, Qian L, Luo H, et al., 2022, The significance of NLRP inflammasome in neuropsychiatric disorders. Brain Sci 12: 1057. https://doi.org/10.3390/brainsci12081057

[82]

Barrio C, Arias-Sanchez S, Martin-Monzon I, 2022, The gut microbiota-brain axis, psychobiotics and its influence on brain and behaviour: A systematic review. Psychoneuroendocrinology, 137: 105640. https://doi.org/10.1016/j.psyneuen.2021.105640

[83]

Cheng F, Vivacqua G, Yu S, 2011, The role of α-synuclein in neurotransmission and synaptic plasticity. J Chem Neuroanat, 42: 242–248. https://doi.org/10.1016/j.jchemneu.2010.12.001 

[84]

Jeong W, Kim H, Joo JH, et al., 2021, Association between depression and risk of Parkinson’s disease in South Korean adults. J Affect Disord, 292: 75–80. https://doi.org/10.1016/j.jad.2021.05.038

[85]

Schrag A, Anastasiou Z, Ambler G, et al., 2019, Predicting diagnosis of Parkinson’s disease: A risk algorithm based on primary care presentations. Mov Disord, 34: 480–486. https://doi.org/10.1002/mds.27616

[86]

Kim D, Wang R, Kiss A, et al., 2021, Depression and increased risk of Alzheimer’s dementia: Longitudinal analyses of modifiable risk and sex-related factors. Am J Geriatr Psychiatry, 29: 917–926. https://doi.org/10.1016/j.jagp.2020.12.031

[87]

Canton-Habas V, Rich-Ruiz M, Romero-Saldana M, et al., 2020, Depression as a risk factor for dementia and Alzheimer’s disease. Biomedicines, 8: 457. https://doi.org/10.3390/biomedicines8110457

[88]

Peng H, Lin J, Guan W, 2021, Letter to the editor-association between depression and risk of Parkinson’s disease in South Korean adults. J Affect Disord, 295: 1151–1152. https://doi.org/10.1016/j.jad.2021.09.006

[89]

Wingo TS, Gerasimov ES, Canon SM, et al., 2022, Alzheimer’s disease genetic burden is associated with mid-life depression among persons with normal cognition. Alzheimers Dement. https://doi.org/10.1002/alz.12716

[90]

Harerimana NV, Liu Y, Gerasimov ES, et al., 2022, Genetic evidence supporting a causal role of depression in Alzheimer’s disease. Biol Psychiatry, 92: 25–33. https://doi.org/10.1016/j.biopsych.2021.11.025

[91]

Dalle E, Mabandla MV, 2018, Early life stress, depression and Parkinson’s disease: A new approach. Mol Brain, 11: 18. https://doi.org/10.1186/s13041-018-0356-9

[92]

Dolotov OV, Inozemtseva LS, Myasoedov NF, et al., 2022, Stress-induced depression and Alzheimer’s disease: Focus on astrocytes. Int J Mol Sci, 23: 4999. https://doi.org/10.3390/ijms23094999

[93]

Calderon-Garciduenas L, Ayala A, 2022, Air pollution, ultrafine particles, and your brain: Are combustion nanoparticle emissions and engineered nanoparticles causing preventable fatal neurodegenerative diseases and common neuropsychiatric outcomes? Environ Sci Technol, 56: 6847–6856. https://doi.org/10.1021/acs.est.1c04706

[94]

Jacobs BM, Belete D, Bestwick J, et al., 2020, Parkinson’s disease determinants, prediction and gene-environment interactions in the UK Biobank. J Neurol Neurosurg Psychiatry, 91: 1046–1054. https://doi.org/10.1136/jnnp-2020-324472 

[95]

Akiskal HS, 1994, Dysthymia: Clinical and external validity. Acta Psychiatr Scand Suppl, 383: 19–23. https://doi.org/10.1111/j.1600-0447.1994.tb05879.x

[96]

Brunello N, Akiskal H, Boyer P, et al., 1999, Dysthymia: Clinical picture, extent of overlap with chronic fatigue syndrome, neuropharmacological considerations, and new therapeutic vistas. J Affect Disord, 52: 275–290. https://doi.org/10.1016/s0165-0327(98)00163-3

[97]

Reijnders JS, Ehrt U, Weber WE, et al., 2008, A systematic review of prevalence studies of depression in Parkinson’s disease. Mov Disord, 23: 183–189. https://doi.org/10.1002/mds.21803

[98]

Migliorelli R, Teson A, Sabe L, et al., 1995, Prevalence and correlates of dysthymia and major depression among patients with Alzheimer’s disease. Am J Psychiatry, 152: 37–44. https://doi.org/10.1176/ajp.152.1.37

[99]

Kazmi H, Walker Z, Booij J, et al., 2021, Late onset depression: Dopaminergic deficit and clinical features of prodromal Parkinson’s disease: A cross-sectional study. J Neurol Neurosurg Psychiatry, 92: 158–164. https://doi.org/10.1136/jnnp-2020-324266 

[100]

Lehto SM, Tolmunen T, Kuikka J, et al., 2008, Midbrain serotonin and striatum dopamine transporter binding in double depression: A one-year follow-up study. Neurosci Lett, 441: 291–295. https://doi.org/10.1016/j.neulet.2008.06.042

[101]

Ishizaki J, Mimura M, 2011, Dysthymia and apathy: Diagnosis and treatment. Depress Res Treat, 2011: 893905. https://doi.org/10.1155/2011/893905

[102]

Chen CY, Hung HJ, Chang KH, et al., 2017, Long-term exposure to air pollution and the incidence of Parkinson’s disease: A nested case-control study. PLoS One, 12, e0182834. https://doi.org/10.1371/journal.pone.0182834

[103]

Guo P, Gong W, Li Y, et al., 2022, Pinpointing novel risk loci for Lewy body dementia and the shared genetic etiology with Alzheimer’s disease and Parkinson’s disease: A large-scale multi-trait association analysis. BMC Med, 20: 214. https://doi.org/10.1186/s12916-022-02404-2

[104]

Khan SS, LaCroix M, Boyle G, et al., 2018, Bidirectional modulation of Alzheimer phenotype by alpha-synuclein in mice and primary neurons. Acta Neuropathol, 136: 589–605. https://doi.org/10.1007/s00401-018-1886-z

[105]

Monge-Garcia V, Garcia-Ayllon MS, Saez-Valero J, et al., 2021, Relation between alpha-synuclein and core CSF biomarkers of Alzheimer’s disease. Medicina, 57: 954. https://doi.org/10.3390/medicina57090954

[106]

Eisenberg DT, Hayes MG, 2011, Testing the null hypothesis: Comments on ‘Culture-gene coevolution of individualism-collectivism and the serotonin transporter gene’. Proc Biol Sci, 278: 329–332. https://doi.org/10.1098/rspb.2010.0714

[107]

Lee PC, Raaschou-Nielsen O, Lill CM, et al., 2016, Gene-environment interactions linking air pollution and inflammation in Parkinson’s disease. Environ Res, 151: 713–720. https://doi.org/10.1016/j.envres.2016.09.006

[108]

Borrageiro G, Haylett W, Seedat S, et al., 2018, A review of genome-wide transcriptomics studies in Parkinson’s disease. Eur J Neurosci, 47: 1–16. https://doi.org/10.1111/ejn.13760 

[109]

Paul KC, Kusters C, Furlong M, et al., 2022, Immune system disruptions implicated in whole blood epigenome-wide association study of depression among Parkinson’s disease patients. Brain Behav Immun Health, 26: 100530. https://doi.org/10.1016/j.bbih.2022.100530

[110]

Tirozzi A, Quiccione MS, Cerletti C, et al., 2023, A multi-trait association analysis of brain disorders and platelet traits identifies novel susceptibility loci for major depression, Alzheimer’s and Parkinson’s disease. Cells, 12: 245. https://doi.org/10.3390/cells12020245

[111]

Vanni S, Baldeschi AC, Zattoni M, et al., 2020, Brain aging: A Ianus-faced player between health and neurodegeneration. J Neurosci Res, 98: 299–311. https://doi.org/10.1002/jnr.24379

Conflict of interest
The author declares that he has no competing interests.
Share
Back to top
Advanced Neurology, Electronic ISSN: 2810-9619 Published by AccScience Publishing