Skip to main content

Advertisement

Log in

CSF Biomarkers for Early Diagnosis of Synucleinopathies: Focus on Idiopathic RBD

  • Sleep (M Thorpy and M Billiard, Section Editors)
  • Published:
Current Neurology and Neuroscience Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Idiopathic REM sleep behavior disorder (iRBD) is one of the most significant prodromal manifestations of synucleinopathies. Different predictive biomarkers for iRBD conversion have been investigated, but scarce data are present in literature about the predictive role of cerebrospinal fluid (CSF) biomarkers. In this review, we focus on CSF biomarkers in patients with both iRBD and RBD associated with synucleinopathies to explore their potential predictive power.

Recent Findings

Recent studies revealed that CSF α-synuclein levels are higher in Parkinson’s disease (PD) patients with RBD compared to those without RBD, even if α-synuclein does not seem to predict conversion of iRBD into PD. In the Parkinson Progression Marker Initiative (PPMI) cohort, early PD patients with RBD show lower CSF Aβ42 levels, which predict faster cognitive decline. CSF prion protein and inflammatory biomarkers have been also investigated in RBD and synucleinopathies with controversial results.

Summary

A variety of CSF biomarkers are promising candidate for predicting iRBD conversion into synucleinopathies. Further studies are needed in iRBD patients followed for several years in order to observe the phenoconversion in synucleinopathies and to elucidate the possible role of CSF biomarkers as predictive biomarkers of conversion.

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

Access this article

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

Instant access to the full article PDF.

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Barone DA, Henchcliffe C. Rapid eye movement sleep behavior disorders and link to alpha-synucleinopathies. Clin Neurophysiol. 2018;129:1551–64.

    Article  Google Scholar 

  2. Mahlknecht P, Seppi K, Poewe W. The concept of prodromal Parkinson’s disease. J Parkinsons Dis. 2015;5(4):681–97.

    Article  Google Scholar 

  3. Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder: a 16-year update on a previously reported series. Sleep Med. 2013;14(8):744–8.

    Article  Google Scholar 

  4. Schenck C, Bundlie S, Ettinger MG, Mahowald MW. Chronic behavioral disorders of human rem sleep: a new category of parasomnia. Sleep. 1986;9(2):293–308.

    Article  CAS  Google Scholar 

  5. Stiasny-Kolster K, Mayer G, Schäfer S, Möller JC, Heinzel-Gutenbrunner M, Oertel WH. The REM sleep behavior disorder screening questionnaire—a new diagnostic instrument. Mov Disord. 2007 Dec;22(16):2386–93.

    Article  Google Scholar 

  6. American Academy of Sleep Medicine. International Classification of Sleep Disorders. 3rd edition Darien. 2014.

  7. Ohayon MM, Caulet M, Priest RG. Violent behavior during sleep. J Clin Psychiatry. 1997;58(8):369–76.

    Article  CAS  Google Scholar 

  8. Peever J, Luppi PH, Montplaisir J. Breakdown in REM sleep circuitry underlies REM sleep behavior disorder. Trends Neurosci. 2014;37(5):279–88.

    Article  Google Scholar 

  9. Boeve BF, Silber MH, Ferman TJ, Lin SC, Benarroch EE, Schmeichel AM, et al. Clinicopathologic correlations in 172 cases of rapid eye movement sleep behavior disorder with or without a coexisting neurologic disorder. Sleep Med. 2013;14(8):754–62.

    Article  CAS  Google Scholar 

  10. 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. PLoS One. 2014;9(2):e89741.

    Article  Google Scholar 

  11. Claassen DO, Josephs KA, Ahlskog JE, Silber MH, Tippmann-Peikert M, Boeve BF. REM sleep behavior disorder preceding other aspects of synucleinopathies by up to half a century. Neurology. 2010;75(6):494–9.

    Article  CAS  Google Scholar 

  12. Iranzo A, Tolosa E, Gelpi E, Molinuevo JL, Valldeoriola F, Serradell M, et al. Neurodegenerative disease status and post-mortem pathology in idiopathic rapid-eye-movement sleep behaviour disorder: an observational cohort study. Lancet Neurol. 2013;12:443–53.

    Article  Google Scholar 

  13. De Cock VC. Restoration of normal motor control in Parkinson’s disease during REM sleep. Brain. 2007;130:450–6.

    Article  Google Scholar 

  14. Postuma RB. REM sleep behavior disorders in Parkinson’s disease is associated with specific motor features. J Neurol Neurosurg Psychiatry. 2008;79:1117–21.

    Article  CAS  Google Scholar 

  15. Palma JA, Fernandez-Cordon C, Coon EA, Low PA, Miglis MG, Jaradeh S, et al. Prevalence of REM sleep behavior disorder in multiple system atrophy: a multicenter study and meta-analysis. Clin Auton Res Off J Clin Auton Res Soc. 2015;25:69–75.

    Article  Google Scholar 

  16. Ferman TJ, Boeve BF, Smith GE, Lin SC, Silber MH, Pedraza O, et al. Inclusion of RBD improves the diagnostic classification of dementia with Lewy bodies. Neurology. 2011;77:875–82.

    Article  CAS  Google Scholar 

  17. Ferini-Strambi L. Does idiopathic REM sleep behavior disorder (iRBD) really exist? What are the potential markers of neurodegeneration in iRBD? Sleep Med. 2011;12(Suppl 2):S43–9.

    Article  Google Scholar 

  18. •• Postuma RB, Gagnon JF, Bertrand JA, Génier Marchand D, Montplaisir JY. Parkinson risk in idiopathic REM sleep behavior disorder: preparing for neuroprotective trials. Neurology. 2015;84(11):1104–13 This paper documented that the use of simply assessed markers can identify patients with RBD to be included in neuroprotective trials against Parkinson disease, multiple system atrophy, and dementia with Lewy bodies.

    Article  Google Scholar 

  19. Rocchi C, Placidi F, Liguori C, Del Bianco C, Lauretti B, Diomedi M, et al. Daytime autonomic activity in idiopathic rapid eye movement sleep behavior disorder: a preliminary study. Sleep Med. 2018;52:163–7.

    Article  Google Scholar 

  20. •• Li Y, Kang W, Yang Q, Zhang L, Zhang L, Dong F, et al. Predictive markers for early conversion of iRBD to neurodegenerative synucleinopathy diseases. Neurology. 2017;88:1493–500 This research article suggested the predictive value of autonomic dysfunction and DAT uptake in identifying patients with iRBD at a high risk of progressing into neurodegenerative synucleinopathy diseases.

    Article  Google Scholar 

  21. •• Iranzo A, Santamaria J, Valldeoriola F, Serradell M, Salamero M, Gaig C, et al. Dopamine transporter imaging deficit predicts early transition to synucleinopathy in idiopathic rapid eye movement sleep behavior disorder. Ann Neurol. 2017;82:419–28 This paper quantified the decreased FP-CIT putamen uptake as greater than 25% able to predict the conversion to synucleinopathy after 3 years' follow-up in idiopathic RBD patients.

    Article  CAS  Google Scholar 

  22. Rodrigues Brazete J, Gagnon JF, Postuma RB, Bertrand JA, Petit D, Montplaisir J. Electroencephalogram slowing predicts neurodegeneration in rapid eye movement sleep behavior disorder. Neurobiol Aging. 2016;37:74–81.

    Article  Google Scholar 

  23. •• Rolinski M, Griffanti L, Piccini P, Roussakis AA, Szewczyk-Krolikowski K, Menke RA, et al. Basal ganglia dysfunction in idiopathic REM sleep behaviour disorder parallels that in early Parkinson’s disease. Brain. 2016;139:2224–34 Here Authors proved the very early dysfunction of basal ganglia connectivity in RBD, which was comparable to that of PD, despite obvious differences on dopamine transported single photon emission computerized tomography.

    Article  Google Scholar 

  24. Frosini D, Cosottini M, Donatelli G, Costagli M, Biagi L, Pacchetti C, et al. Seven tesla MRI of the substantia nigra in patients with rapid eye movement sleep behavior disorder. Parkinsonism Relat Disord. 2017;43:105–9.

    Article  Google Scholar 

  25. Marques O, Outeiro TF. Alpha-synuclein: from secretion to dysfunction and death. Cell Death Dis. 2012;3(7):e350.

    Article  CAS  Google Scholar 

  26. Shi M, Bradner J, Hancock AM, Chung KA, Quinn JF, Peskind ER, et al. Cerebrospinal fluid biomarkers for Parkinson’s disease diagnosis and progression. Ann Neuol. 2011;69:570–80.

    Article  CAS  Google Scholar 

  27. Parnetti L, Chiasserini D, Bellomo G, Giannandrea D, de Carlo C, Qureshi MM, et al. Cerebrospinal fluid tau/αsynuclein ratio in Parkinson’s disease and degenerative dementias. Mov Disord. 2011;26:1428–35.

    Article  Google Scholar 

  28. Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Döring F, Trenkwalder C, Schlossmacher MG. α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol. 2011;10(3):230–40.

    Article  CAS  Google Scholar 

  29. Parnetti L, Chiasserini D, Persichetti E, Eusebi P, Varghese S, Qureshi MM, et al. Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson’s disease. Mov Disord. 2014;29(8):1019–27.

    Article  CAS  Google Scholar 

  30. Parnetti L, Farotti L, Eusebi P, et al. Differential role of CSF alphasynuclein species, tau, and Aβ42 in Parkinson’s disease. Front Aging Neurosci Internet. 2014;6(53).

  31. Tokuda T, Qureshi MM, Ardah MT, Varghese S, Shehab SAS, Kasai T, et al. Detection of elevated levels of α-synuclein oligomers in CSF from patients with Parkinson’s disease. Neurology. 2010;75:1766–70.

    Article  CAS  Google Scholar 

  32. Wang Y, Shi M, Chung KA, et al. Phosphorylated synuclein in Parkinson’s disease. Sci Transl Med. 2012;4(121):121ra20–121ra20.

    Google Scholar 

  33. Foulds PG, Yojota O, Thurston A, Davidson Y, Ahmed Z, Holton J, et al. Post mortem cerebrospinal fluid α-synuclein levels are raised in multiple system atrophy and distinguish this from the other α-synucleinopathies, Parkinson’s disease and dementia with lewy bodies. Neurobiol Dis. 2012;45:188–95.

    Article  CAS  Google Scholar 

  34. Aasly JO, Johansen KK, Brønstad G, et al. Elevated levels of cerebrospinal fluid α-synuclein oligomers in healthy asymptomatic LRRK2 mutation carriers. Front Aging Neurosci. 2014;6:248.

    Article  Google Scholar 

  35. Kang J-H, Irwin DJ, Chen-Plotkin AS, et al. Association of cerebrospinal fluidβ-amyloid1-42,T-tau,P-tau181,andα-synucleinlevelswithclinical features of drug-naive patients with early Parkinson disease. JAMA Neurol Internet 2013;70:1277–1287.

  36. Jellinger KA. Neuropathological aspects of Alzheimer disease, Parkinson disease and frontotemporal dementia. Neurodegener Dis. 2008;5(3–4):118–21.

    Article  CAS  Google Scholar 

  37. Compta Y, Martí MJ, Ibarretxe-Bilbao N, et al. Cerebrospinal tau, phospho-tau, and beta-amyloid and neuropsychological functions in Parkinson’s disease. Mov Disord. 2009;24(15):2203–10.

    Article  Google Scholar 

  38. Montine TJ, Shi M, Quinn JF, Peskind ER, Craft S, Ginghina C, et al. CSF Aβ 42 and tau in Parkinson’s disease with cognitive impairment. Mov Disord. 2010;25(15):2682–5.

    Article  Google Scholar 

  39. Leverenz JB, Stennis Watson G, Shofer J, Zabetian CP, Zhang J, Montine TJ. Cerebrospinal fluid biomarkers and cognitive performance in non-demented patients with Parkinson’s disease. Parkinsonism Relat Disord. 2011;17(1):61–4.

    Article  Google Scholar 

  40. • Compta Y, Valente T, Saura J, Segura B, Iranzo Á, Serradell M, et al. Correlates of cerebrospinal fluid levels of oligomeric- and total-α-synuclein in premotor, motor and dementia stages of Parkinson’s disease. J Neurol. 2015;262(2):294–306 The Authors documented that CSF total-α-synuclein levels correlate with CSF tau and Aβ 42 levels, possibly suggesting that tau pathology may influence the presence of high (instead of low) CSF total-α-synuclein levels in the setting of PD-related dementia.

    Article  CAS  Google Scholar 

  41. Waragai M, Wei J, Fujita M, Nakai M, Ho GJ, Masliah E, et al. Increased level of DJ-1 in the cerebrospinal fluids of sporadic Parkinson’s disease. Biochem Biophys Res Commun Internet. 2006;345:967–72.

    Article  CAS  Google Scholar 

  42. Magdalinou NK, Paterson RW, Schott JM, Fox NC, Mummery C, Blennow K, et al. A panel of nine cerebrospinal fluid biomarkers may identify patients with atypical parkinsonian syndromes. J Neurol Neurosurg Psychiatry. 2015;86(11):1240–7.

    Article  CAS  Google Scholar 

  43. Bech S, Hjermind LE, Salvesen L, Nielsen JE, Heegaard NH, Jørgensen HL, et al. Amyloid-related biomarkers and axonal damage proteins in parkinsonian syndromes. Parkinsonism Relat Disord. 2012 Jan;18(1):69–72.

    Article  Google Scholar 

  44. • Hansson O, Janelidze S, Hall S, Magdalinou N, Lees AJ, Andreasson U, et al. Swedish BioFINDER study. Blood-based NfL: a biomarker for differential diagnosis of parkinsonian disorder. Neurology. 2017;88(10):930–7 Authors firstly identified a blood biomarker (NfL) which can be used to distinguish PD from atypical parkinsonisms.

    Article  CAS  Google Scholar 

  45. • Hu Y, Yu SY, Zuo LJ, Cao CJ, Wang F, Chen ZJ, et al. Parkinson disease with REM sleep behavior disorder: features, α-synuclein, and inflammation. Neurology. 2015;84(9):888–94 Here, it was documented that elevated α-synuclein levels in CSF and serum may be correlated with RBD in PD patients through central and peripheral nervous systems inflammatory mechanisms.

    Article  CAS  Google Scholar 

  46. Dolatshahi M, Pourmirbabaei S, Kamalian A, Ashraf-Ganjouei A, Yaseri M, Aarabi MH. Longitudinal alterations of alpha-synuclein, amyloid Beta, total, and phosphorylated tau in cerebrospinal fluid and correlations between their changes in Parkinson’s disease. Front Neurol. 2018;9:560.

    Article  Google Scholar 

  47. • Pagano G, De Micco R, Yousaf T, Wilson H, Chandra A, Politis M. REM behavior disorder predicts motor progression and cognitive decline in Parkinson disease. Neurology. 2018;91(10):e894–905 Pagano and co-Authors proved that the presence of RBD in PD patients is associated with faster motor progression when greater synuclein and dopaminergic pathologies are present, and with higher risk of cognitive decline when greater synuclein and amyloid pathologies are present.

    Article  Google Scholar 

  48. • Ba M, Yu G, Kong M, Liang H, Yu L. CSF Aβ1–42 level is associated with cognitive decline in early Parkinson’s disease with rapid eye movement sleep behavior disorder. Transl Neurodegener. 2018;7:22 This paper suggested that the addition of CSF Aβ 42 assessment to RBD increases the likelihood of identifying early PD patients at high risk for cognitive decline.

    Article  Google Scholar 

  49. Goldman JG, Andrews H, Amara A, Naito A, Alcalay RN, Shaw LM, et al. Cerebrospinal fluid, plasma, and saliva in the BioFIND study: relationships among biomarkers and Parkinson’s disease features. Mov Disord. 2018;33(2):282–8.

    Article  CAS  Google Scholar 

  50. Hall S, Janelidze S, Surova Y, Widner H, Zetterberg H, Hansson O. Cerebrospinal fluid concentrations of inflammatory markers in Parkinson’s disease and atypical parkinsonian disorders. Sci Rep. 2018;8(1):13276.

    Article  Google Scholar 

  51. Zhang WJ, Shang XL, Peng J, Zhou MH, Sun WJ. Expression of prion protein in the cerebrospinal fluid of patients with Parkinson’s disease complicated with rapid eye movement sleep behavior disorder. Genet Mol Res. 2017;23:16 (1).

    Google Scholar 

  52. •• Stokholm MG, Iranzo A, Østergaard K, Serradell M, Otto M, Svendsen KB, et al. Assessment of neuroinflammation in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a case-control study. Lancet Neurol. 2017;16(10):789–96 This original research documented that increased microglial activation (detected by PET scans) in the substantia nigra along with reduced dopaminergic function in the putamen is present in patients affected by idiopathic RBD.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudio Liguori.

Ethics declarations

Conflict of Interest

Paolo Eusepi, Claudio Liguori, Nicola Biagio Mercuri, Federico Paolini Paoletti, Lucilla Parnetti, Fabio Placidi, Roberta Ruffini and Giulia Maria Sancesario each declare no potential conflicts of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Sleep

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liguori, C., Paoletti, F.P., Placidi, F. et al. CSF Biomarkers for Early Diagnosis of Synucleinopathies: Focus on Idiopathic RBD. Curr Neurol Neurosci Rep 19, 3 (2019). https://doi.org/10.1007/s11910-019-0918-y

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11910-019-0918-y

Keywords

Navigation