Skip to main content
Log in

Sleep Disturbances in the Prodromal Stage of Parkinson Disease

  • Sleep Disorders (A Iranzo, Section Editor)
  • Published:
Current Treatment Options in Neurology Aims and scope Submit manuscript

Opinion statement

Decades of research on neurodegenerative diseases such as Parkinson’s disease (PD) have shifted our focus from clinical symptomatic stages to one step earlier, the prodromal stage. There is convincing evidence showing that sleep disorders such as excessive daytime somnolence, insomnia, obstructive sleep apnea, and particularly REM sleep behavior disorder (RBD) are potential prodromal features starting years and decades before conversion to PD. This review aims to provide an overview on various sleep disorders as prodromal features of PD. We start with a discussion of how neuroanatomy of sleep might link to PD pathology. Afterwards, specific sleep disorders that are either known or suspected to be a prodromal symptom for PD, namely insomnia, sleep apnea, somnolence, and RBD, are discussed. Disturbances in serotonergic, cholinergic, dopaminergic, and glutamatergic neurotransmitter systems have made sleep disorders potential candidates for prodromal PD. Nevertheless, the strength of such linkage varies considerably for different entities. While RBD is by far the strongest prodromal manifestation for PD with a high likelihood of phenoconversion and there is reasonable evidence that somnolence can predict PD, data on the prodromal role of sleep apnea and insomnia is limited. Treatment options are available for symptomatic relief of prodromal sleep disorders; however, there is a dearth of knowledge on neuroprotective agents to be used on people with prodromal sleep disorders to either prevent or postpone conversion to PD.

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.

Fig. 1
Fig. 2

Similar content being viewed by others

References and Recommended Reading

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

  1. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386(9996):896–912. doi:10.1016/S0140-6736(14)61393-3.

    Article  CAS  PubMed  Google Scholar 

  2. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197–211.

    Article  PubMed  Google Scholar 

  3. Davie CA. A review of Parkinson’s disease. Br Med Bull. 2008;86:109–27. doi:10.1093/bmb/ldn013.

    Article  CAS  PubMed  Google Scholar 

  4. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167(6):1469–80 e12. doi:10.1016/j.cell.2016.11.018.

    Article  CAS  PubMed  Google Scholar 

  5. Postuma RB, Aarsland D, Barone P, Burn DJ, Hawkes CH, Oertel W, et al. Identifying prodromal Parkinson’s disease: pre-motor disorders in Parkinson’s disease. Mov Disord. 2012;27(5):617–26. doi:10.1002/mds.24996.

    Article  PubMed  Google Scholar 

  6. Jones BE. Reticular formation: cytoarchitecture, transmitters, and projections. In: Paxinos G, editor. The rat nervous sytem. 2nd ed. San Diego: Academic Press; 1995. p. 155–71.

    Google Scholar 

  7. Espana RA, Scammell TE. Sleep neurobiology for the clinician. Sleep. 2004;27(4):811–20.

    PubMed  Google Scholar 

  8. Popa D, Lena C, Fabre V, Prenat C, Gingrich J, Escourrou P, et al. Contribution of 5-HT2 receptor subtypes to sleep-wakefulness and respiratory control, and functional adaptations in knock-out mice lacking 5-HT2A receptors. J Neurosci. 2005;25(49):11231–8. doi:10.1523/JNEUROSCI.1724-05.2005.

    Article  CAS  PubMed  Google Scholar 

  9. Halliday GM, Blumbergs PC, Cotton RG, Blessing WW, Geffen LB. Loss of brainstem serotonin- and substance P-containing neurons in Parkinson’s disease. Brain Res. 1990;510(1):104–7.

    Article  CAS  PubMed  Google Scholar 

  10. Qamhawi Z, Towey D, Shah B, Pagano G, Seibyl J, Marek K, et al. Clinical correlates of raphe serotonergic dysfunction in early Parkinson’s disease. Brain. 2015;138(Pt 10):2964–73. doi:10.1093/brain/awv215.

    Article  PubMed  Google Scholar 

  11. Kotagal V, Albin RL, Muller ML, Koeppe RA, Chervin RD, Frey KA, et al. Symptoms of rapid eye movement sleep behavior disorder are associated with cholinergic denervation in Parkinson disease. Ann Neurol. 2012;71(4):560–8. doi:10.1002/ana.22691.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lelieveld IM, Muller ML, Bohnen NI, Koeppe RA, Chervin RD, Frey KA, et al. The role of serotonin in sleep disordered breathing associated with Parkinson disease: a correlative [11C]DASB PET imaging study. PLoS One. 2012;7(7):e40166. doi:10.1371/journal.pone.0040166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Haas HL, Sergeeva OA, Selbach O. Histamine in the nervous system. Physiol Rev. 2008;88(3):1183–241. doi:10.1152/physrev.00043.2007.

    Article  CAS  PubMed  Google Scholar 

  14. Shan L, Liu CQ, Balesar R, Hofman MA, Bao AM, Swaab DF. Neuronal histamine production remains unaltered in Parkinson’s disease despite the accumulation of Lewy bodies and Lewy neurites in the tuberomamillary nucleus. Neurobiol Aging. 2012;33(7):1343–4. doi:10.1016/j.neurobiolaging.2011.01.004.

    Article  CAS  PubMed  Google Scholar 

  15. Lee MG, Hassani OK, Jones BE. Discharge of identified orexin/hypocretin neurons across the sleep-waking cycle. J Neurosci. 2005;25(28):6716–20. doi:10.1523/JNEUROSCI.1887-05.2005.

    Article  CAS  PubMed  Google Scholar 

  16. Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe JG, et al. Neurons containing hypocretin (orexin) project to multiple neuronal systems. J Neurosci. 1998;18(23):9996–10015.

    CAS  PubMed  Google Scholar 

  17. Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E. Hypocretin (orexin) deficiency in human narcolepsy. Lancet. 2000;355(9197):39–40. doi:10.1016/S0140-6736(99)05582-8.

    Article  CAS  PubMed  Google Scholar 

  18. Thannickal TC, Lai YY, Siegel JM. Hypocretin (orexin) cell loss in Parkinson’s disease. Brain. 2007;130(Pt 6):1586–95. doi:10.1093/brain/awm097.

    Article  PubMed  Google Scholar 

  19. Mesulam MM, Mufson EJ, Wainer BH, Levey AI. Central cholinergic pathways in the rat: an overview based on an alternative nomenclature (Ch1-Ch6). Neuroscience. 1983;10(4):1185–201.

    Article  CAS  PubMed  Google Scholar 

  20. Duque A, Balatoni B, Detari L, Zaborszky L. EEG correlation of the discharge properties of identified neurons in the basal forebrain. J Neurophysiol. 2000;84(3):1627–35.

    CAS  PubMed  Google Scholar 

  21. Marrosu F, Portas C, Mascia MS, Casu MA, Fa M, Giagheddu M, et al. Microdialysis measurement of cortical and hippocampal acetylcholine release during sleep-wake cycle in freely moving cats. Brain Res. 1995;671(2):329–32.

    Article  CAS  PubMed  Google Scholar 

  22. Karczmar AG, Longo A, de Carolis S. A pharmacological model of paradoxical sleep: the role of cholinergic and monoaminergic neurons. Physiol Behav. 1970;5:175–82.

    Article  CAS  PubMed  Google Scholar 

  23. Torontali ZA, Grace KP, Horner RL, Peever JH. Cholinergic involvement in control of REM sleep paralysis. J Physiol. 2014;592(7):1425–6. doi:10.1113/jphysiol.2014.271304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Pavese N, Metta V, Simpson BS, Marphy TA, Ramlackhansingh A, Chaudhuri KR, et al. Sleep regulatory centres dysfunction in Parkinson’s disease patients with excessive daytime sleepiness. Parkinsonism Relat Disord. 2012;18:S24–S5.

    Article  Google Scholar 

  25. Lindvall O, Bjorklund A. Neuroanatomy of central dopamine pathways: review of recent progress. In: Keal M, editor. Advances in dopamine research. New York: Pergamon Press; 1982. p. 297–311.

    Chapter  Google Scholar 

  26. Rye DB, Bliwise DL, Dihenia B, Gurecki P. FAST TRACK: daytime sleepiness in Parkinson’s disease. J Sleep Res. 2000;9(1):63–9.

    Article  CAS  PubMed  Google Scholar 

  27. Happe S, Baier PC, Helmschmied K, Meller J, Tatsch K, Paulus W. Association of daytime sleepiness with nigrostriatal dopaminergic degeneration in early Parkinson’s disease. J Neurol. 2007;254(8):1037–43. doi:10.1007/s00415-006-0483-6.

    Article  PubMed  Google Scholar 

  28. Eisensehr I, Linke R, Tatsch K, Kharraz B, Gildehaus JF, Wetter CT, et al. Increased muscle activity during rapid eye movement sleep correlates with decrease of striatal presynaptic dopamine transporters. IPT and IBZM SPECT imaging in subclinical and clinically manifest idiopathic REM sleep behavior disorder, Parkinson’s disease, and controls. Sleep. 2003;26(5):507–12.

    Article  PubMed  Google Scholar 

  29. Kim YK, Yoon IY, Kim JM, Jeong SH, Kim KW, Shin YK, et al. The implication of nigrostriatal dopaminergic degeneration in the pathogenesis of REM sleep behavior disorder. Eur J Neurol. 2010;17(3):487–92. doi:10.1111/j.1468-1331.2009.02854.x.

    Article  CAS  PubMed  Google Scholar 

  30. Dash MB, Douglas CL, Vyazovskiy VV, Cirelli C, Tononi G. Long-term homeostasis of extracellular glutamate in the rat cerebral cortex across sleep and waking states. J Neurosci. 2009;29(3):620–9. doi:10.1523/JNEUROSCI.5486-08.2009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. John J, Ramanathan L, Siegel JM. Rapid changes in glutamate levels in the posterior hypothalamus across sleep-wake states in freely behaving rats. Am J Physiol Regul Integr Comp Physiol. 2008;295(6):R2041–9. doi:10.1152/ajpregu.90541.2008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. •• Luppi PH, Clement O, Valencia Garcia S, Brischoux F, Fort P. New aspects in the pathophysiology of rapid eye movement sleep behavior disorder: the potential role of glutamate, gamma-aminobutyric acid, and glycine. Sleep Med. 2013;14(8):714–8. doi:10.1016/j.sleep.2013.02.004.This paper provides an updated review on the latest discoveries on the pathophysiology of RBD, as the main prodromal sleep disorder in PD.

    Article  PubMed  Google Scholar 

  33. Garcia-Lorenzo D, Longo-Dos Santos C, Ewenczyk C, Leu-Semenescu S, Gallea C, Quattrocchi G, et al. The coeruleus/subcoeruleus complex in rapid eye movement sleep behaviour disorders in Parkinson’s disease. Brain. 2013;136(Pt 7):2120–9. doi:10.1093/brain/awt152.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Del Tredici K, Braak H. Dysfunction of the locus coeruleus-norepinephrine system and related circuitry in Parkinson’s disease-related dementia. J Neurol Neurosurg Psychiatry. 2013;84(7):774–83. doi:10.1136/jnnp-2011-301817.

    Article  PubMed  Google Scholar 

  35. • Ehrminger M, Latimier A, Pyatigorskaya N, Garcia-Lorenzo D, Leu-Semenescu S, Vidailhet M, et al. The coeruleus/subcoeruleus complex in idiopathic rapid eye movement sleep behaviour disorder. Brain. 2016;139(Pt 4):1180–8. doi:10.1093/brain/aww006.Using magnetic resonance imaging, authors have investigated the pathophysiology of RBD, which is the strongest prodromal feature of PD.

    Article  PubMed  Google Scholar 

  36. Von Economo C. Sleep as a problem of localization. J Nerv Ment Dis. 1930;71:249–59.

    Article  Google Scholar 

  37. Saper CB, Chou TC, Scammell TE. The sleep switch: hypothalamic control of sleep and wakefulness. Trends Neurosci. 2001;24(12):726–31.

    Article  CAS  PubMed  Google Scholar 

  38. Kalaitzakis ME, Gentleman SM, Pearce RK. Disturbed sleep in Parkinson’s disease: anatomical and pathological correlates. Neuropathol Appl Neurobiol. 2013;39(6):644–53. doi:10.1111/nan.12024.

    Article  CAS  PubMed  Google Scholar 

  39. Brown RE, McKenna JT. Turning a negative into a positive: ascending GABAergic control of cortical activation and arousal. Front Neurol. 2015;6:135. doi:10.3389/fneur.2015.00135.

    PubMed  PubMed Central  Google Scholar 

  40. Zhu K, van Hilten JJ, Marinus J. Course and risk factors for excessive daytime sleepiness in Parkinson’s disease. Parkinsonism Relat Disord. 2016;24:34–40. doi:10.1016/j.parkreldis.2016.01.020.

    Article  PubMed  Google Scholar 

  41. Arnulf I. Excessive daytime sleepiness in parkinsonism. Sleep Med Rev. 2005;9(3):185–200. doi:10.1016/j.smrv.2005.01.001.

    Article  PubMed  Google Scholar 

  42. Abbott RD, Ross GW, White LR, Tanner CM, Masaki KH, Nelson JS, et al. Excessive daytime sleepiness and subsequent development of Parkinson disease. Neurology. 2005;65(9):1442–6. doi:10.1212/01.wnl.0000183056.89590.0d.

    Article  CAS  PubMed  Google Scholar 

  43. Gao J, Huang X, Park Y, Hollenbeck A, Blair A, Schatzkin A, et al. Daytime napping, nighttime sleeping, and Parkinson disease. Am J Epidemiol. 2011;173(9):1032–8. doi:10.1093/aje/kwq478.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Simuni T, Caspell-Garcia C, Coffey C, Chahine LM, Lasch S, Oertel WH, et al. Correlates of excessive daytime sleepiness in de novo Parkinson’s disease: a case control study. Mov Disord. 2015;30(10):1371–81. doi:10.1002/mds.26248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mollenhauer B, Trautmann E, Sixel-Doring F, Wicke T, Ebentheuer J, Schaumburg M, et al. Nonmotor and diagnostic findings in subjects with de novo Parkinson disease of the DeNoPa cohort. Neurology. 2013;81(14):1226–34. doi:10.1212/WNL.0b013e3182a6cbd5.

    Article  PubMed  Google Scholar 

  46. • Arnulf I, Neutel D, Herlin B, Golmard JL, Leu-Semenescu S, Cochen de Cock V, et al. Sleepiness in idiopathic REM sleep behavior disorder and Parkinson disease. Sleep. 2015;38(10):1529–35. doi:10.5665/sleep.5040.Findings from this study showed that people with RBD who also had sleepiness converted faster to a neurodegenerative status.

    Article  PubMed  PubMed Central  Google Scholar 

  47. • Zhou J, Zhang J, Lam SP, Chan JW, Mok V, Chan A, et al. Excessive daytime sleepiness predicts neurodegeneration in idiopathic REM sleep behavior disorder. Sleep. 2017; doi:10.1093/sleep/zsx041.In this article, authors have shown that individuals with both RBD and excessive daytime somnolence are at a higher risk for conversion to PD.

    Google Scholar 

  48. Postuma RB, Gagnon JF, Pelletier A, Montplaisir J. Insomnia and somnolence in idiopathic RBD: a prospective cohort study. npj Parkinson’s Disease 2017;3(9).

  49. Fereshtehnejad SM, Romenets SR, Anang JB, Latreille V, Gagnon JF, Postuma RB. New clinical subtypes of Parkinson disease and their longitudinal progression: a prospective cohort comparison with other phenotypes. JAMA Neurol. 2015;72(8):863–73. doi:10.1001/jamaneurol.2015.0703.

    Article  PubMed  Google Scholar 

  50. Fereshtehnejad SM, Zeighami Y, Dagher A, Postuma RB. Clinical criteria for subtyping parkinson’s disease: comparison of imaging, CSF and genetic biomarkers and longitudinal progression. Brain. 2017:[Submitted].

  51. Barone P, Antonini A, Colosimo C, Marconi R, Morgante L, Avarello TP, et al. The PRIAMO study: a multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson’s disease. Mov Disord. 2009;24(11):1641–9. doi:10.1002/mds.22643.

    Article  PubMed  Google Scholar 

  52. Ratti PL, Negre-Pages L, Perez-Lloret S, Manni R, Damier P, Tison F, et al. Subjective sleep dysfunction and insomnia symptoms in Parkinson’s disease: insights from a cross-sectional evaluation of the French CoPark cohort. Parkinsonism Relat Disord. 2015;21(11):1323–9. doi:10.1016/j.parkreldis.2015.09.025.

    Article  PubMed  Google Scholar 

  53. Zhu K, van Hilten JJ, Marinus J. The course of insomnia in Parkinson’s disease. Parkinsonism Relat Disord. 2016;33:51–7. doi:10.1016/j.parkreldis.2016.09.010.

    Article  PubMed  Google Scholar 

  54. Schrag A, Horsfall L, Walters K, Noyce A, Petersen I. Prediagnostic presentations of Parkinson’s disease in primary care: a case-control study. Lancet Neurol. 2015;14(1):57–64. doi:10.1016/S1474-4422(14)70287-X.

    Article  PubMed  Google Scholar 

  55. Braga-Neto P, da Silva-Junior FP, Sueli Monte F, de Bruin PF, de Bruin VM. Snoring and excessive daytime sleepiness in Parkinson’s disease. J Neurol Sci. 2004;217(1):41–5.

    Article  PubMed  Google Scholar 

  56. Maria B, Sophia S, Michalis M, Charalampos L, Andreas P, John ME, et al. Sleep breathing disorders in patients with idiopathic Parkinson’s disease. Respir Med. 2003;97(10):1151–7.

    Article  PubMed  Google Scholar 

  57. Shpirer I, Miniovitz A, Klein C, Goldstein R, Prokhorov T, Theitler J, et al. Excessive daytime sleepiness in patients with Parkinson’s disease: a polysomnography study. Mov Disord. 2006;21(9):1432–8. doi:10.1002/mds.21002.

    Article  PubMed  Google Scholar 

  58. Zeng J, Wei M, Li T, Chen W, Feng Y, Shi R, et al. Risk of obstructive sleep apnea in Parkinson’s disease: a meta-analysis. PLoS One. 2013;8(12):e82091. doi:10.1371/journal.pone.0082091.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Cochen De Cock V, Benard-Serre N, Driss V, Granier M, Charif M, Carlander B, et al. Supine sleep and obstructive sleep apnea syndrome in Parkinson’s disease. Sleep Med. 2015;16(12):1497–501. doi:10.1016/j.sleep.2014.09.014.

    Article  PubMed  Google Scholar 

  60. Beland SG, Postuma RB, Latreille V, Bertrand JA, Panisset M, Chouinard S, et al. Observational study of the relation between Parkinson’s disease and sleep apnea. J Parkinsons Dis. 2015;5(4):805–11. doi:10.3233/JPD-150602.

    Article  PubMed  Google Scholar 

  61. Yeh NC, Tien KJ, Yang CM, Wang JJ, Weng SF. Increased risk of Parkinson’s disease in patients with obstructive sleep apnea: a population-based, propensity score-matched, longitudinal follow-up study. Medicine (Baltimore). 2016;95(2):e2293. doi:10.1097/MD.0000000000002293.

    Article  Google Scholar 

  62. Chen JC, Tsai TY, Li CY, Hwang JH. Obstructive sleep apnea and risk of Parkinson’s disease: a population-based cohort study. J Sleep Res. 2015;24(4):432–7. doi:10.1111/jsr.12289.

    Article  PubMed  Google Scholar 

  63. Sheu JJ, Lee HC, Lin HC, Kao LT, Chung SD. A 5-year follow-up study on the relationship between obstructive sleep apnea and Parkinson disease. J Clin Sleep Med. 2015;11(12):1403–8. doi:10.5664/jcsm.5274.

    Article  PubMed  PubMed Central  Google Scholar 

  64. White C, Hill EA, Morrison I, Riha RL. Diagnostic delay in REM sleep behavior disorder (RBD). J Clin Sleep Med. 2012;8(2):133–6. doi:10.5664/jcsm.1762.

    PubMed  PubMed Central  Google Scholar 

  65. Medicine. AAoS. International classification of sleep disorders. 3rd ed. Darien, USA American Academy of Sleep Medicine; 2014.

  66. Boeve BF, Molano JR, Ferman TJ, Smith GE, Lin SC, Bieniek K, et al. Validation of the Mayo Sleep Questionnaire to screen for REM sleep behavior disorder in an aging and dementia cohort. Sleep Med. 2011;12(5):445–53. doi:10.1016/j.sleep.2010.12.009.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Boeve BF, Molano JR, Ferman TJ, Lin SC, Bieniek K, Tippmann-Peikert M, et al. Validation of the Mayo Sleep Questionnaire to screen for REM sleep behavior disorder in a community-based sample. J Clin Sleep Med. 2013;9(5):475–80. doi:10.5664/jcsm.2670.

    PubMed  PubMed Central  Google Scholar 

  68. Bolitho SJ, Naismith SL, Terpening Z, Grunstein RR, Melehan K, Yee BJ, et al. Investigating rapid eye movement sleep without atonia in Parkinson’s disease using the rapid eye movement sleep behavior disorder screening questionnaire. Mov Disord. 2014;29(6):736–42. doi:10.1002/mds.25832.

    Article  PubMed  Google Scholar 

  69. Louter M, Arends JB, Bloem BR, Overeem S. Actigraphy as a diagnostic aid for REM sleep behavior disorder in Parkinson’s disease. BMC Neurol. 2014;14:76. doi:10.1186/1471-2377-14-76.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Postuma RB, Montplaisir JY. Large-scale population screening for prodromal PD: a way forward becomes clear. Sleep Med. 2016;24:148. doi:10.1016/j.sleep.2016.07.004.

    Article  PubMed  Google Scholar 

  71. Ohayon MM, Caulet M, Priest RG. Violent behavior during sleep. J Clin Psychiatry 1997;58(8):369–376; quiz 77.

  72. Chiu HF, Wing YK, Lam LC, Li SW, Lum CM, Leung T, et al. Sleep-related injury in the elderly—an epidemiological study in Hong Kong. Sleep. 2000;23(4):513–7.

    Article  CAS  PubMed  Google Scholar 

  73. Kang SH, Yoon IY, Lee SD, Han JW, Kim TH, Kim KW. REM sleep behavior disorder in the Korean elderly population: prevalence and clinical characteristics. Sleep. 2013;36(8):1147–52. doi:10.5665/sleep.2874.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Zhang J, Xu CY, Liu J. Meta-analysis on the prevalence of REM sleep behavior disorder symptoms in Parkinson’s disease. BMC Neurol. 2017;17(1):23. doi:10.1186/s12883-017-0795-4.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Chen H, Zhao EJ, Zhang W, Lu Y, Liu R, Huang X, et al. Meta-analyses on prevalence of selected Parkinson’s nonmotor symptoms before and after diagnosis. Transl Neurodegener. 2015;4(1):1. doi:10.1186/2047-9158-4-1.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Boeve BF, Silber MH, Ferman TJ. REM sleep behavior disorder in Parkinson’s disease and dementia with Lewy bodies. J Geriatr Psychiatry Neurol. 2004;17(3):146–57. doi:10.1177/0891988704267465.

    Article  PubMed  Google Scholar 

  77. Plazzi G, Corsini R, Provini F, Pierangeli G, Martinelli P, Montagna P, et al. REM sleep behavior disorders in multiple system atrophy. Neurology. 1997;48(4):1094–7.

    Article  CAS  PubMed  Google Scholar 

  78. Postuma RB, Adler CH, Dugger BN, Hentz JG, Shill HA, Driver-Dunckley E, et al. REM sleep behavior disorder and neuropathology in Parkinson’s disease. Mov Disord. 2015;30(10):1413–7. doi:10.1002/mds.26347.

    Article  CAS  PubMed  Google Scholar 

  79. Kunz D, Mahlberg R. A two-part, double-blind, placebo-controlled trial of exogenous melatonin in REM sleep behaviour disorder. J Sleep Res. 2010;19(4):591–6. doi:10.1111/j.1365-2869.2010.00848.x.

    Article  PubMed  Google Scholar 

  80. McCarter SJ, Boswell CL, St Louis EK, Dueffert LG, Slocumb N, Boeve BF, et al. Treatment outcomes in REM sleep behavior disorder. Sleep Med. 2013;14(3):237–42. doi:10.1016/j.sleep.2012.09.018.

    Article  PubMed  PubMed Central  Google Scholar 

  81. • Aurora RN, Zak RS, Maganti RK, Auerbach SH, Casey KR, Chowdhuri S, et al. Best practice guide for the treatment of REM sleep behavior disorder (RBD). J Clin Sleep Med. 2010;6(1):85–95.This review provides evidence-based guide for symptomatic treatment of RBD with critical appraisal of the clinical trials using different medications.

    PubMed  PubMed Central  Google Scholar 

  82. •• Postuma RB, Iranzo A, Hogl B, Arnulf I, Ferini-Strambi L, Manni R, et al. Risk factors for neurodegeneration in idiopathic rapid eye movement sleep behavior disorder: a multicenter study. Ann Neurol. 2015;77(5):830–9. doi:10.1002/ana.24385.In this article, original data has been presented on a large multicenter study from 12 different sites on the risk factors of phenoconversion to neurodegenerative state in people with prodromal RBD.

    Article  PubMed  Google Scholar 

  83. Sasai T, Inoue Y, Matsuura M. Effectiveness of pramipexole, a dopamine agonist, on rapid eye movement sleep behavior disorder. Tohoku J Exp Med. 2012;226(3):177–81.

    Article  CAS  PubMed  Google Scholar 

  84. Kashihara K, Nomura T, Maeda T, Tsuboi Y, Mishima T, Takigawa H, et al. Beneficial effects of ramelteon on rapid eye movement sleep behavior disorder associated with Parkinson’s disease—results of a multicenter open trial. Intern Med. 2016;55(3):231–6. doi:10.2169/internalmedicine.55.5464.

    Article  CAS  PubMed  Google Scholar 

  85. Esaki Y, Kitajima T, Koike S, Fujishiro H, Iwata Y, Tsuchiya A, et al. An open-labeled trial of ramelteon in idiopathic rapid eye movement sleep behavior disorder. J Clin Sleep Med. 2016;12(5):689–93. doi:10.5664/jcsm.5796.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Iranzo A, Fernandez-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. doi:10.1371/journal.pone.0089741.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Postuma RB, Gagnon JF, Bertrand JA, Genier Marchand D, Montplaisir JY. Parkinson risk in idiopathic REM sleep behavior disorder: preparing for neuroprotective trials. Neurology. 2015;84(11):1104–13. doi:10.1212/WNL.0000000000001364.

    Article  PubMed  PubMed Central  Google Scholar 

  88. 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. doi:10.1016/j.sleep.2012.10.009.

    Article  PubMed  Google Scholar 

  89. Wing YK, Li SX, Mok V, Lam SP, Tsoh J, Chan A, et al. Prospective outcome of rapid eye movement sleep behaviour disorder: psychiatric disorders as a potential early marker of Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2012;83(4):470–2. doi:10.1136/jnnp-2011-301232.

    Article  PubMed  Google Scholar 

  90. Mahlknecht P, Iranzo A, Hogl B, Frauscher B, Muller C, Santamaria J, et al. Olfactory dysfunction predicts early transition to a Lewy body disease in idiopathic RBD. Neurology. 2015;84(7):654–8. doi:10.1212/WNL.0000000000001265.

    Article  PubMed  Google Scholar 

  91. Li Y, Kang Y, Yang Q, Zhang L, Zhang L, Dong F et al. Predictive markers for early conversion of iRBD to neurodegenerative synucleinopathy diseases. Neurology. 2017;[in press].

  92. Fereshtehnejad SM, Montplaisir JY, Pelletier A, Gagnon JF, Berg D, Postuma RB. Validation of the MDS research criteria for prodromal Parkinson’s disease: longitudinal assessment in a REM sleep behavior disorder (RBD) cohort. Mov Disord. 2017:[Submitted].

  93. • Berg D, Postuma RB, Adler CH, Bloem BR, Chan P, Dubois B, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30(12):1600–11. doi:10.1002/mds.26431.In this methodologic article, the Movement Disorders Society has introduced the research criteria for prodromal PD, consisting of two sleep disorders, RBD and excessive daytime somnolence.

    Article  PubMed  Google Scholar 

  94. Boeve BF. REM sleep behavior disorder: updated review of the core features, the REM sleep behavior disorder-neurodegenerative disease association, evolving concepts, controversies, and future directions. Ann N Y Acad Sci. 2010;1184:15–54. doi:10.1111/j.1749-6632.2009.05115.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Siegel JM. The neurobiology of sleep. Semin Neurol. 2009;29(4):277–96. doi:10.1055/s-0029-1237118.

    Article  PubMed  Google Scholar 

  96. Vetrivelan R, Fuller PM, Tong Q, Lu J. Medullary circuitry regulating rapid eye movement sleep and motor atonia. J Neurosci. 2009;29(29):9361–9. doi:10.1523/JNEUROSCI.0737-09.2009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ronald B. Postuma MD MSc.

Ethics declarations

Conflict of Interest

Ahmed Al-Qassabi declares that he has no conflict of interest.

Seyed-Mohammad Fereshtehnejad has received Richard and Edith Strauss Postdoctoral Fellowship (McGill University, Canada) and Preston Robb Fellowship (Montreal Neurological Institute, Canada).

Ronald B. Postuma has received personal compensation for travel, speaker fees, and consultation from Biotie, Biogen, Boehringer-Ingelheim, Roche, and Teva Neurosciences and is funded by grants from the Fonds de Recherche du Québec–Santé, the Michael J. Fox Foundation, the W. Garfield Weston Foundation, and the Canadian Institutes of Health Research.

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 Disorders

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Al-Qassabi, A., Fereshtehnejad, SM. & Postuma, R.B. Sleep Disturbances in the Prodromal Stage of Parkinson Disease. Curr Treat Options Neurol 19, 22 (2017). https://doi.org/10.1007/s11940-017-0458-1

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11940-017-0458-1

Keywords

Navigation