Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

'The clocks that time us'—circadian rhythms in neurodegenerative disorders

Key Points

  • Sleep and circadian disruption are common in Alzheimer disease, Parkinson disease and Huntington disease

  • Symptoms of circadian disruption range from subjective sleep complaints and alterations of sleep timing to severe disruptions in rest–activity cycles and sleep architecture

  • Behavioural, physiological and molecular markers of circadian system function suggest progressive deterioration of circadian rhythmicity as the disease progresses

  • The relationship between circadian dysfunction and neurodegenerative processes might be bidirectional: circadian dysfunction might exacerbate the disease processes

  • Behavioural or pharmaceutical interventions that target sleep–wake cycles might reverse circadian disruption

Abstract

Circadian rhythms are physiological and behavioural cycles generated by an endogenous biological clock, the suprachiasmatic nucleus. The circadian system influences the majority of physiological processes, including sleep–wake homeostasis. Impaired sleep and alertness are common symptoms of neurodegenerative disorders, and circadian dysfunction might exacerbate the disease process. The pathophysiology of sleep–wake disturbances in these disorders remains largely unknown, and is presumably multifactorial. Circadian rhythm dysfunction is often observed in patients with Alzheimer disease, in whom it has a major impact on quality of life and represents one of the most important factors leading to institutionalization of patients. Similarly, sleep and circadian problems represent common nonmotor features of Parkinson disease and Huntington disease. Clinical studies and experiments in animal models of neurodegenerative disorders have revealed the progressive nature of circadian dysfunction throughout the course of neurodegeneration, and suggest strategies for the restoration of circadian rhythmicity involving behavioural and pharmacological interventions that target the sleep–wake cycle. In this Review, we discuss the role of the circadian system in the regulation of the sleep–wake cycle, and outline the implications of disrupted circadian timekeeping in neurodegenerative diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: A simplified scheme of the circadian system.
Figure 2: Molecular organization of the circadian system.

Similar content being viewed by others

References

  1. Mohawk, J. A., Green, C. B. & Takahashi, J. S. Central and peripheral circadian clocks in mammals. Annu. Rev. Neurosci. 35, 445–462 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Swaab, D. F., Fliers, E. & Partiman, T. S. The suprachiasmatic nucleus of the human brain in relation to sex, age and senile dementia. Brain Res. 342, 37–44 (1985).

    Article  CAS  PubMed  Google Scholar 

  3. Cassone, V. M., Speh, J. C., Card, J. P. & Moore, R. Y. Comparative anatomy of the mammalian hypothalamic suprachiasmatic nucleus. J. Biol. Rhythms 3, 71–91 (1988).

    Article  CAS  PubMed  Google Scholar 

  4. Golombek, D. A. & Rosenstein, R. E. Physiology of circadian entrainment. Physiol. Rev. 90, 1063–1102 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Dardente, H. & Cermakian, N. Molecular circadian rhythms in central and peripheral clocks in mammals. Chronobiol. Int. 24, 195–213 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Moore, R. Y. The suprachiasmatic nucleus and the circadian timing system. Prog. Mol. Biol. Transl. Sci. 119, 1–28 (2013).

    Article  PubMed  Google Scholar 

  7. Ballard, C. et al. Alzheimer's disease. Lancet 377, 1019–1031 (2011).

    Article  PubMed  Google Scholar 

  8. Ittner, L. M. & Götz, J. Amyloid-β and tau-—a toxic pas de deux in Alzheimer's disease. Nat. Rev. Neurosci. 12, 65–72 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  10. Bianchetti, A. et al. Predictors of mortality and institutionalization in Alzheimer disease patients 1 year after discharge from an Alzheimer dementia unit. Dementia 6, 108–112 (1995).

    CAS  PubMed  Google Scholar 

  11. Pollak, C. P. & Perlick, D. Sleep problems and institutionalization of the elderly. J. Geriatr. Psychiatry Neurol. 4, 204–210 (1991).

    CAS  PubMed  Google Scholar 

  12. Harper, D. G. et al. Dementia severity and Lewy bodies affect circadian rhythms in Alzheimer disease. Neurobiol. Aging 25, 771–781 (2004).

    Article  CAS  PubMed  Google Scholar 

  13. Satlin, A., Volicer, L., Stopa, E. G. & Harper, D. Circadian locomotor activity and core-body temperature rhythms in Alzheimer's disease. Neurobiol. Aging 16, 765–771 (1995).

    Article  CAS  PubMed  Google Scholar 

  14. Harper, D. G. et al. Disturbance of endogenous circadian rhythm in aging and Alzheimer disease. Am. J. Geriatr Psychiatry 13, 359–368 (2005).

    Article  PubMed  Google Scholar 

  15. Most, E. I., Scheltens, P. & Van Someren, E. J. Increased skin temperature in Alzheimer's disease is associated with sleepiness. J. Neural Transm. 119, 1185–1194 (2012).

    Article  PubMed  Google Scholar 

  16. Witting, W., Kwa, I. H., Eikelenboom, P., Mirmiran, M. & Swaab, D. F. Alterations in the circadian rest–activity rhythm in aging and Alzheimer's disease. Biol. Psychiatry 27, 563–572 (1990).

    Article  CAS  PubMed  Google Scholar 

  17. van Someren, E. J. et al. Circadian rest–activity rhythm disturbances in Alzheimer's disease. Biol. Psychiatry 40, 259–270 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Tranah, G. J. et al. Circadian activity rhythms and risk of incident dementia and mild cognitive impairment in older women. Ann. Neurol. 70, 722–732 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Hatfield, C. F., Herbert, J., van Someren, E. J., Hodges, J. R. & Hastings, M. H. Disrupted daily activity/rest cycles in relation to daily cortisol rhythms of home-dwelling patients with early Alzheimer's dementia. Brain 127, 1061–1074 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Manconi, M., Hutchins, W., Feroah, T. R., Zucconi, M. & Ferini-Strambi, L. On the pathway of an animal model for restless legs syndrome. Neurol. Sci. 28 (Suppl. 1), S53–S60 (2007).

    Article  PubMed  Google Scholar 

  21. Hu, K., Van Someren, E. J., Shea, S. A. & Scheer, F. A. Reduction of scale invariance of activity fluctuations with aging and Alzheimer's disease: Involvement of the circadian pacemaker. Proc. Natl Acad. Sci. USA 106, 2490–2494 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Skene, D. J. & Swaab, D. F. Melatonin rhythmicity: effect of age and Alzheimer's disease. Exp. Gerontol. 38, 199–206 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Rosales-Corral, S. A. et al. Alzheimer's disease: pathological mechanisms and the beneficial role of melatonin. J. Pineal Res. 52, 167–202 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Lin, L. et al. Melatonin in Alzheimer's disease. Int. J. Mol. Sci. 14, 14575–14593 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu, R. Y., Zhou, J. N., van Heerikhuize, J., Hofman, M. A. & Swaab, D. F. Decreased melatonin levels in postmortem cerebrospinal fluid in relation to aging, Alzheimer's disease, and apolipoprotein E-ε4/4 genotype. J. Clin. Endocrinol. Metab. 84, 323–327 (1999).

    CAS  PubMed  Google Scholar 

  26. Zhou, J. N., Liu, R. Y., Kamphorst, W., Hofman, M. A. & Swaab, D. F. Early neuropathological Alzheimer's changes in aged individuals are accompanied by decreased cerebrospinal fluid melatonin levels. J. Pineal Res. 35, 125–130 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Wu, Y. H. et al. Molecular changes underlying reduced pineal melatonin levels in Alzheimer disease: alterations in preclinical and clinical stages. J. Clin. Endocrinol. Metab. 88, 5898–5906 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Mishima, K. et al. Melatonin secretion rhythm disorders in patients with senile dementia of Alzheimer's type with disturbed sleep–waking. Biol. Psychiatry 45, 417–421 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Shan, L., Bossers, K., Unmehopa, U., Bao, A. M. & Swaab, D. F. Alterations in the histaminergic system in Alzheimer's disease: a postmortem study. Neurobiol. Aging 33, 2585–2598 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Shan, L. et al. Diurnal fluctuation in histidine decarboxylase expression, the rate limiting enzyme for histamine production, and its disorder in neurodegenerative diseases. Sleep 35, 713–715 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Wu, Y. H., Zhou, J. N., Van Heerikhuize, J., Jockers, R. & Swaab, D. F. Decreased MT1 melatonin receptor expression in the suprachiasmatic nucleus in aging and Alzheimer's disease. Neurobiol. Aging 28, 1239–1247 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Stopa, E. G. et al. Pathologic evaluation of the human suprachiasmatic nucleus in severe dementia. J. Neuropathol. Exp. Neurol. 58, 29–39 (1999).

    Article  CAS  PubMed  Google Scholar 

  33. Coogan, A. N., Rawlings, N., Luckman, S. M. & Piggins, H. D. Effects of neurotensin on discharge rates of rat suprachiasmatic nucleus neurons in vitro. Neuroscience 103, 663–672 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Zhou, J. N., Hofman, M. A. & Swaab, D. F. VIP neurons in the human SCN in relation to sex, age, and Alzheimer's disease. Neurobiol. Aging 16, 571–576 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Liu, R. Y. et al. Decreased vasopressin gene expression in the biological clock of Alzheimer disease patients with and without depression. J. Neuropathol. Exp. Neurol. 59, 314–322 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Coogan, A. N. et al. The circadian system in Alzheimer's disease: disturbances, mechanisms, and opportunities. Biol. Psychiatry 74, 333–339 (2013).

    Article  PubMed  Google Scholar 

  37. Friedland, R. P., Luxenberg, J. S. & Koss, E. A quantitative study of intracranial calcification in dementia of the Alzheimer type. Int. Psychogeriatr. 2, 36–43 (1990).

    Article  CAS  PubMed  Google Scholar 

  38. Wu, Y. H. et al. Pineal clock gene oscillation is disturbed in Alzheimer's disease, due to functional disconnection from the “master clock”. FASEB J. 20, 1874–1876 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Cermakian, N., Lamont, E. W., Boudreau, P. & Boivin, D. B. Circadian clock gene expression in brain regions of Alzheimer's disease patients and control subjects. J. Biol. Rhythms 26, 160–170 (2011).

    Article  PubMed  Google Scholar 

  40. Sterniczuk, R., Dyck, R. H., Laferla, F. M. & Antle, M. C. Characterization of the 3xTg-AD mouse model of Alzheimer's disease: part 1. Circadian changes. Brain Res. 1348, 139–148 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Knight, E. M. et al. Age-related changes in core body temperature and activity in triple-transgenic Alzheimer's disease (3xTgAD) mice. Dis. Model Mech. 6, 160–170 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Gorman, M. R. & Yellon, S. Lifespan daily locomotor activity rhythms in a mouse model of amyloid-induced neuropathology. Chronobiol. Int. 27, 1159–1177 (2010).

    Article  PubMed  Google Scholar 

  43. Wisor, J. P. et al. Sleep and circadian abnormalities in a transgenic mouse model of Alzheimer's disease: a role for cholinergic transmission. Neuroscience 131, 375–385 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Ambree, O. et al. Activity changes and marked stereotypic behavior precede Aβ pathology in TgCRND8 Alzheimer mice. Neurobiol. Aging 27, 955–964 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Slats, D., Claassen, J. A., Verbeek, M. M. & Overeem, S. Reciprocal interactions between sleep, circadian rhythms and Alzheimer's disease: focus on the role of hypocretin and melatonin. Ageing Res. Rev. 12, 188–200 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Wang, J. Z. & Wang, Z. F. Role of melatonin in Alzheimer-like neurodegeneration. Acta Pharmacol. Sin. 27, 41–49 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Ju, Y. E., Lucey, B. P. & Holtzman, D. M. Sleep and Alzheimer disease pathology—a bidirectional relationship. Nat. Rev. Neurol. 10, 115–119 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Kang, J. E. et al. Amyloid-β dynamics are regulated by orexin and the sleep–wake cycle. Science 326, 1005–1007 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Xie, L. et al. Sleep drives metabolite clearance from the adult brain. Science 342, 373–377 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Ooms, S. et al. Effect of 1 night of total sleep deprivation on cerebrospinal fluid β-amyloid 42 in healthy middle-aged men: a randomized clinical trial. JAMA Neurol. 71, 971–977 (2014).

    Article  PubMed  Google Scholar 

  51. Pappolla, M. et al. Inhibition of Alzheimer β-fibrillogenesis by melatonin. J. Biol. Chem. 273, 7185–7188 (1998).

    Article  CAS  PubMed  Google Scholar 

  52. Lahiri, D. K., Chen, D., Ge, Y. W., Bondy, S. C. & Sharman, E. H. Dietary supplementation with melatonin reduces levels of amyloid beta-peptides in the murine cerebral cortex. J. Pineal Res. 36, 224–231 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Matsubara, E. et al. Melatonin increases survival and inhibits oxidative and amyloid pathology in a transgenic model of Alzheimer's disease. J. Neurochem. 85, 1101–1108 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Asayama, K. et al. Double blind study of melatonin effects on the sleep–wake rhythm, cognitive and non-cognitive functions in Alzheimer type dementia. J. Nippon Med. Sch. 70, 334–341 (2003).

    Article  PubMed  Google Scholar 

  55. Cardinali, D. P., Furio, A. M. & Brusco, L. I. Clinical aspects of melatonin intervention in Alzheimer's disease progression. Curr. Neuropharmacol. 8, 218–227 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dowling, G. A. et al. Melatonin and bright-light treatment for rest–activity disruption in institutionalized patients with Alzheimer's disease. J. Am. Geriatr Soc. 56, 239–246 (2008).

    Article  PubMed  Google Scholar 

  57. Mahlberg, R. & Walther, S. Actigraphy in agitated patients with dementia. Monitoring treatment outcomes. Z. Gerontol. Geriatr. 40, 178–184 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Singer, C. et al. A multicenter, placebo-controlled trial of melatonin for sleep disturbance in Alzheimer's disease. Sleep 26, 893–901 (2003).

    Article  PubMed  Google Scholar 

  59. Gehrman, P. R. et al. Melatonin fails to improve sleep or agitation in double-blind randomized placebo-controlled trial of institutionalized patients with Alzheimer disease. Am. J. Geriatr Psychiatry 17, 166–169 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Jansen, S. L., Forbes, D. A., Duncan, V. & Morgan, D. G. Melatonin for cognitive impairment. Cochrane Database of Systematic Reviews, Issue 1. Art. No.: CD003802. http://dx.doi.org/10.1002/14651858.CD003802.pub3.

  61. Lyketsos, C. G., Lindell Veiel, L., Baker, A. & Steele, C. A randomized, controlled trial of bright light therapy for agitated behaviors in dementia patients residing in long-term care. Int. J. Geriatr Psychiatry 14, 520–525 (1999).

    Article  CAS  PubMed  Google Scholar 

  62. Ancoli-Israel, S. et al. Increased light exposure consolidates sleep and strengthens circadian rhythms in severe Alzheimer's disease patients. Behav. Sleep Med. 1, 22–36 (2003).

    Article  PubMed  Google Scholar 

  63. Burns, A., Allen, H., Tomenson, B., Duignan, D. & Byrne, J. Bright light therapy for agitation in dementia: a randomized controlled trial. Int. Psychogeriatr. 21, 711–721 (2009).

    Article  PubMed  Google Scholar 

  64. McCurry, S. M. et al. Increasing walking and bright light exposure to improve sleep in community-dwelling persons with Alzheimer's disease: results of a randomized, controlled trial. J. Am. Geriatr. Soc. 59, 1393–1402 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Dowling, G. A. et al. Effect of morning bright light treatment for rest–activity disruption in institutionalized patients with severe Alzheimer's disease. Int. Psychogeriatr. 17, 221–236 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Dowling, G. A., Mastick, J., Hubbard, E. M., Luxenberg, J. S. & Burr, R. L. Effect of timed bright light treatment for rest–activity disruption in institutionalized patients with Alzheimer's disease. Int. J. Geriatr. Psychiatry 20, 738–743 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Alves, G., Forsaa, E. B., Pedersen, K. F., Dreetz Gjerstad, M. & Larsen, J. P. Epidemiology of Parkinson's disease. J. Neurol. 255 (Suppl. 5), 18–32 (2008).

    Article  PubMed  Google Scholar 

  68. Dorsey, E. R. et al. Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology 68, 384–386 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Bruguerolle, B. & Simon, N. Biologic rhythms and Parkinson's disease: a chronopharmacologic approach to considering fluctuations in function. Clin. Neuropharmacol. 25, 194–201 (2002).

    Article  PubMed  Google Scholar 

  70. Willis, G. L. Parkinson's disease as a neuroendocrine disorder of circadian function: dopamine-melatonin imbalance and the visual system in the genesis and progression of the degenerative process. Rev. Neurosci. 19, 245–316 (2008).

    Article  CAS  PubMed  Google Scholar 

  71. Videnovic, A. & Golombek, D. Circadian and sleep disorders in Parkinson's disease. Exp. Neurol. 243, 45–56 (2013).

    Article  PubMed  Google Scholar 

  72. Bonuccelli, U. et al. Diurnal motor variations to repeated doses of levodopa in Parkinson's disease. Clin. Neuropharmacol. 23, 28–33 (2000).

    Article  CAS  PubMed  Google Scholar 

  73. Nutt, J. G., Woodward, W. R., Carter, J. H. & Trotman, T. L. Influence of fluctuations of plasma large neutral amino acids with normal diets on the clinical response to levodopa. J. Neurol. Neurosurg. Psychiatry 52, 481–487 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. van Hilten, J. J. et al. Assessment of response fluctuations in Parkinson's disease by ambulatory wrist activity monitoring. Acta Neurol. Scand. 87, 171–177 (1993).

    Article  CAS  PubMed  Google Scholar 

  75. van Hilten, J. J., Middelkoop, H. A., Kerkhof, G. A. & Roos, R. A. A new approach in the assessment of motor activity in Parkinson's disease. J. Neurol. Neurosurg. Psychiatry 54, 976–979 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Mihci, E., Kardelen, F., Dora, B. & Balkan, S. Orthostatic heart rate variability analysis in idiopathic Parkinson's disease. Acta Neurol. Scand. 113, 288–293 (2006).

    Article  CAS  PubMed  Google Scholar 

  77. Pathak, A. & Senard, J. M. Blood pressure disorders during Parkinson's disease: epidemiology, pathophysiology and management. Expert Rev. Neurother. 6, 1173–1180 (2006).

    Article  PubMed  Google Scholar 

  78. Pursiainen, V. et al. Circadian heart rate variability in Parkinson's disease. J. Neurol. 249, 1535–1540 (2002).

    Article  PubMed  Google Scholar 

  79. Comella, C. L. Sleep disorders in Parkinson's disease: an overview. Mov Disord. 22 (Suppl. 17), S367–S373 (2007).

    Article  PubMed  Google Scholar 

  80. Porter, B., Macfarlane, R. & Walker, R. The frequency and nature of sleep disorders in a community-based population of patients with Parkinson's disease. Eur. J. Neurol. 15, 50–54 (2008).

    CAS  PubMed  Google Scholar 

  81. Verbaan, D., van Rooden, S. M., Visser, M., Marinus, J. & van Hilten, J. J. Nighttime sleep problems and daytime sleepiness in Parkinson's disease. Mov. Disord. 23, 35–41 (2008).

    Article  PubMed  Google Scholar 

  82. Struck, L. K., Rodnitzky, R. L. & Dobson, J. K. Circadian fluctuations of contrast sensitivity in Parkinson's disease. Neurology 40, 467–470 (1990).

    Article  CAS  PubMed  Google Scholar 

  83. Piccini, P. et al. Diurnal worsening in Parkinson patients treated with levodopa [Italian]. Riv. Neurol. 61, 219–224 (1991).

    CAS  PubMed  Google Scholar 

  84. van Hilten, B. et al. Sleep disruption in Parkinson's disease. Assessment by continuous activity monitoring. Arch. Neurol. 51, 922–928 (1994).

    Article  CAS  PubMed  Google Scholar 

  85. Whitehead, D. L., Davies, A. D., Playfer, J. R. & Turnbull, C. J. Circadian rest–activity rhythm is altered in Parkinson's disease patients with hallucinations. Mov. Disord. 23, 1137–1145 (2008).

    Article  PubMed  Google Scholar 

  86. Kallio, M. et al. Heart rate variability in patients with untreated Parkinson's disease. Eur. J. Neurol. 7, 667–672 (2000).

    Article  CAS  PubMed  Google Scholar 

  87. Senard, J. M., Chamontin, B., Rascol, A. & Montastruc, J. L. Ambulatory blood pressure in patients with Parkinson's disease without and with orthostatic hypotension. Clin. Auton. Res. 2, 99–104 (1992).

    Article  CAS  PubMed  Google Scholar 

  88. Devos, D. et al. Heart rate variability and Parkinson's disease severity. J. Neural Transm. 110, 997–1011 (2003).

    Article  CAS  PubMed  Google Scholar 

  89. Wakabayashi, K. & Takahashi, H. Neuropathology of autonomic nervous system in Parkinson's disease. Eur. Neurol. 38 (Suppl. 2), 2–7 (1997).

    Article  PubMed  Google Scholar 

  90. Langston, J. The hypothalamus in Parkinson's disease. Ann. Neurol. 3, 129–133 (1978).

    Article  CAS  PubMed  Google Scholar 

  91. Wirz-Justice, A., Da Prada, M. & Reme, C. Circadian rhythm in rat retinal dopamine. Neurosci. Lett. 45, 21–25 (1984).

    Article  CAS  PubMed  Google Scholar 

  92. Dearry, A. & Burnside, B. Dopaminergic regulation of cone retinomotor movement in isolated teleost retinas: I. Induction of cone contraction is mediated by D2 receptors. J. Neurochem. 46, 1006–1021 (1986).

    Article  CAS  PubMed  Google Scholar 

  93. Raggi, A., Bella, R., Pennisi, G., Neri, W. & Ferri, R. Sleep disorders in Parkinson's disease: a narrative review of the literature. Rev. Neurosci. 24, 279–291 (2013).

    Article  PubMed  Google Scholar 

  94. Braak, H. & Del Tredici, K. Invited Article: Nervous system pathology in sporadic Parkinson disease. Neurology 70, 1916–1925 (2008).

    Article  PubMed  Google Scholar 

  95. Bordet, R. et al. Study of circadian melatonin secretion pattern at different stages of Parkinson's disease. Clin. Neuropharmacol. 26, 65–72 (2003).

    Article  CAS  PubMed  Google Scholar 

  96. Fertl, E., Auff, E., Doppelbauer, A. & Waldhauser, F. Circadian secretion pattern of melatonin in de novo parkinsonian patients: evidence for phase-shifting properties of l-dopa. J. Neural Transm. Park. Dis. Dement. Sect. 5, 227–234 (1993).

    Article  CAS  PubMed  Google Scholar 

  97. Hartmann, A., Veldhuis, J. D., Deuschle, M., Standhardt, H. & Heuser, I. Twenty-four hour cortisol release profiles in patients with Alzheimer's and Parkinson's disease compared to normal controls: ultradian secretory pulsatility and diurnal variation. Neurobiol. Aging 18, 285–289 (1997).

    Article  CAS  PubMed  Google Scholar 

  98. Aziz, N. A., Pijl, H., Frolich, M., Roelfsema, F. & Roos, R. A. Diurnal secretion profiles of growth hormone, thyrotrophin and prolactin in Parkinson's disease. J. Neuroendocrinol. 23, 519–524 (2011).

    Article  CAS  PubMed  Google Scholar 

  99. Pierangeli, G. et al. Nocturnal body core temperature falls in Parkinson's disease but not in multiple-system atrophy. Mov. Disord. 16, 226–232 (2001).

    Article  CAS  PubMed  Google Scholar 

  100. Cagnacci, A. et al. Effect of naloxone on body temperature in postmenopausal women with Parkinson's disease. Life Sci. 46, 1241–1247 (1990).

    Article  CAS  PubMed  Google Scholar 

  101. Suzuki, K. et al. Circadian variation of core body temperature in Parkinson disease patients with depression: a potential biological marker for depression in Parkinson disease. Neuropsychobiology 56, 172–179 (2007).

    Article  PubMed  Google Scholar 

  102. Videnovic, A. et al. Circadian melatonin rhythm and excessive daytime sleepiness in Parkinson disease. JAMA Neurol. 71, 463–469 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Breen, D. P. et al. Sleep and circadian rhythm regulation in early Parkinson disease. JAMA Neurol. 71, 589–595 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  104. Bolitho, S. J. et al. Disturbances in melatonin secretion and circadian sleep–wake regulation in Parkinson disease. Sleep Med. 15, 342–347 (2014).

    Article  CAS  PubMed  Google Scholar 

  105. Boivin, D. B. et al. Circadian clock genes oscillate in human peripheral blood mononuclear cells. Blood 102, 4143–4145 (2003).

    Article  CAS  PubMed  Google Scholar 

  106. Takimoto, M. et al. Daily expression of clock genes in whole blood cells in healthy subjects and a patient with circadian rhythm sleep disorder. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289, R1273–R1279 (2005).

    Article  CAS  PubMed  Google Scholar 

  107. Teboul, M. et al. Atypical patterns of circadian clock gene expression in human peripheral blood mononuclear cells. J. Mol. Med. 83, 693–699 (2005).

    Article  CAS  PubMed  Google Scholar 

  108. Cai, Y., Liu, S., Sothern, R. B., Xu, S. & Chan, P. Expression of clock genes Per1 and Bmal1 in total leukocytes in health and Parkinson's disease. Eur. J. Neurol. 17, 550–554 (2009).

    Article  PubMed  Google Scholar 

  109. Witkovsky, P. Dopamine and retinal function. Doc. Ophthalmol. 108, 17–40 (2004).

    Article  PubMed  Google Scholar 

  110. Harrell, L. E. & Balagura, S. The effects of dark and light on the functional recovery following lateral hypothalamic lesions. Life Sci. 15, 2079–2087 (1974).

    Article  CAS  PubMed  Google Scholar 

  111. Willis, G. L. & Turner, E. J. Primary and secondary features of Parkinson's disease improve with strategic exposure to bright light: a case series study. Chronobiol. Int. 24, 521–537 (2007).

    Article  PubMed  Google Scholar 

  112. Willis, G. L., Moore, C. & Armstrong, S. M. A historical justification for and retrospective analysis of the systematic application of light therapy in Parkinson's disease. Rev. Neurosci. 23, 199–226 (2012).

    CAS  PubMed  Google Scholar 

  113. Paus, S. et al. Bright light therapy in Parkinson's disease: a pilot study. Mov. Disord. 22, 1495–1498 (2007).

    Article  PubMed  Google Scholar 

  114. Barraud, Q. et al. Sleep disorders in Parkinson's disease: the contribution of the MPTP non-human primate model. Exp. Neurol. 219, 574–582 (2009).

    Article  CAS  PubMed  Google Scholar 

  115. Fifel, K. et al. Alteration of daily and circadian rhythms following dopamine depletion in MPTP treated non-human primates. PLoS ONE 9, e86240 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Vezoli, J. et al. Early presymptomatic and long-term changes of rest activity cycles and cognitive behavior in a MPTP-monkey model of Parkinson's disease. PLoS ONE 6, e23952 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Ben, V. & Bruguerolle, B. Effects of bilateral striatal 6-OHDA lesions on circadian rhythms in the rat: a radiotelemetric study. Life Sci. 67, 1549–1558 (2000).

    Article  CAS  PubMed  Google Scholar 

  118. Isobe, Y. & Nishino, H. Circadian rhythm of drinking and running-wheel activity in rats with 6-hydroxydopamine lesions of the ventral tegmental area. Brain Res. 899, 187–192 (2001).

    Article  CAS  PubMed  Google Scholar 

  119. Baier, P. C. et al. Circadian distribution of motor-activity in unilaterally 6-hydroxy-dopamine lesioned rats. Exp. Brain Res. 169, 283–288 (2006).

    Article  CAS  PubMed  Google Scholar 

  120. Gravotta, L., Gavrila, A. M., Hood, S. & Amir, S. Global depletion of dopamine using intracerebroventricular 6-hydroxydopamine injection disrupts normal circadian wheel-running patterns and PERIOD2 expression in the rat forebrain. J. Mol. Neurosci. 45, 162–171 (2011).

    Article  CAS  PubMed  Google Scholar 

  121. Kudo, T., Loh, D. H., Truong, D., Wu, Y. & Colwell, C. S. Circadian dysfunction in a mouse model of Parkinson's disease. Exp. Neurol. 232, 66–75 (2011).

    Article  PubMed  Google Scholar 

  122. Willison, L. D., Kudo, T., Loh, D. H., Kuljis, D. & Colwell, C. S. Circadian dysfunction may be a key component of the non-motor symptoms of Parkinson's disease: insights from a transgenic mouse model. Exp. Neurol. 243, 57–66 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Morrison, P. J. Accurate prevalence and uptake of testing for Huntington's disease. Lancet Neurol. 9, 1147 (2010).

    Article  PubMed  Google Scholar 

  124. Harper, P. S. The epidemiology of Huntington's disease. Hum. Genet. 89, 365–376 (1992).

    Article  CAS  PubMed  Google Scholar 

  125. Snell, R. G. et al. Relationship between trinucleotide repeat expansion and phenotypic variation in Huntington's disease. Nat. Genet. 4, 393–397 (1993).

    Article  CAS  PubMed  Google Scholar 

  126. Gusella, J. F. et al. A polymorphic DNA marker genetically linked to Huntington's disease. Nature 306, 234–238 (1983).

    Article  CAS  PubMed  Google Scholar 

  127. Gilliam, T. C. et al. A DNA segment encoding two genes very tightly linked to Huntington's disease. Science 238, 950–952 (1987).

    Article  CAS  PubMed  Google Scholar 

  128. [No authors listed] A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. The Huntington's Disease Collaborative Research Group. Cell 72, 971–983 (1993).

  129. Bamford, K. A., Caine, E. D., Kido, D. K., Cox, C. & Shoulson, I. A prospective evaluation of cognitive decline in early Huntington's disease: functional and radiographic correlates. Neurology 45, 1867–1873 (1995).

    Article  CAS  PubMed  Google Scholar 

  130. Paulsen, J. S., Ready, R. E., Hamilton, J. M., Mega, M. S. & Cummings, J. L. Neuropsychiatric aspects of Huntington's disease. J. Neurol. Neurosurg. Psychiatry 71, 310–314 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Stoy, N. & McKay, E. Weight loss in Huntington's disease. Ann. Neurol. 48, 130–131 (2000).

    Article  CAS  PubMed  Google Scholar 

  132. Goodman, A. O. et al. Asymptomatic sleep abnormalities are a common early feature in patients with Huntington's disease. Curr. Neurol. Neurosci. Rep. 11, 211–217 (2011).

    Article  PubMed  Google Scholar 

  133. Hansotia, P., Wall, R. & Berendes, J. Sleep disturbances and severity of Huntington's disease. Neurology 35, 1672–1674 (1985).

    Article  CAS  PubMed  Google Scholar 

  134. Emser, W., Brenner, M., Stober, T. & Schimrigk, K. Changes in nocturnal sleep in Huntington's and Parkinson's disease. J. Neurol. 235, 177–179 (1988).

    Article  CAS  PubMed  Google Scholar 

  135. Silvestri, R. et al. Sleep features in Tourette's syndrome, neuroacanthocytosis and Huntington's chorea. Neurophysiol. Clin. 25, 66–77 (1995).

    Article  CAS  PubMed  Google Scholar 

  136. Cuturic, M., Abramson, R. K., Vallini, D., Frank, E. M. & Shamsnia, M. Sleep patterns in patients with Huntington's disease and their unaffected first-degree relatives: a brief report. Behav. Sleep Med. 7, 245–254 (2009).

    Article  PubMed  Google Scholar 

  137. Arnulf, I. et al. Rapid eye movement sleep disturbances in Huntington disease. Arch. Neurol. 65, 482–488 (2008).

    Article  PubMed  Google Scholar 

  138. Morton, A. J. et al. Disintegration of the sleep–wake cycle and circadian timing in Huntington's disease. J. Neurosci. 25, 157–163 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Kudo, T. et al. Dysfunctions in circadian behavior and physiology in mouse models of Huntington's disease. Exp. Neurol. 228, 80–90 (2011).

    Article  CAS  PubMed  Google Scholar 

  140. Lazar, A. S. et al. Sleep, diurnal preference, health, and psychological well-being: a prospective single-allelic-variation study. Chronobiol. Int. 29, 131–146 (2012).

    Article  CAS  PubMed  Google Scholar 

  141. Epping, E. A. & Paulsen, J. S. Depression in the early stages of Huntington disease. Neurodegener. Dis. Manag. 1, 407–414 (2011).

    Article  PubMed  Google Scholar 

  142. Goodman, A. O., Morton, A. J. & Barker, R. A. Identifying sleep disturbances in Huntington's disease using a simple disease-focused questionnaire. PLoS Curr. 2, RRN1189 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  143. Aziz, N. A., Anguelova, G. V., Marinus, J., Lammers, G. J. & Roos, R. A. Sleep and circadian rhythm alterations correlate with depression and cognitive impairment in Huntington's disease. Parkinsonism Relat. Disord. 16, 345–350 (2010).

    Article  PubMed  Google Scholar 

  144. Taylor, N. & Bramble, D. Sleep disturbance and Huntingdon's disease. Br. J. Psychiatry 171, 393 (1997).

    Article  CAS  PubMed  Google Scholar 

  145. Videnovic, A., Leurgans, S., Fan, W., Jaglin, J. & Shannon, K. M. Daytime somnolence and nocturnal sleep disturbances in Huntington disease. Parkinsonism Relat. Disord. 15, 471–474 (2009).

    Article  PubMed  Google Scholar 

  146. Starr, A. A disorder of rapid eye movements in Huntington's chorea. Brain 90, 545–564 (1967).

    Article  CAS  PubMed  Google Scholar 

  147. Wiegand, M. et al. Nocturnal sleep in Huntington's disease. J. Neurol. 238, 203–208 (1991).

    Article  CAS  PubMed  Google Scholar 

  148. van Vugt, J. P., van Hilten, B. J. & Roos, R. A. Hypokinesia in Huntington's disease. Mov. Disord. 11, 384–388 (1996).

    Article  CAS  PubMed  Google Scholar 

  149. van Vugt, J. P. et al. Quantitative assessment of daytime motor activity provides a responsive measure of functional decline in patients with Huntington's disease. Mov. Disord. 16, 481–488 (2001).

    Article  CAS  PubMed  Google Scholar 

  150. Hurelbrink, C. B., Lewis, S. J. & Barker, R. A. The use of the Actiwatch-Neurologica system to objectively assess the involuntary movements and sleep–wake activity in patients with mild-moderate Huntington's disease. J. Neurol. 252, 642–647 (2005).

    Article  PubMed  Google Scholar 

  151. Pouladi, M. A., Morton, A. J. & Hayden, M. R. Choosing an animal model for the study of Huntington's disease. Nat. Rev. Neurosci. 14, 708–721 (2013).

    Article  CAS  PubMed  Google Scholar 

  152. Shirbin, C. A. et al. Cortisol and depression in pre-diagnosed and early stage Huntington's disease. Psychoneuroendocrinology 38, 2439–2447 (2013).

    Article  CAS  PubMed  Google Scholar 

  153. Kantor, S., Szabo, L., Varga, J., Cuesta, M. & Morton, A. J. Progressive sleep and electroencephalogram changes in mice carrying the Huntington's disease mutation. Brain 136, 2147–2158 (2013).

    Article  PubMed  Google Scholar 

  154. Fisher, S. P. et al. Longitudinal analysis of the electroencephalogram and sleep phenotype in the R6/2 mouse model of Huntington's disease. Brain 136, 2159–2172 (2013).

    Article  PubMed  Google Scholar 

  155. Fahrenkrug, J., Popovic, N., Georg, B., Brundin, P. & Hannibal, J. Decreased VIP and VPAC2 receptor expression in the biological clock of the R6/2 Huntington's disease mouse. J. Mol. Neurosci. 31, 139–148 (2007).

    CAS  PubMed  Google Scholar 

  156. Rudenko, O., Tkach, V., Berezin, V. & Bock, E. Detection of early behavioral markers of Huntington's disease in R6/2 mice employing an automated social home cage. Behav. Brain Res. 203, 188–199 (2009).

    Article  PubMed  Google Scholar 

  157. Bode, F. J. et al. Increased numbers of motor activity peaks during light cycle are associated with reductions in adrenergic α-receptor levels in a transgenic Huntington's disease rat model. Behav. Brain Res. 205, 175–182 (2009).

    Article  CAS  PubMed  Google Scholar 

  158. Pietropaolo, S., Delage, P., Cayzac, S., Crusio, W. E. & Cho, Y. H. Sex-dependent changes in social behaviors in motor pre-symptomatic R6/1 mice. PLoS ONE 6, e19965 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Loh, D. H., Kudo, T., Truong, D., Wu, Y. & Colwell, C. S. The Q175 mouse model of Huntington's disease shows gene dosage- and age-related decline in circadian rhythms of activity and sleep. PLoS ONE 8, e69993 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Heuser, I. J., Chase, T. N. & Mouradian, M. M. The limbic-hypothalamic-pituitary-adrenal axis in Huntington's disease. Biol. Psychiatry 30, 943–952 (1991).

    Article  CAS  PubMed  Google Scholar 

  161. Pallier, P. N. et al. Pharmacological imposition of sleep slows cognitive decline and reverses dysregulation of circadian gene expression in a transgenic mouse model of Huntington's disease. J. Neurosci. 27, 7869–7878 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Pallier, P. N. & Morton, A. J. Management of sleep/wake cycles improves cognitive function in a transgenic mouse model of Huntington's disease. Brain Res. 1279, 90–98 (2009).

    Article  CAS  PubMed  Google Scholar 

  163. Wood, N. I. et al. Responses to environmental enrichment differ with sex and genotype in a transgenic mouse model of Huntington's disease. PLoS ONE 5, e9077 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Aziz, N. A. et al. Delayed onset of the diurnal melatonin rise in patients with Huntington's disease. J. Neurol. 256, 1961–1965 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Kalliolia, E. et al. Plasma melatonin is reduced in Huntington's disease. Mov. Disord. (2014).

  166. Saleh, N. et al. Neuroendocrine disturbances in Huntington's disease. PLoS ONE 4, e4962 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Aziz, N. A. et al. Increased hypothalamic–pituitary–adrenal axis activity in Huntington's disease. J. Clin. Endocrinol. Metab. 94, 1223–1228 (2009).

    Article  CAS  PubMed  Google Scholar 

  168. Kalsbeek, A., Buijs, R. M., van Heerikhuize, J. J., Arts, M. & van der Woude, T. P. Vasopressin-containing neurons of the suprachiasmatic nuclei inhibit corticosterone release. Brain Res. 580, 62–67 (1992).

    Article  CAS  PubMed  Google Scholar 

  169. van Wamelen, D. J. et al. Suprachiasmatic nucleus neuropeptide expression in patients with Huntington's Disease. Sleep 36, 117–125 (2013).

    PubMed  PubMed Central  Google Scholar 

  170. Kudo, T. et al. Circadian dysfunction in response to in vivo treatment with the mitochondrial toxin 3-nitropropionic acid. ASN Neuro 6, e00133 (2014).

    PubMed  PubMed Central  Google Scholar 

  171. Robinson, I. & Reddy, A. B. Molecular mechanisms of the circadian clockwork in mammals. FEBS Lett. 588, 2477–2483 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A.V. has received research grant support from the NIH (K23 NS072283). A.S.L. has received research grant support from the CHDI Foundation (RG50786). R.A.B. has received research grant support from the CHDI Foundation (RG50786), Evelyn Trust (RG66030), Butterfield Trust (RG68592), and the NIHR Biomedical Research Centre and Biomedical Research Unit Award to Addenbrooke's Hospital/University of Cambridge (RG68592). S.O. has received research grant support from the Netherlands Organization for Scientific Research (grant no. 016.116.371).

Author information

Authors and Affiliations

Authors

Contributions

A.V., A.S.L. and S.O. researched data for the article. All authors provided substantial contributions to discussion of the content, wrote the article and reviewed and/or edited the manuscript before and after submission.

Corresponding author

Correspondence to Aleksandar Videnovic.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Videnovic, A., Lazar, A., Barker, R. et al. 'The clocks that time us'—circadian rhythms in neurodegenerative disorders. Nat Rev Neurol 10, 683–693 (2014). https://doi.org/10.1038/nrneurol.2014.206

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2014.206

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing