Skip to main content
Erschienen in: Zeitschrift für Gerontologie und Geriatrie 7/2023

07.09.2023 | Themenschwerpunkt

Schlaf und Demenz

verfasst von: Prof. Dr. Geert Mayer, Carla Stenmanns, Thorsten R. Doeppner, Dirk M. Hermann, Janine Gronewold

Erschienen in: Zeitschrift für Gerontologie und Geriatrie | Ausgabe 7/2023

Einloggen, um Zugang zu erhalten

Zusammenfassung

Altern geht mit Änderungen der Schlafstruktur sowie zerebraler Ablagerung von Amyloid‑β und Tau-Proteinen einher. Schlafstörungen gehen dem Beginn einer Demenz um Jahre voraus. Komorbide Schlafstörungen wie Insomnien und schlafbezogene Atmungsstörungen, familiäre Demenzbelastung und epigenetische Faktoren können zur Demenzentwicklung beitragen. Dieser Beitrag geht der Frage nach der Interaktion zwischen Schlaf und Demenz anhand von bisher existierender Literatur nach. Veränderungen von „slow wave sleep“ führen zu Änderungen der glymphatischen Clearance von Amyloid‑β, Tau- und anderen Proteinen. Transiente und chronifizierte Schlafstörungen verursachen Veränderungen in Gehirnarealen, die für Kognition und Verhalten verantwortlich sind. Schlafregulierende Hirnareale sind als erste im neurodegenerativen Prozess betroffen und beschleunigen das Demenzrisiko. Zirkadiane altersbedingte Änderungen von Amyloid‑β und Tau-Protein beeinflussen die Schlafmenge bzw. -tiefe und umgekehrt. Die Liquor-Aβ-Konzentration weist eine inverse Korrelation mit dem Schlaf auf. Orexine modulieren Amyloid‑β und Schlaf.
Literatur
1.
Zurück zum Zitat Aizenstein HJ, Nebes RD, Saxton JA et al (2008) Frequent amyloid deposition without significant cognitive impairment among the elderly. Arch Neurol 65:1509–1517PubMedPubMedCentralCrossRef Aizenstein HJ, Nebes RD, Saxton JA et al (2008) Frequent amyloid deposition without significant cognitive impairment among the elderly. Arch Neurol 65:1509–1517PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Barthélemy NR, Li Y, Joseph-Mathurin N et al (2020) A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat Med 26:398–407PubMedPubMedCentralCrossRef Barthélemy NR, Li Y, Joseph-Mathurin N et al (2020) A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat Med 26:398–407PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Borbély AA (1982) A two process model of sleep regulation. Hum Neurobiol 1:195–204PubMed Borbély AA (1982) A two process model of sleep regulation. Hum Neurobiol 1:195–204PubMed
5.
Zurück zum Zitat Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82:239–259PubMedCrossRef Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82:239–259PubMedCrossRef
6.
Zurück zum Zitat Braak H, Braak E (1995) Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol Aging 16:271–278 (discussion 278–84)PubMedCrossRef Braak H, Braak E (1995) Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol Aging 16:271–278 (discussion 278–84)PubMedCrossRef
7.
Zurück zum Zitat Bubu OM, Pirraglia E, Andrade AG et al (2019) Obstructive sleep apnea and longitudinal Alzheimer’s disease biomarker changes. Sleep 42(6):zsz48PubMedPubMedCentralCrossRef Bubu OM, Pirraglia E, Andrade AG et al (2019) Obstructive sleep apnea and longitudinal Alzheimer’s disease biomarker changes. Sleep 42(6):zsz48PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Chong MS, Tay L, Chan M et al (2015) Prospective longitudinal study of frailty transitions in a community-dwelling cohort of older adults with cognitive impairment. BMC Geriatr 15:175PubMedPubMedCentralCrossRef Chong MS, Tay L, Chan M et al (2015) Prospective longitudinal study of frailty transitions in a community-dwelling cohort of older adults with cognitive impairment. BMC Geriatr 15:175PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Christensen J, Li C, Mychasiuk R (2022) Choroid plexus function in neurological homeostasis and disorders: The awakening of the circadian clocks and orexins. J Cereb Blood Flow Metab 42:1163–1175PubMedPubMedCentralCrossRef Christensen J, Li C, Mychasiuk R (2022) Choroid plexus function in neurological homeostasis and disorders: The awakening of the circadian clocks and orexins. J Cereb Blood Flow Metab 42:1163–1175PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat D’Atri A, Scarpelli S, Gorgoni M et al (2021) EEG alterations during wake and sleep in mild cognitive impairment and Alzheimer’s disease. iScience 24:102386PubMedPubMedCentralCrossRef D’Atri A, Scarpelli S, Gorgoni M et al (2021) EEG alterations during wake and sleep in mild cognitive impairment and Alzheimer’s disease. iScience 24:102386PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Feng R, Li L, Yu H et al (2016) Melanopsin retinal ganglion cell loss and circadian dysfunction in Alzheimer’s disease (Review). Mol Med Rep 13:3397–3400PubMedPubMedCentralCrossRef Feng R, Li L, Yu H et al (2016) Melanopsin retinal ganglion cell loss and circadian dysfunction in Alzheimer’s disease (Review). Mol Med Rep 13:3397–3400PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Gildner TE, Salinas-Rodríguez A, Manrique-Espinoza B et al (2019) Does poor sleep impair cognition during aging? Longitudinal associations between changes in sleep duration and cognitive performance among older Mexican adults. Arch Gerontol Geriatr 83:161–168PubMedPubMedCentralCrossRef Gildner TE, Salinas-Rodríguez A, Manrique-Espinoza B et al (2019) Does poor sleep impair cognition during aging? Longitudinal associations between changes in sleep duration and cognitive performance among older Mexican adults. Arch Gerontol Geriatr 83:161–168PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Gozzi A, Turrini G, Piccoli L et al (2011) Functional magnetic resonance imaging reveals different neural substrates for the effects of orexin‑1 and orexin‑2 receptor antagonists. Plos One 6:e16406PubMedPubMedCentralCrossRef Gozzi A, Turrini G, Piccoli L et al (2011) Functional magnetic resonance imaging reveals different neural substrates for the effects of orexin‑1 and orexin‑2 receptor antagonists. Plos One 6:e16406PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Grau-Rivera O, Operto G, Falcón C et al (2020) Association between insomnia and cognitive performance, gray matter volume, and white matter microstructure in cognitively unimpaired adults. Alzheimers Res Ther 12:4PubMedPubMedCentralCrossRef Grau-Rivera O, Operto G, Falcón C et al (2020) Association between insomnia and cognitive performance, gray matter volume, and white matter microstructure in cognitively unimpaired adults. Alzheimers Res Ther 12:4PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Guarnieri B, Adorni F, Musicco M et al (2012) Prevalence of sleep disturbances in mild cognitive impairment and dementing disorders: a multicenter Italian clinical cross-sectional study on 431 patients. Dement Geriatr Cogn Disord 33:50–58PubMedPubMedCentralCrossRef Guarnieri B, Adorni F, Musicco M et al (2012) Prevalence of sleep disturbances in mild cognitive impairment and dementing disorders: a multicenter Italian clinical cross-sectional study on 431 patients. Dement Geriatr Cogn Disord 33:50–58PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Herring WJ, Ceesay P, Snyder E et al (2020) Polysomnographic assessment of suvorexant in patients with probable Alzheimer’s disease dementia and insomnia: a randomized trial. Alzheimers Dement 16:541–551PubMedPubMedCentralCrossRef Herring WJ, Ceesay P, Snyder E et al (2020) Polysomnographic assessment of suvorexant in patients with probable Alzheimer’s disease dementia and insomnia: a randomized trial. Alzheimers Dement 16:541–551PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Iranzo A, Fairfoul G, Ayudhaya ACN et al (2021) Detection of α‑synuclein in CSF by RT-QuIC in patients with isolated rapid-eye-movement sleep behaviour disorder: a longitudinal observational study. Lancet Neurol 20:203–212PubMedCrossRef Iranzo A, Fairfoul G, Ayudhaya ACN et al (2021) Detection of α‑synuclein in CSF by RT-QuIC in patients with isolated rapid-eye-movement sleep behaviour disorder: a longitudinal observational study. Lancet Neurol 20:203–212PubMedCrossRef
19.
Zurück zum Zitat Josephs KA, Whitwell JL, Ahmed Z et al (2008) Beta-amyloid burden is not associated with rates of brain atrophy. Ann Neurol 63:204–212PubMedPubMedCentralCrossRef Josephs KA, Whitwell JL, Ahmed Z et al (2008) Beta-amyloid burden is not associated with rates of brain atrophy. Ann Neurol 63:204–212PubMedPubMedCentralCrossRef
20.
21.
Zurück zum Zitat Kant S, Stopa EG, Johanson CE et al (2018) Choroid plexus genes for CSF production and brain homeostasis are altered in Alzheimer’s disease. Fluids Barriers CNS 15:34PubMedPubMedCentralCrossRef Kant S, Stopa EG, Johanson CE et al (2018) Choroid plexus genes for CSF production and brain homeostasis are altered in Alzheimer’s disease. Fluids Barriers CNS 15:34PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Kent BA, Feldman HH, Nygaard HB (2021) Sleep and its regulation: An emerging pathogenic and treatment frontier in Alzheimer’s disease. Prog Neurobiol 197:101902PubMedCrossRef Kent BA, Feldman HH, Nygaard HB (2021) Sleep and its regulation: An emerging pathogenic and treatment frontier in Alzheimer’s disease. Prog Neurobiol 197:101902PubMedCrossRef
24.
Zurück zum Zitat Liguori C, Romigi A, Nuccetelli M et al (2014) Orexinergic system dysregulation, sleep impairment, and cognitive decline in Alzheimer disease. JAMA Neurol 71:1498–1505PubMedCrossRef Liguori C, Romigi A, Nuccetelli M et al (2014) Orexinergic system dysregulation, sleep impairment, and cognitive decline in Alzheimer disease. JAMA Neurol 71:1498–1505PubMedCrossRef
25.
Zurück zum Zitat Lucey BP, Wisch J, Boerwinkle AH et al (2021) Sleep and longitudinal cognitive performance in preclinical and early symptomatic Alzheimer’s disease. Brain 144:2852–2862PubMedPubMedCentralCrossRef Lucey BP, Wisch J, Boerwinkle AH et al (2021) Sleep and longitudinal cognitive performance in preclinical and early symptomatic Alzheimer’s disease. Brain 144:2852–2862PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Mander BA, Dave A, Lui KK et al (2022) Inflammation, tau pathology, and synaptic integrity associated with sleep spindles and memory prior to β‑amyloid positivity. Sleep 45(9):zsac135PubMedPubMedCentralCrossRef Mander BA, Dave A, Lui KK et al (2022) Inflammation, tau pathology, and synaptic integrity associated with sleep spindles and memory prior to β‑amyloid positivity. Sleep 45(9):zsac135PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Miglis MG, Adler CH, Antelmi E et al (2021) Biomarkers of conversion to α‑synucleinopathy in isolated rapid-eye-movement sleep behaviour disorder. Lancet Neurol 20:671–684PubMedPubMedCentralCrossRef Miglis MG, Adler CH, Antelmi E et al (2021) Biomarkers of conversion to α‑synucleinopathy in isolated rapid-eye-movement sleep behaviour disorder. Lancet Neurol 20:671–684PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Mintun MA, Larossa GN, Sheline YI et al (2006) 11CPIB in a nondemented population: potential antecedent marker of Alzheimer disease. Neurology 67:446–452PubMedCrossRef Mintun MA, Larossa GN, Sheline YI et al (2006) 11CPIB in a nondemented population: potential antecedent marker of Alzheimer disease. Neurology 67:446–452PubMedCrossRef
30.
Zurück zum Zitat Mormino EC, Kluth JT, Madison CM et al (2009) Episodic memory loss is related to hippocampal-mediated beta-amyloid deposition in elderly subjects. Brain 132:1310–1323PubMedCrossRef Mormino EC, Kluth JT, Madison CM et al (2009) Episodic memory loss is related to hippocampal-mediated beta-amyloid deposition in elderly subjects. Brain 132:1310–1323PubMedCrossRef
31.
Zurück zum Zitat Neikrug AB, Ancoli-Israel S (2010) Sleep disorders in the older adult—a mini-review. Gerontology 56:181–189PubMedCrossRef Neikrug AB, Ancoli-Israel S (2010) Sleep disorders in the older adult—a mini-review. Gerontology 56:181–189PubMedCrossRef
33.
Zurück zum Zitat Ohayon MM, Carskadon MA, Guilleminault C et al (2004) Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: developing normative sleep values across the human lifespan. Sleep 27:1255–1273PubMedCrossRef Ohayon MM, Carskadon MA, Guilleminault C et al (2004) Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: developing normative sleep values across the human lifespan. Sleep 27:1255–1273PubMedCrossRef
34.
Zurück zum Zitat Pase MP, Himali JJ, Grima NA et al (2017) Sleep architecture and the risk of incident dementia in the community. Neurology 89:1244–1250PubMedPubMedCentralCrossRef Pase MP, Himali JJ, Grima NA et al (2017) Sleep architecture and the risk of incident dementia in the community. Neurology 89:1244–1250PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Riemann D, Nissen C, Palagini L et al (2015) The neurobiology, investigation, and treatment of chronic insomnia. Lancet Neurol 14:547–558PubMedCrossRef Riemann D, Nissen C, Palagini L et al (2015) The neurobiology, investigation, and treatment of chronic insomnia. Lancet Neurol 14:547–558PubMedCrossRef
36.
Zurück zum Zitat Shokri-Kojori E, Wang G‑J, Wiers CE et al (2018) β‑Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A 115:4483–4488PubMedPubMedCentralCrossRef Shokri-Kojori E, Wang G‑J, Wiers CE et al (2018) β‑Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A 115:4483–4488PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Spira AP, Gamaldo AA, An Y et al (2013) Self-reported sleep and beta-amyloid deposition in community-dwelling older adults. JAMA Neurol 70:1537–1543PubMedPubMedCentral Spira AP, Gamaldo AA, An Y et al (2013) Self-reported sleep and beta-amyloid deposition in community-dwelling older adults. JAMA Neurol 70:1537–1543PubMedPubMedCentral
39.
Zurück zum Zitat Sun X, He G, Qing H et al (2006) Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc Natl Acad Sci U S A 103:18727–18732PubMedPubMedCentralCrossRef Sun X, He G, Qing H et al (2006) Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc Natl Acad Sci U S A 103:18727–18732PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Szymusiak R, Alam N, McGinty D (2000) Discharge patterns of neurons in cholinergic regions of the basal forebrain during waking and sleep. Behav Brain Res 115:171–182PubMedCrossRef Szymusiak R, Alam N, McGinty D (2000) Discharge patterns of neurons in cholinergic regions of the basal forebrain during waking and sleep. Behav Brain Res 115:171–182PubMedCrossRef
41.
Zurück zum Zitat Valencia Garcia S, Brischoux F, Clément O et al (2018) Ventromedial medulla inhibitory neuron inactivation induces REM sleep without atonia and REM sleep behavior disorder. Nat Commun 9:504PubMedPubMedCentralCrossRef Valencia Garcia S, Brischoux F, Clément O et al (2018) Ventromedial medulla inhibitory neuron inactivation induces REM sleep without atonia and REM sleep behavior disorder. Nat Commun 9:504PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Westerberg CE, Lundgren EM, Florczak SM et al (2010) Sleep influences the severity of memory disruption in amnestic mild cognitive impairment: results from sleep self-assessment and continuous activity monitoring. Alzheimer Dis Assoc Disord 24:325–333PubMedPubMedCentralCrossRef Westerberg CE, Lundgren EM, Florczak SM et al (2010) Sleep influences the severity of memory disruption in amnestic mild cognitive impairment: results from sleep self-assessment and continuous activity monitoring. Alzheimer Dis Assoc Disord 24:325–333PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Westerberg CE, Mander BA, Florczak SM et al (2012) Concurrent impairments in sleep and memory in amnestic mild cognitive impairment. J Int Neuropsychol Soc 18:490–500PubMedPubMedCentralCrossRef Westerberg CE, Mander BA, Florczak SM et al (2012) Concurrent impairments in sleep and memory in amnestic mild cognitive impairment. J Int Neuropsychol Soc 18:490–500PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Winer JR, Deters KD, Kennedy G et al (2021) Association of short and long sleep duration with Amyloid‑β burden and cognition in aging. JAMA Neurol 78:1187–1196PubMedCrossRef Winer JR, Deters KD, Kennedy G et al (2021) Association of short and long sleep duration with Amyloid‑β burden and cognition in aging. JAMA Neurol 78:1187–1196PubMedCrossRef
45.
Zurück zum Zitat Winer JR, Mander BA, Kumar S et al (2020) Sleep disturbance forecasts β‑Amyloid accumulation across subsequent years. Curr Biol 30:4291–4298.e3PubMedPubMedCentralCrossRef Winer JR, Mander BA, Kumar S et al (2020) Sleep disturbance forecasts β‑Amyloid accumulation across subsequent years. Curr Biol 30:4291–4298.e3PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Xie L, Kang H, Xu Q et al (2013) Sleep drives metabolite clearance from the adult brain. Science 342:373–377PubMedCrossRef Xie L, Kang H, Xu Q et al (2013) Sleep drives metabolite clearance from the adult brain. Science 342:373–377PubMedCrossRef
Metadaten
Titel
Schlaf und Demenz
verfasst von
Prof. Dr. Geert Mayer
Carla Stenmanns
Thorsten R. Doeppner
Dirk M. Hermann
Janine Gronewold
Publikationsdatum
07.09.2023
Verlag
Springer Medizin
Erschienen in
Zeitschrift für Gerontologie und Geriatrie / Ausgabe 7/2023
Print ISSN: 0948-6704
Elektronische ISSN: 1435-1269
DOI
https://doi.org/10.1007/s00391-023-02237-5

Weitere Artikel der Ausgabe 7/2023

Zeitschrift für Gerontologie und Geriatrie 7/2023 Zur Ausgabe

Journal Club

Journal Club

Mitteilungen der DGG

Mitteilungen der DGGG