L. Fratiglioni, Incidence of dementia and major subtypes in Europe: A collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group, Neurology, vol.54, issue.11, pp.10-15, 2000.

B. Dubois, Preclinical Alzheimer's disease: Definition, natural history, and diagnostic criteria, Alzheimers Dement, vol.12, issue.3, pp.292-323, 2016.

R. J. Bateman, Clinical and biomarker changes in dominantly inherited Alzheimer's disease, N Engl J Med, vol.367, issue.9, pp.795-804, 2012.

A. M. Fagan, Longitudinal change in CSF biomarkers in autosomal-dominant Alzheimer's disease, Sci Transl Med, vol.6, issue.226, pp.226-256, 2014.

D. J. Selkoe and J. Hardy, The amyloid hypothesis of Alzheimer's disease at 25 years, EMBO Mol Med, vol.8, issue.6, pp.595-608, 2016.

N. Mattsson, Independent information from cerebrospinal fluid amyloid-beta and florbetapir imaging in Alzheimer's disease. Brain, pp.772-83, 2015.

W. C. Orr, Utilization of polysomnography in the assessment of sleep disorders, Med Clin North Am, vol.69, issue.6, pp.1153-67, 1985.

I. Alapin, How is good and poor sleep in older adults and college students related to daytime sleepiness, fatigue, and ability to concentrate?, J Psychosom Res, vol.49, issue.5, pp.381-90, 2000.

J. R. Espiritu, Aging-related sleep changes, Clin Geriatr Med, vol.24, issue.1, pp.1-14, 2008.

M. M. Ohayon, Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: developing normative sleep values across the human lifespan, Sleep, vol.27, issue.7, pp.1255-73, 2004.

E. J. Van-someren, Circadian rhythms and sleep in human aging, Chronobiol Int, vol.17, issue.3, pp.233-276, 2000.

J. Carrier, Sleep slow wave changes during the middle years of life, Eur J Neurosci, vol.33, issue.4, pp.758-66, 2011.

K. Crowley, J. Trinder, and I. M. Colrain, An examination of evoked K-complex amplitude and frequency of occurrence in the elderly, J Sleep Res, vol.11, issue.2, pp.129-169, 2002.

K. Crowley, The effects of normal aging on sleep spindle and K-complex production, Clin Neurophysiol, vol.113, issue.10, pp.1615-1637, 2002.

K. Crowley, J. Trinder, and I. M. Colrain, Evoked K-complex generation: the impact of sleep spindles and age, Clin Neurophysiol, vol.115, issue.2, pp.471-477, 2004.

G. J. Landry and T. Liu-ambrose, Buying time: a rationale for examining the use of circadian rhythm and sleep interventions to delay progression of mild cognitive impairment to Alzheimer's disease. Front Aging Neurosci, vol.6, p.325, 2014.

D. Petit, Sleep and quantitative EEG in neurodegenerative disorders, J Psychosom Res, vol.56, issue.5, pp.487-96, 2004.

K. Jost, A gerontopsychological pilot study on the sleep of the very elderly--initial experiences with the Nurnberg Gerontopsychologic Inventory

, Z Gerontol, vol.26, issue.6, pp.471-476, 1993.

J. Montplaisir, Sleep in Alzheimer's disease: further considerations on the role of brainstem and forebrain cholinergic populations in sleep-wake mechanisms, Sleep, vol.18, issue.3, pp.145-153, 1995.

M. F. Vecchierini,

, Psychol Neuropsychiatr Vieil, vol.8, issue.1, pp.15-23, 2010.

S. Ancoli-israel, Use of wrist activity for monitoring sleep/wake in demented nursing-home patients, Sleep, vol.20, issue.1, pp.24-31, 1997.

J. Martin, Circadian rhythms of agitation in institutionalized patients with Alzheimer's disease, Chronobiol Int, vol.17, issue.3, pp.405-423, 2000.

D. G. Harper, Disturbance of endogenous circadian rhythm in aging and Alzheimer disease, Am J Geriatr Psychiatry, vol.13, issue.5, pp.359-68, 2005.

D. J. Buysse, Rapid eye movement sleep deprivation in elderly patients with concurrent symptoms of depression and dementia, J Neuropsychiatry Clin Neurosci, vol.4, issue.3, pp.249-56, 1992.

D. L. Bliwise, Observed sleep/wakefulness and severity of dementia in an Alzheimer's disease special care unit, J Gerontol A Biol Sci Med Sci, vol.50, issue.6, pp.303-309, 1995.

M. Moran, Sleep disturbance in mild to moderate Alzheimer's disease, Sleep Med, vol.6, issue.4, pp.347-52, 2005.

P. Gehrman, The relationship between dementia severity and rest/activity circadian rhythms, Neuropsychiatr Dis Treat, vol.1, issue.2, pp.155-63, 2005.

S. M. Mccurry, Treatment of sleep disturbance in Alzheimer's disease, Sleep Med Rev, vol.4, issue.6, pp.603-628, 2000.

G. Tononi and C. Cirelli, Sleep and synaptic homeostasis: a hypothesis, Brain Res Bull, vol.62, issue.2, pp.143-50, 2003.

S. Beaulieu-bonneau and C. Hudon, Sleep disturbances in older adults with mild cognitive impairment, Int Psychogeriatr, vol.21, issue.4, pp.654-66, 2009.

E. Hita-yanez, M. Atienza, and J. L. Cantero, Polysomnographic and subjective sleep markers of mild cognitive impairment, Sleep, vol.36, issue.9, pp.1327-1361, 2013.

L. Bidzan,

, Psychiatr Pol, vol.45, issue.6, pp.851-60, 2011.

G. E. Schlosser-covell, Disrupted daytime activity and altered sleep-wake patterns may predict transition to mild cognitive impairment or dementia: a critically appraised topic, Neurologist, vol.18, issue.6, pp.426-435, 2012.

O. M. Bubu, Cognitive impairment, and Alzheimer's disease: A Systematic Review and Meta-Analysis, Sleep, issue.1, p.40, 2017.

L. Shi, Sleep disturbances increase the risk of dementia: A systematic review and meta-analysis, Sleep Med Rev, vol.40, pp.4-16, 2018.

Y. E. Ju, Sleep quality and preclinical Alzheimer disease, JAMA Neurol, vol.70, issue.5, pp.587-93, 2013.

J. R. Molano, C. M. Roe, and Y. S. Ju, The interaction of sleep and amyloid deposition on cognitive performance, J Sleep Res, vol.26, issue.3, pp.288-292, 2017.

K. E. Sprecher, Poor sleep is associated with CSF biomarkers of amyloid pathology in cognitively normal adults, Neurology, vol.89, issue.5, pp.445-453, 2017.

A. M. Fjell, Neuroinflammation and Tau Interact with Amyloid in Predicting Sleep Problems in Aging Independently of Atrophy, Cereb Cortex, vol.28, issue.8, pp.2775-2785, 2018.

C. Benedict, Self-reported sleep disturbance is associated with Alzheimer's disease risk in men, Alzheimers Dement, vol.11, issue.9, pp.1090-1097, 2015.

M. P. Sanchez-espinosa, M. Atienza, and J. L. Cantero, Sleep deficits in mild cognitive impairment are related to increased levels of plasma amyloid-beta and cortical thinning, Neuroimage, vol.98, pp.395-404, 2014.

J. B. Toledo, L. M. Shaw, and J. Q. Trojanowski, Plasma amyloid beta measurementsa desired but elusive Alzheimer's disease biomarker, Alzheimers Res Ther, vol.5, issue.2, p.8, 2013.

E. N. Minakawa, Chronic sleep fragmentation exacerbates amyloid beta deposition in Alzheimer's disease model mice, Neurosci Lett, vol.653, pp.362-369, 2017.

A. P. Spira, Self-reported sleep and beta-amyloid deposition in communitydwelling older adults, JAMA Neurol, vol.70, issue.12, pp.1537-1580, 2013.

B. M. Brown, The Relationship between Sleep Quality and Brain Amyloid Burden. Sleep, vol.39, pp.1063-1071, 2016.

P. Branger, Relationships between sleep quality and brain volume, metabolism, and amyloid deposition in late adulthood, Neurobiol Aging, vol.41, pp.107-114, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01295793

J. E. Kang, Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle, Science, vol.326, issue.5955, pp.1005-1012, 2009.

J. R. Cirrito, Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo, Neuron, vol.48, issue.6, pp.913-935, 2005.

G. F. Gilestro, G. Tononi, and C. Cirelli, Widespread changes in synaptic markers as a function of sleep and wakefulness in Drosophila, Science, vol.324, issue.5923, pp.109-121, 2009.

S. G. Horovitz, Decoupling of the brain's default mode network during deep sleep, Proc Natl Acad Sci, vol.106, issue.27, pp.11376-81, 2009.

V. I. Spoormaker, Development of a large-scale functional brain network during human non-rapid eye movement sleep, J Neurosci, vol.30, issue.34, pp.11379-87, 2010.

C. W. Wu, Variations in connectivity in the sensorimotor and default-mode networks during the first nocturnal sleep cycle, Brain Connect, vol.2, issue.4, pp.177-90, 2012.

R. J. Bateman, Human amyloid-beta synthesis and clearance rates as measured in cerebrospinal fluid in vivo, Nat Med, vol.12, issue.7, pp.856-61, 2006.

Y. Huang, beta-amyloid dynamics in human plasma, Arch Neurol, vol.69, issue.12, pp.1591-1598, 2012.

T. Saido and M. A. Leissring, Proteolytic degradation of amyloid beta-protein. Cold Spring Harb Perspect Med, vol.2, p.6379, 2012.

D. A. Dewitt, Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer's disease, Exp Neurol, vol.149, issue.2, pp.329-369, 1998.

R. D. Bell and B. V. Zlokovic, Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer's disease, Acta Neuropathol, vol.118, issue.1, pp.103-116, 2009.

J. J. Iliff, A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta, Sci Transl Med, vol.4, issue.147, pp.147-111, 2012.

L. Xie, Sleep drives metabolite clearance from the adult brain, Science, vol.342, issue.6156, pp.373-380, 2013.

J. Cedernaes, Candidate mechanisms underlying the association between sleepwake disruptions and Alzheimer's disease, Sleep Med Rev, vol.31, pp.102-111, 2017.

L. I. Kiyashchenko, Release of hypocretin (orexin) during waking and sleep states, J Neurosci, vol.22, issue.13, pp.5282-5288, 2002.

J. H. Roh, Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer's disease, J Exp Med, vol.211, issue.13, pp.2487-96, 2014.

C. Liguori, CSF beta-amyloid levels are altered in narcolepsy: a link with the inflammatory hypothesis?, J Sleep Res, vol.23, issue.4, pp.420-424, 2014.

A. Gabelle, Cerebrospinal fluid levels of orexin-A and histamine, and sleep profile within the Alzheimer process, Neurobiol Aging, vol.53, pp.59-66, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01781465

M. Wennstrom, Altered CSF orexin and alpha-synuclein levels in dementia patients, J Alzheimers Dis, vol.29, issue.1, pp.125-157, 2012.

V. D'almeida, Sleep deprivation induces brain region-specific decreases in glutathione levels, Neuroreport, vol.9, issue.12, pp.2853-2859, 1998.

D. C. Mathangi, R. Shyamala, and A. S. Subhashini, Effect of REM sleep deprivation on the antioxidant status in the brain of Wistar rats, Ann Neurosci, vol.19, issue.4, pp.161-165, 2012.

L. Ramanathan, Sleep deprivation decreases superoxide dismutase activity in rat hippocampus and brainstem, vol.13, pp.1387-90, 2002.

B. Gomez-gonzalez, REM sleep loss and recovery regulates blood-brain barrier function, Curr Neurovasc Res, vol.10, issue.3, pp.197-207, 2013.

J. He, Sleep restriction impairs blood-brain barrier function, J Neurosci, vol.34, issue.44, pp.14697-706, 2014.

A. M. Swomley, Abeta, oxidative stress in Alzheimer disease: evidence based on proteomics studies, Biochim Biophys Acta, vol.1842, issue.8, pp.1248-57, 2014.

E. L. Goldwaser, Breakdown of the Cerebrovasculature and Blood-Brain Barrier: A Mechanistic Link Between Diabetes Mellitus and Alzheimer's Disease, J Alzheimers Dis, vol.54, issue.2, pp.445-56, 2016.

J. Zhang, Extended wakefulness: compromised metabolics in and degeneration of locus ceruleus neurons, J Neurosci, vol.34, issue.12, pp.4418-4449, 2014.

R. H. Silva, Role of hippocampal oxidative stress in memory deficits induced by sleep deprivation in mice, Neuropharmacology, vol.46, issue.6, pp.895-903, 2004.

A. Gopalakrishnan, L. L. Ji, and C. Cirelli, Sleep deprivation and cellular responses to oxidative stress. Sleep, vol.27, pp.27-35, 2004.

E. A. Winkler, A. P. Sagare, and B. V. Zlokovic, The pericyte: a forgotten cell type with important implications for Alzheimer's disease?, Brain Pathol, vol.24, issue.4, pp.371-86, 2014.

Y. Huang, Effects of age and amyloid deposition on Abeta dynamics in the human central nervous system, Arch Neurol, vol.69, issue.1, pp.51-59, 2012.

J. Altman, Are new neurons formed in the brains of adult mammals? Science, vol.135, pp.1127-1135, 1962.

J. B. Aimone, Regulation and function of adult neurogenesis: from genes to cognition, Physiol Rev, vol.94, issue.4, pp.991-1026, 2014.

M. Carlen, Functional integration of adult-born neurons, Curr Biol, vol.12, issue.7, pp.606-614, 2002.

H. Van-praag, Functional neurogenesis in the adult hippocampus, Nature, vol.415, issue.6875, pp.1030-1034, 2002.

S. Masri, M. Cervantes, and P. Sassone-corsi, The circadian clock and cell cycle: interconnected biological circuits, Curr Opin Cell Biol, vol.25, issue.6, pp.730-734, 2013.

A. Laposky, Deletion of the mammalian circadian clock gene BMAL1/Mop3 alters baseline sleep architecture and the response to sleep deprivation, Sleep, vol.28, issue.4, pp.395-409, 2005.

P. Bouchard-cannon, The circadian molecular clock regulates adult hippocampal neurogenesis by controlling the timing of cell-cycle entry and exit, Cell Rep, vol.5, issue.4, pp.961-73, 2013.

B. D. Rakai, Survival of adult generated hippocampal neurons is altered in circadian arrhythmic mice, PLoS One, vol.9, issue.6, p.99527, 2014.

A. A. Ali, Premature aging of the hippocampal neurogenic niche in adult Bmal1-deficient mice, Aging (Albany NY), issue.7, pp.435-484, 2015.

B. A. Kent and R. E. Mistlberger, Sleep and hippocampal neurogenesis: Implications for Alzheimer's disease. Front Neuroendocrinol, vol.45, pp.35-52, 2017.

R. Guzman-marin, Sleep deprivation suppresses neurogenesis in the adult hippocampus of rats, Eur J Neurosci, vol.22, issue.8, pp.2111-2117, 2005.

R. Guzman-marin, Hippocampal neurogenesis is reduced by sleep fragmentation in the adult rat, Neuroscience, vol.148, issue.1, pp.325-358, 2007.

P. Meerlo, Sleep restriction alters the hypothalamic-pituitary-adrenal response to stress, J Neuroendocrinol, vol.14, issue.5, pp.397-402, 2002.
URL : https://hal.archives-ouvertes.fr/hal-02326238

I. S. Hairston, Sleep restriction suppresses neurogenesis induced by hippocampus-dependent learning, J Neurophysiol, vol.94, issue.6, pp.4224-4257, 2005.

C. Mirescu, Sleep deprivation inhibits adult neurogenesis in the hippocampus by elevating glucocorticoids, Proc Natl Acad Sci, vol.103, issue.50, pp.19170-19175, 2006.

A. D. Mueller, Sleep deprivation can inhibit adult hippocampal neurogenesis independent of adrenal stress hormones, Am J Physiol Regul Integr Comp Physiol, vol.294, issue.5, pp.1693-703, 2008.

C. A. Everson, Clinical assessment of blood leukocytes, serum cytokines, and serum immunoglobulins as responses to sleep deprivation in laboratory rats, Am J Physiol Regul Integr Comp Physiol, vol.289, issue.4, pp.1054-63, 2005.

M. R. Irwin, Sleep deprivation and activation of morning levels of cellular and genomic markers of inflammation, Arch Intern Med, vol.166, issue.16, pp.1756-62, 2006.

M. Haack, E. Sanchez, and J. M. Mullington, Elevated inflammatory markers in response to prolonged sleep restriction are associated with increased pain experience in healthy volunteers, Sleep, vol.30, issue.9, pp.1145-52, 2007.

S. Yehuda, REM sleep deprivation in rats results in inflammation and interleukin-17 elevation, J Interferon Cytokine Res, vol.29, issue.7, pp.393-401, 2009.

L. Vallieres, Reduced hippocampal neurogenesis in adult transgenic mice with chronic astrocytic production of interleukin-6, J Neurosci, vol.22, issue.2, pp.486-92, 2002.

M. L. Monje, H. Toda, and T. D. Palmer, Inflammatory blockade restores adult hippocampal neurogenesis, Science, vol.302, issue.5651, pp.1760-1765, 2003.

C. T. Ekdahl, Inflammation is detrimental for neurogenesis in adult brain, Proc Natl Acad Sci, vol.100, issue.23, pp.13632-13639, 2003.

M. A. Kheirbek, L. Tannenholz, and R. Hen, NR2B-dependent plasticity of adult-born granule cells is necessary for context discrimination, J Neurosci, vol.32, issue.25, pp.8696-702, 2012.

H. A. Cameron, B. S. Mcewen, and E. Gould, Regulation of adult neurogenesis by excitatory input and NMDA receptor activation in the dentate gyrus, J Neurosci, vol.15, issue.6, pp.4687-92, 1995.

M. Tabuchi, Sleep interacts with abeta to modulate intrinsic neuronal excitability, Curr Biol, vol.25, issue.6, pp.702-712, 2015.

S. Liu, Sleep Drive Is Encoded by Neural Plastic Changes in a Dedicated Circuit, Cell, vol.165, issue.6, pp.1347-1360, 2016.

V. V. Vyazovskiy, Molecular and electrophysiological evidence for net synaptic potentiation in wake and depression in sleep, Nat Neurosci, vol.11, issue.2, pp.200-208, 2008.

W. Danysz and C. G. Parsons, Alzheimer's disease, beta-amyloid, glutamate, NMDA receptors and memantine--searching for the connections, Br J Pharmacol, vol.167, issue.2, pp.324-52, 2012.

M. F. Folstein, S. E. Folstein, and P. R. Mchugh, A practical method for grading the cognitive state of patients for the clinician, J Psychiatr Res, vol.12, issue.3, pp.189-98, 1975.

M. Gagnon, Validity of the Mini-Mental State examination as a screening instrument for cognitive impairment and dementia in French elderly community residents, Neuroepidemiology, vol.9, issue.3, pp.143-50, 1990.

P. S. Freedson, E. Melanson, and J. Sirard, Calibration of the Computer Science and Applications, Inc. accelerometer, Med Sci Sports Exerc, vol.30, issue.5, pp.777-81, 1998.

R. J. Cole, Automatic sleep/wake identification from wrist activity, Sleep, vol.15, issue.5, pp.461-470, 1992.

M. Quante, Actigraphy-based sleep estimation in adolescents and adults: a comparison with polysomnography using two scoring algorithms, Nat Sci Sleep, vol.10, pp.13-20, 2018.

N. Cellini, Direct comparison of two actigraphy devices with polysomnographically recorded naps in healthy young adults, Chronobiol Int, vol.30, issue.5, pp.691-699, 2013.

D. R. Samson, What is segmented sleep? Actigraphy field validation for daytime sleep and nighttime wake, Sleep Health, vol.2, issue.4, pp.341-347, 2016.

J. E. Maglione, Actigraphy for the assessment of sleep measures in Parkinson's disease. Sleep, vol.36, pp.1209-1226, 2013.

J. Ashburner and K. J. Friston, Unified segmentation, Neuroimage, vol.26, issue.3, pp.839-51, 2005.

M. Chupin, Automatic segmentation of the hippocampus and the amygdala driven by hybrid constraints: method and validation, Neuroimage, vol.46, issue.3, pp.749-61, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00805390

M. Chupin, Anatomically constrained region deformation for the automated segmentation of the hippocampus and the amygdala: Method and validation on controls and patients with Alzheimer's disease. Neuroimage, vol.34, pp.996-1019, 2007.

B. Dubois, Cognitive and neuroimaging features and brain beta-amyloidosis in individuals at risk of Alzheimer's disease (INSIGHT-preAD): a longitudinal observational study, Lancet Neurol, vol.17, issue.4, pp.335-346, 2018.

C. M. Clark, Use of florbetapir-PET for imaging beta-amyloid pathology, JAMA, vol.305, issue.3, pp.275-83, 2011.

F. Cacciamani, Low Cognitive Awareness, but Not Complaint, is a Good Marker of Preclinical Alzheimer's Disease, J Alzheimers Dis, vol.59, issue.2, pp.753-762, 2017.

F. L. Besson, Cognitive and Brain Profiles Associated with Current Neuroimaging Biomarkers of Preclinical Alzheimer's Disease, J Neurosci, vol.35, issue.29, pp.10402-10413, 2015.

S. M. Landau, Amyloid PET imaging in Alzheimer's disease: a comparison of three radiotracers, Eur J Nucl Med Mol Imaging, vol.41, issue.7, pp.1398-407, 2014.

D. R. Thal, Phases of A beta-deposition in the human brain and its relevance for the development of AD, Neurology, vol.58, issue.12, pp.1791-800, 2002.

K. E. Sprecher, Amyloid burden is associated with self-reported sleep in nondemented late middle-aged adults, Neurobiol Aging, vol.36, issue.9, pp.2568-76, 2015.

M. Lehmann, Diverging patterns of amyloid deposition and hypometabolism in clinical variants of probable Alzheimer's disease. Brain, pp.844-58, 2013.

G. D. Rabinovici, Abeta amyloid and glucose metabolism in three variants of primary progressive aphasia, Ann Neurol, vol.64, issue.4, pp.388-401, 2008.

D. L. Bachman, Prevalence of dementia and probable senile dementia of the Alzheimer type in the Framingham Study, Neurology, vol.42, issue.1, pp.115-124, 1992.

F. Beck, J. B. Richard, and D. Leger, Insomnia and total sleep time in France: prevalence and associated socio-demographic factors in a general population survey

, Rev Neurol (Paris), issue.169, pp.956-64, 2013.

P. P. Chang, Insomnia in young men and subsequent depression. The Johns Hopkins Precursors Study, Am J Epidemiol, vol.146, issue.2, pp.105-119, 1997.

R. L. Ownby, Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis, Arch Gen Psychiatry, vol.63, issue.5, pp.530-538, 2006.

M. Murphy, Source modeling sleep slow waves, Proc Natl Acad Sci, vol.106, issue.5, pp.1608-1621, 2009.

M. Schabus, Hemodynamic cerebral correlates of sleep spindles during human non-rapid eye movement sleep, Proc Natl Acad Sci, vol.104, issue.32, pp.13164-13173, 2007.

J. T. O'brien, Vascular cognitive impairment. Lancet Neurol, vol.2, issue.2, pp.89-98, 2003.

C. Y. Cheng, Sleep disturbance correlates with white matter hyperintensity in patients with subcortical ischemic vascular dementia, J Geriatr Psychiatry Neurol, vol.26, issue.3, pp.158-64, 2013.

M. F. Zhu,

G. Zhong, Association between Benzodiazepine Use and Dementia: A Meta-Analysis, PLoS One, vol.10, issue.5, p.127836, 2015.

D. Shash, Benzodiazepine, psychotropic medication, and dementia: A population-based cohort study, Alzheimers Dement, vol.12, issue.5, pp.604-617, 2016.

S. Gimenez, Effects of olanzapine, risperidone and haloperidol on sleep after a single oral morning dose in healthy volunteers, Psychopharmacology (Berl), vol.190, issue.4, pp.507-523, 2007.

J. Kim, J. M. Basak, and D. M. Holtzman, The role of apolipoprotein E in Alzheimer's disease. Neuron, vol.63, pp.287-303, 2009.

R. S. Osorio, Orexin-A is Associated with Increases in Cerebrospinal Fluid Phosphorylated-Tau in Cognitively Normal Elderly Subjects. Sleep, vol.39, issue.6, pp.1253-60, 2016.

B. Jafari and V. Mohsenin, Chitinase-3-like protein-1 (YKL-40) as a marker of endothelial dysfunction in obstructive sleep apnea, Sleep Med, vol.25, pp.87-92, 2016.

R. Craig-schapiro, YKL-40: a novel prognostic fluid biomarker for preclinical Alzheimer's disease, Biol Psychiatry, vol.68, issue.10, pp.903-915, 2010.

B. A. Mander, beta-amyloid disrupts human NREM slow waves and related hippocampus-dependent memory consolidation, Nat Neurosci, vol.18, issue.7, pp.1051-1058, 2015.

Y. E. Ju, B. P. Lucey, and D. M. Holtzman, Sleep and Alzheimer disease pathology--a bidirectional relationship, Nat Rev Neurol, vol.10, issue.2, pp.115-124, 2014.

J. Dufort-gervais, J. V.-mongrain, and . Brouillette, Bidirectional relationships between sleep and amyloid-beta in the hippocampus, Neurobiol Learn Mem, 2018.

E. Shokri-kojori, beta-Amyloid accumulation in the human brain after one night of sleep deprivation, Proc Natl Acad Sci U S A, vol.115, issue.17, pp.4483-4488, 2018.

C. M. Yuede, Effects of voluntary and forced exercise on plaque deposition, hippocampal volume, and behavior in the Tg2576 mouse model of Alzheimer's disease, Neurobiol Dis, vol.35, issue.3, pp.426-458, 2009.

D. Head, Exercise Engagement as a Moderator of the Effects of APOE Genotype on Amyloid Deposition, Arch Neurol, vol.69, issue.5, pp.636-679, 2012.

J. A. Slater, T. Btosis, J. Walsh, S. King, L. M. Straker et al., Résumé : Les troubles du sommeil sont associés à une augmentation du risque de développer des troubles cognitifs tels que les troubles cognitifs légers et la maladie d'Alzheimer. De nombreuses études récentes suggèrent que l'accumulation du peptide bêta-amyloïde (Aß), Sleep Biol Rythms, vol.13, pp.172-180, 2015.

, Cependant, les mécanismes liant le sommeil et les modifications cérébrales sous-jacentes au déclin cognitif ne sont pas encore clairement élucidées

. Objectif, Analyser objectivement les perturbations du cycle veille-sommeil chez les personnes âgées présentant des troubles cognitifs subjectifs avec (A+) ou sans (A-) dépôt amyloïde cérébral significatif

, Méthode: Les participants (68 individus cognitivement sains avec une plainte subjective issus de la cohorte INSIGHT-preAD) ont été invités à enregistrer leur qualité de sommeil pendant sept nuit à domicile par l'intermédiaire d'un accéléromètre, et ont été divisés en deux sousgroupes (A+ et A-) en fonction de la présence d'un dépôt cérébral amyloïde significatif. Les participants ont également passé une imagerie par résonance magnétique afin de mesurer la trophicité hippocampique

, Résultats: Plusieurs paramètres du sommeil étaient significativement différents entre les deux sous-groupes

, Une latence du sommeil plus longue, des éveils nocturnes plus longs et plus nombreux ainsi qu'une plus mauvaise efficacité du sommeil étaient associés à une charge amyloïde cérébrale plus importante. Cependant, le dépôt d'Aß n'était pas associé à une région cérébrale spécifique

, De plus, aucune association n'a été retrouvée entre la qualité du sommeil et la trophicité hippocampique

, Conclusion: Conformément aux données de la littérature, la qualité du sommeil évaluée objectivement était associée à un niveau plus élevé de dépôt amyloïde cérébral. Ces résultats confirment l'importance de la qualité du sommeil chez les personnes âgées

, Mot clés : Sommeil, amyloïde, Maladie d'Alzheimer, florbetapir