1
|
Tsai CY, Su CL, Huang HT, Lin HW, Lin JW, Hei NC, Cheng WH, Chen YL, Majumdar A, Kang JH, Lee KY, Chen Z, Lin YC, Wu CJ, Kuan YC, Lin YT, Hsu CR, Lee HC, Liu WT. Mediating role of obstructive sleep apnea in altering slow-wave activity and elevating Alzheimer's disease risk: Pilot study from a northern Taiwan cohort. Sleep Health 2024:S2352-7218(24)00188-8. [PMID: 39419711 DOI: 10.1016/j.sleh.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/31/2024] [Accepted: 08/31/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES Obstructive sleep apnea is associated with alterations in slow-wave activity during sleep, potentially increasing the risk of Alzheimer's disease. This study investigated the associations between obstructive sleep apnea manifestations such as respiratory events, hypoxia, arousal, slow-wave patterns, and neurochemical biomarker levels. METHODS Individuals with suspected obstructive sleep apnea underwent polysomnography. Sleep disorder indices, oxygen metrics, and slow-wave activity data were obtained from the polysomnography, and blood samples were taken the following morning to determine the plasma levels of total tau (T-Tau) and amyloid beta-peptide 42 (Aβ42) by using an ultrasensitive immunomagnetic reduction assay. Subsequently, the participants were categorized into groups with low and high Alzheimer's disease risk on the basis of their computed product Aβ42 × T-Tau. Intergroup differences and the associations and mediation effects between sleep-related parameters and neurochemical biomarkers were analyzed. RESULTS Forty-two participants were enrolled, with 21 assigned to each of the low- and high-risk groups. High-risk individuals had a higher apnea-hypopnea index, oxygen desaturation index (≥3%, ODI-3%), fraction of total sleep time with oxygen desaturation (SpO2-90% TST), and arousal index and greater peak-to-peak amplitude and slope in slow-wave activity, with a correspondingly shorter duration, than did low-risk individuals. Furthermore, indices such as the apnea-hypopnea index, ODI-3% and SpO2-90% TST were found to indirectly affect slow-wave activity, thereby raising the Aβ42 × T-Tau level. CONCLUSIONS Obstructive sleep apnea manifestations, such as respiratory events and hypoxia, may influence slow-wave sleep activity (functioning as intermediaries) and may be linked to elevated neurochemical biomarker levels. However, a longitudinal study is necessary to determine causal relationships among these factors. STATEMENT OF SIGNIFICANCE This research aims to bridge gaps in understanding how obstructive sleep apnea is associated with an elevated risk of Alzheimer's disease, providing valuable knowledge for sleep and cognitive health.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Chien-Ling Su
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Research Center of Biomedical Devices, Taipei Medical University, Taipei, Taiwan
| | - Huei-Tyng Huang
- Department of Medical Physics and Bioengineering, University College London, United Kingdom
| | - Hsin-Wei Lin
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Wei Lin
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ng Cheuk Hei
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wun-Hao Cheng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Ling Chen
- Institute of Biomedical Informatics of National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Arnab Majumdar
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Jiunn-Horng Kang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Zhihe Chen
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Yi-Chih Lin
- Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Otolaryngology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Cheng-Jung Wu
- Department of Otolaryngology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yin-Tzu Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Rung Hsu
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Hsin-Chien Lee
- Institute of Medical Humanities, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan.
| | - Wen-Te Liu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| |
Collapse
|
2
|
Elberse JD, Saberi A, Ahmadi R, Changizi M, Bi H, Hoffstaedter F, Mander BA, Eickhoff SB, Tahmasian M, Alzheimer’s Disease Neuroimaging Initiative. The interplay between insomnia symptoms and Alzheimer's disease across three main brain networks. Sleep 2024; 47:zsae145. [PMID: 38934787 PMCID: PMC11467060 DOI: 10.1093/sleep/zsae145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/20/2024] [Indexed: 06/28/2024] Open
Abstract
STUDY OBJECTIVES Insomnia symptoms are prevalent along the trajectory of Alzheimer's disease (AD), but the neurobiological underpinning of their interaction is poorly understood. Here, we assessed structural and functional brain measures within and between the default mode network (DMN), salience network, and central executive network (CEN). METHODS We selected 320 participants from the ADNI database and divided them by their diagnosis: cognitively normal (CN), Mild Cognitive Impairment (MCI), and AD, with and without self-reported insomnia symptoms. We measured the gray matter volume (GMV), structural covariance (SC), degrees centrality (DC), and functional connectivity (FC), testing the effect and interaction of insomnia symptoms and diagnosis on each index. Subsequently, we performed a within-group linear regression across each network and ROI. Finally, we correlated observed abnormalities with changes in cognitive and affective scores. RESULTS Insomnia symptoms were associated with FC alterations across all groups. The AD group also demonstrated an interaction between insomnia and diagnosis. Within-group analyses revealed that in CN and MCI, insomnia symptoms were characterized by within-network hyperconnectivity, while in AD, within- and between-network hypoconnectivity was ubiquitous. SC and GMV alterations were nonsignificant in the presence of insomnia symptoms, and DC indices only showed network-level alterations in the CEN of AD individuals. Abnormal FC within and between DMN and CEN hubs was additionally associated with reduced cognitive function across all groups, and increased depressive symptoms in AD. CONCLUSIONS We conclude that patients with clinical AD present with a unique pattern of insomnia-related functional alterations, highlighting the profound interaction between both conditions.
Collapse
Affiliation(s)
- Jorik D Elberse
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Amin Saberi
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Reihaneh Ahmadi
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Faculty of Medicine, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Monir Changizi
- Department of Neurological Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanwen Bi
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Felix Hoffstaedter
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Masoud Tahmasian
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Nuclear Medicine, University Hospital and Medical Faculty, University of Cologne, Cologne, Germany
| | | |
Collapse
|
3
|
Carpi M, Mercuri NB, Liguori C. Orexin Receptor Antagonists for the Prevention and Treatment of Alzheimer's Disease and Associated Sleep Disorders. Drugs 2024:10.1007/s40265-024-02096-3. [PMID: 39365407 DOI: 10.1007/s40265-024-02096-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/05/2024]
Abstract
Orexins/hypocretins are neuropeptides produced by the hypothalamic neurons, binding two G-protein coupled receptors (orexin 1 and orexin 2 receptors) and playing a critical role in regulating arousal, wakefulness, and various physiological functions. Given the high prevalence of sleep disturbances in Alzheimer's disease (AD) and their reported involvement in AD pathophysiology, the orexin system is hypothesized to contribute to the disease pathogenesis. Specifically, recent evidence suggests that orexin's influence may extend beyond sleep regulation, potentially affecting amyloid-β and tau pathologies. Dual orexin receptor antagonists (DORAs), namely suvorexant, lemborexant, and daridorexant, demonstrated efficacy in treating chronic insomnia disorder across diverse clinical populations. Considering their stabilizing effects on sleep parameters and emerging evidence of a possible neuroprotective role, these agents represent a promising strategy for AD management. This leading article reviews the potential use of orexin receptor antagonists in AD, particularly focusing on their effect in modulating disease-associated sleep disturbances and clinical outcomes. Overall, clinical studies support the use of DORAs to enhance sleep quality in patients with AD with comorbid sleep and circadian sleep-wake rhythm disorders. Preliminary results also suggest that these compounds might influence AD pathology, potentially affecting disease progression. Conversely, research on selective orexin receptor antagonists in AD is currently limited. Further investigation is needed to explore orexin antagonism not only as a symptomatic treatment for sleep disturbances, but also for its broader implications in modifying AD neurodegeneration, emphasizing mechanisms of action and long-term outcomes.
Collapse
Affiliation(s)
- Matteo Carpi
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy
| | - Nicola Biagio Mercuri
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy.
- Department of Systems Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy.
| |
Collapse
|
4
|
Fernandes M, Chiaravalloti A, Cassetta E, Placidi F, Mercuri NB, Liguori C. Sleep Fragmentation and Sleep-Wake Cycle Dysregulation Are Associated with Cerebral Tau Burden in Patients with Mild Cognitive Impairment due to Alzheimer's Disease: A Case Series. J Alzheimers Dis Rep 2024; 8:1275-1283. [PMID: 39434815 PMCID: PMC11491934 DOI: 10.3233/adr-230187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/14/2024] [Indexed: 10/23/2024] Open
Abstract
Background Although disturbed sleep is frequent in patients with mild cognitive impairment (MCI) and dementia due to Alzheimer's disease (AD), the association between sleep and tau pathology is unclear. Objective This case series focused on measuring the sleep-wake rhythm over 7 days through actigraphy in patients diagnosed with MCI due to AD. Further, the association between sleep-wake cycle and tau deposition measured through positron emission tomography (PET) was explored. Methods This case series included 6 MCI due to AD patients (2 women and 4 men, mean age 73.17±5.53 years), who completed neuropsychological testing, 7-day actigraphy, and tau PET imaging with radiolabeled compounds aimed to estimate the density and distribution of aggregated tau neurofibrillary tangles in the brain. Results The case series indicated that patients with MCI due to AD who exhibited greater tau deposition in the frontal, parietal, and limbic regions, as well as in the precuneus and olfactory regions, also showed increased sleep fragmentation, as measured through actigraphy. Conclusion The findings from this case series suggest a potential link between tau deposition in key brain regions associated with AD and both sleep fragmentation and sleep-wake cycle dysregulation in a small sample of patients with MCI due to AD. These preliminary results warrant further investigation in larger, more comprehensive studies to confirm and expand upon these findings.
Collapse
Affiliation(s)
- Mariana Fernandes
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Emanuele Cassetta
- Fatebenefratelli Foundation, Associazione Fatebenefratelli Per la Ricerca Division, Fatebenefratelli Hospital, Rome, Italy
| | - Fabio Placidi
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome “Tor Vergata”, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome “Tor Vergata”, Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
5
|
Borges CR, Poyares DLR, Studart-Neto A, Coutinho AM, Cassimiro L, Avolio I, Piovezan R, Trés ES, Teixeira TBM, Barbosa BJAP, Tufik S, Brucki SMD. Amyloid profile is associated with sleep quality in preclinical but not in prodromal Alzheimer's disease older adults. Sleep Med 2024; 121:359-364. [PMID: 39079370 DOI: 10.1016/j.sleep.2024.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Few studies have assessed whether neuropathological markers of AD in the preclinical and prodromal stages are associated with polysomnographic changes and obstructive sleep apnea (OSA). METHODS This was a cross-sectional, case-control study of older adults (≥60 years) without relevant clinical and psychiatric comorbidities selected randomly from a cohort of individuals without dementia in a tertiary university hospital in São Paulo, Brazil. They underwent neuropsychological evaluation for clinical diagnosis and were allocated into two samples: cognitively unimpaired (CU) and mild cognitive impairment (MCI). Also, they underwent PET-PiB to determine the amyloid profile and all-night in-lab polysomnography. For each sample, we compared polysomnographic parameters according to the amyloid profile (A+ vs A-). RESULTS We allocated 67 participants (mean age 73 years, SD 10,1), 70 % females, 14 ± 5 years of education, into two samples: CU (n = 28, 42.4 %) and MCI (n = 39, 57.6 %). In the CU sample, the group A+ (n = 9) showed worse sleep parameters than A- (n = 19) (lower total sleep time (p = 0.007), and sleep efficiency (p = 0.005); higher sleep onset latency (p = 0.025), wake time after sleep onset (p = 0.011), and arousal index (AI) (p = 0.007)), and changes in sleep structure: higher %N1 (p = 0.005), and lower %REM (p = 0.006). In the MCI sample, MCI A-had higher AI (p = 0.013), respiratory disturbance index (p = 0.025, controlled for age), and higher rates of severe OSA than A+. DISCUSSION The amyloid profile was associated with polysomnographic markers of worse sleep quality in individuals with preclinical AD but not with prodromal AD, probably due to the higher frequencies of severe OSA.
Collapse
Affiliation(s)
- Conrado Regis Borges
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil.
| | - Dalva L R Poyares
- Universidade Federal de São Paulo - Escola Paulista de Medicina, R. Botucatu, 862, São Paulo (SP), Brazil
| | - Adalberto Studart-Neto
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Artur M Coutinho
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Luciana Cassimiro
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Isabela Avolio
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Ronaldo Piovezan
- Universidade Federal de São Paulo - Escola Paulista de Medicina, R. Botucatu, 862, São Paulo (SP), Brazil
| | - Eduardo S Trés
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Thiago B M Teixeira
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Breno J A P Barbosa
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| | - Sergio Tufik
- Universidade Federal de São Paulo - Escola Paulista de Medicina, R. Botucatu, 862, São Paulo (SP), Brazil
| | - Sonia M D Brucki
- Universidade de São Paulo - Faculdade de Medicina, R. Dr.Enéas de Carvalho Aguiar, 255, São Paulo (SP), Brazil
| |
Collapse
|
6
|
Falgàs N, Peña‐González M, Val‐Guardiola A, Pérez‐Millan A, Guillén N, Sarto J, Esteller D, Bosch B, Fernández‐Villullas G, Tort‐Merino A, Mayà G, Augé JM, Iranzo A, Balasa M, Lladó A, Morales‐Ruiz M, Bargalló N, Muñoz‐Moreno E, Grinberg LT, Sánchez‐Valle R. Locus coeruleus integrity and neuropsychiatric symptoms in a cohort of early- and late-onset Alzheimer's disease. Alzheimers Dement 2024; 20:6351-6364. [PMID: 39051173 PMCID: PMC11497680 DOI: 10.1002/alz.14131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Early-onset Alzheimer's disease (EOAD) shows a higher burden of neuropsychiatric symptoms than late-onset Alzheimer's disease (LOAD). We aim to determine the differences in the severity of neuropsychiatric symptoms and locus coeruleus (LC) integrity between EOAD and LOAD accounting for disease stage. METHODS One hundred four subjects with AD diagnosis and 32 healthy controls were included. Participants underwent magnetic resonance imaging (MRI) to measure LC integrity, measures of noradrenaline levels in cerebrospinal fluid (CSF) and Neuropsychiatric Inventory (NPI). We analyzed LC-noradrenaline measurements and clinical and Alzheimer's disease (AD) biomarker associations. RESULTS EOAD showed higher NPI scores, lower LC integrity, and similar levels of CSF noradrenaline compared to LOAD. Notably, EOAD exhibited lower LC integrity independently of disease stage. LC integrity negatively correlated with neuropsychiatric symptoms. Noradrenaline levels were increased in AD correlating with AD biomarkers. DISCUSSION Decreased LC integrity negatively contributes to neuropsychiatric symptoms. The higher LC degeneration in EOAD compared to LOAD could explain the more severe neuropsychiatric symptoms in EOAD. HIGHLIGHTS LC degeneration is greater in early-onset AD (EOAD) compared to late-onset AD. Tau-derived LC degeneration drives a higher severity of neuropsychiatric symptoms. EOAD harbors a more profound selective vulnerability of the LC system. LC degeneration is associated with an increase of cerebrospinal fluid noradrenaline levels in AD.
Collapse
Affiliation(s)
- Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
- Global Brain Health InstituteUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Marta Peña‐González
- Magnetic Resonance Imaging Core FacilityInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
| | - Andrea Val‐Guardiola
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Agnès Pérez‐Millan
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Núria Guillén
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Diana Esteller
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Guadalupe Fernández‐Villullas
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Adrià Tort‐Merino
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Gerard Mayà
- Neurology ServiceHospital Clínic de BarcelonaIDIBAPSCIBERNEDUniversitat de BarcelonaBarcelonaSpain
| | - Josep Maria Augé
- Biochemistry and Molecular Genetics Department‐CDBHospital ClinicIDIBAPSCIBERehdBarcelonaSpain
| | - Alex Iranzo
- Neurology ServiceHospital Clínic de BarcelonaIDIBAPSCIBERNEDUniversitat de BarcelonaBarcelonaSpain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| | - Manuel Morales‐Ruiz
- Biochemistry and Molecular Genetics Department‐CDBHospital ClinicIDIBAPSCIBERehdBarcelonaSpain
| | - Núria Bargalló
- Magnetic Resonance Imaging Core FacilityInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
| | - Emma Muñoz‐Moreno
- Magnetic Resonance Imaging Core FacilityInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
| | - Lea T. Grinberg
- Global Brain Health InstituteUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyMemory & Aging CenterWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of Sao Paulo Medical SchoolSao PauloBrazil
| | - Raquel Sánchez‐Valle
- Alzheimer's Disease and Other Cognitive Disorders UnitHospital Clínic de BarcelonaFundació de Recerca Clínic Barcelona‐IDIBAPSUniversitat de BarcelonaBarcelonaCataloniaSpain
| |
Collapse
|
7
|
Zhang Z, Xue P, Bendlin BB, Zetterberg H, De Felice F, Tan X, Benedict C. Melatonin: A potential nighttime guardian against Alzheimer's. Mol Psychiatry 2024:10.1038/s41380-024-02691-6. [PMID: 39128995 DOI: 10.1038/s41380-024-02691-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
In the context of the escalating global health challenge posed by Alzheimer's disease (AD), this comprehensive review considers the potential of melatonin in both preventive and therapeutic capacities. As a naturally occurring hormone and robust antioxidant, accumulating evidence suggests melatonin is a compelling candidate to consider in the context of AD-related pathologies. The review considers several mechanisms, including potential effects on amyloid-beta and pathologic tau burden, antioxidant defense, immune modulation, and regulation of circadian rhythms. Despite its promise, several gaps need to be addressed prior to clinical translation. These include conducting additional randomized clinical trials in patients with or at risk for AD dementia, determining optimal dosage and timing, and further determining potential side effects, particularly of long-term use. This review consolidates existing knowledge, identifies gaps, and suggests directions for future research to better understand the potential of melatonin for neuroprotection and disease mitigation within the landscape of AD.
Collapse
Affiliation(s)
- Zefan Zhang
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Pei Xue
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Barbara B Bendlin
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Fernanda De Felice
- Centre for Neurosciences Studies, Departments of Biomedical and Molecular Sciences, and Psychiatry, Queen's University, Kingston, ON, K7L 3N6, Canada
- D'Or Institute for Research and Education, Rio de Janeiro RJ, 22281-100, Brazil
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, 21941-902, Rio de Janeiro RJ, Brazil
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Christian Benedict
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
André C, Martineau-Dussault MÈ, Baril AA, Marchi NA, Daneault V, Lorrain D, Hudon C, Bastien CH, Petit D, Thompson C, Poirier J, Montplaisir J, Gosselin N, Carrier J. Reduced rapid eye movement sleep in late middle-aged and older apolipoprotein E ɛ4 allele carriers. Sleep 2024; 47:zsae094. [PMID: 38634644 PMCID: PMC11236949 DOI: 10.1093/sleep/zsae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
STUDY OBJECTIVES Apolipoprotein E ɛ4 (APOE4) is the strongest genetic risk factor for Alzheimer's disease (AD). In addition, APOE4 carriers may exhibit sleep disturbances, but conflicting results have been reported, such that there is no clear consensus regarding which aspects of sleep are impacted. Our objective was to compare objective sleep architecture between APOE4 carriers and non-carriers, and to investigate the modulating impact of age, sex, cognitive status, and obstructive sleep apnea (OSA). METHODS A total of 198 dementia-free participants aged >55 years old (mean age: 68.7 ± 8.08 years old, 40.91% women, 41 APOE4 carriers) were recruited in this cross-sectional study. They underwent polysomnography, APOE4 genotyping, and a neuropsychological evaluation. ANCOVAs assessed the effect of APOE4 status on sleep architecture, controlling for age, sex, cognitive status, and the apnea-hypopnea index. Interaction terms were added between APOE4 status and covariates. RESULTS Rapid eye movement (REM) sleep percentage (F = 9.95, p = .002, ηp2 = 0.049) and duration (F = 9.23, p = .003, ηp2 = 0.047) were lower in APOE4 carriers. The results were replicated in a subsample of 112 participants without moderate-to-severe OSA. There were no significant interactions between APOE4 status and age, sex, cognitive status, and OSA in the whole sample. CONCLUSIONS Our results show that APOE4 carriers exhibit lower REM sleep duration, including in cognitively unimpaired individuals, possibly resulting from early neurodegenerative processes in regions involved in REM sleep generation and maintenance.
Collapse
Affiliation(s)
- Claire André
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Marie-Ève Martineau-Dussault
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Andrée-Ann Baril
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Nicola Andrea Marchi
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
- Center for Investigation and Research in Sleep, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Véronique Daneault
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Dominique Lorrain
- Research Centre on Aging, University Institute of Geriatrics of Sherbrooke, CIUSSS de l’Estrie-CHUS, Sherbrooke, QC, Canada
- Department of Psychology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Carol Hudon
- CERVO Brain Research Centre, Institut Universitaire en Santé Mentale de Québec, Québec City, QC, Canada
- School of Psychology, Université Laval, Québec City, QC, Canada
| | - Célyne H Bastien
- CERVO Brain Research Centre, Institut Universitaire en Santé Mentale de Québec, Québec City, QC, Canada
- School of Psychology, Université Laval, Québec City, QC, Canada
| | - Dominique Petit
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychiatry, Université de Montréal, Montréal, QC, Canada
| | - Cynthia Thompson
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
| | - Judes Poirier
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, CIUSSS de l’Ouest-de-l’Ile-de-Montréal, Verdun, QC, Canada
| | - Jacques Montplaisir
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychiatry, Université de Montréal, Montréal, QC, Canada
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Julie Carrier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
9
|
Deantoni M, Reyt M, Dourte M, de Haan S, Lesoinne A, Vandewalle G, Phillips C, Berthomier C, Maquet P, Muto V, Hammad G, Schmidt C, Baillet M. Circadian rapid eye movement sleep expression is associated with brain microstructural integrity in older adults. Commun Biol 2024; 7:758. [PMID: 38909162 PMCID: PMC11193799 DOI: 10.1038/s42003-024-06415-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/05/2024] [Indexed: 06/24/2024] Open
Abstract
Rapid eye movement sleep (REMS) is increasingly suggested as a discriminant sleep state for subtle signs of age-related neurodegeneration. While REMS expression is under strong circadian control and circadian dysregulation increases with age, the association between brain aging and circadian REMS regulation has not yet been assessed. Here, we measure the circadian amplitude of REMS through a 40-h in-lab multiple nap protocol in controlled laboratory conditions, and brain microstructural integrity with quantitative multi-parameter mapping (MPM) imaging in 86 older individuals. We show that reduced circadian REMS amplitude is related to lower magnetization transfer saturation (MTsat), longitudinal relaxation rate (R1) and effective transverse relaxation rate (R2*) values in several white matter regions mostly located around the lateral ventricles, and with lower R1 values in grey matter clusters encompassing the hippocampus, parahippocampus, thalamus and hypothalamus. Our results further highlight the importance of considering circadian regulation for understanding the association between sleep and brain structure in older individuals.
Collapse
Affiliation(s)
| | - Mathilde Reyt
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Faculty of Psychology and Educational Sciences, University of Liège, Liège, Belgium
| | - Marine Dourte
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Faculty of Psychology and Educational Sciences, University of Liège, Liège, Belgium
| | - Stella de Haan
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
| | | | | | - Christophe Phillips
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
- GIGA-In Silico Medicine, University of Liège, Liège, Belgium
| | | | - Pierre Maquet
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
- Department of Neurology, University Hospital of Liège, University of Liège, Liège, Belgium
| | - Vincenzo Muto
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
| | - Grégory Hammad
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium
- Human Chronobiology and Sleep, University of Surrey, Guildford, England
| | - Christina Schmidt
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium.
- Psychology and Neuroscience of Cognition Research Unit (PsyNCog), Faculty of Psychology and Educational Sciences, University of Liège, Liège, Belgium.
| | - Marion Baillet
- GIGA-CRC Human Imaging, University of Liège, Liège, Belgium.
| |
Collapse
|
10
|
Chen Y, Al-Nusaif M, Li S, Tan X, Yang H, Cai H, Le W. Progress on early diagnosing Alzheimer's disease. Front Med 2024; 18:446-464. [PMID: 38769282 PMCID: PMC11391414 DOI: 10.1007/s11684-023-1047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 05/22/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects both cognition and non-cognition functions. The disease follows a continuum, starting with preclinical stages, progressing to mild cognitive and behavioral impairment, ultimately leading to dementia. Early detection of AD is crucial for better diagnosis and more effective treatment. However, the current AD diagnostic tests of biomarkers using cerebrospinal fluid and/or brain imaging are invasive or expensive, and mostly are still not able to detect early disease state. Consequently, there is an urgent need to develop new diagnostic techniques with higher sensitivity and specificity during the preclinical stages of AD. Various non-cognitive manifestations, including behavioral abnormalities, sleep disturbances, sensory dysfunctions, and physical changes, have been observed in the preclinical AD stage before occurrence of notable cognitive decline. Recent research advances have identified several biofluid biomarkers as early indicators of AD. This review focuses on these non-cognitive changes and newly discovered biomarkers in AD, specifically addressing the preclinical stages of the disease. Furthermore, it is of importance to explore the potential for developing a predictive system or network to forecast disease onset and progression at the early stage of AD.
Collapse
Affiliation(s)
- Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
11
|
Hitrec T, Squarcio F, Piscitiello E, Cerri M, Martelli D, Occhinegro A, Taddei L, Tupone D, Amici R, Luppi M. Sleep deprivation soon after recovery from synthetic torpor enhances tau protein dephosphorylation in the rat brain. J Comp Physiol B 2024; 194:347-368. [PMID: 37812305 DOI: 10.1007/s00360-023-01516-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/11/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023]
Abstract
Neuronal Tau protein hyperphosphorylation (PPtau) is a hallmark of tauopathic neurodegeneration. However, a reversible brain PPtau occurs in mammals during either natural or "synthetic" torpor (ST), a transient deep hypothermic state that can be pharmacologically induced in rats. Since in both conditions a high sleep pressure builds up during the regaining of euthermia, the aim of this work was to assess the possible role of post-ST sleep in PPtau dephosphorylation. Male rats were studied at the hypothermic nadir of ST, and 3-6 h after the recovery of euthermia, after either normal sleep (NS) or total sleep deprivation (SD). The effects of SD were studied by assessing: (i) deep brain temperature (Tb); (ii) immunofluorescent staining for AT8 (phosphorylated Tau) and Tau-1 (non-phosphorylated Tau), assessed in 19 brain structures; (iii) different phosphorylated forms of Tau and the main cellular factors involved in Tau phospho-regulation, including pro- and anti-apoptotic markers, assessed through western blot in the parietal cortex and hippocampus; (iv) systemic factors which are involved in natural torpor; (v) microglia activation state, by considering morphometric variations. Unexpectedly, the reversibility of PPtau was more efficient in SD than in NS animals, and was concomitant with a higher Tb, higher melatonin plasma levels, and a higher frequency of the microglia resting phenotype. Since the reversibility of ST-induced PPtau was previously shown to be driven by a latent physiological molecular mechanism triggered by deep hypothermia, short-term SD soon after the regaining of euthermia seems to boost the possible neuroprotective effects of this mechanism.
Collapse
Affiliation(s)
- Timna Hitrec
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
| | - Fabio Squarcio
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Emiliana Piscitiello
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy
| | - Matteo Cerri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
| | - Davide Martelli
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy
| | - Alessandra Occhinegro
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy
| | - Ludovico Taddei
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy
| | - Domenico Tupone
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Roberto Amici
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy
| | - Marco Luppi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
- Centre for Applied Biomedical Research - CRBA, University of Bologna, St. Orsola Hospital, Bologna, Italy.
| |
Collapse
|
12
|
Qin Y, Zhao Y, Hu X, Chen X, Jiang YP, Jin XJ, Li G, Li ZH, Yang JH, Zhang GL, Cui SY, Zhang YH. Ganoderma lucidum spore extract improves sleep disturbances in a rat model of sporadic Alzheimer's disease. Front Pharmacol 2024; 15:1390294. [PMID: 38720773 PMCID: PMC11076761 DOI: 10.3389/fphar.2024.1390294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction: Ganoderma lucidum (G. lucidum, Lingzhi) has long been listed as a premium tonic that can be used to improve restlessness, insomnia, and forgetfulness. We previously reported that a rat model of sporadic Alzheimer's disease (sAD) that was induced by an intracerebroventricular injection of streptozotocin (ICV-STZ) showed significant learning and cognitive deficits and sleep disturbances. Treatment with a G. lucidum spore extract with the sporoderm removed (RGLS) prevented learning and memory impairments in sAD model rats. Method: The present study was conducted to further elucidate the preventive action of RGLS on sleep disturbances in sAD rats by EEG analysis, immunofluorescence staining, HPLC-MS/MS and Western blot. Results: Treatment with 720 mg/kg RGLS for 14 days significantly improved the reduction of total sleep time, rapid eye movement (REM) sleep time, and non-REM sleep time in sAD rats. The novelty recognition experiment further confirmed that RGLS prevented cognitive impairments in sAD rats. We also found that RGLS inhibited the nuclear factor-κB (NF-κB)/Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammatory pathway in the medial prefrontal cortex (mPFC) in sAD rats and ameliorated the lower activity of γ-aminobutyric acid (GABA)-ergic neurons in the parabrachial nucleus (PBN). Discussion: These results suggest that inhibiting the neuroinflammatory response in the mPFC may be a mechanism by which RGLS improves cognitive impairment. Additionally, improvements in PBN-GABAergic activity and the suppression of neuroinflammation in the mPFC in sAD rats might be a critical pathway to explain the preventive effects of RGLS on sleep disturbances in sAD.
Collapse
Affiliation(s)
- Yu Qin
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yan Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Xiao Hu
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Xi Chen
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yan-Ping Jiang
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Xue-Jun Jin
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Gao Li
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Zhen-Hao Li
- Zhejiang ShouXianGu Pharmaceutical Co., Ltd., Wuyi, Zhejiang, China
| | - Ji-Hong Yang
- Zhejiang ShouXianGu Pharmaceutical Co., Ltd., Wuyi, Zhejiang, China
| | - Guo-Liang Zhang
- Zhejiang ShouXianGu Pharmaceutical Co., Ltd., Wuyi, Zhejiang, China
| | - Su-Ying Cui
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yong-He Zhang
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| |
Collapse
|
13
|
Zadrozny M, Drapich P, Gasiorowska-Bien A, Niewiadomski W, Harrington CR, Wischik CM, Riedel G, Niewiadomska G. Neuroprotection of Cholinergic Neurons with a Tau Aggregation Inhibitor and Rivastigmine in an Alzheimer's-like Tauopathy Mouse Model. Cells 2024; 13:642. [PMID: 38607082 PMCID: PMC11011792 DOI: 10.3390/cells13070642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Basal forebrain cholinergic dysfunction, most likely linked with tau protein aggregation, is a characteristic feature of Alzheimer's disease (AD). Recent evidence suggests that tau protein is a putative target for the treatment of dementia, and the tau aggregation inhibitor, hydromethylthionine mesylate (HMTM), has emerged as a potential disease-modifying treatment. However, its efficacy was diminished in patients already receiving approved acetylcholinesterase inhibitors. In this study, we ask whether this negative interaction can also be mimicked in experimental tau models of AD and whether the underlying mechanism can be understood. From a previous age profiling study, 6-month-old line 1 (L1) tau transgenic mice were characterized by a severe reduction in several cholinergic markers. We therefore assessed whether long-term pre-exposure with the acetylcholinesterase inhibitor rivastigmine alone and in conjunction with the tau aggregation inhibitor HMTM can reverse cholinergic deficits in L1. Rivastigmine and HMTM, and combinations of the two compounds were administered orally for 11 weeks to both L1 and wild-type mice. The brains were sectioned with a focus on the basal forebrain, motor cortex and hippocampus. Immunohistochemical staining and quantification of choline acetyltransferase (ChAT), tyrosine kinase A (TrkA)-positive neurons and relative optical intensity (ROI) for vesicular acetylcholine transporter (VAChT), and acetylcholinesterase (AChE) reactivity confirmed reversal of the diminished cholinergic phenotype of interneurons (nucleus accumbens, striatum) and projection neurons (medial septum, nucleus basalis magnocellularis) by HMTM, to a greater extent than by rivastigmine alone in L1 mice. Combined administration did not yield additivity but, in most proxies, led to antagonistic effects in which rivastigmine decreased the benefits shown with HMTM alone. Local markers (VAChT and AChE) in target structures of the basal forebrain, motor cortex and hippocampal CA3 seemed to be normalized by HMTM, but not by rivastigmine or the combination of both drugs. HMTM, which was developed as a tau aggregation inhibitor, strongly decreased the tau load in L1 mice, however, not in combination with rivastigmine. Taken together, these data confirm a cholinergic phenotype in L1 tau transgenic mice that resembles the deficits observed in AD patients. This phenotype is reversible by HMTM, but at the same time appears to be subject to a homeostatic regulation induced by chronic pre-treatment with an acetylcholinesterase inhibitor, which interferes with the efficacy of HMTM. The strongest phenotypic reversal coincided with a normalization of the tau load in the cortex and hippocampus of L1, suggesting that tau accumulation underpins the loss of cholinergic markers in the basal forebrain and its projection targets.
Collapse
Affiliation(s)
- Maciej Zadrozny
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Patrycja Drapich
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Anna Gasiorowska-Bien
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Wiktor Niewiadomski
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Charles R. Harrington
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
- TauRx Therapeutics Ltd., Aberdeen AB24 3FX, UK
| | - Claude M. Wischik
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
- TauRx Therapeutics Ltd., Aberdeen AB24 3FX, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
| | - Grazyna Niewiadomska
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
- Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| |
Collapse
|
14
|
Van Egroo M, van Someren EJW, Grinberg LT, Bennett DA, Jacobs HIL. Associations of 24-Hour Rest-Activity Rhythm Fragmentation, Cognitive Decline, and Postmortem Locus Coeruleus Hypopigmentation in Alzheimer's Disease. Ann Neurol 2024; 95:653-664. [PMID: 38407546 DOI: 10.1002/ana.26880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVE While studies suggested that locus coeruleus (LC) neurodegeneration contributes to sleep-wake dysregulation in Alzheimer's disease (AD), the association between LC integrity and circadian rest-activity patterns remains unknown. Here, we investigated the relationships between 24-hour rest-activity rhythms, cognitive trajectories, and autopsy-derived LC integrity in older adults with and without cortical AD neuropathology. METHODS This retrospective study leveraged multi-modal data from participants of the longitudinal clinical-pathological Rush Memory and Aging Project. Indices of 24-hour rest-activity rhythm fragmentation (intradaily variability) and stability (interdaily stability) were extracted from annual actigraphic recordings, and cognitive trajectories were computed from annual cognitive evaluations. At autopsy, LC neurodegeneration was determined by the presence of hypopigmentation, and cortical AD neuropathology was assessed. Contributions of comorbid pathologies (Lewy bodies, cerebrovascular pathology) were evaluated. RESULTS Among the 388 cases included in the study sample (age at death = 92.1 ± 5.9 years; 273 women), 98 (25.3%) displayed LC hypopigmentation, and 251 (64.7%) exhibited cortical AD neuropathology. Logistic regression models showed that higher rest-activity rhythm fragmentation, measured up to ~7.1 years before death, was associated with increased risk to display LC neurodegeneration at autopsy (odds ratio [OR] = 1.46, 95% confidence interval [CI95%]: 1.16-1.84, pBONF = 0.004), particularly in individuals with cortical AD neuropathology (OR = 1.56, CI95%: 1.15-2.15, pBONF = 0.03) and independently of comorbid pathologies. In addition, longitudinal increases in rest-activity rhythm fragmentation partially mediated the association between LC neurodegeneration and cognitive decline (estimate = -0.011, CI95%: -0.023--0.002, pBONF = 0.03). INTERPRETATION These findings highlight the LC as a neurobiological correlate of sleep-wake dysregulation in AD, and further underscore the clinical relevance of monitoring rest-activity patterns for improved detection of at-risk individuals. ANN NEUROL 2024;95:653-664.
Collapse
Affiliation(s)
- Maxime Van Egroo
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Eus J W van Someren
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lea T Grinberg
- Department of Pathology, LIM-22, University of São Paulo Medical School, São Paulo, Brazil
- Memory and Aging Center, Department of Neurology, and Pathology, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Heidi I L Jacobs
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Kim RT, Zhou L, Li Y, Krieger AC, Nordvig AS, Butler T, de Leon MJ, Chiang GC. Impaired sleep is associated with tau deposition on 18F-flortaucipir PET and accelerated cognitive decline, accounting for medications that affect sleep. J Neurol Sci 2024; 458:122927. [PMID: 38341949 PMCID: PMC10947806 DOI: 10.1016/j.jns.2024.122927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Impaired sleep is commonly associated with Alzheimer's disease (AD), although the underlying mechanisms remain unclear. Furthermore, the moderating effects of sleep-affecting medications, which have been linked to AD pathology, are incompletely characterized. Using data from the Alzheimer's Disease Neuroimaging Initiative, we investigated whether a medical history of impaired sleep, informant-reported nighttime behaviors, and sleep-affecting medications are associated with beta-amyloid and tau deposition on PET and cognitive change, cross-sectionally and longitudinally. METHODS We included 964 subjects with 18F-florbetapir PET scans. Measures of sleep impairment and medication use were obtained from medical histories and the Neuropsychiatric Inventory Questionnaire. Multivariate models, adjusted for covariates, were used to assess associations among sleep-related features, beta-amyloid and tau, and cognition. Cortical tau deposition, categorized by Braak stage, was assessed using the standardized uptake value peak alignment (SUVP) method on 18F-flortaucipir PET. RESULTS Medical history of sleep impairment was associated with greater baseline tau in the meta-temporal, Braak 1, and Braak 4 regions (p = 0.04, p < 0.001, p = 0.025, respectively). Abnormal nighttime behaviors were also associated with greater baseline tau in the meta-temporal region (p = 0.024), and greater cognitive impairment, cross-sectionally (p = 0.007) and longitudinally (p < 0.001). Impaired sleep was not associated with baseline beta-amyloid (p > 0.05). Short-term use of selective serotonin reuptake inhibitors and benzodiazepines slightly weakened the sleep-tau relationship. CONCLUSIONS Sleep impairment was associated with tauopathy and cognitive decline, which could be linked to increased tau secretion from neuronal hyperactivity. Clinically, our results help identify high-risk individuals who could benefit from sleep-related interventions aimed to delay cognitive decline and AD.
Collapse
Affiliation(s)
- Ryan T Kim
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Bauer-Sherman Fairchild Complex 7 Divinity Avenue, Cambridge, MA 02138, United States of America.
| | - Liangdong Zhou
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Yi Li
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Ana C Krieger
- From the Departments of Medicine and Neurology, Division of Sleep Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 425 E 61st St., 5th Floor, New York, NY 10065, United States of America.
| | - Anna S Nordvig
- From the Department of Neurology, Alzheimer's Disease and Memory Disorders Program, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 428 East 72(nd) Street Suite 500, New York, NY 10021, United States of America.
| | - Tracy Butler
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Mony J de Leon
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Gloria C Chiang
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America; From the Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, United States of America.
| |
Collapse
|
16
|
Samudra N, Lerner H, Yack L, Walsh CM, Kirsch HE, Kudo K, Yballa C, La Joie R, Gorno‐Tempini ML, Spina S, Seeley WW, Neylan TC, Miller BL, Rabinovici GD, Boxer A, Grinberg LT, Rankin KP, Nagarajan SS, Ranasinghe KG. Spatiotemporal characteristics of neurophysiological changes in patients with four-repeat tauopathies. Ann Clin Transl Neurol 2024; 11:525-535. [PMID: 38226843 PMCID: PMC10863921 DOI: 10.1002/acn3.51974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024] Open
Abstract
INTRODUCTION Progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), are the most common four-repeat tauopathies (4RT), and both frequently occur with varying degree of Alzheimer's disease (AD) copathology. Intriguingly, patients with 4RT and patients with AD are at opposite ends of the wakefulness spectrum-AD showing reduced wakefulness and excessive sleepiness whereas 4RT showing decreased homeostatic sleep. The neural mechanisms underlying these distinct phenotypes in the comorbid condition of 4RT and AD are unknown. The objective of the current study was to define the alpha oscillatory spectrum, which is prominent in the awake resting-state in the human brain, in patients with primary 4RT, and how it is modified in comorbid AD-pathology. METHOD In an autopsy-confirmed case series of 4R-tauopathy patients (n = 10), whose primary neuropathological diagnosis was either PSP (n = 7) or CBD (n = 3), using high spatiotemporal resolution magnetoencephalography (MEG), we quantified the spectral power density within alpha-band (8-12 Hz) and examined how this pattern was modified in increasing AD-copathology. For each patient, their regional alpha power was compared to an age-matched normative control cohort (n = 35). RESULT Patients with 4RT showed increased alpha power but in the presence of AD-copathology alpha power was reduced. CONCLUSIONS Alpha power increase in PSP-tauopathy and reduction in the presence of AD-tauopathy is consistent with the observation that neurons activating wakefulness-promoting systems are preserved in PSP but degenerated in AD. These results highlight the selectively vulnerable impacts in 4RT versus AD-tauopathy that may have translational significance on disease-modifying therapies for specific proteinopathies.
Collapse
Affiliation(s)
- Niyatee Samudra
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Hannah Lerner
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Leslie Yack
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of PsychiatrySan Francisco Veterans Affairs, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Christine M. Walsh
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Heidi E. Kirsch
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
- Epilepsy Center, Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kiwamu Kudo
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
- Medical Imaging Business CenterRicoh CompanyKanazawaJapan
| | - Claire Yballa
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Renaud La Joie
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Maria L. Gorno‐Tempini
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Salvatore Spina
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - William W. Seeley
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Thomas C. Neylan
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of PsychiatrySan Francisco Veterans Affairs, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Bruce L. Miller
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Gil D. Rabinovici
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
| | - Adam Boxer
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of PathologyUniversity of CaliforniaSan FranciscoCalifornia94158USA
- Department of PathologyUniversity of Sao Paulo Medical SchoolSao PauloBrazil
| | - Katherine P. Rankin
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Srikantan S. Nagarajan
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
| | - Kamalini G. Ranasinghe
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| |
Collapse
|
17
|
Kron JOZJ, Keenan RJ, Hoyer D, Jacobson LH. Orexin Receptor Antagonism: Normalizing Sleep Architecture in Old Age and Disease. Annu Rev Pharmacol Toxicol 2024; 64:359-386. [PMID: 37708433 DOI: 10.1146/annurev-pharmtox-040323-031929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Sleep is essential for human well-being, yet the quality and quantity of sleep reduce as age advances. Older persons (>65 years old) are more at risk of disorders accompanied and/or exacerbated by poor sleep. Furthermore, evidence supports a bidirectional relationship between disrupted sleep and Alzheimer's disease (AD) or related dementias. Orexin/hypocretin neuropeptides stabilize wakefulness, and several orexin receptor antagonists (ORAs) are approved for the treatment of insomnia in adults. Dysregulation of the orexin system occurs in aging and AD, positioning ORAs as advantageous for these populations. Indeed, several clinical studies indicate that ORAs are efficacious hypnotics in older persons and dementia patients and, as in adults, are generally well tolerated. ORAs are likely to be more effective when administered early in sleep/wake dysregulation to reestablish good sleep/wake-related behaviors and reduce the accumulation of dementia-associated proteinopathic substrates. Improving sleep in aging and dementia represents a tremendous opportunity to benefit patients, caregivers, and health systems.
Collapse
Affiliation(s)
- Jarrah O-Z J Kron
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
| | - Ryan J Keenan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia;
| |
Collapse
|
18
|
Son G, Neylan TC, Grinberg LT. Neuronal and glial vulnerability of the suprachiasmatic nucleus in tauopathies: evidence from human studies and animal models. Mol Neurodegener 2024; 19:4. [PMID: 38195580 PMCID: PMC10777507 DOI: 10.1186/s13024-023-00695-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/08/2023] [Indexed: 01/11/2024] Open
Abstract
Tauopathies, a group of neurodegenerative diseases that includes Alzheimer's disease, commonly lead to disturbances in sleep-wake patterns and circadian rhythm disorders. The circadian rhythm, a recurring 24-hour cycle governing human biological activity, is regulated by the hypothalamic suprachiasmatic nucleus (SCN) and endogenous transcriptional-translational feedback loops. Surprisingly, little attention has been given to investigating tauopathy-driven neuropathology in the SCN and the repercussions of SCN and circadian gene dysfunction in the human brain affected by tauopathies. This review aims to provide an overview of the current literature on the vulnerability of the SCN in tauopathies in humans. Emphasis is placed on elucidating the neuronal and glial changes contributing to the widespread disruption of the molecular circadian clock. Furthermore, this review identifies areas of knowledge requiring further investigation.
Collapse
Affiliation(s)
- Gowoon Son
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas C Neylan
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
19
|
Baril A, Picard C, Labonté A, Sanchez E, Duclos C, Mohammediyan B, Ashton NJ, Zetterberg H, Blennow K, Breitner JCS, Villeneuve S, Poirier J. Day-to-day sleep variability with Alzheimer's biomarkers in at-risk elderly. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12521. [PMID: 38371359 PMCID: PMC10870017 DOI: 10.1002/dad2.12521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Measuring day-to-day sleep variability might reveal unstable sleep-wake cycles reflecting neurodegenerative processes. We evaluated the association between Alzheimer's disease (AD) fluid biomarkers with day-to-day sleep variability. METHODS In the PREVENT-AD cohort, 203 dementia-free participants (age: 68.3 ± 5.4; 78 males) with a parental history of sporadic AD were tested with actigraphy and fluid biomarkers. Day-to-day variability (standard deviations over a week) was assessed for sleep midpoint, duration, efficiency, and nighttime activity count. RESULTS Lower cerebrospinal fluid (CSF) ApoE, higher CSF p-tau181/amyloid-β (Aβ)42, and higher plasma p-tau231/Aβ42 were associated with higher variability of sleep midpoint, sleep duration, and/or activity count. The associations between fluid biomarkers with greater sleep duration variability were especially observed in those that carried the APOE4 allele, mild cognitive impairment converters, or those with gray matter atrophy. DISCUSSION Day-to-day sleep variability were associated with biomarkers of AD in at-risk individuals, suggesting that unstable sleep promotes neurodegeneration or, conversely, that AD neuropathology disrupts sleep-wake cycles.
Collapse
Affiliation(s)
- Andrée‐Ann Baril
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Cynthia Picard
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Anne Labonté
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Erlan Sanchez
- Sunnybrook Research InstituteUniversity of TorontoTorontoOntarioCanada
| | - Catherine Duclos
- Hôpital du Sacré‐Coeur de MontréalCIUSSS‐NIMMontréalQuébecCanada
- Department of Anesthesiology and Pain MedicineUniversité de MontréalMontréalQuébecCanada
| | - Béry Mohammediyan
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- King's College LondonInstitute of PsychiatryPsychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - John C. S. Breitner
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Sylvia Villeneuve
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Judes Poirier
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | | |
Collapse
|
20
|
Yang Y, Kim WS, Michaelian JC, Lewis SJG, Phillips CL, D'Rozario AL, Chatterjee P, Martins RN, Grunstein R, Halliday GM, Naismith SL. Predicting neurodegeneration from sleep related biofluid changes. Neurobiol Dis 2024; 190:106369. [PMID: 38049012 DOI: 10.1016/j.nbd.2023.106369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023] Open
Abstract
Sleep-wake disturbances are common in neurodegenerative diseases and may occur years before the clinical diagnosis, potentially either representing an early stage of the disease itself or acting as a pathophysiological driver. Therefore, discovering biomarkers that identify individuals with sleep-wake disturbances who are at risk of developing neurodegenerative diseases will allow early diagnosis and intervention. Given the association between sleep and neurodegeneration, the most frequently analyzed fluid biomarkers in people with sleep-wake disturbances to date include those directly associated with neurodegeneration itself, such as neurofilament light chain, phosphorylated tau, amyloid-beta and alpha-synuclein. Abnormalities in these biomarkers in patients with sleep-wake disturbances are considered as evidence of an underlying neurodegenerative process. Levels of hormonal sleep-related biomarkers such as melatonin, cortisol and orexin are often abnormal in patients with clinical neurodegenerative diseases, but their relationships with the more standard neurodegenerative biomarkers remain unclear. Similarly, it is unclear whether other chronobiological/circadian biomarkers, such as disrupted clock gene expression, are causal factors or a consequence of neurodegeneration. Current data would suggest that a combination of fluid biomarkers may identify sleep-wake disturbances that are most predictive for the risk of developing neurodegenerative disease with more optimal sensitivity and specificity.
Collapse
Affiliation(s)
- Yue Yang
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia.
| | - Woojin Scott Kim
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Johannes C Michaelian
- Healthy Brain Ageing Program, School of Psychology, Brain and Mind Centre & The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2050, Australia.
| | - Simon J G Lewis
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; Parkinson's Disease Research Clinic, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia.
| | - Craig L Phillips
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2109, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Angela L D'Rozario
- Healthy Brain Ageing Program, School of Psychology, Brain and Mind Centre & The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2050, Australia; CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2109, Australia.
| | - Pratishtha Chatterjee
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Ralph N Martins
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia; School of Psychiatry and Clinical Neurosciences, University of Western Australia, Perth, WA 6009, Australia.
| | - Ron Grunstein
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2109, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Glenda M Halliday
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Sharon L Naismith
- Healthy Brain Ageing Program, School of Psychology, Brain and Mind Centre & The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2050, Australia.
| |
Collapse
|
21
|
Paolillo EW, Saloner R, VandeBunte A, Lee S, Bennett DA, Casaletto KB. Multimodal lifestyle engagement patterns support cognitive stability beyond neuropathological burden. Alzheimers Res Ther 2023; 15:221. [PMID: 38111051 PMCID: PMC10726589 DOI: 10.1186/s13195-023-01365-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/03/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Modifiable lifestyle behaviors account for a large proportion of dementia risk. However, the combined contributions of multidomain lifestyle patterns to cognitive aging are poorly understood, as most studies have examined individual lifestyle behaviors in isolation and without neuropathological characterization. This study examined data-driven patterns of lifestyle behaviors across multiple domains among older adults and tested their associations with disease-specific neuropathological burden and cognitive decline. METHODS Participants included 2059 older adults enrolled in the longitudinal Memory and Aging Project (MAP) at the Rush Alzheimer's Disease Center; none of whom had dementia at baseline (73% no cognitive impairment (NCI), 27% mild cognitive impairment [MCI]). All participants completed cognitive testing annually. Lifestyle factors were measured during at least one visit and included (1) actigraphy-measured physical activity, as well as self-reported (2) sleep quality, (3) life space, (4) cognitive activities, (5) social activities, and (6) social network. A subset of participants (n = 791) had autopsy data for which burden of Alzheimer's disease (AD), cerebrovascular disease (CVD), Lewy body disease, and hippocampal sclerosis/TDP-43 was measured. Latent profile analysis across all 2059 participants identified distinct subgroups (i.e., classes) of lifestyle patterns. Linear mixed-effects models examined relationships between lifestyle classes and global cognitive trajectories, with and without covarying for all neuropathologies. Classes were also compared on rates of incident MCI/dementia. RESULTS Five classes were identified: Class 1Low Life Space (lowest lifestyle engagement), Class 2PA (high physical activity), Class 3Low Avg (low to average lifestyle engagement), Class 4Balanced (high average lifestyle engagement), and Class 5Social (large social network). Classes 4Balanced and 5Social had the lowest AD burden, and Class 2PA had the lowest CVD burden. Classes 2-5 had significantly less steep global cognitive decline compared to Class 1Low Life Space, with comparable effect sizes before and after covarying for neuropathological burden. Classes 4Balanced and 5Social exhibited the lowest rates of incident MCI/dementia. CONCLUSIONS Lifestyle behavior patterns among older adults account for differential rates of cognitive decline and clinical progression. Those with at least average engagement across all lifestyle domains exhibit greater cognitive stability after adjustment for neuropathology, highlighting the importance of engagement in multiple healthy lifestyle behaviors for later life cognitive health.
Collapse
Affiliation(s)
- Emily W Paolillo
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
| | - Rowan Saloner
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Anna VandeBunte
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Shannon Lee
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
| | - Kaitlin B Casaletto
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
22
|
Yoon SH, Kim HK, Lee JH, Chun JH, Sohn YH, Lee PH, Ryu YH, Cho H, Yoo HS, Lyoo CH. Association of Sleep Disturbances With Brain Amyloid and Tau Burden, Cortical Atrophy, and Cognitive Dysfunction Across the AD Continuum. Neurology 2023; 101:e2162-e2171. [PMID: 37813585 PMCID: PMC10663023 DOI: 10.1212/wnl.0000000000207917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/24/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Patients with Alzheimer disease (AD) frequently suffer from various sleep disturbances. However, how sleep disturbance is associated with AD and its progression remains poorly investigated. We investigated the association of total sleep time with brain amyloid and tau burden, cortical atrophy, cognitive dysfunction, and their longitudinal changes in the AD spectrum. METHODS In this retrospective cohort study, we enrolled participants on the AD spectrum who were positive on 18F-florbetaben (FBB) PET. All participants underwent the Pittsburgh Sleep Quality Index, brain MRI, FBB PET, 18F-flortaucipir (FTP) PET, and detailed neuropsychological testing. In addition, a subset of participants completed follow-up assessments. We analyzed the association of total sleep time with the baseline and longitudinal FBB-standardized uptake value ratio (SUVR), FTP-SUVR, cortical thickness, and cognitive domain composite scores. RESULTS We examined 138 participants on the AD spectrum (15 with preclinical AD, 62 with prodromal AD, and 61 with AD dementia; mean age 73.4 ± 8.0 years; female 58.7%). Total sleep time was longer in the AD dementia group (7.4 ± 1.6 hours) compared with the preclinical (6.5 ± 1.4 hours; p = 0.026) and prodromal groups (6.6 ± 1.4 hours; p = 0.001), whereas other sleep parameters did not differ between groups. Longer total sleep time was not associated with amyloid accumulation but rather with tau accumulation, especially in the amygdala, hippocampus, basal forebrain, insular, cingulate, occipital, inferior temporal cortices, and precuneus. Longer total sleep time predicted faster tau accumulation in Braak regions V-VI (β = 0.016, p = 0.007) and disease progression to mild cognitive impairment or dementia (hazard ratio = 1.554, p = 0.024). Longer total sleep time was also associated with memory deficit (β = -0.19, p = 0.008). DISCUSSION Prolonged total sleep time was associated with tau accumulation in sleep-related cortical and subcortical areas as well as memory dysfunction. It also predicted faster disease progression with tau accumulation. Our study highlights the clinical importance of assessing total sleep time as a marker for disease severity and prognosis in the AD spectrum.
Collapse
Affiliation(s)
- So Hoon Yoon
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han-Kyeol Kim
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Hoon Lee
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong-Hyun Chun
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young H Sohn
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Phil Hyu Lee
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Ryu
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hanna Cho
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Han Soo Yoo
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Chul Hyoung Lyoo
- From the Department of Neurology (S.H.Y.), International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon; Departments of Neurology (H.-K.K., H.C., H.S.Y., C.H.L.) and Nuclear Medicine (J.-H.L., Y.H.R.), Gangnam Severance Hospital; Departments of Nuclear Medicine (J.-H.C.) and Neurology (Y.H.S., P.H.L.), Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Schaffer Aguzzoli C, Ferreira PCL, Povala G, Ferrari-Souza JP, Bellaver B, Soares Katz C, Zalzale H, Lussier FZ, Rohden F, Abbas S, Leffa DT, Scop Medeiros M, Therriault J, Benedet AL, Tissot C, Servaes S, Rahmouni N, Cassa Macedo A, Bezgin G, Kang MS, Stevenson J, Pallen V, Cohen A, Lopez OL, Tudorascu DL, Klunk WE, Villemagne VL, Soucy JP, Zimmer ER, Schilling LP, Karikari TK, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Valcour V, Miller BL, Rosa-Neto P, Pascoal TA. Neuropsychiatric Symptoms and Microglial Activation in Patients with Alzheimer Disease. JAMA Netw Open 2023; 6:e2345175. [PMID: 38010651 PMCID: PMC10682836 DOI: 10.1001/jamanetworkopen.2023.45175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/15/2023] [Indexed: 11/29/2023] Open
Abstract
Importance Neuropsychiatric symptoms are commonly encountered and are highly debilitating in patients with Alzheimer disease. Understanding their underpinnings has implications for identifying biomarkers and treatment for these symptoms. Objective To evaluate whether glial markers are associated with neuropsychiatric symptoms in individuals across the Alzheimer disease continuum. Design, Setting, and Participants This cross-sectional study was conducted from January to June 2023, leveraging data from the Translational Biomarkers in Aging and Dementia cohort at McGill University, Canada. Recruitment was based on referrals of individuals from the community or from outpatient clinics. Exclusion criteria included active substance abuse, major surgery, recent head trauma, safety contraindications for positron emission tomography (PET) or magnetic resonance imaging, being currently enrolled in other studies, and having inadequately treated systemic conditions. Main Outcomes and Measures All individuals underwent assessment for neuropsychiatric symptoms (Neuropsychiatry Inventory Questionnaire [NPI-Q]), and imaging for microglial activation ([11C]PBR28 PET), amyloid-β ([18F]AZD4694 PET), and tau tangles ([18F]MK6240 PET). Results Of the 109 participants, 72 (66%) were women and 37 (34%) were men; the median age was 71.8 years (range, 38.0-86.5 years). Overall, 70 had no cognitive impairment and 39 had cognitive impairment (25 mild; 14 Alzheimer disease dementia). Amyloid-β PET positivity was present in 21 cognitively unimpaired individuals (30%) and in 31 cognitively impaired individuals (79%). The NPI-Q severity score was associated with microglial activation in the frontal, temporal, and parietal cortices (β = 7.37; 95% CI, 1.34-13.41; P = .01). A leave-one-out approach revealed that irritability was the NPI-Q domain most closely associated with the presence of brain microglial activation (β = 6.86; 95% CI, 1.77-11.95; P = .008). Furthermore, we found that microglia-associated irritability was associated with study partner burden measured by NPI-Q distress score (β = 5.72; 95% CI, 0.33-11.10; P = .03). Conclusions and Relevance In this cross-sectional study of 109 individuals across the AD continuum, microglial activation was associated with and a potential biomarker of neuropsychiatric symptoms in Alzheimer disease. Moreover, our findings suggest that the combination of amyloid-β- and microglia-targeted therapies could have an impact on relieving these symptoms.
Collapse
Affiliation(s)
- Cristiano Schaffer Aguzzoli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Global Brain Health Institute, University of California, San Francisco
| | - Pâmela C. L. Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carolina Soares Katz
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Hussein Zalzale
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Firoza Z. Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Francieli Rohden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sarah Abbas
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas T. Leffa
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina Scop Medeiros
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Ann Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dana L. Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William E. Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jean Paul Soucy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas P. Schilling
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Neurology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Victor Valcour
- Global Brain Health Institute, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
| | - Bruce L. Miller
- Global Brain Health Institute, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
24
|
Ariaei A, Ramezani F. The promising impact of Bemcentinib and Repotrectinib on sleep impairment in Alzheimer's disease. J Biomol Struct Dyn 2023:1-17. [PMID: 37909502 DOI: 10.1080/07391102.2023.2276876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disease, demands effective medication to alleviate symptoms. This study focused on sleep impairment as an overt clinical symptom and tauopathy as a prominent molecular symptom of this disease. Multiple compounds from three biomolecule libraries (719 compounds; ChemDiv:366 - ChEMBL:180 - PubChem:173) were evaluated for potential binding affinity and safety using AutoDock Vina and pkCSM, respectively, resulting in the selection of four candidate compounds (Lestaurtinib, Repotrectinib, Bemcentinib, and Zotiraciclib). Due to the similarity of Repotrectinib and Bemcentinib binding sites to ATP, 300 ns Martini 3 coarse-grained molecular dynamics (MD) was performed on these two molecules and ATP by NAMD. The stability of tau protein in the presence of drugs was assessed using a 200 ns Martini 3 MD simulation. Binding site analysis discloses Bemcentinib and Repotrectinib as two inhibitors occupying most amino acids in binding with ATP. The RMSD and RMS average correlation results revealed protein containing Bemcentinib and Repotrectinib to have a more stable state compared to ATP in the first 220 ns simulation. There was only a single detachment of Bemcentinib, while Repotrictinib detached twice at the end of the simulation. Eventually, adding Bemcentinib and Repotrectinib to the enzyme-tau complex significantly increased the number of tau detachments during the 200 ns simulation. We report Bemcentinib and Repotrectinib, formerly prescribed for cancer, as potential inhibitors of the CK1 δ. Besides their high binding affinity compared to ATP, they can inhibit all ATP-binding sites and alter the tau binding stability.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Armin Ariaei
- Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
André C, Martineau-Dussault MÈ, Daneault V, Blais H, Frenette S, Lorrain D, Hudon C, Bastien C, Petit D, Lafrenière A, Thompson C, Montplaisir J, Gosselin N, Carrier J. REM sleep is associated with the volume of the cholinergic basal forebrain in aMCI individuals. Alzheimers Res Ther 2023; 15:151. [PMID: 37684650 PMCID: PMC10485959 DOI: 10.1186/s13195-023-01265-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/29/2023] [Indexed: 09/10/2023]
Abstract
BACKGROUND Rapid-eye movement (REM) sleep highly depends on the activity of cholinergic basal forebrain (BF) neurons and is reduced in Alzheimer's disease. Here, we investigated the associations between the volume of BF nuclei and REM sleep characteristics, and the impact of cognitive status on these links, in late middle-aged and older participants. METHODS Thirty-one cognitively healthy controls (66.8 ± 7.2 years old, 13 women) and 31 participants with amnestic Mild Cognitive Impairment (aMCI) (68.3 ± 8.8 years old, 7 women) were included in this cross-sectional study. All participants underwent polysomnography, a comprehensive neuropsychological assessment and Magnetic Resonance Imaging examination. REM sleep characteristics (i.e., percentage, latency and efficiency) were derived from polysomnographic recordings. T1-weighted images were preprocessed using CAT12 and the DARTEL algorithm, and we extracted the gray matter volume of BF regions of interest using a probabilistic atlas implemented in the JuBrain Anatomy Toolbox. Multiple linear regressions were performed between the volume of BF nuclei and REM sleep characteristics controlling for age, sex and total intracranial volume, in the whole cohort and in subgroups stratified by cognitive status. RESULTS In the whole sample, lower REM sleep percentage was significantly associated to lower nucleus basalis of Meynert (Ch4) volume (β = 0.32, p = 0.009). When stratifying the cohort according to cognitive status, lower REM sleep percentage was significantly associated to both lower Ch4 (β = 0.48, p = 0.012) and total BF volumes (β = 0.44, p = 0.014) in aMCI individuals, but not in cognitively unimpaired participants. No significant associations were observed between the volume of the BF and wake after sleep onset or non-REM sleep variables. DISCUSSION These results suggest that REM sleep disturbances may be an early manifestation of the degeneration of the BF cholinergic system before the onset of dementia, especially in participants with mild memory deficits.
Collapse
Affiliation(s)
- Claire André
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Marie-Ève Martineau-Dussault
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Véronique Daneault
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
- Functional Neuroimaging Unit, University of Montreal Geriatric Institute, 4565 Queen-Mary Road, Montreal, QC, H3W 1W5, Canada
| | - Hélène Blais
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
| | - Sonia Frenette
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
| | - Dominique Lorrain
- Research Centre On Aging, University Institute of Geriatrics of Sherbrooke, CIUSSS de L'Estrie-CHUS, Sherbrooke, QC, Canada
- Department of Psychology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Carol Hudon
- CERVO Research Centre, Québec City, QC, Canada
- School of Psychology, Université Laval, Québec City, QC, Canada
| | - Célyne Bastien
- CERVO Research Centre, Québec City, QC, Canada
- School of Psychology, Université Laval, Québec City, QC, Canada
| | - Dominique Petit
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Département de Psychiatrie, Université de Montréal, Montréal, QC, Canada
| | - Alexandre Lafrenière
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Cynthia Thompson
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
| | - Jacques Montplaisir
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Département de Psychiatrie, Université de Montréal, Montréal, QC, Canada
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Julie Carrier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Recherche CIUSSS NIM, 5400 Boul Gouin O, Montréal, QC, H4J 1C5, Canada.
- Department of Psychology, Université de Montréal, Montreal, QC, Canada.
- Functional Neuroimaging Unit, University of Montreal Geriatric Institute, 4565 Queen-Mary Road, Montreal, QC, H3W 1W5, Canada.
| |
Collapse
|
26
|
Krohn F, Lancini E, Ludwig M, Leiman M, Guruprasath G, Haag L, Panczyszyn J, Düzel E, Hämmerer D, Betts M. Noradrenergic neuromodulation in ageing and disease. Neurosci Biobehav Rev 2023; 152:105311. [PMID: 37437752 DOI: 10.1016/j.neubiorev.2023.105311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
The locus coeruleus (LC) is a small brainstem structure located in the lower pons and is the main source of noradrenaline (NA) in the brain. Via its phasic and tonic firing, it modulates cognition and autonomic functions and is involved in the brain's immune response. The extent of degeneration to the LC in healthy ageing remains unclear, however, noradrenergic dysfunction may contribute to the pathogenesis of Alzheimer's (AD) and Parkinson's disease (PD). Despite their differences in progression at later disease stages, the early involvement of the LC may lead to comparable behavioural symptoms such as preclinical sleep problems and neuropsychiatric symptoms as a result of AD and PD pathology. In this review, we draw attention to the mechanisms that underlie LC degeneration in ageing, AD and PD. We aim to motivate future research to investigate how early degeneration of the noradrenergic system may play a pivotal role in the pathogenesis of AD and PD which may also be relevant to other neurodegenerative diseases.
Collapse
Affiliation(s)
- F Krohn
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Lancini
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - M Ludwig
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - M Leiman
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - G Guruprasath
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - L Haag
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Panczyszyn
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Düzel
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - D Hämmerer
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany; Department of Psychology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - M Betts
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
27
|
Falgàs N, Walsh CM, Yack L, Simon AJ, Allen IE, Kramer JH, Rosen HJ, Joie RL, Rabinovici G, Miller B, Spina S, Seeley WW, Ranasinghe K, Vossel K, Neylan TC, Grinberg LT. Alzheimer's disease phenotypes show different sleep architecture. Alzheimers Dement 2023; 19:3272-3282. [PMID: 36749893 PMCID: PMC10404632 DOI: 10.1002/alz.12963] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Sleep-wake disturbances are a prominent feature of Alzheimer's disease (AD). Atypical (non-amnestic) AD syndromes have different patterns of cortical vulnerability to AD. We hypothesized that atypical AD also shows differential vulnerability in subcortical nuclei that will manifest as different patterns of sleep dysfunction. METHODS Overnight electroencephalography monitoring was performed on 48 subjects, including 15 amnestic, 19 atypical AD, and 14 controls. AD was defined based on neuropathological or biomarker confirmation. We compared sleep architecture by visual scoring and spectral power analysis in each group. RESULTS Overall, AD cases showed increased sleep fragmentation and N1 sleep compared to controls. Compared to atypical AD groups, typical AD showed worse N3 sleep dysfunction and relatively preserved rapid eye movement (REM) sleep. DISCUSSION Results suggest differing effects of amnestic and atypical AD variants on slow wave versus REM sleep, respectively, corroborating the hypothesis of differential selective vulnerability patterns of the subcortical nuclei within variants. Optimal symptomatic treatment for sleep dysfunction in clinical phenotypes may differ. HIGHLIGHTS Alzheimer's disease (AD) variants show distinct patterns of sleep impairment. Amnestic/typical AD has worse N3 slow wave sleep (SWS) impairment compared to atypical AD. Atypical AD shows more rapid eye movement deficits than typical AD. Selective vulnerability patterns in subcortical areas may underlie sleep differences. Relatively preserved SWS may explain better memory scores in atypical versus typical AD.
Collapse
Affiliation(s)
- Neus Falgàs
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Christine M Walsh
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Leslie Yack
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
| | - Alexander J Simon
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Isabel E Allen
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
| | - Joel H Kramer
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Department of Psychiatry, University of California, San Francisco, California, USA
| | - Howard J Rosen
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
| | - Renaud La Joie
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Gil Rabinovici
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Bruce Miller
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
| | - Salvatore Spina
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
| | - William W Seeley
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Kamalini Ranasinghe
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Keith Vossel
- Mary S. Easton Center for Alzheimer's Disease Research, University of California Los Angeles, Los Angeles, California, USA
| | - Thomas C Neylan
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Department of Psychiatry, University of California, San Francisco, California, USA
| | - Lea T Grinberg
- Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, California, USA
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Department of Pathology, University of California, San Francisco, California, USA
| |
Collapse
|
28
|
Zhou M, Li Y. Effect of different doses of almorexant on learning and memory in 8-month-old APP/PS1 (AD) mice. Peptides 2023; 167:171044. [PMID: 37330110 DOI: 10.1016/j.peptides.2023.171044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
OBJECTIVE To explore the effects of different doses of almorexant (an dual orexin receptor antagonist) on learning and memory in Alzheimer's disease (AD) mice. METHODS Forty-four APP/PS1 (model of Alzheimer's disease; AD) mice were randomly divided into 4 groups: the control group (CON) and those that received 10mg/kg almorexant (low dose; LOW), 30mg/kg almorexant (medium dose; MED) and 60mg/kg almorexant (high dose; HIGH). During the 28-day intervention period, mice received an intraperitoneal injection at the beginning of the light period (6:00 am). The effects of different doses of almorexant on learning and memory and 24-hour sleep-wake behaviour were assessed by immunohistochemical staining. The above continuous variables are expressed as the mean ± standard deviation (SD), and then univariate regression analysis and generalized estimating equations were performed to compare the groups; these results are expressed as the mean difference (MD) and 95% confidence interval (CI). The statistical software used STATA 17.0 MP. RESULTS Forty-one mice completed the experiment (3 died: 2 mice in the HIGH group and 1 mouse in the CON group). Compared with the CON group, the LOW group (MD=6,803s, 95% CI: 4,470 to 9,137s), MED group (MD=14,473s, 95% CI: 12,140 to 16,806s) and the HIGH group (MD=24,505s, 95% CI: 22,052 to 26,959s) had significantly longer sleep durations. The Y maze results showed that LOW group (MD=0.14,95%CI: 0.078 to 0.20) and MED group (MD=0.14,95%CI = 0.074 to 0.20) mice compared to the CON group, and the low-medium dose of Almorexant did not damage the short-term learning and memory performance of APP / PS1 (AD) mice.Compared with the CON, LOW, and MED groups, the HIGH group exhibited a significant decrease in the Aβ plaque-positive area in the cortex (MD= -0.030, 95% CI: -0.035 to -0.025; MD=-0.049, 95% CI: -0.054 to -0.044; and MD=-0.07, 95% CI: -0.076 to -0.066, respectively). CONCLUSION The moderate dose of almorexant (30mg/kg) prolonged the sleep duration of APP/PS1 (AD) mice to a greater extent than the low dose (10mg/kg) without altering learning and memory. The MED mice showed a good sleep response and a small residual effect on the next day. High-dose (60mg / kg) almorexant impaired behavioral learning and memory performance in mice.Compared to the CON group and the LOW group, the MED group exhibited improved working memory. Thus, treatment with almorexant may reduce β-amyloid deposition in AD, slowing neurodegeneration. Additional studies are needed to determine the mechanism of action.
Collapse
Affiliation(s)
- Mengzhen Zhou
- Department of Neurology, Qianfo Mountain Hospital affiliated to Shandong First Medical University ,Jinan, Shandong, China.
| | - Yanran Li
- Department of Neurology, Qianfo Mountain Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
29
|
Ehrenberg AJ, Kelberman MA, Liu KY, Dahl MJ, Weinshenker D, Falgàs N, Dutt S, Mather M, Ludwig M, Betts MJ, Winer JR, Teipel S, Weigand AJ, Eschenko O, Hämmerer D, Leiman M, Counts SE, Shine JM, Robertson IH, Levey AI, Lancini E, Son G, Schneider C, Egroo MV, Liguori C, Wang Q, Vazey EM, Rodriguez-Porcel F, Haag L, Bondi MW, Vanneste S, Freeze WM, Yi YJ, Maldinov M, Gatchel J, Satpati A, Babiloni C, Kremen WS, Howard R, Jacobs HIL, Grinberg LT. Priorities for research on neuromodulatory subcortical systems in Alzheimer's disease: Position paper from the NSS PIA of ISTAART. Alzheimers Dement 2023; 19:2182-2196. [PMID: 36642985 PMCID: PMC10182252 DOI: 10.1002/alz.12937] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 01/17/2023]
Abstract
The neuromodulatory subcortical system (NSS) nuclei are critical hubs for survival, hedonic tone, and homeostasis. Tau-associated NSS degeneration occurs early in Alzheimer's disease (AD) pathogenesis, long before the emergence of pathognomonic memory dysfunction and cortical lesions. Accumulating evidence supports the role of NSS dysfunction and degeneration in the behavioral and neuropsychiatric manifestations featured early in AD. Experimental studies even suggest that AD-associated NSS degeneration drives brain neuroinflammatory status and contributes to disease progression, including the exacerbation of cortical lesions. Given the important pathophysiologic and etiologic roles that involve the NSS in early AD stages, there is an urgent need to expand our understanding of the mechanisms underlying NSS vulnerability and more precisely detail the clinical progression of NSS changes in AD. Here, the NSS Professional Interest Area of the International Society to Advance Alzheimer's Research and Treatment highlights knowledge gaps about NSS within AD and provides recommendations for priorities specific to clinical research, biomarker development, modeling, and intervention. HIGHLIGHTS: Neuromodulatory nuclei degenerate in early Alzheimer's disease pathological stages. Alzheimer's pathophysiology is exacerbated by neuromodulatory nuclei degeneration. Neuromodulatory nuclei degeneration drives neuropsychiatric symptoms in dementia. Biomarkers of neuromodulatory integrity would be value-creating for dementia care. Neuromodulatory nuclei present strategic prospects for disease-modifying therapies.
Collapse
Affiliation(s)
- Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, USA
| | - Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London, UK
| | - Martin J Dahl
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Global Brain Health Institute, University of California, San Francisco, San Francisco, California, USA
| | - Shubir Dutt
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Department of Psychology, University of Southern California, Los Angeles, California, USA
| | - Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Department of Psychology, University of Southern California, Los Angeles, California, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Mareike Ludwig
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - Matthew J Betts
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| | - Alexandra J Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, California, USA
| | - Oxana Eschenko
- Department of Computational Neuroscience, Max Planck Institute for Biological Cybernetics, Tuebingen, Germany
| | - Dorothea Hämmerer
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
- Department of Psychology, University of Innsbruck, Innsbruck, Austria
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Marina Leiman
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Scott E Counts
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Michigan Alzheimer's Disease Research Center, Ann Arbor, Michigan, USA
| | - James M Shine
- Brain and Mind Center, The University of Sydney, Sydney, Australia
| | - Ian H Robertson
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, Georgia, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Institute, Emory University, Atlanta, Georgia, USA
| | - Elisa Lancini
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Gowoon Son
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Health, Medicine, and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome Tor Vergata, Rome, Italy
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Agusta University, Agusta, Georgia, USA
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | | | - Lena Haag
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Mark W Bondi
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, California, USA
| | - Sven Vanneste
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- School of Psychology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Whitney M Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, the Netherlands
| | - Yeo-Jin Yi
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Mihovil Maldinov
- Department of Psychiatry and Psychotherapy, University of Rostock, Rostock, Germany
| | - Jennifer Gatchel
- Division of Geriatric Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abhijit Satpati
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "V. Erspamer,", Sapienza University of Rome, Rome, Italy
- Hospital San Raffaele Cassino, Cassino, Italy
| | - William S Kremen
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Health, Medicine, and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, San Francisco, California, USA
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
30
|
Morrone CD, Raghuraman R, Hussaini SA, Yu WH. Proteostasis failure exacerbates neuronal circuit dysfunction and sleep impairments in Alzheimer's disease. Mol Neurodegener 2023; 18:27. [PMID: 37085942 PMCID: PMC10119020 DOI: 10.1186/s13024-023-00617-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
Failed proteostasis is a well-documented feature of Alzheimer's disease, particularly, reduced protein degradation and clearance. However, the contribution of failed proteostasis to neuronal circuit dysfunction is an emerging concept in neurodegenerative research and will prove critical in understanding cognitive decline. Our objective is to convey Alzheimer's disease progression with the growing evidence for a bidirectional relationship of sleep disruption and proteostasis failure. Proteostasis dysfunction and tauopathy in Alzheimer's disease disrupts neurons that regulate the sleep-wake cycle, which presents behavior as impaired slow wave and rapid eye movement sleep patterns. Subsequent sleep loss further impairs protein clearance. Sleep loss is a defined feature seen early in many neurodegenerative disorders and contributes to memory impairments in Alzheimer's disease. Canonical pathological hallmarks, β-amyloid, and tau, directly disrupt sleep, and neurodegeneration of locus coeruleus, hippocampal and hypothalamic neurons from tau proteinopathy causes disruption of the neuronal circuitry of sleep. Acting in a positive-feedback-loop, sleep loss and circadian rhythm disruption then increase spread of β-amyloid and tau, through impairments of proteasome, autophagy, unfolded protein response and glymphatic clearance. This phenomenon extends beyond β-amyloid and tau, with interactions of sleep impairment with the homeostasis of TDP-43, α-synuclein, FUS, and huntingtin proteins, implicating sleep loss as an important consideration in an array of neurodegenerative diseases and in cases of mixed neuropathology. Critically, the dynamics of this interaction in the neurodegenerative environment are not fully elucidated and are deserving of further discussion and research. Finally, we propose sleep-enhancing therapeutics as potential interventions for promoting healthy proteostasis, including β-amyloid and tau clearance, mechanistically linking these processes. With further clinical and preclinical research, we propose this dynamic interaction as a diagnostic and therapeutic framework, informing precise single- and combinatorial-treatments for Alzheimer's disease and other brain disorders.
Collapse
Affiliation(s)
- Christopher Daniel Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
| | - Radha Raghuraman
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
| | - S Abid Hussaini
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
| | - Wai Haung Yu
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Geriatric Mental Health Research Services, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
31
|
Zhou L, Kong J, Li X, Ren Q. Sex differences in the effects of sleep disorders on cognitive dysfunction. Neurosci Biobehav Rev 2023; 146:105067. [PMID: 36716906 DOI: 10.1016/j.neubiorev.2023.105067] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
Sleep is an essential physiological function that sustains human life. Sleep disorders involve problems with the quality, duration, and abnormal behaviour of sleep. Insomnia is the most common sleep disorder, followed by sleep-disordered breathing (SDB). Sleep disorders often occur along with medical conditions or other mental health conditions. Of particular interest to researchers is the role of sleep disorders in cognitive dysfunction. Sleep disorder is a risk factor for cognitive dysfunction, yet the exact pathogenesis is still far from agreement. Little is known about how sex differences influence the changes in cognitive functions caused by sleep disorders. This narrative review examines how sleep disorders might affect cognitive impairment, and then explores the sex-specific consequences of sleep disorders as a risk factor for dementia and the potential underlying mechanisms. Some insights on the direction of further research are also presented.
Collapse
Affiliation(s)
- Lv Zhou
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jingting Kong
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaoli Li
- School of Medicine, Southeast University, Nanjing 210009, China; Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing 210009, China
| | - Qingguo Ren
- School of Medicine, Southeast University, Nanjing 210009, China; Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing 210009, China.
| |
Collapse
|
32
|
Avila J, Perry G. Memory, Sleep, and Tau Function. J Alzheimers Dis 2023; 94:491-495. [PMID: 37248906 DOI: 10.3233/jad-230230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Memory consolidation related to the hippocampal-cortex connection takes place during sleep. This connection may involve at least two steps- one in the NREM phase of sleep (transmission) and the other in the REM phase (consolidation). In this brief report, we comment on the role of tau protein in these two phases of sleep. The absence of tau decreases δ waves in NREM, whereas the overexpression of modified (phosphorylated and/or mutated) tau alters θ waves in REM.
Collapse
Affiliation(s)
- Jesús Avila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - George Perry
- Neurology, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
33
|
Cheng L, Wang F, Li ZH, Wen C, Ding L, Zhang SB, You QY. Study on the active components and mechanism of Suanzaoren decoction in improving cognitive impairment caused by sleep deprivation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115502. [PMID: 35777606 DOI: 10.1016/j.jep.2022.115502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/09/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Suanzaoren Decoction (SZRD) is a traditional and classic prescription for the treatment of insomnia, with a history of more than 1,000 years. It replenishes blood components, calms the nerves, reduces fever and irritability. It is commonly used in the clinical treatment of chronic fatigue syndrome, cardiac neurosis, and menopausal syndromes. Modern pharmacological studies have shown that it improves cognitive impairment; however, its mechanism of action remains unclear. AIM OF THE STUDY This study preliminarily investigated the potential bioactive components and mechanism of SZRD in improving cognitive impairment by exploring network pharmacology, molecular docking, and conducting in vivo experiments. MATERIALS AND METHODS The components of various Chinese herbs in SZRD and their disease-related targets were identified through network pharmacology and literature. Gene ontology (GO) function enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of intersection targets were performed using the relevant database. Next, the "Components-Targets-Pathways" (C-T-P) and "Protein-Protein interaction" networks were constructed using the enrichment analysis results to further identify potential pathways, bioactive components, and hub genes. At the same time, molecular docking was used to further distinguish the key bioactive components and genes of SZRD responsible for improving cognitive impairment. Finally, the potential mechanism of action was further analysed and verified using in vivo experiments. RESULTS A total of 117 potential active components and 138 intersection targets were identified by network pharmacology screening. The key bioactive components, including calycosin, 5-Prenylbutein, licochalcone G, glypallichalcone, and ZINC189892, were identified by analysing the networks and molecular docking results. Hub genes included ACHE, CYP19A1, EGFR, ESR1, and ESR2. The oestrogen signalling pathway was the most important in the enrichment analysis. In vivo experiments further proved that SZRD could improve cognitive impairment by affecting the oestrogen signalling pathway and the expression of ACHE and CYP19A1. CONCLUSIONS Network pharmacology and in vivo experiments demonstrate that SZRD improves cognitive impairment caused by sleep disturbance through estrogen receptor pathway, which provides a basis for its clinical application.
Collapse
Affiliation(s)
- Li Cheng
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Fei Wang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Zi-Heng Li
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Chun Wen
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Ding
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Shun-Bo Zhang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Qiu-Yun You
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
34
|
Neylan TC, Walsh CM. Sleep spindles, tau, and neurodegeneration. Sleep 2022; 45:6633544. [PMID: 35797181 PMCID: PMC9453613 DOI: 10.1093/sleep/zsac161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Thomas C Neylan
- Corresponding author. Thomas C. Neylan, UCSF-VAMC 116P, 4150 Clement Street, San Francisco, CA 94121, USA.
| | - Christine M Walsh
- Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
35
|
Gu L, Yu Q, Shen Y, Wang Y, Xu Q, Zhang H. The role of monoaminergic neurons in modulating respiration during sleep and the connection with SUDEP. Biomed Pharmacother 2022; 150:112983. [PMID: 35453009 DOI: 10.1016/j.biopha.2022.112983] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 11/25/2022] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death among epilepsy patients, occurring even more frequently in cases with anti-epileptic drug resistance. Despite some advancements in characterizing SUDEP, the underlying mechanism remains incompletely understood. This review summarizes the latest advances in our understanding of the pathogenic mechanisms of SUDEP, in order to identify possible targets for the development of new strategies to prevent SUDEP. Based on our previous research along with the current literature, we focus on the role of sleep-disordered breathing (SDB) and its related neural mechanisms to consider the possible roles of monoaminergic neurons in the modulation of respiration during sleep and the occurrence of SUDEP. Overall, this review suggests that targeting the monoaminergic neurons is a promising approach to preventing SUDEP. The proposed roles of SDB and related monoaminergic neural mechanisms in SUDEP provide new insights for explaining the pathogenesis of SUDEP.
Collapse
Affiliation(s)
- LeYuan Gu
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qian Yu
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yue Shen
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - YuLing Wang
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qing Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - HongHai Zhang
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310006, China.
| |
Collapse
|
36
|
Oh JY, Walsh CM, Ranasinghe K, Mladinov M, Pereira FL, Petersen C, Falgàs N, Yack L, Lamore T, Nasar R, Lew C, Li S, Metzler T, Coppola Q, Pandher N, Le M, Heuer HW, Heinsen H, Spina S, Seeley WW, Kramer J, Rabinovici GD, Boxer AL, Miller BL, Vossel K, Neylan TC, Grinberg LT. Subcortical Neuronal Correlates of Sleep in Neurodegenerative Diseases. JAMA Neurol 2022; 79:498-508. [PMID: 35377391 PMCID: PMC8981071 DOI: 10.1001/jamaneurol.2022.0429] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022]
Abstract
Importance Sleep disturbance is common among patients with neurodegenerative diseases. Examining the subcortical neuronal correlates of sleep disturbances is important to understanding the early-stage sleep neurodegenerative phenomena. Objectives To examine the correlation between the number of important subcortical wake-promoting neurons and clinical sleep phenotypes in patients with Alzheimer disease (AD) or progressive supranuclear palsy (PSP). Design, Setting, and Participants This longitudinal cohort study enrolled 33 patients with AD, 20 patients with PSP, and 32 healthy individuals from the Memory and Aging Center of the University of California, San Francisco, between August 22, 2008, and December 31, 2020. Participants received electroencephalographic and polysomnographic sleep assessments. Postmortem neuronal analyses of brainstem hypothalamic wake-promoting neurons were performed and were included in the clinicopathological correlation analysis. No eligible participants were excluded from the study. Exposures Electroencephalographic and polysomnographic assessment of sleep and postmortem immunohistological stereological analysis of 3 wake-promoting nuclei (noradrenergic locus coeruleus [LC], orexinergic lateral hypothalamic area [LHA], and histaminergic tuberomammillary nucleus [TMN]). Main Outcomes and Measures Nocturnal sleep variables, including total sleep time, sleep maintenance, rapid eye movement (REM) latency, and time spent in REM sleep and stages 1, 2, and 3 of non-REM (NREM1, NREM2, and NREM3, respectively) sleep, and wake after sleep onset. Neurotransmitter, tau, and total neuronal counts of LC, LHA, and TMN. Results Among 19 patients included in the clinicopathological correlation analysis, the mean (SD) age at death was 70.53 (7.75) years; 10 patients (52.6%) were female; and all patients were White. After adjusting for primary diagnosis, age, sex, and time between sleep analyses and death, greater numbers of LHA and TMN neurons were correlated with decreased homeostatic sleep drive, as observed by less total sleep time (LHA: r = -0.63; P = .009; TMN: r = -0.62; P = .008), lower sleep maintenance (LHA: r = -0.85; P < .001; TMN: r = -0.78; P < .001), and greater percentage of wake after sleep onset (LHA: r = 0.85; P < .001; TMN: r = 0.78; P < .001). In addition, greater numbers of LHA and TMN neurons were correlated with less NREM2 sleep (LHA: r = -0.76; P < .001; TMN: r = -0.73; P < .001). A greater number of TMN neurons was also correlated with less REM sleep (r = -0.61; P = .01). A greater number of LC neurons was mainly correlated with less total sleep time (r = -0.68; P = .008) and greater REM latency (r = 0.71; P = .006). The AD-predominant group had significantly greater sleep drive, including higher total sleep time (mean [SD], 0.49 [1.18] vs -1.09 [1.37]; P = .03), higher sleep maintenance (mean [SD], 0.18 [1.22] vs -1.53 [1.78]; P = .02), and lower percentage of wake after sleep onset during sleep period time (mean [SD], -0.18 [1.20] vs 1.49 [1.72]; P = .02) than the PSP-predominant group based on unbiased k-means clustering and principal component analyses. Conclusions and Relevance In this cohort study, subcortical wake-promoting neurons were significantly correlated with sleep phenotypes in patients with AD and PSP, suggesting that the loss of wake-promoting neurons among patients with neurodegenerative conditions may disturb the control of sleep-wake homeostasis. These findings suggest that the subcortical system is a primary mechanism associated with sleep disturbances in the early stages of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jun Y. Oh
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- School of Medicine, University of California, San Francisco, San Francisco
| | - Christine M. Walsh
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Kamalini Ranasinghe
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Mihovil Mladinov
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Felipe L. Pereira
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Cathrine Petersen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Neus Falgàs
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Global Brain Health Institute, University of California, San Francisco, San Francisco
| | - Leslie Yack
- Stress and Health Research Program, Department of Mental Health, San Francisco VA Medical Center, San Francisco, California
| | - Tia Lamore
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Rakin Nasar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Caroline Lew
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Song Li
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Thomas Metzler
- Stress and Health Research Program, Department of Mental Health, San Francisco VA Medical Center, San Francisco, California
| | - Quentin Coppola
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Natalie Pandher
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Michael Le
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Hilary W. Heuer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Helmut Heinsen
- Department of Psychiatry, University of Wurzburg, Wurzburg, Germany
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Joel Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Gil D. Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Global Brain Health Institute, University of California, San Francisco, San Francisco
| | - Keith Vossel
- Department of Neurology, University of California, Los Angeles, Los Angeles
| | - Thomas C. Neylan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Stress and Health Research Program, Department of Mental Health, San Francisco VA Medical Center, San Francisco, California
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Global Brain Health Institute, University of California, San Francisco, San Francisco
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Department of Pathology, University of California, San Francisco, San Francisco
| |
Collapse
|
37
|
Osorio-Forero A, Cherrad N, Banterle L, Fernandez LMJ, Lüthi A. When the Locus Coeruleus Speaks Up in Sleep: Recent Insights, Emerging Perspectives. Int J Mol Sci 2022; 23:ijms23095028. [PMID: 35563419 PMCID: PMC9099715 DOI: 10.3390/ijms23095028] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/03/2022] Open
Abstract
For decades, numerous seminal studies have built our understanding of the locus coeruleus (LC), the vertebrate brain’s principal noradrenergic system. Containing a numerically small but broadly efferent cell population, the LC provides brain-wide noradrenergic modulation that optimizes network function in the context of attentive and flexible interaction with the sensory environment. This review turns attention to the LC’s roles during sleep. We show that these roles go beyond down-scaled versions of the ones in wakefulness. Novel dynamic assessments of noradrenaline signaling and LC activity uncover a rich diversity of activity patterns that establish the LC as an integral portion of sleep regulation and function. The LC could be involved in beneficial functions for the sleeping brain, and even minute alterations in its functionality may prove quintessential in sleep disorders.
Collapse
|
38
|
Van Egroo M, Koshmanova E, Vandewalle G, Jacobs HI. Importance of the locus coeruleus-norepinephrine system in sleep-wake regulation: implications for aging and Alzheimer’s disease. Sleep Med Rev 2022; 62:101592. [PMID: 35124476 PMCID: PMC9064973 DOI: 10.1016/j.smrv.2022.101592] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/24/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022]
Abstract
Five decades ago, seminal studies positioned the brainstem locus coeruleus (LC) norepinephrine (NE) system as a key substrate for the regulation of wakefulness and sleep, and this picture has recently been elaborated thanks to methodological advances in the precise investigation and experimental modulation of LC structure and functions. This review presents and discusses findings that support the major role of the LC-NE system at different levels of sleep-wake organization, ranging from its involvement in the overall architecture of the sleep-wake cycle to its associations with sleep microstructure, while accounting for the intricate neuroanatomy surrounding the LC. Given the particular position held by the LC-NE system by being at the intersection of sleep-wake dysregulation and initial pathophysiological processes of Alzheimer's disease (AD), we conclude by examining emerging opportunities to investigate LC-NE mediated relationships between sleep-wake alteration and AD in human aging. We further propose several research perspectives that could support the LC-NE system as a promising target for the identification of at-risk individuals in the preclinical stages of AD, and for the development of novel preventive interventions.
Collapse
|