1
|
Rajendran K, Krishnan UM. Mechanistic insights and emerging therapeutic stratagems for Alzheimer's disease. Ageing Res Rev 2024; 97:102309. [PMID: 38615895 DOI: 10.1016/j.arr.2024.102309] [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: 07/24/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD), a multi-factorial neurodegenerative disorder has affected over 30 million individuals globally and these numbers are expected to increase in the coming decades. Current therapeutic interventions are largely ineffective as they focus on a single target. Development of an effective drug therapy requires a deep understanding of the various factors influencing the onset and progression of the disease. Aging and genetic factors exert a major influence on the development of AD. Other factors like post-viral infections, iron overload, gut dysbiosis, and vascular dysfunction also exacerbate the onset and progression of AD. Further, post-translational modifications in tau, DRP1, CREB, and p65 proteins increase the disease severity through triggering mitochondrial dysfunction, synaptic loss, and differential interaction of amyloid beta with different receptors leading to impaired intracellular signalling. With advancements in neuroscience tools, new inter-relations that aggravate AD are being discovered including pre-existing diseases and exposure to other pathogens. Simultaneously, new therapeutic strategies involving modulation of gene expression through targeted delivery or modulation with light, harnessing the immune response to promote clearance of amyloid deposits, introduction of stem cells and extracellular vesicles to replace the destroyed neurons, exploring new therapeutic molecules from plant, marine and biological sources delivered in the free state or through nanoparticles and use of non-pharmacological interventions like music, transcranial stimulation and yoga. Polypharmacology approaches involving combination of therapeutic agents are also under active investigation for superior therapeutic outcomes. This review elaborates on various disease-causing factors, their underlying mechanisms, the inter-play between different disease-causing players, and emerging therapeutic options including those under clinical trials, for treatment of AD. The challenges involved in AD therapy and the way forward have also been discussed.
Collapse
Affiliation(s)
- Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India.
| |
Collapse
|
2
|
Champetier P, André C, Rehel S, Ourry V, Landeau B, Mézenge F, Roquet D, Vivien D, de La Sayette V, Chételat G, Rauchs G. Multimodal neuroimaging correlates of spectral power in NREM sleep delta sub-bands in cognitively unimpaired older adults. Sleep 2024; 47:zsae012. [PMID: 38227830 PMCID: PMC11009032 DOI: 10.1093/sleep/zsae012] [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: 05/22/2023] [Revised: 11/27/2023] [Indexed: 01/18/2024] Open
Abstract
STUDY OBJECTIVES In aging, reduced delta power (0.5-4 Hz) during N2 and N3 sleep has been associated with gray matter (GM) atrophy and hypometabolism within frontal regions. Some studies have also reported associations between N2 and N3 sleep delta power in specific sub-bands and amyloid pathology. Our objective was to better understand the relationships between spectral power in delta sub-bands during N2-N3 sleep and brain integrity using multimodal neuroimaging. METHODS In-home polysomnography was performed in 127 cognitively unimpaired older adults (mean age ± SD: 69.0 ± 3.8 years). N2-N3 sleep EEG power was calculated in delta (0.5-4 Hz), slow delta (0.5-1 Hz), and fast delta (1-4 Hz) frequency bands. Participants also underwent magnetic resonance imaging and Florbetapir-PET (early and late acquisitions) scans to assess GM volume, brain perfusion, and amyloid burden. Amyloid accumulation over ~21 months was also quantified. RESULTS Higher delta power was associated with higher GM volume mainly in fronto-cingular regions. Specifically, slow delta power was positively correlated with GM volume and perfusion in these regions, while the inverse association was observed with fast delta power. Delta power was neither associated with amyloid burden at baseline nor its accumulation over time, whatever the frequency band considered. CONCLUSIONS Our results show that slow delta is particularly associated with preserved brain structure, and highlight the importance of analyzing delta power sub-bands to better understand the associations between delta power and brain integrity. Further longitudinal investigations with long follow-ups are needed to disentangle the associations among sleep, amyloid pathology, and dementia risk in older populations. CLINICAL TRIAL INFORMATION Name: Study in Cognitively Intact Seniors Aiming to Assess the Effects of Meditation Training (Age-Well). URL: https://clinicaltrials.gov/ct2/show/NCT02977819?term=Age-Well&draw=2&rank=1. See STROBE_statement_AGEWELL in supplemental materials. REGISTRATION EudraCT: 2016-002441-36; IDRCB: 2016-A01767-44; ClinicalTrials.gov Identifier: NCT02977819.
Collapse
Affiliation(s)
- Pierre Champetier
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH, 14000 Caen, France
| | - Claire André
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
| | - Stéphane Rehel
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH, 14000 Caen, France
| | - Valentin Ourry
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH, 14000 Caen, France
| | - Brigitte Landeau
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
| | - Florence Mézenge
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
| | - Daniel Roquet
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
- Département de Recherche Clinique, CHU Caen-Normandie, Caen, France
| | | | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
| | - Géraldine Rauchs
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,” NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, 14000 Caen, France
| |
Collapse
|
3
|
El Hajj R, Al Sagheer T, Ballout N. Optogenetics in chronic neurodegenerative diseases, controlling the brain with light: A systematic review. J Neurosci Res 2024; 102:e25321. [PMID: 38588013 DOI: 10.1002/jnr.25321] [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: 10/24/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
Neurodegenerative diseases are progressive disorders characterized by synaptic loss and neuronal death. Optogenetics combines optical and genetic methods to control the activity of specific cell types. The efficacy of this approach in neurodegenerative diseases has been investigated in many reviews, however, none of them tackled it systematically. Our study aimed to review systematically the findings of optogenetics and its potential applications in animal models of chronic neurodegenerative diseases and compare it with deep brain stimulation and designer receptors exclusively activated by designer drugs techniques. The search strategy was performed based on the PRISMA guidelines and the risk of bias was assessed following the Systematic Review Centre for Laboratory Animal Experimentation tool. A total of 247 articles were found, of which 53 were suitable for the qualitative analysis. Our data revealed that optogenetic manipulation of distinct neurons in the brain is efficient in rescuing memory impairment, alleviating neuroinflammation, and reducing plaque pathology in Alzheimer's disease. Similarly, this technique shows an advanced understanding of the contribution of various neurons involved in the basal ganglia pathways with Parkinson's disease motor symptoms and pathology. However, the optogenetic application using animal models of Huntington's disease, multiple sclerosis, and amyotrophic lateral sclerosis was limited. Optogenetics is a promising technique that enhanced our knowledge in the research of neurodegenerative diseases and addressed potential therapeutic solutions for managing these diseases' symptoms and delaying their progression. Nevertheless, advanced investigations should be considered to improve optogenetic tools' efficacy and safety to pave the way for their translatability to the clinic.
Collapse
Affiliation(s)
- Rojine El Hajj
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Tareq Al Sagheer
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Nissrine Ballout
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| |
Collapse
|
4
|
Almeida VN. Somatostatin and the pathophysiology of Alzheimer's disease. Ageing Res Rev 2024; 96:102270. [PMID: 38484981 DOI: 10.1016/j.arr.2024.102270] [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: 07/18/2023] [Revised: 03/09/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-β plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aβ production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aβ to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aβ release, fibrillation and plaque formation. Aβ plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.
Collapse
Affiliation(s)
- Victor N Almeida
- Institute of Psychiatry, Faculty of Medicine, University of São Paulo (USP), Brazil; Faculty of Languages, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
5
|
Ferini-Strambi L, Liguori C, Lucey BP, Mander BA, Spira AP, Videnovic A, Baumann C, Franco O, Fernandes M, Gnarra O, Krack P, Manconi M, Noain D, Saxena S, Kallweit U, Randerath W, Trenkwalder C, Rosenzweig I, Iranzo A, Bradicich M, Bassetti C. Role of sleep in neurodegeneration: the consensus report of the 5th Think Tank World Sleep Forum. Neurol Sci 2024; 45:749-767. [PMID: 38087143 DOI: 10.1007/s10072-023-07232-7] [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: 10/10/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024]
Abstract
Sleep abnormalities may represent an independent risk factor for neurodegeneration. An international expert group convened in 2021 to discuss the state-of-the-science in this domain. The present article summarizes the presentations and discussions concerning the importance of a strategy for studying sleep- and circadian-related interventions for early detection and prevention of neurodegenerative diseases. An international expert group considered the current state of knowledge based on the most relevant publications in the previous 5 years; discussed the current challenges in the field of relationships among sleep, sleep disorders, and neurodegeneration; and identified future priorities. Sleep efficiency and slow wave activity during non-rapid eye movement (NREM) sleep are decreased in cognitively normal middle-aged and older adults with Alzheimer's disease (AD) pathology. Sleep deprivation increases amyloid-β (Aβ) concentrations in the interstitial fluid of experimental animal models and in cerebrospinal fluid in humans, while increased sleep decreases Aβ. Obstructive sleep apnea (OSA) is a risk factor for dementia. Studies indicate that positive airway pressure (PAP) treatment should be started in patients with mild cognitive impairment or AD and comorbid OSA. Identification of other measures of nocturnal hypoxia and sleep fragmentation could better clarify the role of OSA as a risk factor for neurodegeneration. Concerning REM sleep behavior disorder (RBD), it will be crucial to identify the subset of RBD patients who will convert to a specific neurodegenerative disorder. Circadian sleep-wake rhythm disorders (CSWRD) are strong predictors of caregiver stress and institutionalization, but the absence of recommendations or consensus statements must be considered. Future priorities include to develop and validate existing and novel comprehensive assessments of CSWRD in patients with/at risk for dementia. Strategies for studying sleep-circadian-related interventions for early detection/prevention of neurodegenerative diseases are required. CSWRD evaluation may help to identify additional biomarkers for phenotyping and personalizing treatment of neurodegeneration.
Collapse
Affiliation(s)
- Luigi Ferini-Strambi
- Sleep Disorders Center, Division of Neuroscience, Università Vita-Salute San Raffaele, Milan, Italy.
| | - Claudio Liguori
- Sleep Medicine Center, University of Rome Tor Vergata, Rome, Italy
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aleksandar Videnovic
- Department of Neurology, Division of Sleep Medicine, Massachussets General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Baumann
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Oscar Franco
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | | | - Oriella Gnarra
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Paul Krack
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Mauro Manconi
- Sleep Medicine Unit, Faculty of Biomedical Sciences, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Daniela Noain
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Ulf Kallweit
- Clinical Sleep and Neuroimmunology, University Witten/Herdecke, Witten, Germany
| | | | - C Trenkwalder
- Department of Neurosurgery, Paracelsus-Elena Klinik, University Medical Center, KasselGoettingen, Germany
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, King's College London, London, UK
| | - Alex Iranzo
- Sleep Center, Neurology Service, Hospital Clinic de Barcelona, Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Matteo Bradicich
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
6
|
Wortha SM, Schulz J, Hanna J, Schwarz C, Stubbe B, Frenzel S, Bülow R, Friedrich N, Nauck M, Völzke H, Ewert R, Vogelgesang A, Grabe HJ, Ladenbauer J, Flöel A. Association of spermidine blood levels with microstructure of sleep-implications from a population-based study. GeroScience 2024; 46:1319-1330. [PMID: 37548882 PMCID: PMC10828152 DOI: 10.1007/s11357-023-00886-3] [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: 06/05/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023] Open
Abstract
Deteriorations in slow wave sleep (SWS) have been linked to brain aging and Alzheimer's disease (AD), possibly due to its key role in clearance of amyloid-beta and tau (Aß/tau), two pathogenic hallmarks of AD. Spermidine administration has been shown to improve sleep quality in animal models. So far, the association between spermidine levels in humans and parameters of SWS physiology are unknown but may be valuable for therapeutic strategies. Data from 216 participants (age range 50-81 years) of the population-based Study of Health in Pomerania TREND were included in our analysis. We investigated associations between spermidine plasma levels, key parameters of sleep macroarchitecture and microarchitecture that were previously associated with AD pathology, and brain health measured via a marker of structural brain atrophy (AD score). Higher spermidine levels were significantly associated with lower coupling between slow oscillations and spindle activity. No association was evident for SWS, slow oscillatory, and spindle activity throughout non-rapid eye movement sleep. Furthermore, elevated spermidine blood levels were significantly associated with a higher AD score, while sleep markers revealed no association with AD score. The association between higher spermidine levels and brain health was not mediated by coupling between slow oscillations and spindle activity. We report that higher spermidine blood levels are associated not only with deteriorated brain health but also with less advantageous markers of sleep quality in older adults. Future studies need to evaluate whether sleep, spermidine, and Aß/tau deposition are interrelated and whether sleep may play a mediating role.
Collapse
Affiliation(s)
- Silke M Wortha
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany.
- Centre for Mathematical Cognition, School of Science, Loughborough University, Loughborough, UK.
| | - Juliane Schulz
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Jevri Hanna
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Claudia Schwarz
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Beate Stubbe
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Robin Bülow
- Institute for Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Ralf Ewert
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Antje Vogelgesang
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Julia Ladenbauer
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| |
Collapse
|
7
|
Johnson CE, Duncan MJ, Murphy MP. Sex and Sleep Disruption as Contributing Factors in Alzheimer's Disease. J Alzheimers Dis 2024; 97:31-74. [PMID: 38007653 PMCID: PMC10842753 DOI: 10.3233/jad-230527] [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] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease (AD) affects more women than men, with women throughout the menopausal transition potentially being the most under researched and at-risk group. Sleep disruptions, which are an established risk factor for AD, increase in prevalence with normal aging and are exacerbated in women during menopause. Sex differences showing more disrupted sleep patterns and increased AD pathology in women and female animal models have been established in literature, with much emphasis placed on loss of circulating gonadal hormones with age. Interestingly, increases in gonadotropins such as follicle stimulating hormone are emerging to be a major contributor to AD pathogenesis and may also play a role in sleep disruption, perhaps in combination with other lesser studied hormones. Several sleep influencing regions of the brain appear to be affected early in AD progression and some may exhibit sexual dimorphisms that may contribute to increased sleep disruptions in women with age. Additionally, some of the most common sleep disorders, as well as multiple health conditions that impair sleep quality, are more prevalent and more severe in women. These conditions are often comorbid with AD and have bi-directional relationships that contribute synergistically to cognitive decline and neuropathology. The association during aging of increased sleep disruption and sleep disorders, dramatic hormonal changes during and after menopause, and increased AD pathology may be interacting and contributing factors that lead to the increased number of women living with AD.
Collapse
Affiliation(s)
- Carrie E. Johnson
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
| | - Marilyn J. Duncan
- University of Kentucky, College of Medicine, Department of Neuroscience, Lexington, KY, USA
| | - M. Paul Murphy
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY, USA
| |
Collapse
|
8
|
Miller MR, Lee YF, Kastanenka KV. Calcium sensor Yellow Cameleon 3.6 as a tool to support the calcium hypothesis of Alzheimer's disease. Alzheimers Dement 2023; 19:4196-4203. [PMID: 37154246 PMCID: PMC10524576 DOI: 10.1002/alz.13111] [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: 11/29/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disease with increasing relevance as dementia cases rise. The etiology of AD is widely debated. The Calcium Hypothesis of Alzheimer's disease and brain aging states that the dysfunction of calcium signaling is the final common pathway leading to neurodegeneration. When the Calcium Hypothesis was originally coined, the technology did not exist to test it, but with the advent of Yellow Cameleon 3.6 (YC3.6) we are able to test its validity. METHODS Here we review use of YC3.6 in studying Alzheimer's disease using mouse models and discuss whether these studies support or refute the Calcium Hypothesis. RESULTS YC3.6 studies showed that amyloidosis preceded dysfunction in neuronal calcium signaling and changes in synapse structure. This evidence supports the Calcium Hypothesis. DISCUSSION In vivo YC3.6 studies point to calcium signaling as a promising therapeutic target; however, additional work is necessary to translate these findings to humans.
Collapse
Affiliation(s)
- Morgan R. Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yee Fun Lee
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ksenia V. Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| |
Collapse
|
9
|
Lee YF, Russ AN, Zhao Q, Perle SJ, Maci M, Miller MR, Hou SS, Algamal M, Zhao Z, Li H, Gelwan N, Liu Z, Gomperts SN, Araque A, Galea E, Bacskai BJ, Kastanenka KV. Optogenetic targeting of astrocytes restores slow brain rhythm function and slows Alzheimer's disease pathology. Sci Rep 2023; 13:13075. [PMID: 37567942 PMCID: PMC10421876 DOI: 10.1038/s41598-023-40402-3] [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: 04/13/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
Patients with Alzheimer's disease (AD) exhibit non-rapid eye movement (NREM) sleep disturbances in addition to memory deficits. Disruption of NREM slow waves occurs early in the disease progression and is recapitulated in transgenic mouse models of beta-amyloidosis. However, the mechanisms underlying slow-wave disruptions remain unknown. Because astrocytes contribute to slow-wave activity, we used multiphoton microscopy and optogenetics to investigate whether they contribute to slow-wave disruptions in APP/PS1 mice. The power but not the frequency of astrocytic calcium transients was reduced in APP/PS1 mice compared to nontransgenic controls. Optogenetic activation of astrocytes at the endogenous frequency of slow waves restored slow-wave power, reduced amyloid deposition, prevented neuronal calcium elevations, and improved memory performance. Our findings revealed malfunction of the astrocytic network driving slow-wave disruptions. Thus, targeting astrocytes to restore circuit activity underlying sleep and memory disruptions in AD could ameliorate disease progression.
Collapse
Affiliation(s)
- Yee Fun Lee
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Alyssa N Russ
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Qiuchen Zhao
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Stephen J Perle
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Megi Maci
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Morgan R Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Steven S Hou
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Moustafa Algamal
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Zhuoyang Zhao
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Hanyan Li
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Noah Gelwan
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Zhe Liu
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Stephen N Gomperts
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Elena Galea
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Brian J Bacskai
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Ksenia V Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
10
|
Semyachkina-Glushkovskaya O, Penzel T, Poluektov M, Fedosov I, Tzoy M, Terskov A, Blokhina I, Sidorov V, Kurths J. Phototherapy of Alzheimer's Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review. Int J Mol Sci 2023; 24:10946. [PMID: 37446135 DOI: 10.3390/ijms241310946] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The global number of people with Alzheimer's disease (AD) doubles every 5 years. It has been established that unless an effective treatment for AD is found, the incidence of AD will triple by 2060. However, pharmacological therapies for AD have failed to show effectiveness and safety. Therefore, the search for alternative methods for treating AD is an urgent problem in medicine. The lymphatic drainage and removal system of the brain (LDRSB) plays an important role in resistance to the progression of AD. The development of methods for augmentation of the LDRSB functions may contribute to progress in AD therapy. Photobiomodulation (PBM) is considered to be a non-pharmacological and safe approach for AD therapy. Here, we highlight the most recent and relevant studies of PBM for AD. We focus on emerging evidence that indicates the potential benefits of PBM during sleep for modulation of natural activation of the LDRSB at nighttime, providing effective removal of metabolites, including amyloid-β, from the brain, leading to reduced progression of AD. Our review creates a new niche in the therapy of brain diseases during sleep and sheds light on the development of smart sleep technologies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Department of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
| | - Thomas Penzel
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
- Interdisziplinäres Schlafmedizinisches Zentrum, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Mikhail Poluektov
- Department of Nervous Diseases, Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya 2, Building 4, 119435 Moscow, Russia
| | - Ivan Fedosov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
| | - Maria Tzoy
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
| | - Andrey Terskov
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
| | - Inna Blokhina
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
| | - Viktor Sidorov
- Company "Lazma" for Research and Production Enterprise of Laser Medical Equipment, Kuusinena Str. 11, 123308 Moscow, Russia
| | - Jürgen Kurths
- Department of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
- Department of Complexity Science, Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
11
|
Lee YF, Russ AN, Zhao Q, Maci M, Miller MR, Hou SS, Algamal M, Zhao Z, Li H, Gelwan N, Gomperts SN, Araque A, Galea E, Bacskai BJ, Kastanenka KV. Optogenetic Targeting of Astrocytes Restores Slow Brain Rhythm Function and Slows Alzheimer's Disease Pathology. RESEARCH SQUARE 2023:rs.3.rs-2813056. [PMID: 37163040 PMCID: PMC10168443 DOI: 10.21203/rs.3.rs-2813056/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Patients with Alzheimer's disease (AD) exhibit non-rapid eye movement (NREM) sleep disturbances in addition to memory deficits. Disruption of NREM slow waves occurs early in the disease progression and is recapitulated in transgenic mouse models of beta-amyloidosis. However, the mechanisms underlying slow-wave disruptions remain unknown. Because astrocytes contribute to slow-wave activity, we used multiphoton microscopy and optogenetics to investigate whether they contribute to slow-wave disruptions in APP mice. The power but not the frequency of astrocytic calcium transients was reduced in APP mice compared to nontransgenic controls. Optogenetic activation of astrocytes at the endogenous frequency of slow waves restored slow-wave power, reduced amyloid deposition, prevented neuronal calcium elevations, and improved memory performance. Our findings revealed malfunction of the astrocytic network driving slow-wave disruptions. Thus, targeting astrocytes to restore circuit activity underlying sleep and memory disruptions in AD could ameliorate disease progression.
Collapse
Affiliation(s)
| | - Alyssa N Russ
- Massachusetts General Hospital, Harvard Medical School
| | - Qiuchen Zhao
- Massachusetts General Hospital, Harvard Medical School
| | - Megi Maci
- Massachusetts General Hospital, Harvard Medical School
| | | | - Steven S Hou
- Massachusetts General Hospital, Harvard Medical School
| | | | - Zhuoyang Zhao
- Massachusetts General Hospital, Harvard Medical School
| | - Hanyan Li
- Massachusetts General Hospital, Harvard Medical School
| | - Noah Gelwan
- Massachusetts General Hospital, Harvard Medical School
| | | | | | - Elena Galea
- Massachusetts General Hospital, Harvard Medical School
| | | | | |
Collapse
|
12
|
Soula M, Martín-Ávila A, Zhang Y, Dhingra A, Nitzan N, Sadowski MJ, Gan WB, Buzsáki G. Forty-hertz light stimulation does not entrain native gamma oscillations in Alzheimer's disease model mice. Nat Neurosci 2023; 26:570-578. [PMID: 36879142 PMCID: PMC10839995 DOI: 10.1038/s41593-023-01270-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/31/2023] [Indexed: 03/08/2023]
Abstract
There is a demand for noninvasive methods to ameliorate disease. We investigated whether 40-Hz flickering light entrains gamma oscillations and suppresses amyloid-β in the brains of APP/PS1 and 5xFAD mouse models of Alzheimer's disease. We used multisite silicon probe recording in the visual cortex, entorhinal cortex or the hippocampus and found that 40-Hz flickering simulation did not engage native gamma oscillations in these regions. Additionally, spike responses in the hippocampus were weak, suggesting 40-Hz light does not effectively entrain deep structures. Mice avoided 40-Hz flickering light, associated with elevated cholinergic activity in the hippocampus. We found no reliable changes in plaque count or microglia morphology by either immunohistochemistry or in vivo two-photon imaging following 40-Hz stimulation, nor reduced levels of amyloid-β 40/42. Thus, visual flicker stimulation may not be a viable mechanism for modulating activity in deep structures.
Collapse
Affiliation(s)
- Marisol Soula
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Alejandro Martín-Ávila
- Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, NY, USA
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, USA
| | - Yiyao Zhang
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Annika Dhingra
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Noam Nitzan
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Martin J Sadowski
- Department of Neurology and Psychiatry, Langone Medical Center, New York University, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, Langone Medical Center, New York University, New York, NY, USA
| | - Wen-Biao Gan
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, USA
| | - György Buzsáki
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA.
- Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, NY, USA.
- Department of Neurology and Psychiatry, Langone Medical Center, New York University, New York, NY, USA.
| |
Collapse
|
13
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Abusudah WF, Almohmadi NH, Eldahshan OA, Ahmed EA, Batiha GES. Insights on benzodiazepines' potential in Alzheimer's disease. Life Sci 2023; 320:121532. [PMID: 36858314 DOI: 10.1016/j.lfs.2023.121532] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023]
Abstract
Alzheimer's disease (AD) is the most frequent type of dementia characterized by the deposition of amyloid beta (Aβ) plaque and tau-neurofibrillary tangles (TNTs) in the brain. AD is associated with the disturbances of various neurotransmitters including gamma-aminobutyric acid (GABA). Of note, GABA is reduced in AD, and restoration of GABA effect by benzodiazepines (BDZs) may improve AD outcomes. However, BDZs may adversely affect cognitive functions chiefly in elderly AD patients with sleep disorders. Besides, there is a controversy regarding the use of BDZs in AD. Consequently, the objective of the present review was to disclose the possible role of BDZs on the pathogenesis of AD that might be beneficial, neutral, or detrimental effects on AD. Prolonged use of intermediate-acting BDZ lorazepam exerts amnesic effects due to attenuation of synaptic plasticity and impairment of recognition memory. However, BDZs may have a protective effect against the development of AD by reducing tau phosphorylation, neuroinflammation, and progression of AD neuropathology. On the other side, other findings highlighted that extended use of BDZs was not associated with the development of AD. In conclusion, there are controversial points concerning the use of BDZs and the risk for the progression of AD. Thus, preclinical, and clinical studies are essential in this regard.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department Applied Medical Sciences, College Jazan University, Jazan 82817, Saudi Arabia.
| | - Wafaa Fouzi Abusudah
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Najlaa Hamed Almohmadi
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Omayma A Eldahshan
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Al Beheira, Egypt.
| |
Collapse
|
14
|
Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases. Int J Mol Sci 2023; 24:ijms24043221. [PMID: 36834631 PMCID: PMC9965491 DOI: 10.3390/ijms24043221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Emerging evidence suggests that an important function of the sleeping brain is the removal of wastes and toxins from the central nervous system (CNS) due to the activation of the brain waste removal system (BWRS). The meningeal lymphatic vessels (MLVs) are an important part of the BWRS. A decrease in MLV function is associated with Alzheimer's and Parkinson's diseases, intracranial hemorrhages, brain tumors and trauma. Since the BWRS is activated during sleep, a new idea is now being actively discussed in the scientific community: night stimulation of the BWRS might be an innovative and promising strategy for neurorehabilitation medicine. This review highlights new trends in photobiomodulation of the BWRS/MLVs during deep sleep as a breakthrough technology for the effective removal of wastes and unnecessary compounds from the brain in order to increase the neuroprotection of the CNS as well as to prevent or delay various brain diseases.
Collapse
|
15
|
Zhou H, Li H, Gowravaram N, Quan M, Kausar N, Gomperts SN. Disruption of hippocampal neuronal circuit function depends upon behavioral state in the APP/PS1 mouse model of Alzheimer's disease. Sci Rep 2022; 12:21022. [PMID: 36471155 PMCID: PMC9723144 DOI: 10.1038/s41598-022-25364-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The Alzheimer's disease-associated peptide amyloid-beta (Aβ) has been associated with neuronal hyperactivity under anesthesia, but clinical trials of anticonvulsants or neural system suppressors have, so far, failed to improve symptoms in AD. Using simultaneous hippocampal calcium imaging and electrophysiology in freely moving mice expressing human Aβ, here we show that Aβ aggregates perturbed neural systems in a state-dependent fashion, driving neuronal hyperactivity in exploratory behavior and slow wave sleep (SWS), yet suppressing activity in quiet wakefulness (QW) and REM sleep. In exploratory behavior and REM sleep, Aβ impaired hippocampal theta-gamma phase-amplitude coupling and altered neuronal synchronization with theta. In SWS, Aβ reduced cortical slow oscillation (SO) power, the coordination of hippocampal sharp wave-ripples with both the SO and thalamocortical spindles, and the coordination of calcium transients with the sharp wave-ripple. Physostigmine improved Aβ-associated hyperactivity in exploratory behavior and hypoactivity in QW and expanded the range of gamma that coupled with theta phase, but exacerbated hypoactivity in exploratory behavior. Together, these findings show that the effects of Aβ alone on hippocampal circuit function are profoundly state dependent and suggest a reformulation of therapeutic strategies aimed at Aβ induced hyperexcitability.
Collapse
Affiliation(s)
- Heng Zhou
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Hanyan Li
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Niket Gowravaram
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Moqin Quan
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Naila Kausar
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Stephen N Gomperts
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
16
|
Stephen TL, Breningstall B, Suresh S, McGill CJ, Pike CJ. APOE genotype and biological sex regulate astroglial interactions with amyloid plaques in Alzheimer's disease mice. J Neuroinflammation 2022; 19:286. [PMID: 36457019 PMCID: PMC9714101 DOI: 10.1186/s12974-022-02650-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/20/2022] [Indexed: 12/03/2022] Open
Abstract
The most significant genetic risk factor for developing late-onset Alzheimer's disease (AD) is the ε4 allele of apolipoprotein E (APOE4). APOE genotype and biological sex are key modulators of microglial and astroglial function, which exert multiple effects on AD pathogenesis. Here, we show astroglial interactions with amyloid plaques in the EFAD transgenic mouse model of AD. Using confocal microscopy, we observed significantly lower levels of astrocytic plaque coverage and plaque compaction (beneficial effects of glial barrier formation) with APOE4 genotype and female sex. Conversely, neurite damage and astrocyte activation in the plaque environment were significantly higher in APOE4 carriers and female mice. Astrocyte coverage of plaques was highest in APOE3 males and poorest in APOE4 females. Collectively, our findings provide new insights into the roles of astroglia and highlight the importance of addressing independent and interactive effects of APOE genotype and biological sex in understanding processes contributing to AD pathogenesis.
Collapse
Affiliation(s)
- T. L. Stephen
- grid.42505.360000 0001 2156 6853Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| | - B. Breningstall
- grid.42505.360000 0001 2156 6853Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| | - S. Suresh
- grid.42505.360000 0001 2156 6853Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| | - C. J. McGill
- grid.42505.360000 0001 2156 6853Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| | - C. J. Pike
- grid.42505.360000 0001 2156 6853Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| |
Collapse
|
17
|
Chylinski D, Narbutas J, Balteau E, Collette F, Bastin C, Berthomier C, Salmon E, Maquet P, Carrier J, Phillips C, Lina JM, Vandewalle G, Van Egroo M. Frontal grey matter microstructure is associated with sleep slow waves characteristics in late midlife. Sleep 2022; 45:zsac178. [PMID: 35869626 PMCID: PMC9644125 DOI: 10.1093/sleep/zsac178] [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: 03/24/2022] [Revised: 07/13/2022] [Indexed: 07/25/2023] Open
Abstract
STUDY OBJECTIVES The ability to generate slow waves (SW) during non-rapid eye movement (NREM) sleep decreases as early as the 5th decade of life, predominantly over frontal regions. This decrease may concern prominently SW characterized by a fast switch from hyperpolarized to depolarized, or down-to-up, state. Yet, the relationship between these fast and slow switcher SW and cerebral microstructure in ageing is not established. METHODS We recorded habitual sleep under EEG in 99 healthy late midlife individuals (mean age = 59.3 ± 5.3 years; 68 women) and extracted SW parameters (density, amplitude, frequency) for all SW as well as according to their switcher type (slow vs. fast). We further used neurite orientation dispersion and density imaging (NODDI) to assess microstructural integrity over a frontal grey matter region of interest (ROI). RESULTS In statistical models adjusted for age, sex, and sleep duration, we found that a lower SW density, particularly for fast switcher SW, was associated with a reduced orientation dispersion of neurites in the frontal ROI (p = 0.018, R2β* = 0.06). In addition, overall SW frequency was positively associated with neurite density (p = 0.03, R2β* = 0.05). By contrast, we found no significant relationships between SW amplitude and NODDI metrics. CONCLUSIONS Our findings suggest that the complexity of neurite organization contributes specifically to the rate of fast switcher SW occurrence in healthy middle-aged individuals, corroborating slow and fast switcher SW as distinct types of SW. They further suggest that the density of frontal neurites plays a key role for neural synchronization during sleep. TRIAL REGISTRATION NUMBER EudraCT 2016-001436-35.
Collapse
Affiliation(s)
- Daphne Chylinski
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Justinas Narbutas
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Evelyne Balteau
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Fabienne Collette
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Christine Bastin
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | | | - Eric Salmon
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
- Department of Neurology, University Hospital of Liège, Liège, Belgium
| | - Pierre Maquet
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Department of Neurology, University Hospital of Liège, Liège, Belgium
| | - Julie Carrier
- CARSM, CIUSSS of Nord-de l’Île-de-Montréal, Montreal, Canada
- Department of Psychology, University of Montreal, Canada
| | - Christophe Phillips
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- GIGA-In Silico Medicine, University of Liège, Liège, Belgium
| | - Jean-Marc Lina
- CARSM, CIUSSS of Nord-de l’Île-de-Montréal, Montreal, Canada
- Department of Psychology, University of Montreal, Canada
| | - Gilles Vandewalle
- Corresponding authors. Gilles Vandewalle, GIGA-Cyclotron Research Centre-In Vivo Imaging, Bâtiment B30, Université de Liège, Allée du Six Août, 8, 4000 Liège, Belgium.
| | - Maxime Van Egroo
- Maxime Van Egroo, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, Maastricht, The Netherlands.
| |
Collapse
|
18
|
Blackman J, Stankeviciute L, Arenaza-Urquijo EM, Suárez-Calvet M, Sánchez-Benavides G, Vilor-Tejedor N, Iranzo A, Molinuevo JL, Gispert JD, Coulthard E, Grau-Rivera O. Cross-sectional and longitudinal association of sleep and Alzheimer biomarkers in cognitively unimpaired adults. Brain Commun 2022; 4:fcac257. [PMID: 36337343 PMCID: PMC9630979 DOI: 10.1093/braincomms/fcac257] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/25/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Sleep abnormalities are prevalent in Alzheimer’s disease, with sleep quality already impaired at its preclinical stage. Epidemiological and experimental data point to sleep abnormalities contributing to the risk of Alzheimer’s disease. However, previous studies are limited by either a lack of Alzheimer’s disease biomarkers, reduced sample size or cross-sectional design. Understanding if, when, and how poor sleep contributes to Alzheimer’s disease progression is important so that therapies can be targeted to the right phase of the disease. Using the largest cohort to date, the European Prevention of Alzheimer’s Dementia Longitudinal Cohort Study, we test the hypotheses that poor sleep is associated with core Alzheimer’s disease CSF biomarkers cross-sectionally and predicts future increments of Alzheimer’s disease pathology in people without identifiable symptoms of Alzheimer’s disease at baseline. This study included 1168 adults aged over 50 years with CSF core Alzheimer’s disease biomarkers (total tau, phosphorylated tau and amyloid-beta), cognitive performance, and sleep quality (Pittsburgh sleep quality index questionnaire) data. We used multivariate linear regressions to analyse associations between core Alzheimer’s disease biomarkers and the following Pittsburgh sleep quality index measures: total score of sleep quality, binarized score (poor sleep categorized as Pittsburgh sleep quality index > 5), sleep latency, duration, efficiency and disturbance. On a subsample of 332 participants with CSF taken at baseline and after an average period of 1.5 years, we assessed the effect of baseline sleep quality on change in Alzheimer’s disease biomarkers over time. Cross-sectional analyses revealed that poor sleep quality (Pittsburgh sleep quality index total > 5) was significantly associated with higher CSF t-tau; shorter sleep duration (<7 h) was associated with higher CSF p-tau and t-tau; and a higher degree of sleep disturbance (1–9 versus 0 and >9 versus 0) was associated with lower CSF amyloid-beta. Longitudinal analyses showed that greater sleep disturbances (1–9 versus 0 and >9 versus 0) were associated with a decrease in CSF Aβ42 over time. This study demonstrates that self-reported poor sleep quality is associated with greater Alzheimer’s disease-related pathology in cognitively unimpaired individuals, with longitudinal results further strengthening the hypothesis that disrupted sleep may represent a risk factor for Alzheimer’s disease. This highlights the need for future work to test the efficacy of preventive practices, designed to improve sleep at pre-symptomatic stages of disease, on reducing Alzheimer’s disease pathology.
Collapse
Affiliation(s)
- Jonathan Blackman
- North Bristol NHS Trust , Bristol BS10 5NB , UK
- Bristol Medical School, University of Bristol , Bristol BS8 1UD , UK
| | - Laura Stankeviciute
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- Universitat Pompeu Fabra , Barcelona 08005 , Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
- Servei de Neurologia, Hospital del Mar , Barcelona 08003 , Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
| | - Natalia Vilor-Tejedor
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- Universitat Pompeu Fabra , Barcelona 08005 , Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology , Barcelona 08003 , Spain
- Department of Clinical Genetics, Erasmus University Medical Center , Rotterdam 3015 GD , The Netherlands
| | - Alejandro Iranzo
- Neurology Service, Hospital Clínic de Barcelona and Institut D'Investigacions Biomèdiques , Barcelona 08036 , Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de Barcelona , Barcelona 28029 , Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Madrid 28029 , Spain
| | - Elizabeth Coulthard
- North Bristol NHS Trust , Bristol BS10 5NB , UK
- Bristol Medical School, University of Bristol , Bristol BS8 1UD , UK
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
- Servei de Neurologia, Hospital del Mar , Barcelona 08003 , Spain
| | | |
Collapse
|
19
|
Matera C, Bregestovski P. Light-Controlled Modulation and Analysis of Neuronal Functions. Int J Mol Sci 2022; 23:12921. [PMID: 36361710 PMCID: PMC9657357 DOI: 10.3390/ijms232112921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 04/15/2024] Open
Abstract
Light is an extraordinary tool allowing us to read out and control neuronal functions thanks to its unique properties: it has a great degree of bioorthogonality and is minimally invasive; it can be precisely delivered with high spatial and temporal precision; and it can be used simultaneously or consequently at multiple wavelengths and locations [...].
Collapse
Affiliation(s)
- Carlo Matera
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Piotr Bregestovski
- Institut National de la Santé et de la Recherche Médicale, Institut de Neurosciences des Systèmes, Aix-Marseille University, 13005 Marseille, France
- Institute of Neurosciences, Kazan State Medical University, 420111 Kazan, Russia
- Department of Normal Physiology, Kazan State Medical University, 420111 Kazan, Russia
| |
Collapse
|
20
|
Ko H, Yoon SP. Optogenetic neuromodulation with gamma oscillation as a new strategy for Alzheimer disease: a narrative review. JOURNAL OF YEUNGNAM MEDICAL SCIENCE 2022; 39:269-277. [PMID: 35152662 PMCID: PMC9580057 DOI: 10.12701/jyms.2021.01683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/31/2022]
Abstract
The amyloid hypothesis has been considered a major explanation of the pathogenesis of Alzheimer disease. However, failure of phase III clinical trials with anti-amyloid-beta monoclonal antibodies reveals the need for other therapeutic approaches to treat Alzheimer disease. Compared to its relatively short history, optogenetics has developed considerably. The expression of microbial opsins in cells using genetic engineering allows specific control of cell signals or molecules. The application of optogenetics to Alzheimer disease research or clinical approaches is increasing. When applied with gamma entrainment, optogenetic neuromodulation can improve Alzheimer disease symptoms. Although safety problems exist with optogenetics such as the use of viral vectors, this technique has great potential for use in Alzheimer disease. In this paper, we review the historical applications of optogenetic neuromodulation with gamma entrainment to investigate the mechanisms involved in Alzheimer disease and potential therapeutic strategies.
Collapse
Affiliation(s)
- Haneol Ko
- Medical Course, Jeju National University School of Medicine, Jeju, Korea
| | - Sang-Pil Yoon
- Department of Anatomy, Jeju National University College of Medicine, Jeju, Korea
| |
Collapse
|
21
|
Abstract
Over the past few decades, the importance of sleep has become increasingly recognized for many physiologic functions, including cognition. Many studies have reported the deleterious effect of sleep loss or sleep disruption on cognitive performance. Beyond ensuring adequate sleep quality and duration, discovering methods to enhance sleep to augment its restorative effects is important to improve learning in many populations, such as the military, students, age-related cognitive decline, and cognitive disorders.
Collapse
Affiliation(s)
- Roneil G Malkani
- Division of Sleep Medicine, Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, 710 North Lake Shore Drive, Suite 525, Chicago, IL 60611, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA.
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, 710 North Lake Shore Drive, Suite 520, Chicago, IL 60611, USA
| |
Collapse
|
22
|
Thankachan S, Yang C, Kastanenka KV, Bacskai BJ, Gerashchenko D. Low frequency visual stimulation enhances slow wave activity without disrupting the sleep pattern in mice. Sci Rep 2022; 12:12278. [PMID: 35853986 PMCID: PMC9296645 DOI: 10.1038/s41598-022-16478-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Non-invasive stimulation technologies are emerging as potential treatment options for a range of neurodegenerative disorders. Experimental evidence suggests that stimuli-evoked changes in slow brain rhythms may mitigate or even prevent neuropathological and behavioral impairments. Slow wave activity is prevalent during sleep and can be triggered non-invasively by sensory stimulation targeting the visual system or directly via activation of neurons locally using optogenetics. Here, we developed new tools for delivering visual stimulation using light-emitting diodes in freely moving mice while awake and during sleep. We compared these tools to traditional optogenetic approaches used for local stimulation of neurons in the cerebral cortex. We then used these tools to compare the effects of low-frequency visual versus optogenetic stimulations on the slow wave activity and sleep pattern in mice. Visual stimulation effectively enhanced slow wave activity without disrupting the sleep pattern. Optogenetic stimulation of cortical GABAergic neurons increased NREM sleep. These results suggest that visual stimulation can be effective at boosting slow wave activity without having adverse effects on sleep and thus holds great potential as a non-invasive stimulation treatment strategy.
Collapse
Affiliation(s)
- Stephen Thankachan
- Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, MA, 02132, USA
| | - Chun Yang
- Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, MA, 02132, USA
| | - Ksenia V Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Brian J Bacskai
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Dmitry Gerashchenko
- Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, MA, 02132, USA.
| |
Collapse
|
23
|
Katsuki F, Gerashchenko D, Brown RE. Alterations of sleep oscillations in Alzheimer's disease: A potential role for GABAergic neurons in the cortex, hippocampus, and thalamus. Brain Res Bull 2022; 187:181-198. [PMID: 35850189 DOI: 10.1016/j.brainresbull.2022.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/01/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
Sleep abnormalities are widely reported in patients with Alzheimer's disease (AD) and are linked to cognitive impairments. Sleep abnormalities could be potential biomarkers to detect AD since they are often observed at the preclinical stage. Moreover, sleep could be a target for early intervention to prevent or slow AD progression. Thus, here we review changes in brain oscillations observed during sleep, their connection to AD pathophysiology and the role of specific brain circuits. Slow oscillations (0.1-1 Hz), sleep spindles (8-15 Hz) and their coupling during non-REM sleep are consistently reduced in studies of patients and in AD mouse models although the timing and magnitude of these alterations depends on the pathophysiological changes and the animal model studied. Changes in delta (1-4 Hz) activity are more variable. Animal studies suggest that hippocampal sharp-wave ripples (100-250 Hz) are also affected. Reductions in REM sleep amount and slower oscillations during REM are seen in patients but less consistently in animal models. Thus, changes in a variety of sleep oscillations could impact sleep-dependent memory consolidation or restorative functions of sleep. Recent mechanistic studies suggest that alterations in the activity of GABAergic neurons in the cortex, hippocampus and thalamic reticular nucleus mediate sleep oscillatory changes in AD and represent a potential target for intervention. Longitudinal studies of the timing of AD-related sleep abnormalities with respect to pathology and dysfunction of specific neural networks are needed to identify translationally relevant biomarkers and guide early intervention strategies to prevent or delay AD progression.
Collapse
Affiliation(s)
- Fumi Katsuki
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA.
| | - Dmitry Gerashchenko
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| | - Ritchie E Brown
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| |
Collapse
|
24
|
Mirzayi P, Shobeiri P, Kalantari A, Perry G, Rezaei N. Optogenetics: implications for Alzheimer's disease research and therapy. Mol Brain 2022; 15:20. [PMID: 35197102 PMCID: PMC8867657 DOI: 10.1186/s13041-022-00905-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD), a critical neurodegenerative condition, has a wide range of effects on brain activity. Synaptic plasticity and neuronal circuits are the most vulnerable in Alzheimer’s disease, but the exact mechanism is unknown. Incorporating optogenetics into the study of AD has resulted in a significant leap in this field during the last decades, kicking off a revolution in our knowledge of the networks that underpin cognitive functions. In Alzheimer's disease, optogenetics can help to reduce and reverse neural circuit and memory impairments. Here we review how optogenetically driven methods have helped expand our knowledge of Alzheimer's disease, and how optogenetic interventions hint at a future translation into therapeutic possibilities for further utilization in clinical settings. In conclusion, neuroscience has witnessed one of its largest revolutions following the introduction of optogenetics into the field.
Collapse
Affiliation(s)
- Parsa Mirzayi
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Kalantari
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
25
|
Duncan M, Guerriero L, Kohler K, Beechem L, Gillis B, Salisbury F, Wessel C, Wang J, Sunderam S, Bachstetter A, O’Hara B, Murphy M. Chronic Fragmentation of the Daily Sleep-Wake Rhythm Increases Amyloid-beta Levels and Neuroinflammation in the 3xTg-AD Mouse Model of Alzheimer's Disease. Neuroscience 2022; 481:111-122. [PMID: 34856352 PMCID: PMC8941625 DOI: 10.1016/j.neuroscience.2021.11.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/17/2023]
Abstract
Fragmentation of the daily sleep-wake rhythm with increased nighttime awakenings and more daytime naps is correlated with the risk of development of Alzheimer's disease (AD). To explore whether a causal relationship underlies this correlation, the present study tested the hypothesis that chronic fragmentation of the daily sleep-wake rhythm stimulates brain amyloid-beta (Aβ) levels and neuroinflammation in the 3xTg-AD mouse model of AD. Female 3xTg-AD mice were allowed to sleep undisturbed or were subjected to chronic sleep fragmentation consisting of four daily sessions of enforced wakefulness (one hour each) evenly distributed during the light phase, five days a week for four weeks. Piezoelectric sleep recording revealed that sleep fragmentation altered the daily sleep-wake rhythm to resemble the pattern observed in AD. Levels of amyloid-beta (Aβ40 and Aβ42) determined by ELISA were higher in hippocampal tissue collected from sleep-fragmented mice than from undisturbed controls. In contrast, hippocampal levels of tau and phospho-tau differed minimally between sleep fragmented and undisturbed control mice. Sleep fragmentation also stimulated neuroinflammation as shown by increased expression of markers of microglial activation and proinflammatory cytokines measured by q-RT-PCR analysis of hippocampal samples. No significant effects of sleep fragmentation on Aβ, tau, or neuroinflammation were observed in the cerebral cortex. These studies support the concept that improving sleep consolidation in individuals at risk for AD may be beneficial for slowing the onset or progression of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- M.J. Duncan
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| | - L.E. Guerriero
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - K. Kohler
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - L.E. Beechem
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536
| | - B.D. Gillis
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536
| | - F. Salisbury
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - C. Wessel
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - J. Wang
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - S. Sunderam
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - A.D. Bachstetter
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536
| | - B.F. O’Hara
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - M.P. Murphy
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| |
Collapse
|
26
|
Coulthard E, Blackman J. Sleep and future cognitive decline. Brain 2021; 144:2568-2570. [PMID: 34687209 PMCID: PMC8536927 DOI: 10.1093/brain/awab315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
This scientific commentary refers to ‘Sleep and longitudinal cognitive performance in preclinical and early symptomatic Alzheimer disease’ by Lucey et al. (doi:10.1093/brain/awab272).
Collapse
Affiliation(s)
- Elizabeth Coulthard
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jonathan Blackman
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
27
|
Keshmiri Neghab H, Soheilifar MH, Grusch M, Ortega MM, Esmaeeli Djavid G, Saboury AA, Goliaei B. The state of the art of biomedical applications of optogenetics. Lasers Surg Med 2021; 54:202-216. [PMID: 34363230 DOI: 10.1002/lsm.23463] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Optogenetics has opened new insights into biomedical research with the ability to manipulate and control cellular activity using light in combination with genetically engineered photosensitive proteins. By stimulating with light, this method provides high spatiotemporal and high specificity resolution, which is in contrast to conventional pharmacological or electrical stimulation. Optogenetics was initially introduced to control neural activities but was gradually extended to other biomedical fields. STUDY DESIGN In this paper, firstly, we summarize the current optogenetic tools stimulated by different light sources, including lasers, light-emitting diodes, and laser diodes. Second, we outline the variety of biomedical applications of optogenetics not only for neuronal circuits but also for various kinds of cells and tissues from cardiomyocytes to ganglion cells. Furthermore, we highlight the potential of this technique for treating neurological disorders, cardiac arrhythmia, visual impairment, hearing loss, and urinary bladder diseases as well as clarify the mechanisms underlying cancer progression and control of stem cell differentiation. CONCLUSION We sought to summarize the various types of promising applications of optogenetics to treat a broad spectrum of disorders. It is conceivable to expect that optogenetics profits a growing number of patients suffering from a range of different diseases in the near future.
Collapse
Affiliation(s)
- Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | | | - Michael Grusch
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Manoela Marques Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, São Paulo, Brazil
| | - Gholamreza Esmaeeli Djavid
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Ali Akbar Saboury
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Bahram Goliaei
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
28
|
Byron N, Semenova A, Sakata S. Mutual Interactions between Brain States and Alzheimer's Disease Pathology: A Focus on Gamma and Slow Oscillations. BIOLOGY 2021; 10:707. [PMID: 34439940 PMCID: PMC8389330 DOI: 10.3390/biology10080707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 12/26/2022]
Abstract
Brain state varies from moment to moment. While brain state can be defined by ongoing neuronal population activity, such as neuronal oscillations, this is tightly coupled with certain behavioural or vigilant states. In recent decades, abnormalities in brain state have been recognised as biomarkers of various brain diseases and disorders. Intriguingly, accumulating evidence also demonstrates mutual interactions between brain states and disease pathologies: while abnormalities in brain state arise during disease progression, manipulations of brain state can modify disease pathology, suggesting a therapeutic potential. In this review, by focusing on Alzheimer's disease (AD), the most common form of dementia, we provide an overview of how brain states change in AD patients and mouse models, and how controlling brain states can modify AD pathology. Specifically, we summarise the relationship between AD and changes in gamma and slow oscillations. As pathological changes in these oscillations correlate with AD pathology, manipulations of either gamma or slow oscillations can modify AD pathology in mouse models. We argue that neuromodulation approaches to target brain states are a promising non-pharmacological intervention for neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicole Byron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Anna Semenova
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Shuzo Sakata
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
29
|
Hector A, Brouillette J. Hyperactivity Induced by Soluble Amyloid-β Oligomers in the Early Stages of Alzheimer's Disease. Front Mol Neurosci 2021; 13:600084. [PMID: 33488358 PMCID: PMC7817907 DOI: 10.3389/fnmol.2020.600084] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Soluble amyloid-beta oligomers (Aβo) start to accumulate in the human brain one to two decades before any clinical symptoms of Alzheimer's disease (AD) and are implicated in synapse loss, one of the best predictors of memory decline that characterize the illness. Cognitive impairment in AD was traditionally thought to result from a reduction in synaptic activity which ultimately induces neurodegeneration. More recent evidence indicates that in the early stages of AD synaptic failure is, at least partly, induced by neuronal hyperactivity rather than hypoactivity. Here, we review the growing body of evidence supporting the implication of soluble Aβo on the induction of neuronal hyperactivity in AD animal models, in vitro, and in humans. We then discuss the impact of Aβo-induced hyperactivity on memory performance, cell death, epileptiform activity, gamma oscillations, and slow wave activity. We provide an overview of the cellular and molecular mechanisms that are emerging to explain how Aβo induce neuronal hyperactivity. We conclude by providing an outlook on the impact of hyperactivity for the development of disease-modifying interventions at the onset of AD.
Collapse
Affiliation(s)
- Audrey Hector
- Department of Pharmacology and Physiology, Hôpital du Sacré-Cœur de Montréal Research Center, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de-Montréal (CIUSSS-NIM), Université de Montréal, Montreal, QC, Canada
| | - Jonathan Brouillette
- Department of Pharmacology and Physiology, Hôpital du Sacré-Cœur de Montréal Research Center, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de-Montréal (CIUSSS-NIM), Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
30
|
Winer JR, Mander BA, Kumar S, Reed M, Baker SL, Jagust WJ, Walker MP. Sleep Disturbance Forecasts β-Amyloid Accumulation across Subsequent Years. Curr Biol 2020; 30:4291-4298.e3. [PMID: 32888482 PMCID: PMC7642104 DOI: 10.1016/j.cub.2020.08.017] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/08/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
Experimental sleep-wake disruption in rodents and humans causally modulates β-amyloid (Aβ) dynamics (e.g., [1-3]). This leads to the hypothesis that, beyond cross-sectional associations, impaired sleep structure and physiology could represent prospective biomarkers of the speed with which Aβ accumulates over time. Here, we test the hypothesis that initial baseline measures of non-rapid eye movement (NREM) sleep slow-wave activity (SWA) and sleep quality (efficiency) provide future forecasting sensitivity to the rate of Aβ accumulation over subsequent years. A cohort of clinically normal older adults was assessed using objective sleep polysomnography in combination with longitudinal tracking of Aβ accumulation with [11C]PiB positron emission tomography (PET) imaging. Both the proportion of NREM SWA below 1 Hz and the measure of sleep efficiency predicted the speed (slope) of subsequent Aβ deposition over time, and these associations remained robust when taking into account additional cofactors of interest (e.g., age, sex, sleep apnea). Moreover, these measures were specific, such that no other macro- and microphysiological architecture metrics of sleep demonstrated such sensitivity. Our data support the proposal that objective sleep markers could be part of a set of biomarkers that statistically forecast the longitudinal trajectory of cortical Aβ deposition in the human brain. Sleep may therefore represent a potentially affordable, scalable, repeatable, and non-invasive tool for quantifying of Aβ pathological progression, prior to cognitive symptoms of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Joseph R Winer
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, 101 The City Drive, Orange, CA 92697, USA
| | - Samika Kumar
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA
| | - Mark Reed
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - William J Jagust
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, 132 Barker Hall, Berkeley, CA 94720, USA
| | - Matthew P Walker
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, 132 Barker Hall, Berkeley, CA 94720, USA.
| |
Collapse
|
31
|
Mander BA. Local Sleep and Alzheimer's Disease Pathophysiology. Front Neurosci 2020; 14:525970. [PMID: 33071726 PMCID: PMC7538792 DOI: 10.3389/fnins.2020.525970] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
Even prior to the onset of the prodromal stages of Alzheimer's disease (AD), a constellation of sleep disturbances are apparent. A series of epidemiological studies indicate that multiple forms of these sleep disturbances are associated with increased risk for developing mild cognitive impairment (MCI) and AD, even triggering disease onset at an earlier age. Through the combination of causal manipulation studies in humans and rodents, as well as targeted examination of sleep disturbance with respect to AD biomarkers, mechanisms linking sleep disturbance to AD are beginning to emerge. In this review, we explore recent evidence linking local deficits in brain oscillatory function during sleep with local AD pathological burden and circuit-level dysfunction and degeneration. In short, three deficits in the local expression of sleep oscillations have been identified in relation to AD pathophysiology: (1) frequency-specific frontal deficits in slow wave expression during non-rapid eye movement (NREM) sleep, (2) deficits in parietal sleep spindle expression, and (3) deficits in the quality of electroencephalographic (EEG) desynchrony characteristic of REM sleep. These deficits are noteworthy since they differ from that seen in normal aging, indicating the potential presence of an abnormal aging process. How each of these are associated with β-amyloid (Aβ) and tau pathology, as well as neurodegeneration of circuits sensitive to AD pathophysiology, are examined in the present review, with a focus on the role of dysfunction within fronto-hippocampal and subcortical sleep-wake circuits. It is hypothesized that each of these local sleep deficits arise from distinct network-specific dysfunctions driven by regionally-specific accumulation of AD pathologies, as well as their associated neurodegeneration. Overall, the evolution of these local sleep deficits offer unique windows into the circuit-specific progression of distinct AD pathophysiological processes prior to AD onset, as well as their impact on brain function. This includes the potential erosion of sleep-dependent memory mechanisms, which may contribute to memory decline in AD. This review closes with a discussion of the remaining critical knowledge gaps and implications of this work for future mechanistic studies and studies implementing sleep-based treatment interventions.
Collapse
Affiliation(s)
- Bryce A. Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
32
|
Stylianou M, Zaaimi B, Thomas A, Taylor JP, LeBeau FEN. Early Disruption of Cortical Sleep-Related Oscillations in a Mouse Model of Dementia With Lewy Bodies (DLB) Expressing Human Mutant (A30P) Alpha-Synuclein. Front Neurosci 2020; 14:579867. [PMID: 33041770 PMCID: PMC7527476 DOI: 10.3389/fnins.2020.579867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/25/2020] [Indexed: 01/03/2023] Open
Abstract
Changes in sleep behavior and sleep-related cortical activity have been reported in conditions associated with abnormal alpha-synuclein (α-syn) expression, in particular Parkinson’s disease (PD) and dementia with Lewy bodies (DLB). Notably, changes can occur in patients years before the onset of cognitive decline. Sleep-related network oscillations play a key role in memory function, but how abnormal α-syn impacts the generation of such activity is currently unclear. To determine whether early changes in sleep-related network activity could also be observed, prior to any previously reported cognitive dysfunction, we used mice that over-express human mutant α-syn (A30P). Recordings in vivo were performed under urethane anesthesia in the medial prefrontal cortex (mPFC) and CA1 region of the hippocampus in young male (2.5 – 4 months old) A30P and age-matched wild type (WT) mice. We found that the slow oscillation (SO) < 1 Hz frequency was significantly faster in both the mPFC and hippocampus in A30P mice, and Up-state-associated fast oscillations at beta (20 – 30 Hz) and gamma (30 – 80 Hz) frequencies were delayed relative to the onset of the Up-state. Spindle (8 – 15 Hz) activity in the mPFC was also altered in A30P mice, as spindles were shorter in duration and had reduced density compared to WT. These changes demonstrate that dysregulation of sleep-related oscillations occurs in young A30P mice long before the onset of cognitive dysfunction. Our data suggest that, as seen in patients, changes in sleep-related oscillations are an early consequence of abnormal α-syn aggregation in A30P mice.
Collapse
Affiliation(s)
- Myrto Stylianou
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Boubker Zaaimi
- School of Life & Health Sciences, Aston University, Birmingham, United Kingdom
| | - Alan Thomas
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fiona E N LeBeau
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
33
|
Semyachkina-Glushkovskaya O, Postnov D, Penzel T, Kurths J. Sleep as a Novel Biomarker and a Promising Therapeutic Target for Cerebral Small Vessel Disease: A Review Focusing on Alzheimer's Disease and the Blood-Brain Barrier. Int J Mol Sci 2020; 21:ijms21176293. [PMID: 32878058 PMCID: PMC7504101 DOI: 10.3390/ijms21176293] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/14/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is a leading cause of cognitive decline in elderly people and development of Alzheimer’s disease (AD). Blood–brain barrier (BBB) leakage is a key pathophysiological mechanism of amyloidal CSVD. Sleep plays a crucial role in keeping health of the central nervous system and in resistance to CSVD. The deficit of sleep contributes to accumulation of metabolites and toxins such as beta-amyloid in the brain and can lead to BBB disruption. Currently, sleep is considered as an important informative platform for diagnosis and therapy of AD. However, there are no effective methods for extracting of diagnostic information from sleep characteristics. In this review, we show strong evidence that slow wave activity (SWA) (0–0.5 Hz) during deep sleep reflects glymphatic pathology, the BBB leakage and memory deficit in AD. We also discuss that diagnostic and therapeutic targeting of SWA in AD might lead to be a novel era in effective therapy of AD. Moreover, we demonstrate that SWA can be pioneering non-invasive and bed–side technology for express diagnosis of the BBB permeability. Finally, we review the novel data about the methods of detection and enhancement of SWA that can be biomarker and a promising therapy of amyloidal CSVD and CSVD associated with the BBB disorders.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Department of Human and Animal Physiology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (D.P.); (T.P.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Correspondence: ; Tel.: +7-927-115-5157
| | - Dmitry Postnov
- Department of Human and Animal Physiology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (D.P.); (T.P.); (J.K.)
| | - Thomas Penzel
- Department of Human and Animal Physiology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (D.P.); (T.P.); (J.K.)
- Advanced Sleep Research GmbH, 12489 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Sleep Medicine Center, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Kurths
- Department of Human and Animal Physiology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (D.P.); (T.P.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
34
|
Lee YF, Gerashchenko D, Timofeev I, Bacskai BJ, Kastanenka KV. Slow Wave Sleep Is a Promising Intervention Target for Alzheimer's Disease. Front Neurosci 2020; 14:705. [PMID: 32714142 PMCID: PMC7340158 DOI: 10.3389/fnins.2020.00705] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is the major cause of dementia, characterized by the presence of amyloid-beta plaques and neurofibrillary tau tangles. Plaques and tangles are associated with sleep-wake cycle disruptions, including the disruptions in non-rapid eye movement (NREM) slow wave sleep (SWS). Alzheimer's patients spend less time in NREM sleep and exhibit decreased slow wave activity (SWA). Consistent with the critical role of SWS in memory consolidation, reduced SWA is associated with impaired memory consolidation in AD patients. The aberrant SWA can be modeled in transgenic mouse models of amyloidosis and tauopathy. Animal models exhibited slow wave impairments early in the disease progression, prior to the deposition of amyloid-beta plaques, however, in the presence of abundant oligomeric amyloid-beta. Optogenetic rescue of SWA successfully halted the amyloid accumulation and restored intraneuronal calcium levels in mice. On the other hand, optogenetic acceleration of slow wave frequency exacerbated amyloid deposition and disrupted neuronal calcium homeostasis. In this review, we summarize the evidence and the mechanisms underlying the existence of a positive feedback loop between amyloid/tau pathology and SWA disruptions that lead to further accumulations of amyloid and tau in AD. Moreover, since SWA disruptions occur prior to the plaque deposition, SWA disruptions may provide an early biomarker for AD. Finally, we propose that therapeutic targeting of SWA in AD might lead to an effective treatment for Alzheimer's patients.
Collapse
Affiliation(s)
- Yee Fun Lee
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Dmitry Gerashchenko
- Harvard Medical School/VA Boston Healthcare System, West Roxbury, MA, United States
| | - Igor Timofeev
- Department of Psychiatry and Neuroscience, School of Medicine, Université Laval, Québec, QC, Canada
- CERVO Brain Research Center, Québec, QC, Canada
| | - Brian J. Bacskai
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Ksenia V. Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|