51
|
Nick H, Fenik P, Zhu Y, Veasey S. Hypocretin/orexin influences chronic sleep disruption injury in the hippocampus. Front Aging Neurosci 2022; 14:1025402. [PMID: 36275002 PMCID: PMC9582517 DOI: 10.3389/fnagi.2022.1025402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/20/2022] [Indexed: 01/13/2023] Open
Abstract
Chronic sleep disruption is a risk factor for Alzheimer's disease (AD), yet mechanisms by which sleep disturbances might promote or exacerbate AD are not understood. Short-term sleep loss acutely increases hippocampal amyloid β (Aβ) in wild type (WT) mice and long-term sleep loss increases amyloid plaque in AD transgenic mouse models. Both effects can be influenced by the wake-promoting neuropeptide, hypocretin (HCRT), but whether HCRT influences amyloid accumulation independent of sleep and wake timing modulation remains unclear. Here, we induced chronic fragmentation of sleep (CFS) in WT and HCRT-deficient mice to elicit similar arousal indices, sleep bout lengths and sleep bout numbers in both genotypes. We then examined the roles of HCRT in CFS-induced hippocampal Aβ accumulation and injury. CFS in WT mice resulted in increased Aβ42 in the hippocampus along with loss of cholinergic projections and loss of locus coeruleus neurons. Mice with HCRT deficiency conferred resistance to CFS Aβ42 accumulation and loss of cholinergic projections in the hippocampus yet evidenced similar CFS-induced loss of locus coeruleus neurons. Collectively, the findings demonstrate specific roles for orexin in sleep disruption hippocampal injury. Significance statement Chronic fragmentation of sleep (CFS) occurs in common conditions, including sleep apnea syndromes and chronic pain disorders, yet CFS can induce neural injury. Our results demonstrate that under conditions of sleep fragmentation, hypocretin/orexin is essential for the accumulation of amyloid-β and loss of cholinergic projections in the hippocampus observed in response to CFS yet does not influence locus coeruleus neuron response to CFS.
Collapse
Affiliation(s)
- Henry Nick
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| | - Polina Fenik
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| | - Yan Zhu
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| | - Sigrid Veasey
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
52
|
Gao F, Liu T, Tuo M, Chi S. The role of orexin in Alzheimer disease: From sleep-wake disturbance to therapeutic target. Neurosci Lett 2021; 765:136247. [PMID: 34530113 DOI: 10.1016/j.neulet.2021.136247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/01/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
Accumulating evidence has shown that sleep disturbance is a common symptom in Alzheimer's disease (AD), which is regarded as a modifiable risk factor for AD. Orexin is a key modulator of the sleep-wake cycle and has been found to be dysregulated in AD patients. The increased orexin in cerebrospinal fluid (CSF) is associated with decreased sleep efficiency and REM sleep, as well as cognitive impairment in AD patients. The orexin system has profuse projections to brain regions that are implicated in arousal and cognition and has been found to participate in the progression of AD pathology. Conversely the orexin receptor antagonists are able to consolidate sleep and reduce AD pathology. Therefore, improved understanding of the mechanisms linking orexin system, sleep disturbance and AD could make orexin receptor antagonists a promising target for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Fan Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miao Tuo
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
53
|
Gan J, Chen Z, Han J, Ma L, Liu S, Wang XD, Ji Y. Orexin-A in Patients With Lewy Body Disease: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:765701. [PMID: 34867809 PMCID: PMC8635768 DOI: 10.3389/fendo.2021.765701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Background Abnormal orexin-A levels in cerebrospinal fluid (CSF) have been identified in Alzheimer's disease (AD) and other neurodegenerative diseases. However, few studies have focused on Lewy body disease (LBD) and often with debatable outcomes. Thus, we performed this systematic review and meta-analysis to investigate orexin-A levels in LBD by incorporating data from different studies. Methods We gathered studies comparing orexin-A levels in patients with LBD and controls (including healthy controls and other dementia subtypes). In the initial search, 117 relevant articles were identified. After a selection process, seven studies, conducted in Japan, USA, Spain, Switzerland, France, Italy, and Netherlands, were chosen. Results In total, 179 patients with LBD and 253 controls were included. Patients with LBD had significantly lower mean orexin-A CSF levels when compared with patients with AD [standard mean difference (SMD): -0.35, 95% confidence interval (CI): -0.70 to -0.00, Z = 1.96, P = 0.05], whereas mean orexin-A levels were significantly higher when compared with patients with frontotemporal lobar degeneration (FTLD) (SMD: 0.61, 95% CI: 0.23-0.99, Z = 3.12, P = 0.002). Orexin-A CSF levels in LBD patients were approximately equal to levels in healthy elderly individuals, whereas they were significantly decreased in LBD patients with excessive daytime sleepiness (EDS) (SMD: -0.15, 95% CI: -0.59 to 0.29, Z = 0.67, P = 0.50). Conclusions We showed that orexin-A levels in patients with LBD were not very different from those in normal elderly individuals, whereas they were lower than those in AD patients and higher than those in FTLD patients. The influence of hypersomnia on orexin-A levels should be carefully interpreted. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42021265900.
Collapse
Affiliation(s)
- Jinghuan Gan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhichao Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiuyan Han
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lingyun Ma
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Xiao-Dan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
54
|
Jagirdar R, Fu CH, Park J, Corbett BF, Seibt FM, Beierlein M, Chin J. Restoring activity in the thalamic reticular nucleus improves sleep architecture and reduces Aβ accumulation in mice. Sci Transl Med 2021; 13:eabh4284. [PMID: 34731016 PMCID: PMC8985235 DOI: 10.1126/scitranslmed.abh4284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sleep disruptions promote increases of amyloid β (Aβ) and tau in the brain and increase Alzheimer’s disease (AD) risk, but the precise mechanisms that give rise to sleep disturbances have yet to be defined. The thalamic reticular nucleus (TRN) is essential for sleep maintenance and for the regulation of slow-wave sleep (SWS). We examined the TRN in transgenic mice that express mutant human amyloid precursor protein (APP) and found reduced neuronal activity, increased sleep fragmentation, and decreased SWS time as compared to nontransgenic littermates. Selective activation of the TRN using excitatory DREADDs restored sleep maintenance, increased time in SWS, and reduced amyloid plaque load in both hippocampus and cortex. Our findings suggest that the TRN may play a major role in symptoms associated with AD. Enhancing TRN activity might be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Rohan Jagirdar
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Chia-Hsuan Fu
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Jin Park
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Brian F. Corbett
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Frederik M. Seibt
- Department of Neurobiology and Anatomy, McGovern Medical School at UTHealth, Houston, TX 77030
| | - Michael Beierlein
- Department of Neurobiology and Anatomy, McGovern Medical School at UTHealth, Houston, TX 77030
| | - Jeannie Chin
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
55
|
Matsumoto S, Tsunematsu T. Association between Sleep, Alzheimer's, and Parkinson's Disease. BIOLOGY 2021; 10:1127. [PMID: 34827122 PMCID: PMC8614785 DOI: 10.3390/biology10111127] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 01/09/2023]
Abstract
The majority of neurodegenerative diseases are pathologically associated with protein misfolding and aggregation. Alzheimer's disease (AD) is a type of dementia that slowly affects memory and cognitive function, and is characterized by the aggregation of the β-amyloid protein and tau neurofibrillary tangles in the brain. Parkinson's disease (PD) is a movement disorder typically resulting in rigidity and tremor, which is pathologically linked to the aggregation of α-synuclein, particularly in dopaminergic neurons in the midbrain. Sleep disorders commonly occur in AD and PD patients, and it can precede the onset of these diseases. For example, cognitively normal older individuals who have highly fragmented sleep had a 1.5-fold increased risk of subsequently developing AD. This suggests that sleep abnormalities may be a potential biomarker of these diseases. In this review, we describe the alterations of sleep in AD and PD, and discuss their potential in the early diagnosis of these diseases. We further discuss whether sleep disturbance could be a target for the treatment of these diseases.
Collapse
Affiliation(s)
- Sumire Matsumoto
- Advanced Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Tomomi Tsunematsu
- Advanced Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan;
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi 332-0012, Japan
| |
Collapse
|
56
|
Kuang H, Zhu YG, Zhou ZF, Yang MW, Hong FF, Yang SL. Sleep disorders in Alzheimer's disease: the predictive roles and potential mechanisms. Neural Regen Res 2021; 16:1965-1972. [PMID: 33642368 PMCID: PMC8343328 DOI: 10.4103/1673-5374.308071] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/12/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Sleep disorders are common in patients with Alzheimer's disease, and can even occur in patients with amnestic mild cognitive impairment, which appears before Alzheimer's disease. Sleep disorders further impair cognitive function and accelerate the accumulation of amyloid-β and tau in patients with Alzheimer's disease. At present, sleep disorders are considered as a risk factor for, and may be a predictor of, Alzheimer's disease development. Given that sleep disorders are encountered in other types of dementia and psychiatric conditions, sleep-related biomarkers to predict Alzheimer's disease need to have high specificity and sensitivity. Here, we summarize the major Alzheimer's disease-specific sleep changes, including abnormal non-rapid eye movement sleep, sleep fragmentation, and sleep-disordered breathing, and describe their ability to predict the onset of Alzheimer's disease at its earliest stages. Understanding the mechanisms underlying these sleep changes is also crucial if we are to clarify the role of sleep in Alzheimer's disease. This paper therefore explores some potential mechanisms that may contribute to sleep disorders, including dysregulation of the orexinergic, glutamatergic, and γ-aminobutyric acid systems and the circadian rhythm, together with amyloid-β accumulation. This review could provide a theoretical basis for the development of drugs to treat Alzheimer's disease based on sleep disorders in future work.
Collapse
Affiliation(s)
- Huang Kuang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yu-Ge Zhu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhi-Feng Zhou
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Mei-Wen Yang
- Department of Nurse, Nanchang University Hospital, Nanchang, Jiangxi Province, China
| | - Fen-Fang Hong
- Department of Experimental Teaching Center, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
57
|
Wang Q, Cao F, Wu Y. Orexinergic System in Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:713201. [PMID: 34483883 PMCID: PMC8416170 DOI: 10.3389/fnagi.2021.713201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023] Open
Abstract
Orexinergic system consisting of orexins and orexin receptors plays an essential role in regulating sleep–wake states, whereas sleep disruption is a common symptom of a number of neurodegenerative diseases. Emerging evidence reveals that the orexinergic system is disturbed in various neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and multiple sclerosis (MS), whereas the dysregulation of orexins and/or orexin receptors contributes to the pathogenesis of these diseases. In this review, we summarized advanced knowledge of the orexinergic system and its role in sleep, and reviewed the dysregulation of the orexinergic system and its role in the pathogenesis of AD, PD, HD, and MS. Moreover, the therapeutic potential of targeting the orexinergic system for the treatment of these diseases was discussed.
Collapse
Affiliation(s)
- Qinqin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment & Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Fei Cao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment & Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Yili Wu
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory, Wenzhou, China
| |
Collapse
|
58
|
Hopkins K, Mukherjee S, Ponce D, Mangum J, Jacobson LH, Hoyer D. Development of a LC-ESI-MRM method for the absolute quantification of orexin A in the CSF of individual mice. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2021.100102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
59
|
Nur İH, Keleş H, Ünlükal N, Solmaz M, Erdoğan E, Pérez W. A new definition about the relationship of intercellular fluid in the brain with the mandibular and parotid lymph nodes. Microsc Res Tech 2021; 85:220-232. [PMID: 34369631 DOI: 10.1002/jemt.23898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 11/12/2022]
Abstract
This study was carried out to reveal the relationship of the brain with both the mandibular lymph node (MLN) and parotid lymph node (PLN) by the hyperspectral fluorescence imaging techniques of Qdot 800 (QD) nanoparticles using in vivo. This relationship of the brain with both lymph nodes offers the preliminary morphological definition of lymphatic drainage. QD was injected into the left parietal brain lobe of each rat at a depth of 2.50 mm. In 65% of the rats that were imaged in vivo, signals were received first from the right MLN and PLN, and then from the left MLN and PLN. In contrast, in two female rats, the first signal was received from the right PLN. There was no difference between the female and male rats overall. The most noteworthy finding of this study was that the tracer injected into the left parietal lobe reached the right mandibular and parotid lymph nodules earlier. This result indicates a different and unknown pathway in the brain that communicates with the lymph nodes. Moreover, this study shows that these lymph nodes pathways can be used in the treatment of diseases such as brain trauma, cerebral edema, and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- İsmail Hakkı Nur
- Department of Anatomy, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Hacı Keleş
- Department of Anatomy, Faculty of Medicine, Nigde Omer Halisdemir University, Nigde, Turkey
| | - Nejat Ünlükal
- Department of Histology and Embryology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Merve Solmaz
- Department of Histology and Embryology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ender Erdoğan
- Department of Histology and Embryology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - William Pérez
- Unidad de Anatomia, Facultad de Veterinaria, Universidad de la Republica, Montevideo, Uruguay
| |
Collapse
|
60
|
Cerebrospinal fluid orexin in Alzheimer's disease: a systematic review and meta-analysis. Sleep Med 2021; 85:230-238. [PMID: 34364094 DOI: 10.1016/j.sleep.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/13/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVE/BACKGROUND A growing body of evidence suggests that sleep and Alzheimer's disease (AD) have a bi-directional relationship. Emerging research also suggests that orexin, a key neurotransmitter involved in sleep-wake regulation, may be altered in persons with AD, however results have not been consistent across prior studies. This investigation was conducted to both evaluate the aggregate literature to minimize the risk of bias and identify potential factors associated with heterogeneity across studies. METHODS Systematic review identified relevant investigations that compared cerebrospinal fluid orexin in persons with AD and controls. Meta-analysis (random effects model) compared effect size (Hedge's g) for orexin between AD and controls. Meta-regression was additionally performed for key variables of interest to evaluate potential causes of heterogeneity among studies. RESULTS 17 studies were identified that met inclusion/exclusion criteria. Evidence of publication bias was not identified. Non-significant increases in orexin were observed in AD relative to controls, with moderate to large heterogeneity among studies (Hedge's g = 0.20, p = 0.136, I2 = 72.6%). Meta-regression demonstrated both year of publication (β = 0.055, p = 0.020) and effect size for phosphorylated tau in AD versus controls (β = 0.417, p = 0.031) were associated with differences in orexin. CONCLUSIONS Results do not support broad differences in orexin in AD compared to controls, however, evolving diagnostic criteria may have affected findings across studies. Future research that examines orexin in AD over the longitudinal course of the disorder and explores potential links between phosphorylated tau and orexin are indicated.
Collapse
|
61
|
Kim E, Nohara K, Wirianto M, Escobedo G, Lim JY, Morales R, Yoo SH, Chen Z. Effects of the Clock Modulator Nobiletin on Circadian Rhythms and Pathophysiology in Female Mice of an Alzheimer's Disease Model. Biomolecules 2021; 11:biom11071004. [PMID: 34356628 PMCID: PMC8301787 DOI: 10.3390/biom11071004] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder and the most common cause of dementia. Various pathogenic mechanisms have been proposed to contribute to disease progression, and recent research provided evidence linking dysregulated circadian rhythms/sleep and energy metabolism with AD. Previously, we found that the natural compound Nobiletin (NOB) can directly activate circadian cellular oscillators to promote metabolic health in disease models and healthy aging in naturally aged mice. In the current study, using the amyloid-β AD model APP/PS1, we investigated circadian, metabolic and amyloid characteristics of female mice and the effects of NOB. Female APP/PS1 mice showed reduced sleep bout duration, and NOB treatment exhibited a trend to improve it. While glucose tolerance was unchanged, female APP/PS1 mice displayed exaggerated oxygen consumption and CO2 production, which was mitigated by NOB. Likewise, cold tolerance in APP/PS1 was impaired relative to WT, and interestingly was markedly enhanced in NOB-treated APP/PS1 mice. Although circadian behavioral rhythms were largely unchanged, real-time qPCR analysis revealed altered expression of several core clock genes by NOB in the cerebral cortex, notably Bmal1, Npas2, and Rora. Moreover, NOB was also able to activate various clock-controlled metabolic genes involved in insulin signaling and mitochondrial function, including Igf1, Glut1, Insr, Irs1, Ucp2, and Ucp4. Finally, we observed that NOB attenuated the expression of several AD related genes including App, Bace1, and ApoE, reduced APP protein levels, and strongly ameliorated Aβ pathology in the cortex. Collectively, these results reveal novel genotype differences and importantly beneficial effects of a natural clock-enhancing compound in biological rhythms and related pathophysiology, suggesting the circadian clock as a modifiable target for AD.
Collapse
Affiliation(s)
- Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Kazunari Nohara
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Gabriel Escobedo
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (G.E.J.); (R.M.)
| | - Ji Ye Lim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (G.E.J.); (R.M.)
- Centro Integrativo de Biologia y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
- Correspondence:
| |
Collapse
|
62
|
Fronczek R, Schinkelshoek M, Shan L, Lammers GJ. The orexin/hypocretin system in neuropsychiatric disorders: Relation to signs and symptoms. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:343-358. [PMID: 34225940 DOI: 10.1016/b978-0-12-820107-7.00021-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hypocretin-1 and 2 (or orexin A and B) are neuropeptides exclusively produced by a group of neurons in the lateral and dorsomedial hypothalamus that project throughout the brain. In accordance with this, the two different hypocretin receptors are also found throughout the brain. The hypocretin system is mainly involved in sleep-wake regulation, but also in reward mechanisms, food intake and metabolism, autonomic regulation including thermoregulation, and pain. The disorder most strongly linked to the hypocretin system is the primary sleep disorder narcolepsy type 1 caused by a lack of hypocretin signaling, which is most likely due to an autoimmune process targeting the hypocretin-producing neurons. However, the hypocretin system may also be affected, but to a lesser extent and less specifically, in various other neurological disorders. Examples are neurodegenerative diseases such as Alzheimer's, Huntington's and Parkinson's disease, immune-mediated disorders such as multiple sclerosis, neuromyelitis optica, and anti-Ma2 encephalitis, and genetic disorders such as type 1 diabetus mellitus and Prader-Willi Syndrome. A partial hypocretin deficiency may contribute to the sleep features of these disorders.
Collapse
Affiliation(s)
- Rolf Fronczek
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands.
| | - Mink Schinkelshoek
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands
| | - Ling Shan
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands; Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Gert Jan Lammers
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands
| |
Collapse
|
63
|
Patel S, Howard D, Chowdhury N, Derieux C, Wellslager B, Yilmaz Ö, French L. Characterization of Human Genes Modulated by Porphyromonas gingivalis Highlights the Ribosome, Hypothalamus, and Cholinergic Neurons. Front Immunol 2021; 12:646259. [PMID: 34194426 PMCID: PMC8236716 DOI: 10.3389/fimmu.2021.646259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Porphyromonas gingivalis, a bacterium associated with periodontal disease, is a suspected cause of Alzheimer's disease. This bacterium is reliant on gingipain proteases, which cleave host proteins after arginine and lysine residues. To characterize gingipain susceptibility, we performed enrichment analyses of arginine and lysine proportion proteome-wide. Genes differentially expressed in brain samples with detected P. gingivalis reads were also examined. Genes from these analyses were tested for functional enrichment and specific neuroanatomical expression patterns. Proteins in the SRP-dependent cotranslational protein targeting to membrane pathway were enriched for these residues and previously associated with periodontal and Alzheimer's disease. These ribosomal genes are up-regulated in prefrontal cortex samples with detected P. gingivalis sequences. Other differentially expressed genes have been previously associated with dementia (ITM2B, MAPT, ZNF267, and DHX37). For an anatomical perspective, we characterized the expression of the P. gingivalis associated genes in the mouse and human brain. This analysis highlighted the hypothalamus, cholinergic neurons, and the basal forebrain. Our results suggest markers of neural P. gingivalis infection and link the cholinergic and gingipain hypotheses of Alzheimer's disease.
Collapse
Affiliation(s)
- Sejal Patel
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Derek Howard
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nityananda Chowdhury
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Casey Derieux
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Bridgette Wellslager
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Özlem Yilmaz
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Leon French
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute for Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
64
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
65
|
Cordone S, Scarpelli S, Alfonsi V, De Gennaro L, Gorgoni M. Sleep-Based Interventions in Alzheimer's Disease: Promising Approaches from Prevention to Treatment along the Disease Trajectory. Pharmaceuticals (Basel) 2021; 14:ph14040383. [PMID: 33921870 PMCID: PMC8073746 DOI: 10.3390/ph14040383] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
The multifactorial nature of Alzheimer’s disease (AD) has led scientific researchers to focus on the modifiable and treatable risk factors of AD. Sleep fits into this context, given the bidirectional relationship with AD confirmed by several studies over the last years. Sleep disorders appear at an early stage of AD and continue throughout the entire course of the pathology. Specifically, sleep abnormalities, such as more fragmented sleep, increase in time of awakenings, worsening of sleep quality and primary sleep disorders raise with the severity and progression of AD. Intervening on sleep, therefore, means acting both with prevention strategies in the pre-clinical phase and with treatments during the course of the disease. This review explores sleep disturbances in the different stages of AD, starting from the pre-clinical stage. Particular attention is given to the empirical evidence investigating obstructive sleep apnea (OSA) disorder and the mechanisms overlapping and sharing with AD. Next, we discuss sleep-based intervention strategies in the healthy elderly population, mild cognitive impairment (MCI) and AD patients. We mention interventions related to behavioral strategies, combination therapies, and bright light therapy, leaving extensive space for new and raising evidence on continuous positive air pressure (CPAP) treatment effectiveness. Finally, we clarify the role of NREM sleep across the AD trajectory and consider the most recent studies based on the promising results of NREM sleep enhancement, which use innovative experimental designs and techniques.
Collapse
Affiliation(s)
- Susanna Cordone
- UniCamillus, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Serena Scarpelli
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| | | | - Luigi De Gennaro
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
- Correspondence:
| | - Maurizio Gorgoni
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| |
Collapse
|
66
|
Trotti LM, Bliwise DL, Keating GL, Rye DB, Hu WT. Cerebrospinal Fluid Hypocretin and Nightmares in Dementia Syndromes. Dement Geriatr Cogn Dis Extra 2021; 11:19-25. [PMID: 33790936 PMCID: PMC7989783 DOI: 10.1159/000509585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/08/2023] Open
Abstract
Background/Aims Hypocretin promotes wakefulness and modulates REM sleep. Alterations in the hypocretin system are increasingly implicated in dementia. We evaluated relationships among hypocretin, dementia biomarkers, and sleep symptoms in elderly participants, most of whom had dementia. Methods One-hundred twenty-six adults (mean age 66.2 ± 8.4 years) were recruited from the Emory Cognitive Clinic. Diagnoses were Alzheimer disease (AD; n = 60), frontotemporal dementia (FTD; n = 21), and dementia with Lewy bodies (DLB; n = 20). We also included cognitively normal controls (n = 25). Participants and/or caregivers completed sleep questionnaires and lumbar puncture was performed for cerebrospinal fluid (CSF) assessments. Results Except for sleepiness (worst in DLB) and nocturia (worse in DLB and FTD) sleep symptoms did not differ by diagnosis. CSF hypocretin concentrations were available for 87 participants and normal in 70, intermediate in 16, and low in 1. Hypocretin levels did not differ by diagnosis. Hypocretin levels correlated with CSF total τ levels only in men (r = 0.34; p = 0.02). Lower hypocretin levels were related to frequency of nightmares (203.9 ± 29.8 pg/mL in those with frequent nightmares vs. 240.4 ± 46.1 pg/mL in those without; p = 0.05) and vivid dreams (209.1 ± 28.3 vs. 239.5 ± 47.8 pg/mL; p = 0.014). Cholinesterase inhibitor use was not associated with nightmares or vivid dreaming. Conclusion Hypocretin levels did not distinguish between dementia syndromes. Disturbing dreams in dementia patients may be related to lower hypocretin concentrations in CSF.
Collapse
Affiliation(s)
- Lynn Marie Trotti
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Sleep Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Donald L Bliwise
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Sleep Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Glenda L Keating
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David B Rye
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Sleep Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
67
|
Delic V, Ratliff WA, Citron BA. Sleep Deprivation, a Link Between Post-Traumatic Stress Disorder and Alzheimer's Disease. J Alzheimers Dis 2021; 79:1443-1449. [PMID: 33459652 DOI: 10.3233/jad-201378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
An estimated 5 million Americans are living with Alzheimer's disease (AD), and there is also a significant impact on caregivers, with an additional 16 million Americans providing unpaid care for individuals with AD and other dementias. These numbers are projected to increase in the coming years. While AD is still without a cure, continued research efforts have led to better understanding of pathology and potential risk factors that could be exploited to slow disease progression. A bidirectional relationship between sleep deprivation and AD has been suggested and is well supported by both human and animal studies. Even brief episodes of inadequate sleep have been shown to cause an increase in amyloidβ and tau proteins, both well-established contributors toAD pathology. Sleep deprivation is also the most common consequence of post-traumatic stress disorder (PTSD). Patients with PTSD frequently present with sleep disturbances and also develop dementia at twice the rate of the general population accounting for a disproportionate representation of AD among U.S. Veterans. The goal of this review is to highlight the relationship triad between sleep deprivation, AD, and PTSD as well as their impact on molecular mechanisms driving AD pathology.
Collapse
Affiliation(s)
- Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development, East Orange, NJ, USA
| | - Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development, Bay Pines, FL, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development, East Orange, NJ, USA.,Department of Pharmacology, Physiology, & Neuroscience ,Rutgers-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
68
|
Kent BA, Feldman HH, Nygaard HB. Sleep and its regulation: An emerging pathogenic and treatment frontier in Alzheimer's disease. Prog Neurobiol 2021; 197:101902. [PMID: 32877742 PMCID: PMC7855222 DOI: 10.1016/j.pneurobio.2020.101902] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/19/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
A majority of patients with Alzheimer's disease (AD) experience some form of sleep disruption, including nocturnal sleep fragmentation, increased daytime napping, decreased slow-wave sleep (SWS, stage N3), and decreased rapid-eye-movement sleep (REM). Clinical studies are investigating whether such sleep disturbances are a consequence of the underlying disease, and whether they also contribute to the clinical and pathological manifestations of AD. Emerging research has provided a direct link between several of these sleep disruptions and AD pathophysiology, suggesting that treating sleep disorders in this population may target basic mechanisms of the disease. Here, we provide a comprehensive review of sleep disturbances associated with the spectrum of AD, ranging from the preclinical stages through dementia. We discuss how sleep interacts with AD pathophysiology and, critically, whether sleep impairments can be targeted to modify the disease course in a subgroup of affected AD patients. Ultimately, larger studies that fully utilize new diagnostic and experimental tools will be required to better define the most relevant sleep disturbance to target in AD, the interventions that best modulate this target symptom, and whether successful early intervention can modify AD risk and prevent dementia.
Collapse
Affiliation(s)
- Brianne A Kent
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Division of Neurology, University of British Columbia, Vancouver, Canada.
| | - Howard H Feldman
- Division of Neurology, University of British Columbia, Vancouver, Canada; Department of Neurosciences, University of California, San Diego, La Jolla, USA
| | - Haakon B Nygaard
- Division of Neurology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
69
|
Sleep disorders and late-onset epilepsy of unknown origin: Understanding new trajectories to brain amyloidopathy. Mech Ageing Dev 2021; 194:111434. [PMID: 33444630 DOI: 10.1016/j.mad.2021.111434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/16/2022]
Abstract
The intertwining between epilepsy, sleep disorders and beta amyloid pathology has been progressively highlighted, as early identification and stratification of patients at high risk of cognitive decline is the need of the hour. Modification of the sleep-wake activity, such as sleep impairment or excessive daytime sleepiness, can critically affect cerebral beta amyloid levels. Both mice models and human studies have demonstrated a substantial increase in the burden of beta amyloid pathology after sleep-deprivation, with potential negative effects partially restored by sleep recovery. The accumulation of beta amyloid has been shown to be an early event in the course of Alzheimer's disease dementia. Beta amyloid accumulation has been linked to epileptic seizures epileptic seizures, with beta amyloid being itself pro-epileptogenic in mice models already at oligomeric stage, well before plaque deposition. Further supporting a potential relationship between beta amyloid and epilepsy: i) seizures happen in 1 out of oofut 10 patients with Alzheimer's disease in the prodromal stage, ii) epileptic activity accelerates cognitive decline in Alzheimer's disease, iii) people with late-onset epilepsy present a critically high risk of developing dementia. In this Review we highlight the role of beta amyloid as a potential shared mechanisms between sleep disorders, late-onset epilepsy, and cognitive decline.
Collapse
|
70
|
Harris SS, Schwerd-Kleine T, Lee BI, Busche MA. The Reciprocal Interaction Between Sleep and Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:169-188. [PMID: 34773232 DOI: 10.1007/978-3-030-81147-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
It is becoming increasingly recognized that patients with a variety of neurodegenerative diseases exhibit disordered sleep/wake patterns. While sleep impairments have typically been thought of as sequelae of underlying neurodegenerative processes in sleep-wake cycle regulating brain regions, including the brainstem, hypothalamus, and basal forebrain, emerging evidence now indicates that sleep deficits may also act as pathophysiological drivers of brain-wide disease progression. Specifically, recent work has indicated that impaired sleep can impact on neuronal activity, brain clearance mechanisms, pathological build-up of proteins, and inflammation. Altered sleep patterns may therefore be novel (potentially reversible) dynamic functional markers of proteinopathies and modifiable targets for early therapeutic intervention using non-invasive stimulation and behavioral techniques. Here we highlight research describing a potentially reciprocal interaction between impaired sleep and circadian patterns and the accumulation of pathological signs and features in Alzheimer's disease, the most prevalent neurodegenerative disease in the elderly.
Collapse
Affiliation(s)
| | | | - Byung Il Lee
- UK Dementia Research Institute at UCL, London, UK
| | | |
Collapse
|
71
|
Djonlagic I, Mariani S, Fitzpatrick AL, Van Der Klei VMGTH, Johnson DA, Wood AC, Seeman T, Nguyen HT, Prerau MJ, Luchsinger JA, Dzierzewski JM, Rapp SR, Tranah GJ, Yaffe K, Burdick KE, Stone KL, Redline S, Purcell SM. Macro and micro sleep architecture and cognitive performance in older adults. Nat Hum Behav 2021; 5:123-145. [PMID: 33199858 PMCID: PMC9881675 DOI: 10.1038/s41562-020-00964-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 09/15/2020] [Indexed: 01/31/2023]
Abstract
We sought to determine which facets of sleep neurophysiology were most strongly linked to cognitive performance in 3,819 older adults from two independent cohorts, using whole-night electroencephalography. From over 150 objective sleep metrics, we identified 23 that predicted cognitive performance, and processing speed in particular, with effects that were broadly independent of gross changes in sleep quality and quantity. These metrics included rapid eye movement duration, features of the electroencephalography power spectra derived from multivariate analysis, and spindle and slow oscillation morphology and coupling. These metrics were further embedded within broader associative networks linking sleep with aging and cardiometabolic disease: individuals who, compared with similarly aged peers, had better cognitive performance tended to have profiles of sleep metrics more often seen in younger, healthier individuals. Taken together, our results point to multiple facets of sleep neurophysiology that track coherently with underlying, age-dependent determinants of cognitive and physical health trajectories in older adults.
Collapse
Affiliation(s)
- Ina Djonlagic
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sara Mariani
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | - Alexis C Wood
- USDA/ARS Children's Nutrition Center, Baylor College of Medicine, Houston, TX, USA
| | - Teresa Seeman
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Ha T Nguyen
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Michael J Prerau
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Stephen R Rapp
- Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Kristine Yaffe
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Katherine E Burdick
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Susan Redline
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Shaun M Purcell
- Harvard Medical School, Boston, MA, USA.
- Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
72
|
Wunderlin M, Züst MA, Fehér KD, Klöppel S, Nissen C. The role of slow wave sleep in the development of dementia and its potential for preventative interventions. Psychiatry Res Neuroimaging 2020; 306:111178. [PMID: 32919869 DOI: 10.1016/j.pscychresns.2020.111178] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 07/02/2020] [Accepted: 09/03/2020] [Indexed: 12/17/2022]
Abstract
The increasing incidence rate of dementia underlines the necessity to identify early biomarkers of imminent cognitive decline. Recent findings suggest that cognitive decline and the pathophysiology of Alzheimer's disease are closely linked to disruptions in slow wave sleep (SWS) - the deepest sleep stage. SWS is essential for memory functions and displays a potentially causal and bidirectional link to the accumulation of amyloid beta deposition. Accordingly, improving SWS in older adults - especially when at risk for dementia - might slow down the rate of cognitive decline. Recent work suggests that SWS can be improved by specifically targeting the electrophysiological peaks of the slow waves with acoustic stimulation. In older adults, this approach is still fairly new and accompanied by challenges posed by the specific complexity of their sleep physiology, like lower amplitude slow waves and fragmented sleep architecture. We suggest an approach that tackles these issues and attempts to re-instate a sleep physiology that resembles a younger, healthier brain. With enough SWS of high quality, metabolic clearance and memory functions could benefit and help slowing the process of cognitive aging. Ultimately, acoustic stimulation to enhance SWS could serve as a cost-effective, non-invasive tool to combat cognitive decline.
Collapse
Affiliation(s)
- Marina Wunderlin
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Switzerland, Bolligenstrasse 111, 3000 Bern 60, Switzerland.
| | - Marc Alain Züst
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Switzerland, Bolligenstrasse 111, 3000 Bern 60, Switzerland
| | - Kristoffer Daniel Fehér
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Switzerland, Bolligenstrasse 111, 3000 Bern 60, Switzerland
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Switzerland, Bolligenstrasse 111, 3000 Bern 60, Switzerland
| | - Christoph Nissen
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Switzerland, Bolligenstrasse 111, 3000 Bern 60, Switzerland
| |
Collapse
|
73
|
Noorani AA, Yamashita H, Gao Y, Islam S, Sun Y, Nakamura T, Enomoto H, Zou K, Michikawa M. High temperature promotes amyloid β-protein production and γ-secretase complex formation via Hsp90. J Biol Chem 2020; 295:18010-18022. [PMID: 33067321 PMCID: PMC7939388 DOI: 10.1074/jbc.ra120.013845] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by neuronal loss and accumulation of β-amyloid-protein (Aβ) in the brain parenchyma. Sleep impairment is associated with AD and affects about 25-40% of patients in the mild-to-moderate stages of the disease. Sleep deprivation leads to increased Aβ production; however, its mechanism remains largely unknown. We hypothesized that the increase in core body temperature induced by sleep deprivation may promote Aβ production. Here, we report temperature-dependent regulation of Aβ production. We found that an increase in temperature, from 37 °C to 39 °C, significantly increased Aβ production in amyloid precursor protein-overexpressing cells. We also found that high temperature (39 °C) significantly increased the expression levels of heat shock protein 90 (Hsp90) and the C-terminal fragment of presenilin 1 (PS1-CTF) and promoted γ-secretase complex formation. Interestingly, Hsp90 was associated with the components of the premature γ-secretase complex, anterior pharynx-defective-1 (APH-1), and nicastrin (NCT) but was not associated with PS1-CTF or presenilin enhancer-2. Hsp90 knockdown abolished the increased level of Aβ production and the increased formation of the γ-secretase complex at high temperature in culture. Furthermore, with in vivo experiments, we observed increases in the levels of Hsp90, PS1-CTF, NCT, and the γ-secretase complex in the cortex of mice housed at higher room temperature (30 °C) compared with those housed at standard room temperature (23 °C). Our results suggest that high temperature regulates Aβ production by modulating γ-secretase complex formation through the binding of Hsp90 to NCT/APH-1.
Collapse
Affiliation(s)
- Arshad Ali Noorani
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Yuan Gao
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Sadequl Islam
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Yang Sun
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Tomohisa Nakamura
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Hiroyuki Enomoto
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Kun Zou
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
74
|
Berteotti C, Liguori C, Pace M. Dysregulation of the orexin/hypocretin system is not limited to narcolepsy but has far-reaching implications for neurological disorders. Eur J Neurosci 2020; 53:1136-1154. [PMID: 33290595 DOI: 10.1111/ejn.15077] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Neuropeptides orexin A and B (OX-A/B, also called hypocretin 1 and 2) are released selectively by a population of neurons which projects widely into the entire central nervous system but is localized in a restricted area of the tuberal region of the hypothalamus, caudal to the paraventricular nucleus. The OX system prominently targets brain structures involved in the regulation of wake-sleep state switching, and also orchestrates multiple physiological functions. The degeneration and dysregulation of the OX system promotes narcoleptic phenotypes both in humans and animals. Hence, this review begins with the already proven involvement of OX in narcolepsy, but it mainly discusses the new pre-clinical and clinical insights of the role of OX in three major neurological disorders characterized by sleep impairment which have been recently associated with OX dysfunction, such as Alzheimer's disease, stroke and Prader Willi syndrome, and have been emerged over the past 10 years to be strongly associated with the OX dysfunction and should be more considered in the future. In the light of the impairment of the OX system in these neurological disorders, it is conceivable to speculate that the integrity of the OX system is necessary for a healthy functioning body.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marta Pace
- Genetics and Epigenetics of Behaviour Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
75
|
Liguori C, Maestri M, Spanetta M, Placidi F, Bonanni E, Mercuri NB, Guarnieri B. Sleep-disordered breathing and the risk of Alzheimer's disease. Sleep Med Rev 2020; 55:101375. [PMID: 33022476 DOI: 10.1016/j.smrv.2020.101375] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/15/2022]
Abstract
Sleep-disordered breathing is highly prevalent in the elderly population. Obstructive sleep apnea (OSA) represents the most common sleep disorder among the adult and elderly population. Recently, OSA diagnosis has been associated with an increased risk of developing cognitive decline and dementia, including vascular dementia and Alzheimer's disease (AD). Subsequently, there have been studies on AD biomarkers investigating cerebrospinal fluid, blood, neuroimaging, and nuclear medicine biomarkers in patients with OSA. Furthermore, studies have attempted to assess the possible effects of continuous positive airway pressure (CPAP) treatment on the cognitive trajectory and AD biomarkers in patients with OSA. This review summarizes the findings of studies on each AD biomarker (cognitive, biofluid, neuroimaging, and nuclear medicine imaging) in patients with OSA, also accounting for the related effects of CPAP treatment. In addition, the hypothetical model connecting OSA to AD in a bi-directional interplay is analyzed. Finally, the sex-based differences in prevalence and clinical symptoms of OSA between men and women have been investigated in relation to AD risk. Further studies investigating AD biomarkers changes in patients with OSA and the effect of CPAP treatment should be auspicated in future for identifying strategies to prevent the development of AD.
Collapse
Affiliation(s)
- Claudio Liguori
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Neurology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Michelangelo Maestri
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Matteo Spanetta
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Fabio Placidi
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Neurology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Enrica Bonanni
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Nicola B Mercuri
- Neurology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Santa Lucia Foundation, Rome, Italy
| | - Biancamaria Guarnieri
- Center of Sleep Medicine, Department of Neurology, Villa Serena Hospital, Città S. Angelo, Pescara, Italy; Villa Serena Foundation for the Research, Città S. Angelo, Pescara, Italy
| |
Collapse
|
76
|
Impact of circadian and diurnal rhythms on cellular metabolic function and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:393-412. [PMID: 32739012 DOI: 10.1016/bs.irn.2020.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The 24-h rotational period of the earth has driven evolution of biological systems that serve to synchronize organismal physiology and behavior to this predictable environmental event. In mammals, the circadian (circa, "about" and dia, "a day") clock keeps 24-h time at the organismal and cellular level, optimizing biological function for a given time of day. The most obvious circadian output is the sleep-wake cycle, though countless bodily functions, ranging from hormone levels to cognitive function, are influenced by the circadian clock. Here we discuss the regulation of metabolic pathways by the circadian clock, discuss the evidence implicating circadian and sleep disruption in neurodegenerative diseases, and suggest some possible connections between the clock, metabolism, and neurodegenerative disease.
Collapse
|
77
|
Romanella SM, Roe D, Paciorek R, Cappon D, Ruffini G, Menardi A, Rossi A, Rossi S, Santarnecchi E. Sleep, Noninvasive Brain Stimulation, and the Aging Brain: Challenges and Opportunities. Ageing Res Rev 2020; 61:101067. [PMID: 32380212 PMCID: PMC8363192 DOI: 10.1016/j.arr.2020.101067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/26/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023]
Abstract
As we age, sleep patterns undergo severe modifications of their micro and macrostructure, with an overall lighter and more fragmented sleep structure. In general, interventions targeting sleep represent an excellent opportunity not only to maintain life quality in the healthy aging population, but also to enhance cognitive performance and, when pathology arises, to potentially prevent/slow down conversion from e.g. Mild Cognitive Impairment (MCI) to Alzheimer's Disease (AD). Sleep abnormalities are, in fact, one of the earliest recognizable biomarkers of dementia, being also partially responsible for a cascade of cortical events that worsen dementia pathophysiology, including impaired clearance systems leading to build-up of extracellular amyloid-β (Aβ) peptide and intracellular hyperphosphorylated tau proteins. In this context, Noninvasive Brain Stimulation (NiBS) techniques, such as transcranial electrical stimulation (tES) and transcranial magnetic stimulation (TMS), may help investigate the neural substrates of sleep, identify sleep-related pathology biomarkers, and ultimately help patients and healthy elderly individuals to restore sleep quality and cognitive performance. However, brain stimulation applications during sleep have so far not been fully investigated in healthy elderly cohorts, nor tested in AD patients or other related dementias. The manuscript discusses the role of sleep in normal and pathological aging, reviewing available evidence of NiBS applications during both wakefulness and sleep in healthy elderly individuals as well as in MCI/AD patients. Rationale and details for potential future brain stimulation studies targeting sleep alterations in the aging brain are discussed, including enhancement of cognitive performance, overall quality of life as well as protein clearance.
Collapse
Affiliation(s)
- Sara M Romanella
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy
| | - Daniel Roe
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rachel Paciorek
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Davide Cappon
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Arianna Menardi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Padova Neuroscience Center, Department of Neuroscience, University of Padova, Padova, Italy
| | - Alessandro Rossi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Human Physiology Section, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Simone Rossi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Human Physiology Section, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy; Siena Robotics and Systems Lab (SIRS-Lab), Engineering and Mathematics Department, University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
78
|
It's complicated: The relationship between sleep and Alzheimer's disease in humans. Neurobiol Dis 2020; 144:105031. [PMID: 32738506 DOI: 10.1016/j.nbd.2020.105031] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/21/2020] [Accepted: 07/26/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an asymptomatic period of amyloid-β (Aβ) deposition as insoluble extracellular plaque, intracellular tau aggregation, neuronal and synaptic loss, and subsequent cognitive dysfunction and dementia. A growing public health crisis, the worldwide prevalence of AD is expected to rise from 46.8 million individuals affected in 2015 to 131.5 million in 2050. Sleep disturbances have been associated with increased future risk of AD. A bi-directional relationship is hypothesized between sleep and AD with sleep disturbances as either markers for AD pathology and/or a mechanism mediating increased risk of AD. In this review, the evidence in humans supporting this complex relationship between sleep and AD will be discussed as well as the therapeutic potential and challenges of treating sleep disturbances to prevent or delay the onset of AD.
Collapse
|
79
|
Robust light-dark patterns and reduced amyloid load in an Alzheimer's disease transgenic mouse model. Sci Rep 2020; 10:11436. [PMID: 32651420 PMCID: PMC7351709 DOI: 10.1038/s41598-020-68199-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/05/2020] [Indexed: 12/01/2022] Open
Abstract
Circadian disruption resulting from exposure to irregular light–dark patterns and sleep deprivation has been associated with beta amyloid peptide (Aβ) aggregation, which is a major event in Alzheimer’s disease (AD) pathology. We exposed 5XFAD mice and littermate controls to dim-light vs. bright-light photophases to investigate the effects of altering photophase strength on AD-associated differences in cortical Aβ42 levels, wheel-running activity, and circadian free-running period (tauDD). We found that increasing light levels significantly reduced cortical Aβ42 accumulation and activity levels during the light phase of the light:dark cycle, the latter being consistent with decreased sleep fragmentation and increased sleep duration for mice exposed to the more robust light–dark pattern. No significant changes were observed for tauDD. Our results are consistent with circadian pacemaker period being relatively unaffected by Aβ pathology in AD, and with reductions in cortical Aβ loads in AD through tailored lighting interventions.
Collapse
|
80
|
Um YH, Lim HK. Orexin and Alzheimer's Disease: A New Perspective. Psychiatry Investig 2020; 17:621-626. [PMID: 32517419 PMCID: PMC7385219 DOI: 10.30773/pi.2020.0136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Orexin's role in human cognition has recently been emphasized and emerging evidences indicate its close relationship with Alzheimer's disease (AD). This review aimed to demonstrate recent research on the relationship between orexin and AD. Orexin's role in stress regulation and memory is discussed, with significant findings related to sexual disparities in stress response, with potential clinical implications pertaining to AD pathology. There are controversies regarding the orexin levels in AD patients, but the role of orexin in the trajectory of AD is still emphasized in recent literatures. Orexin is also accentuated in the context of tau pathology, and orexin as a potential therapeutic target for AD is frequently discussed. Future directions with regard to the relationship between orexin and AD are suggested: 1) consideration for AD trajectory in the measurement of orexin levels, 2) the need for objective measure such as polysomnography and actigraphy, 3) the need for close observation of cognitive profiles of orexin-deficient narcolepsy patients, 4) the need for validation studies by neuroimaging 5) the need for taking account sexual disparities in orexinergic activiation, and 6) consideration for orexin's role as a stress regulator. The aforementioned new perspectives could help unravel the relationship between orexin and AD.
Collapse
Affiliation(s)
- Yoo Hyun Um
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
81
|
Li T, Xu W, Ouyang J, Lu X, Sherchan P, Lenahan C, Irio G, Zhang JH, Zhao J, Zhang Y, Tang J. Orexin A alleviates neuroinflammation via OXR2/CaMKKβ/AMPK signaling pathway after ICH in mice. J Neuroinflammation 2020; 17:187. [PMID: 32539736 PMCID: PMC7294616 DOI: 10.1186/s12974-020-01841-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
Background Orexins are two neuropeptides (orexin A, OXA; orexin B, OXB) secreted mainly from the lateral hypothalamus, which exert a wide range of physiological effects by activating two types of receptors (orexin receptor 1, OXR1; orexin receptor 2, OXR2). OXA has equal affinity for OXR1 and OXR2, whereas OXB binds preferentially to OXR2. OXA rapidly crosses the blood-brain barrier by simple diffusion. Many studies have reported OXA’s protective effect on neurological diseases via regulating inflammatory response which is also a fundamental pathological process in intracerebral hemorrhage (ICH). However, neuroprotective mechanisms of OXA have not been explored in ICH. Methods ICH models were established using stereotactic injection of autologous arterial blood into the right basal ganglia of male CD-1 mice. Exogenous OXA was administered intranasally; CaMKKβ inhibitor (STO-609), OXR1 antagonist (SB-334867), and OXR2 antagonist (JNJ-10397049) were administered intraperitoneally. Neurobehavioral tests, hematoma volume, and brain water content were evaluated after ICH. Western blot and ELISA were utilized to evaluate downstream mechanisms. Results OXA, OXR1, and OXR2 were expressed moderately in microglia and astrocytes and abundantly in neurons. Expression of OXA decreased whereas OXR1 and OXR2 increased after ICH. OXA treatment significantly improved not only short-term but also long-term neurofunctional outcomes and reduced brain edema in ipsilateral hemisphere. OXA administration upregulated p-CaMKKβ, p-AMPK, and anti-inflammatory cytokines while downregulated p-NFκB and pro-inflammatory cytokines after ICH; this effect was reversed by STO-609 or JNJ-10397049 but not SB-334867. Conclusions OXA improved neurofunctional outcomes and mitigated brain edema after ICH, possibly through alleviating neuroinflammation via OXR2/CaMKKβ/AMPK pathway.
Collapse
Affiliation(s)
- Tao Li
- Department of Neurosurgery, The First People's Hospital of Yunnan Province (Kunhua Hospital/The Affiliated Hospital of Kunming University of Science and Technology), Yunnan, 650032, China.,Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA.,Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China
| | - Weilin Xu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA.,Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China
| | - Jinsong Ouyang
- Department of Neurosurgery, The First People's Hospital of Yunnan Province (Kunhua Hospital/The Affiliated Hospital of Kunming University of Science and Technology), Yunnan, 650032, China
| | - Xiaoyang Lu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA.,Burrell College of Osteopathic Medicine, 3501 Arrowhead Dr, Las Cruces, NM, 88001, USA
| | - Giselle Irio
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA.,Burrell College of Osteopathic Medicine, 3501 Arrowhead Dr, Las Cruces, NM, 88001, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA
| | - Jianhua Zhao
- Department of Neurosurgery, The First People's Hospital of Yunnan Province (Kunhua Hospital/The Affiliated Hospital of Kunming University of Science and Technology), Yunnan, 650032, China
| | - Yongfa Zhang
- Department of Neurosurgery, The First People's Hospital of Yunnan Province (Kunhua Hospital/The Affiliated Hospital of Kunming University of Science and Technology), Yunnan, 650032, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus St, Loma Linda, CA, 92354, USA.
| |
Collapse
|
82
|
Owen JE, Veasey SC. Impact of sleep disturbances on neurodegeneration: Insight from studies in animal models. Neurobiol Dis 2020; 139:104820. [PMID: 32087293 PMCID: PMC7593848 DOI: 10.1016/j.nbd.2020.104820] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 02/18/2020] [Indexed: 01/18/2023] Open
Abstract
Chronic short sleep or extended wake periods are commonly observed in most industrialized countries. Previously neurobehavioral impairment following sleep loss was considered to be a readily reversible occurrence, normalized upon recovery sleep. Recent clinical studies suggest that chronic short sleep and sleep disruption may be risk factors for neurodegeneration. Animal models have been instrumental in determining whether disturbed sleep can injure the brain. We now understand that repeated periods of extended wakefulness across the typical sleep period and/or sleep fragmentation can have lasting effects on neurogenesis and select populations of neurons and glia. Here we provide a comprehensive overview of the advancements made using animal models of sleep loss to understand the extent and mechanisms of chronic short sleep induced neural injury.
Collapse
Affiliation(s)
- Jessica E Owen
- Chronobiology and Sleep Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sigrid C Veasey
- Chronobiology and Sleep Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
83
|
La AL, Walsh CM, Neylan TC, Vossel KA, Yaffe K, Krystal AD, Miller BL, Karageorgiou E. Long-Term Trazodone Use and Cognition: A Potential Therapeutic Role for Slow-Wave Sleep Enhancers. J Alzheimers Dis 2020; 67:911-921. [PMID: 30689583 PMCID: PMC6398835 DOI: 10.3233/jad-181145] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent studies reveal an association between slow-wave sleep (SWS), amyloid-β aggregation, and cognition. OBJECTIVE This retrospective study examines whether long-term use of trazodone, an SWS enhancer, is associated with delayed cognitive decline. METHODS We identified 25 regular trazodone users (mean age 75.4±7.5; 9 women, 16 men) who carried a diagnosis of Alzheimer's dementia, mild cognitive impairment, or normal cognition, and 25 propensity-matched trazodone non-users (mean age 74.5±8.0; 13 women, 12 men), accounting for age, sex, education, type of sleep deficit (hypersomnia, insomnia, parasomnia), diagnosis, and baseline Mini-Mental State Examination (MMSE). Longitudinal group differences in cognitive testing were evaluated through repeated measures tests over an average inter-evaluation interval of four years. RESULTS Trazodone non-users had 2.6-fold faster decline MMSE (primary outcome) compared to trazodone users, 0.27 (95% confidence interval [CI]: 0.07-0.48) versus 0.70 (95% CI: 0.50-0.90) points per year (p = 0.023). The observed effects were especially associated with subjective improvement of sleep complaints in post-hoc analyses (p = 0.0006). Secondary outcomes of other cognitive and functional scores had variable worsening in non-users and varied in significance when accounting for co-administered medications and multiple comparisons. Trazodone effects on MMSE remained significant within participants with AD-predicted pathology, with 2.4-fold faster decline in non-users (p = 0.038). CONCLUSIONS These results suggest an association between trazodone use and delayed cognitive decline, adding support for a potentially attractive and cost-effective intervention in dementia. Whether the observed relationship of trazodone to cognitive function is causal or an indirect marker of other effects, such as treated sleep disruption, and if such effects are mediated through SWS enhancement requires confirmation through prospective studies.
Collapse
Affiliation(s)
- Alice L. La
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA
| | - Christine M. Walsh
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA
| | - Thomas C. Neylan
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA,San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Keith A. Vossel
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA,
Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Kristine Yaffe
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA,San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA,
Department of Epidemiology, University of California San Francisco, San Francisco, CA, USA
| | - Andrew D. Krystal
- University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA,
Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA
| | - Bruce L. Miller
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA
| | - Elissaios Karageorgiou
- University of California San Francisco, Memory and Aging Center, Weill Institute for the Neurosciences, San Francisco, CA, USA,University of California San Francisco, Weill Institute for the Neurosciences, San Francisco, CA, USA,Neurological Institute of Athens, Athens, Greece,Stanford Sleep Medicine Center, Redwood City, CA, USA,Correspondence to: Elissaios Karageorgiou, MD, PhD, 675 Nelson Rising Lane Suite 190, San Francisco, CA 94158, USA. Tel.: +1 415 502 0588; Fax: +1 415 476 4800; E-mail:
| |
Collapse
|
84
|
Orexins role in neurodegenerative diseases: From pathogenesis to treatment. Pharmacol Biochem Behav 2020; 194:172929. [PMID: 32315694 DOI: 10.1016/j.pbb.2020.172929] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Orexin is a neurotransmitter that mainly regulates sleep/wake cycle. In addition to its sleep cycle regulatory role, it is involved in regulation of attention, energy homeostasis, neurogenesis and cognition. Several evidences has shown the involvement of orexin in narcolepsy, but there are also growing evidences that shows the disturbance in orexin system in neurodegenerative diseases including Alzheimer's, Parkinson's, Epilepsy, Huntington's diseases and Amyotrophic lateral sclerosis. Pathogenesis and clinical symptoms of these disorders can be partly attributed from orexin system imbalance. However, there are controversial reports on the exact relationship between orexin and these neurodegenerative diseases. Therefore, the aim of this review is to summarize the current evidences regarding the role of orexin in these neurodegenerative diseases.
Collapse
|
85
|
Van Egroo M, Narbutas J, Chylinski D, Villar González P, Maquet P, Salmon E, Bastin C, Collette F, Vandewalle G. Sleep-wake regulation and the hallmarks of the pathogenesis of Alzheimer's disease. Sleep 2020; 42:5289316. [PMID: 30649520 DOI: 10.1093/sleep/zsz017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/26/2018] [Indexed: 01/23/2023] Open
Abstract
While efficient treatments for Alzheimer's disease (AD) remain elusive, a growing body of research has highlighted sleep-wake regulation as a potential modifiable factor to delay disease progression. Evidence accumulated in recent years is pointing toward a tight link between sleep-wake disruption and the three main hallmarks of the pathogenesis of AD, i.e. abnormal amyloid-beta (Aβ) and tau proteins accumulation, and neurodegeneration. However, all three hallmarks are rarely considered together in the same study. In this review, we gather and discuss findings in favor of an association between sleep-wake disruption and each AD hallmark in animal models and in humans, with a focus on the preclinical stages of the disease. We emphasize that these relationships are likely bidirectional for each of these hallmarks. Altogether, current findings provide strong support for considering sleep-wake disruption as a true risk factor in the early unfolding of AD, but more research integrating recent technical advances is needed, particularly with respect to tau protein and neurodegeneration. Interventional longitudinal studies among cognitively healthy older individuals should assess the practical use of improving sleep-wake regulation to slow down the progression of AD pathogenesis.
Collapse
Affiliation(s)
- Maxime Van Egroo
- 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
| | - Daphne Chylinski
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University 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
| | - 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
| | - 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
| | - 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
| | - Gilles Vandewalle
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
| |
Collapse
|
86
|
Cassar M, Law AD, Chow ES, Giebultowicz JM, Kretzschmar D. Disease-Associated Mutant Tau Prevents Circadian Changes in the Cytoskeleton of Central Pacemaker Neurons. Front Neurosci 2020; 14:232. [PMID: 32292325 PMCID: PMC7118733 DOI: 10.3389/fnins.2020.00232] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/02/2020] [Indexed: 01/10/2023] Open
Abstract
A hallmark feature of Alzheimer's disease (AD) and other Tauopathies, like Frontotemporal Dementia with Parkinsonism linked to chromosome 17 (FTDP-17), is the accumulation of neurofibrillary tangles composed of the microtubule-associated protein Tau. As in AD, symptoms of FTDP-17 include cognitive decline, neuronal degeneration, and disruptions of sleep patterns. However, mechanisms by which Tau may lead to these disturbances in sleep and activity patterns are unknown. To identify such mechanisms, we have generated novel Drosophila Tauopathy models by replacing endogenous fly dTau with normal human Tau (hTau) or the FTDP-17 causing hTauV337M mutation. This mutation is localized in one of the microtubule-binding domains of hTau and has a dominant effect. Analyzing heterozygous flies, we found that aged hTauV337M flies show neuronal degeneration and locomotion deficits when compared to wild type or hTauWT flies. Furthermore, hTauV337M flies are hyperactive and they show a fragmented sleep pattern. These changes in the sleep/activity pattern are accompanied by morphological changes in the projection pattern of the central pacemaker neurons. These neurons show daily fluctuations in their connectivity, whereby synapses are increased during the day and reduced during sleep. Synapse formation requires cytoskeletal changes that can be detected by the accumulation of the end-binding protein 1 (EB1) at the site of synapse formation. Whereas, hTauWT flies show the normal day/night changes in EB1 accumulation, hTauV337M flies do not show this fluctuation. This suggests that hTauV337M disrupts sleep patterns by interfering with the cytoskeletal changes that are required for the synaptic homeostasis of central pacemaker neurons.
Collapse
Affiliation(s)
- Marlène Cassar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Alexander D Law
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Eileen S Chow
- Department of Integrative Biology, Oregon State University, Corvallis, OR, United States
| | - Jadwiga M Giebultowicz
- Department of Integrative Biology, Oregon State University, Corvallis, OR, United States
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
87
|
Barnett S, Li A. Orexin in Respiratory and Autonomic Regulation, Health and Diseases. Compr Physiol 2020; 10:345-363. [DOI: 10.1002/cphy.c190013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
88
|
Zhou F, Yan XD, Wang C, He YX, Li YY, Zhang J, Wang ZJ, Cai HY, Qi JS, Wu MN. Suvorexant ameliorates cognitive impairments and pathology in APP/PS1 transgenic mice. Neurobiol Aging 2020; 91:66-75. [PMID: 32224066 DOI: 10.1016/j.neurobiolaging.2020.02.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/28/2020] [Accepted: 02/22/2020] [Indexed: 02/06/2023]
Abstract
Cognitive impairments and circadian rhythm disorders are the main clinical manifestations of Alzheimer's disease (AD). Orexin has been reported as abnormally elevated in the cerebrospinal fluid of AD patients, accompanied with cognitive impairments. Our recent research revealed that suvorexant, a dual orexin receptor antagonist, could improve behavioral circadian rhythm disorders in 9-month-old APP/PS1 mice. Here we further observed whether suvorexant could ameliorate the cognitive decline in APP/PS1 mice by using behavioral tests, and investigated the possible mechanisms by in vivo electrophysiological recording, western blot, and immunochemistry. The results showed that suvorexant treatment effectively ameliorated the cognitive impairments, alleviated in vivo hippocampal long-term potentiation suppression, restored the circadian phosphorylated CREB expression in the hippocampus, and reduced amyloid-β protein deposition in the hippocampus and cortex in APP/PS1 mice. These results indicate that the neuroprotective effects of suvorexant against AD are involved in the reduction of amyloid-β plaques, improvement of synaptic plasticity, and circadian expression of phosphorylated CREB, suggesting that suvorexant could be beneficial to the prevention and treatment of AD.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xu-Dong Yan
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Chun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ye-Xin He
- Department of Radiology, Shanxi Provincial People's Hospital, Taiyuan, People's Republic of China
| | - Yi-Ying Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jun Zhang
- Functional Laboratory Center, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
89
|
Kosel F, Pelley JMS, Franklin TB. Behavioural and psychological symptoms of dementia in mouse models of Alzheimer's disease-related pathology. Neurosci Biobehav Rev 2020; 112:634-647. [PMID: 32070692 DOI: 10.1016/j.neubiorev.2020.02.012] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/08/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Transgenic mouse models have been used extensively to model the cognitive impairments arising from Alzheimer's disease (AD)-related pathology. However, less is known about the relationship between AD-related pathology and the behavioural and psychological symptoms of dementia (BPSD) commonly presented by patients. This review discusses the BPSD-like behaviours recapitulated by several mouse models of AD-related pathology, including the APP/PS1, Tg2576, 3xTg-AD, 5xFAD, and APP23 models. Current evidence suggests that social withdrawal and depressive-like behaviours increase with progressive neuropathology, and increased aggression and sleep-wake disturbances are present even at early stages; however, there is no clear evidence to support increased anxiety-like behaviours, agitation (hyperactivity), or general apathy. Overall, transgenic mouse models of AD-related pathology recapitulate some of the BPSD-like behaviours associated with AD, but these behaviours vary by model. This reflects the patient population, where AD patients typically exhibit one or more BPSD, but rarely all symptoms at once. As a result, we suggest that transgenic mouse models are an important tool to investigate the pathology underlying BPSD in human AD patients.
Collapse
Affiliation(s)
- Filip Kosel
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Jessica M S Pelley
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Tamara B Franklin
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada.
| |
Collapse
|
90
|
Uslaner JM, Herring WJ, Coleman PJ. The Discovery of Suvorexant: Lessons Learned That Can Be Applied to Other CNS Drug Development Efforts. ACS Pharmacol Transl Sci 2020; 3:161-168. [PMID: 32259095 PMCID: PMC7088936 DOI: 10.1021/acsptsci.9b00110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Indexed: 11/29/2022]
Abstract
The development of therapeutics for central nervous system (CNS) disorders has many challenges that result in low probability of success and longer-than-typical development timelines. Suvorexant (Belsomra), the first dual orexin receptor antagonist used for insomnia, was approved by the United States Food and Drug Administration ∼10 years after the initial high-throughput screen was conducted to identify orexin receptor antagonists. What accounted for this success and speed? Here we suggest that this program was unique and set up for success by (1) having a robust and high-throughput pharmacodynamic readout that was translatable across species, including humans, (2) a well-validated target with a defined product profile, resulting in a highly energized team with a can-do attitude, and (3) a highly executable and streamlined clinical strategy. The utility of Belsomra for insomnia, as well as other neurological and psychiatric diseases, continues to be explored, most recently for insomnia associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Jason M. Uslaner
- Discovery Neuroscience, Clinical Neuroscience, and Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - William J. Herring
- Discovery Neuroscience, Clinical Neuroscience, and Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Paul J. Coleman
- Discovery Neuroscience, Clinical Neuroscience, and Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
91
|
Li SB, de Lecea L. The hypocretin (orexin) system: from a neural circuitry perspective. Neuropharmacology 2020; 167:107993. [PMID: 32135427 DOI: 10.1016/j.neuropharm.2020.107993] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| |
Collapse
|
92
|
Ferini-Strambi L, Galbiati A, Casoni F, Salsone M. Therapy for Insomnia and Circadian Rhythm Disorder in Alzheimer Disease. Curr Treat Options Neurol 2020; 22:4. [PMID: 32025925 DOI: 10.1007/s11940-020-0612-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF THE REVIEW There is strong evidence for a bidirectional association between sleep disorders and Alzheimer's disease (AD). In particular, insomnia may be a potentially modifiable risk factor for AD. The present review summarizes recent advances in treatment of sleep disorders in AD. RECENT FINDINGS Some studies investigated the efficacy and safety of hypnotic agents as ramelteon and mirtazapine to treat sleep disorders in AD but no significant therapeutic effects have been observed. Benzodiazepines are the most frequently used medication for treatment of insomnia but they may cause significant side effects in old subjects. Suvorexant, an orexin receptor antagonist, showed a positive effect on AD insomnia. Recent report suggests an association between trazodone use and delayed cognitive decline in AD. With respect to circadian rhythm disorders, non-pharmacological treatments, especially bright light therapy, could be useful and safe options for treatment in AD. Some pharmacological and non-pharmacological treatments might have benefits in AD patients with sleep disturbances, but further well-designed controlled trials are needed.
Collapse
Affiliation(s)
- Luigi Ferini-Strambi
- Department of Clinical Neurosciences, "Vita-Salute" San Raffaele University, Milan, Italy. .,Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Andrea Galbiati
- Department of Clinical Neurosciences, "Vita-Salute" San Raffaele University, Milan, Italy
| | - Francesca Casoni
- Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Salsone
- National Research Council, Institute of Molecular Bioimaging and Physiology, Catanzaro, Italy
| |
Collapse
|
93
|
Lee J, Kim DE, Griffin P, Sheehan PW, Kim D, Musiek ES, Yoon S. Inhibition of REV-ERBs stimulates microglial amyloid-beta clearance and reduces amyloid plaque deposition in the 5XFAD mouse model of Alzheimer's disease. Aging Cell 2020; 19:e13078. [PMID: 31800167 PMCID: PMC6996949 DOI: 10.1111/acel.13078] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 07/30/2019] [Accepted: 10/06/2019] [Indexed: 12/21/2022] Open
Abstract
A promising new therapeutic target for the treatment of Alzheimer's disease (AD) is the circadian system. Although patients with AD are known to have abnormal circadian rhythms and suffer sleep disturbances, the role of the molecular clock in regulating amyloid-beta (Aβ) pathology is still poorly understood. Here, we explored how the circadian repressors REV-ERBα and β affected Aβ clearance in mouse microglia. We discovered that, at Circadian time 4 (CT4), microglia expressed higher levels of the master clock protein BMAL1 and more rapidly phagocytosed fibrillary Aβ1-42 (fAβ1-42 ) than at CT12. BMAL1 directly drives transcription of REV-ERB proteins, which are implicated in microglial activation. Interestingly, pharmacological inhibition of REV-ERBs with the small molecule antagonist SR8278 or genetic knockdown of REV-ERBs-accelerated microglial uptake of fAβ1-42 and increased transcription of BMAL1. SR8278 also promoted microglia polarization toward a phagocytic M2-like phenotype with increased P2Y12 receptor expression. Finally, constitutive deletion of Rev-erbα in the 5XFAD model of AD decreased amyloid plaque number and size and prevented plaque-associated increases in disease-associated microglia markers including TREM2, CD45, and Clec7a. Altogether, our work suggests a novel strategy for controlling Aβ clearance and neuroinflammation by targeting REV-ERBs and provides new insights into the role of REV-ERBs in AD.
Collapse
MESH Headings
- ARNTL Transcription Factors/metabolism
- Alzheimer Disease/pathology
- Amyloid beta-Peptides/chemical synthesis
- Amyloid beta-Peptides/metabolism
- Animals
- CLOCK Proteins/metabolism
- Cell Line
- Circadian Clocks/genetics
- Disease Models, Animal
- Isoquinolines/pharmacology
- Macrophages/metabolism
- Mice
- Mice, Knockout
- Microglia/metabolism
- Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Plaque, Amyloid/genetics
- Plaque, Amyloid/metabolism
- Plaque, Amyloid/pathology
- RNA, Small Interfering
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Purinergic P2Y12/drug effects
- Receptors, Purinergic P2Y12/metabolism
- Repressor Proteins/antagonists & inhibitors
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Synapses/genetics
- Synapses/metabolism
- Thiophenes/pharmacology
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Brain ScienceAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Department of NeurologyHope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Do Eon Kim
- Department of Brain ScienceAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Percy Griffin
- Department of NeurologyHope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Patrick W. Sheehan
- Department of NeurologyHope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Dong‐Hou Kim
- Department of Brain ScienceAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Erik S Musiek
- Department of NeurologyHope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Seung‐Yong Yoon
- Department of Brain ScienceAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| |
Collapse
|
94
|
Hoyer D, Allen A, Jacobson LH. Hypnotics with novel modes of action. Br J Clin Pharmacol 2020; 86:244-249. [PMID: 31756268 PMCID: PMC7015741 DOI: 10.1111/bcp.14180] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/19/2022] Open
Abstract
Insomnia and, more generally, lack of sleep are on the rise. Traditionally treated by classical hypnotics, such as benzodiazepines and Z drugs, which both act on the GABAA receptor, and other modalities, including nondrug therapies, such as cognitive behavioural therapy, there is a range of new hypnotics which are being developed or have recently received market approval. Suvorexant and the like target the orexin/hypocretin system: they should have less side effects in terms of drug-drug interactions with e.g. alcohol, less memory impairment and dependence potential compared to classical hypnotics.
Collapse
Affiliation(s)
- Daniel Hoyer
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health SciencesThe University of MelbourneParkvilleVictoriaAustralia
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCAUSA
| | - Andrew Allen
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health SciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Laura H. Jacobson
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health SciencesThe University of MelbourneParkvilleVictoriaAustralia
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| |
Collapse
|
95
|
Shimizu S, Takenoshita N, Inagawa Y, Tsugawa A, Hirose D, Kaneko Y, Ogawa Y, Serisawa S, Sakurai S, Hirao K, Kanetaka H, Kanbayashi T, Imanishi A, Sakurai H, Hanyu H. Positive Association Between Cognitive Function and Cerebrospinal Fluid Orexin A Levels in Alzheimer’s Disease. J Alzheimers Dis 2020; 73:117-123. [DOI: 10.3233/jad-190958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Naoto Takenoshita
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yuta Inagawa
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Akito Tsugawa
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Hirose
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yoshitsugu Kaneko
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yusuke Ogawa
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Shuntaro Serisawa
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Shu Sakurai
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kentaro Hirao
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hidekazu Kanetaka
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Takashi Kanbayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aya Imanishi
- Department of Neuropsychiatry, Akita University School of Medicine, Akita, Japan
| | - Hirofumi Sakurai
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Haruo Hanyu
- Department of Geriatric Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
96
|
Liguori C, Placidi F, Izzi F, Spanetta M, Mercuri NB, Di Pucchio A. Sleep dysregulation, memory impairment, and CSF biomarkers during different levels of neurocognitive functioning in Alzheimer's disease course. ALZHEIMERS RESEARCH & THERAPY 2020; 12:5. [PMID: 31901236 PMCID: PMC6942389 DOI: 10.1186/s13195-019-0571-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/23/2019] [Indexed: 12/18/2022]
Abstract
Background Alzheimer's disease (AD) is frequently accompanied by sleep impairment, which can induce AD-related neurodegeneration. We herein investigated the sleep architecture, cognition, and cerebrospinal fluid (CSF) biomarkers (tau proteins and β-amyloid42) during AD progression from subjective cognitive impairment (SCI) to mild cognitive impairment (MCI) and eventually to AD dementia, and compared the results with cognitively normal (CN) subjects. Methods We included patients affected by SCI, MCI, mild AD, and moderate-to-severe AD in our study along with CN subjects as controls. All the subjects underwent nocturnal polysomnography to investigate sleep, neuropsychological testing to evaluate cognition, and lumbar puncture for CSF AD biomarkers assessment. Results Sleep (both rapid eye movement (REM) and non-REM sleep) and memory function are both progressively impaired during the course of AD from SCI to mild and subsequently to moderate AD. Further, sleep dysregulation appears earlier than cognitive deterioration, with a reduction of CSF β-amyloid42 level. Conclusion Sleep, memory, and CSF AD biomarkers are closely interrelated in AD progression from the earliest asymptomatic and preclinical stages of the disease related in AD since the earliest and preclinical stages of the disease.
Collapse
Affiliation(s)
- Claudio Liguori
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy. .,Neurology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata", Viale Oxford, 81 00133, Rome, Italy.
| | - Fabio Placidi
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Francesca Izzi
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Matteo Spanetta
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata", Viale Oxford, 81 00133, Rome, Italy.,Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Alessandra Di Pucchio
- Training Office, Italian National Institute of Health (Istituto Superiore di Sanità), Rome, Italy
| |
Collapse
|
97
|
Murillo-Rodríguez E, Budde H, Veras AB, Rocha NB, Telles-Correia D, Monteiro D, Cid L, Yamamoto T, Machado S, Torterolo P. The Endocannabinoid System May Modulate Sleep Disorders in Aging. Curr Neuropharmacol 2020; 18:97-108. [PMID: 31368874 PMCID: PMC7324886 DOI: 10.2174/1570159x17666190801155922] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is an inevitable process that involves changes across life in multiple neurochemical, neuroanatomical, hormonal systems, and many others. In addition, these biological modifications lead to an increase in age-related sickness such as cardiovascular diseases, osteoporosis, neurodegenerative disorders, and sleep disturbances, among others that affect activities of daily life. Demographic projections have demonstrated that aging will increase its worldwide rate in the coming years. The research on chronic diseases of the elderly is important to gain insights into this growing global burden. Novel therapeutic approaches aimed for treatment of age-related pathologies have included the endocannabinoid system as an effective tool since this biological system shows beneficial effects in preclinical models. However, and despite these advances, little has been addressed in the arena of the endocannabinoid system as an option for treating sleep disorders in aging since experimental evidence suggests that some elements of the endocannabinoid system modulate the sleep-wake cycle. This article addresses this less-studied field, focusing on the likely perspective of the implication of the endocannabinoid system in the regulation of sleep problems reported in the aged. We conclude that beneficial effects regarding the putative efficacy of the endocannabinoid system as therapeutic tools in aging is either inconclusive or still missing.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, México
- Intercontinental Neuroscience Research Group
| | - Henning Budde
- Intercontinental Neuroscience Research Group
- Faculty of Human Sciences, Medical School Hamburg, Hamburg, Germany
| | - André Barciela Veras
- Intercontinental Neuroscience Research Group
- Dom Bosco Catholic University, Campo Grande, Mato Grosso do Sul, Brazil
| | - Nuno Barbosa Rocha
- Intercontinental Neuroscience Research Group
- School of Health, Polytechnic Institute of Porto, Porto, Portugal
| | - Diogo Telles-Correia
- Intercontinental Neuroscience Research Group
- University of Lisbon, Faculty of Medicine, Lisbon, Portugal
| | - Diogo Monteiro
- Intercontinental Neuroscience Research Group
- Sport Science School of Rio Maior-Polytechnic Institute of Santarém, Rio Maior, Portugal
- Research Center in Sport, Health and Human Development-CIDESD, Vila Real, Portugal
| | - Luis Cid
- Intercontinental Neuroscience Research Group
- Sport Science School of Rio Maior-Polytechnic Institute of Santarém, Rio Maior, Portugal
- Research Center in Sport, Health and Human Development-CIDESD, Vila Real, Portugal
| | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Sérgio Machado
- Intercontinental Neuroscience Research Group
- Laboratory of Physical Activity Neuroscience, Physical Activity Sciences Postgraduate Program, Salgado de Oliveira University, Niterói, Brazil
| | - Pablo Torterolo
- Intercontinental Neuroscience Research Group
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
98
|
Li M, Meng Y, Chu B, Shen Y, Liu X, Ding M, Song C, Cao X, Wang P, Xu L, Wang Y, Xu S, Bi J, Xie Z. Orexin-A aggravates cytotoxicity and mitochondrial impairment in SH-SY5Y cells transfected with APPswe via p38 MAPK pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:5. [PMID: 32055596 DOI: 10.21037/atm.2019.11.68] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Alzheimer's disease (AD) is one of the common neurodegenerative diseases and is characterized by the accumulation of amyloid-β (Aβ). Orexin-A is a neuropeptide produced in the hypothalamus and thought to be involved in the pathogenesis of AD. However, its underlying mechanism and signaling pathway remains unclear. The aim of this work was to investigate the effect of Orexin-A on AD, and to explore its potential mechanism and signaling pathway. Methods SH-SY5Y cells that were stably transfected with the Swedish mutant amyloid precursor protein (APPswe), a cell model of AD with excessive Aβ production, were used in this study. Cells were treated with Orexin-A, and with or without SB203580, an inhibitor of the p38 mitogen-activated protein kinase (MAPK) pathway, one of the key MAPK pathways associated with cell death. Following treatment, cells were collected and analyzed by western blotting, ELISA, electron microscopy, real-time PCR, fluorescence microscopy, and other biochemical assays. Results Orexin-A increased the level of Aβ1-40 and Aβ1-42 in the cell medium, and activated the p38 MAPK pathway. As evidenced by the CCK-8 and ELISA BrdU assays, Orexin-A decreased cell viability and cell proliferation. Electron microscopic analysis used to observe the morphology of mitochondria, showed that Orexin-A increased the percentage of abnormal mitochondria. Further, decreased activity of cytochrome c oxidase (CCO), level of ATP, and mitochondrial DNA (mtDNA) copy number following Orexin-A treatment showed that Orexin-A exacerbated mitochondrial dysfunction. The level of intracellular reactive oxygen species (ROS), which is mainly generated in mitochondria and reflects mitochondrial dysfunction, was also increased by Orexin-A. SB203580 blocked the cytotoxicity and mitochondrial impairment aggravated by Orexin-A. Conclusions These findings demonstrate that Orexin-A aggravates cytotoxicity and mitochondrial impairment in SH-SY5Y cells transfected with APPswe through the p38 MAPK pathway, and suggest that Orexin-A participates in the pathogenesis of AD, which may provide a new treatment target in the future.
Collapse
Affiliation(s)
- Maoyu Li
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Yao Meng
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Bingcong Chu
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Yang Shen
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Xiangtian Liu
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Mao Ding
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Chaoyuan Song
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Xi Cao
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Ping Wang
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Linlin Xu
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Yun Wang
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Shunliang Xu
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Jianzhong Bi
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| | - Zhaohong Xie
- Department of Neurology, Second Hospital of Shandong University, Jinan 250033, China
| |
Collapse
|
99
|
Wang C, Holtzman DM. Bidirectional relationship between sleep and Alzheimer's disease: role of amyloid, tau, and other factors. Neuropsychopharmacology 2020; 45:104-120. [PMID: 31408876 PMCID: PMC6879647 DOI: 10.1038/s41386-019-0478-5] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/28/2019] [Accepted: 08/02/2019] [Indexed: 01/04/2023]
Abstract
As we age, we experience changes in our nighttime sleep and daytime wakefulness. Individuals afflicted with Alzheimer's disease (AD) can develop sleep problems even before memory and other cognitive deficits are reported. As the disease progresses and cognitive changes ensue, sleep disturbances become even more debilitating. Thus, it is imperative to gain a better understanding of the relationship between sleep and AD pathogenesis. We postulate a bidirectional relationship between sleep and the neuropathological hallmarks of AD; in particular, the accumulation of amyloid-β (Aβ) and tau. Our research group has shown that extracellular levels of both Aβ and tau fluctuate during the normal sleep-wake cycle. Disturbed sleep and increased wakefulness acutely lead to increased Aβ production and decreased Aβ clearance, whereas Aβ aggregation and deposition is enhanced by chronic increased wakefulness in animal models. Once Aβ accumulates, there is evidence in both mice and humans that this results in disturbed sleep. New findings from our group reveal that acute sleep deprivation increases levels of tau in mouse brain interstitial fluid (ISF) and human cerebrospinal fluid (CSF) and chronic sleep deprivation accelerates the spread of tau protein aggregates in neural networks. Finally, recent evidence also suggests that accumulation of tau aggregates in the brain correlates with decreased nonrapid eye movement (NREM) sleep slow wave activity. In this review, we first provide a brief overview of the AD and sleep literature and then highlight recent advances in the understanding of the relationship between sleep and AD pathogenesis. Importantly, the effects of the bidirectional relationship between the sleep-wake cycle and tau have not been previously discussed in other reviews on this topic. Lastly, we provide possible directions for future studies on the role of sleep in AD.
Collapse
Affiliation(s)
- Chanung Wang
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
100
|
Kaur S, DasGupta G, Singh S. Altered Neurochemistry in Alzheimer’s Disease: Targeting Neurotransmitter Receptor Mechanisms and Therapeutic Strategy. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09823-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|