1
|
Turton SM, Padgett S, Maisel MT, Johnson CE, Buzinova VA, Barth SE, Kohler K, Spearman HM, Macheda T, Manauis EC, Guo LZ, Whitlock HR, Bachstetter AD, Sunderam S, O'Hara BF, Duncan MJ, Murphy MP. Interactions between daily sleep-wake rhythms, γ-secretase, and amyloid-β peptide pathology point to complex underlying relationships. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167840. [PMID: 40222459 DOI: 10.1016/j.bbadis.2025.167840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Disrupted or insufficient sleep is a well-documented risk factor for Alzheimer's disease (AD) and related dementias. Previous studies in our lab and others have shown that chronic fragmentation of the daily sleep-wake rhythm in mice can accelerate the development of AD-related neuropathology in the brain, including increases in the levels of amyloid-β (Aβ). Although sleep is known to increase clearance of Aβ via the glymphatic system, little is known about the effect of sleep on Aβ production and the role this might play in amyloid deposition. To examine the relationship of Aβ production and its interaction with sleep and sleep dysfunction, we treated mice from an APP × PS1 mutant knock-in line (APPΔNLh/ΔNLh × PS1P264L/P264L) with an inhibitor of γ-secretase (LY-450,139; Semagacestat®) during a protocol of mild sleep fragmentation (SF). Compared to the male mice, the female mice slept less, and had more Aβ pathology. Semagacestat treatment reduced Aβ, but only in the most soluble extractable fraction. Although the female mice showed an increase in the amount of Aβ following SF, this effect was blocked by Semagacestat, an effect that was not seen in the male mice. SF also led to a significant, sex-dependent changes in the relative amounts of C-terminal fragments of the amyloid precursor protein, the immediate substrate of the γ-secretase enzyme. These findings indicate that the relationship between disruption of the daily sleep-wake rhythm and the development of AD-related pathology is complex, and may involve unappreciated interactions with biological sex. Consideration of these factors is necessary for a better understanding of AD risk, especially the elevated risk in women.
Collapse
Affiliation(s)
| | | | | | - Carrie E Johnson
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA
| | - Valeria A Buzinova
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA
| | | | | | | | | | | | | | | | - Adam D Bachstetter
- The Sanders-Brown Center on Aging, USA; The Spinal Cord and Brain Injury Research Center, USA; Department of Neuroscience, USA
| | | | | | | | - M Paul Murphy
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA.
| |
Collapse
|
2
|
Mao JQ, Cheng L, Zhang YD, Xie GJ, Wang P. Chinese formula Guben-Jiannao Ye alleviates the dysfunction of circadian and sleep rhythms in APP/PS1 mice implicated in activation of the PI3K/AKT/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118696. [PMID: 39151711 DOI: 10.1016/j.jep.2024.118696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/24/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Chinese formula Guben-Jiannao Ye (GBJNY) formula has a long history of usage in traditional Chinese medicine (TCM) for the treatment of learning and memory disorders as well as senile insomnia. This formulation is derived from Sun Simiao's five tonic pills. Furthermore, modern pharmacological investigations have revealed its ability to improve cognitive impairment and ameliorate sleep-wake circadian rhythm disorders. However, the precise mechanism underlying its efficacy remains elusive. AIM OF THE STUDY The current research explored the modulatory effects and possible mechanisms of GBJNY in circadian rhythm sleep-wake disorders and cognitive dysfunction in Alzheimer's disease using transcriptome sequencing and experimental validation. MATERIALS AND METHODS The LC-MS/MS tandem technology was utilized to qualitatively discern the active components present in GBJNY. The APP/PS1 mice received continuous treatment with GBJNY or Melatonin for 3 months. The learning and memory abilities of mice were assessed utilizing the Morris water maze (MWM) test, while sleep changes were studied utilizing the electroencephalogram (EEG) and electromyogram (EMG). Concurrently, mice's hippocampus clock gene rhythmicity was investigated. Subsequently, we employed HE staining, Golgi staining, and immunofluorescence to observe GBJNY's impact on synaptic damage and neuronal loss. We performed high-throughput sequencing to analyze the mRNA expression profiles of mice, aiming to identify differentially expressed genes (DEGs). Subsequently, we conducted GO and KEGG enrichment analyses to explore associated signaling pathways. Furthermore, we evaluated the expression levels of proteins involved in the PI3K/AKT/mTOR pathway and Aβ deposition in the hippocampus of mice. Through this comprehensive approach, we sought to elucidate and validate the potential mechanisms of action of GBJNY in APP/PS1 mice. RESULTS Results showed 216 DEGs. Following this, we conducted GO enrichment and KEGG pathway analyses to delve deeper into the distinctions and fundamental functions of the mRNA target genes. The enrichment analysis underscored the prominence of the PI3K/Akt/mTOR signaling pathway as the most pivotal among them. Through in vivo experiments, it was further demonstrated that the administration of GBJNY enhanced memory and learning capacities in APP/PS1 mice. Additionally, GBJNY treatment resulted in alterations in the sleep-wake circadian rhythm, characterized by reduced wakefulness and an increase in non-rapid eye movement (NREM) sleep. Moreover, alterations in the peak expression of Per1, Per2, Clock, Cry1, Cry2, and Bmal1 mRNA were noted in the hippocampus of treated mice. Particularly noteworthy were the observed reductions in amyloid-beta (Aβ) deposition within the hippocampus, improvements in neuronal synaptic integrity, and upregulation of mTOR, Akt, and PI3K protein expression in the hippocampal region. These findings underscore the critical involvement of the PI3K/Akt/mTOR signaling pathway in mitigating disturbances in sleep-wake circadian rhythms. CONCLUSIONS GBJNY enhanced the cognitive performance of APP/PS1 mice and altered clock gene expression patterns, alleviating sleep-wake circadian rhythm disruptions. The fundamental mechanism appears to be linked to the PI3K/Akt/mTOR pathway regulation, offering a foundation for potential clinical applications.
Collapse
Affiliation(s)
- Jian-Qin Mao
- Basic Medicine College, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Cheng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Yu-Dan Zhang
- Health Medical Center, Hubei Minzu University, Enshi, 445000, China.
| | - Guang-Jing Xie
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| | - Ping Wang
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| |
Collapse
|
3
|
Long DM, Cravetchi O, Chow ES, Allen C, Kretzschmar D. The amyloid precursor protein intracellular domain induces sleep disruptions and its nuclear localization fluctuates in circadian pacemaker neurons in Drosophila and mice. Neurobiol Dis 2024; 192:106429. [PMID: 38309627 DOI: 10.1016/j.nbd.2024.106429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 11/17/2023] [Accepted: 02/01/2024] [Indexed: 02/05/2024] Open
Abstract
The most prominent symptom of Alzheimer's disease (AD) is cognitive decline; however, sleep and other circadian disruptions are also common in AD patients. Sleep disruptions have been connected with memory problems and therefore the changes in sleep patterns observed in AD patients may also actively contribute to cognitive decline. However, the underlying molecular mechanisms that connect sleep disruptions and AD are unclear. A characteristic feature of AD is the formation of plaques consisting of Amyloid-β (Aβ) peptides generated by cleavage of the Amyloid Precursor Protein (APP). Besides Aβ, APP cleavage generates several other fragments, including the APP intracellular domain (AICD) that has been linked to transcriptional regulation and neuronal homeostasis. Here we show that overexpression of the AICD reduces the early evening expression of two core clock genes and disrupts the sleep pattern in flies. Analyzing the subcellular localization of the AICD in pacemaker neurons, we found that the AICD levels in the nucleus are low during daytime but increase at night. While this pattern of nuclear AICD persisted with age, the nighttime levels were higher in aged flies. Increasing the cleavage of the fly APP protein also disrupted AICD nuclear localization. Lastly, we show that the day/nighttime nuclear pattern of the AICD is also detectable in neurons in the suprachiasmatic nucleus of mice and that it also changes with age. Together, these data suggest that AD-associated changes in APP processing and the subsequent changes in AICD levels may cause sleep disruptions in AD.
Collapse
Affiliation(s)
- Dani M Long
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Olga Cravetchi
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Eileen S Chow
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA
| | - Charles Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
4
|
Zhu B, Parsons T, Stensen W, Mjøen Svendsen JS, Fugelli A, Hodge JJL. DYRK1a Inhibitor Mediated Rescue of Drosophila Models of Alzheimer’s Disease-Down Syndrome Phenotypes. Front Pharmacol 2022; 13:881385. [PMID: 35928283 PMCID: PMC9345315 DOI: 10.3389/fphar.2022.881385] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease which is becoming increasingly prevalent due to ageing populations resulting in huge social, economic, and health costs to the community. Despite the pathological processing of genes such as Amyloid Precursor Protein (APP) into Amyloid-β and Microtubule Associated Protein Tau (MAPT) gene, into hyperphosphorylated Tau tangles being known for decades, there remains no treatments to halt disease progression. One population with increased risk of AD are people with Down syndrome (DS), who have a 90% lifetime incidence of AD, due to trisomy of human chromosome 21 (HSA21) resulting in three copies of APP and other AD-associated genes, such as DYRK1A (Dual specificity tyrosine-phosphorylation-regulated kinase 1A) overexpression. This suggests that blocking DYRK1A might have therapeutic potential. However, it is still not clear to what extent DYRK1A overexpression by itself leads to AD-like phenotypes and how these compare to Tau and Amyloid-β mediated pathology. Likewise, it is still not known how effective a DYRK1A antagonist may be at preventing or improving any Tau, Amyloid-β and DYRK1a mediated phenotype. To address these outstanding questions, we characterised Drosophila models with targeted overexpression of human Tau, human Amyloid-β or the fly orthologue of DYRK1A, called minibrain (mnb). We found targeted overexpression of these AD-associated genes caused degeneration of photoreceptor neurons, shortened lifespan, as well as causing loss of locomotor performance, sleep, and memory. Treatment with the experimental DYRK1A inhibitor PST-001 decreased pathological phosphorylation of human Tau [at serine (S) 262]. PST-001 reduced degeneration caused by human Tau, Amyloid-β or mnb lengthening lifespan as well as improving locomotion, sleep and memory loss caused by expression of these AD and DS genes. This demonstrated PST-001 effectiveness as a potential new therapeutic targeting AD and DS pathology.
Collapse
Affiliation(s)
- Bangfu Zhu
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Science, University of Bristol, Bristol, United Kingdom
| | - Tom Parsons
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Science, University of Bristol, Bristol, United Kingdom
| | - Wenche Stensen
- Department of Chemistry, The Arctic University of Norway, Tromsø, Norway
- Pharmasum Therapeutics AS, ShareLab, Forskningsparken i Oslo, Oslo, Norway
| | - John S. Mjøen Svendsen
- Department of Chemistry, The Arctic University of Norway, Tromsø, Norway
- Pharmasum Therapeutics AS, ShareLab, Forskningsparken i Oslo, Oslo, Norway
| | - Anders Fugelli
- Pharmasum Therapeutics AS, ShareLab, Forskningsparken i Oslo, Oslo, Norway
| | - James J. L. Hodge
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Science, University of Bristol, Bristol, United Kingdom
- *Correspondence: James J. L. Hodge,
| |
Collapse
|
5
|
Abstract
Endogenous biological clocks, orchestrated by the suprachiasmatic nucleus, time the circadian rhythms that synchronize physiological and behavioural functions in humans. The circadian system influences most physiological processes, including sleep, alertness and cognitive performance. Disruption of circadian homeostasis has deleterious effects on human health. Neurodegenerative disorders involve a wide range of symptoms, many of which exhibit diurnal variations in frequency and intensity. These disorders also disrupt circadian homeostasis, which in turn has negative effects on symptoms and quality of life. Emerging evidence points to a bidirectional relationship between circadian homeostasis and neurodegeneration, suggesting that circadian function might have an important role in the progression of neurodegenerative disorders. Therefore, the circadian system has become an attractive target for research and clinical care innovations. Studying circadian disruption in neurodegenerative disorders could expand our understanding of the pathophysiology of neurodegeneration and facilitate the development of novel, circadian-based interventions for these disabling disorders. In this Review, we discuss the alterations to the circadian system that occur in movement (Parkinson disease and Huntington disease) and cognitive (Alzheimer disease and frontotemporal dementia) neurodegenerative disorders and provide directions for future investigations in this field.
Collapse
|
6
|
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: 1.8] [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
|
7
|
Carter B, Justin HS, Gulick D, Gamsby JJ. The Molecular Clock and Neurodegenerative Disease: A Stressful Time. Front Mol Biosci 2021; 8:644747. [PMID: 33889597 PMCID: PMC8056266 DOI: 10.3389/fmolb.2021.644747] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Circadian rhythm dysfunction occurs in both common and rare neurodegenerative diseases. This dysfunction manifests as sleep cycle mistiming, alterations in body temperature rhythms, and an increase in symptomatology during the early evening hours known as Sundown Syndrome. Disruption of circadian rhythm homeostasis has also been implicated in the etiology of neurodegenerative disease. Indeed, individuals exposed to a shifting schedule of sleep and activity, such as health care workers, are at a higher risk. Thus, a bidirectional relationship exists between the circadian system and neurodegeneration. At the heart of this crosstalk is the molecular circadian clock, which functions to regulate circadian rhythm homeostasis. Over the past decade, this connection has become a focal point of investigation as the molecular clock offers an attractive target to combat both neurodegenerative disease pathogenesis and circadian rhythm dysfunction, and a pivotal role for neuroinflammation and stress has been established. This review summarizes the contributions of molecular clock dysfunction to neurodegenerative disease etiology, as well as the mechanisms by which neurodegenerative diseases affect the molecular clock.
Collapse
Affiliation(s)
- Bethany Carter
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Hannah S Justin
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Danielle Gulick
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joshua J Gamsby
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
8
|
Wang YY, Ma WW, Peng IF. Screening of sleep assisting drug candidates with a Drosophila model. PLoS One 2020; 15:e0236318. [PMID: 32726319 PMCID: PMC7390450 DOI: 10.1371/journal.pone.0236318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
Lately, Drosophila has been favored as a model in sleep and circadian rhythm research due to its conserved mechanism and easily manageable operation. These studies have revealed the sophisticated parameters in whole-day sleep profiles of Drosophila, drawing connections between Drosophila sleep and human sleep. In this study, we tested several sleep deprivation protocols (mechanical shakes and light interruptions) on Drosophila and delineated their influences on Drosophila sleep. We applied a daytime light-deprivation protocol (DD) mimicking jet-lag to screen drugs that alleviate sleep deprivation. Characteristically, classical sleep-aid compounds exhibited different forms of influence: phenobarbital and pentobarbital modified total sleep time, while melatonin only shortened the latency to sleep. Such results construct the basis for further research on sleep benefits in other treatments in Drosophila. We screened seven herb extracts, and found very diverse results regarding their effect on sleep regulation. For instance, Panax notoginseng and Withania somnifera extracts displayed potent influence on total sleep time, while Melissa officinalis increased the number of sleep episodes. By comparing these treatments, we were able to rank drug potency in different aspects of sleep regulation. Notably, we also confirmed the presence of sleep difficulties in a Drosophila Alzheimer’s disease (AD) model with an overexpression of human Abeta, and recognized clear differences between the portfolios of drug screening effects in AD flies and in the control group. Overall, potential drug candidates and receipts for sleep problems can be identified separately for normal and AD Drosophila populations, outlining Drosophila’s potential in drug screening tests in other populations if combined with the use of other genetic disease tools.
Collapse
Affiliation(s)
- Yan-Ying Wang
- Research Department, Suzhou Joekai Biotech LLC, Kunshan City, Jiangsu, China
| | - Wei-Wei Ma
- Research Department, Suzhou Joekai Biotech LLC, Kunshan City, Jiangsu, China
- School of Life Science, Tsinghua University, Beijing, China
| | - I-Feng Peng
- Research Department, Suzhou Joekai Biotech LLC, Kunshan City, Jiangsu, China
- * E-mail:
| |
Collapse
|
9
|
De Nobrega AK, Lyons LC. Aging and the clock: Perspective from flies to humans. Eur J Neurosci 2020; 51:454-481. [PMID: 30269400 PMCID: PMC6441388 DOI: 10.1111/ejn.14176] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022]
Abstract
Endogenous circadian oscillators regulate molecular, cellular and physiological rhythms, synchronizing tissues and organ function to coordinate activity and metabolism with environmental cycles. The technological nature of modern society with round-the-clock work schedules and heavy reliance on personal electronics has precipitated a striking increase in the incidence of circadian and sleep disorders. Circadian dysfunction contributes to an increased risk for many diseases and appears to have adverse effects on aging and longevity in animal models. From invertebrate organisms to humans, the function and synchronization of the circadian system weakens with age aggravating the age-related disorders and pathologies. In this review, we highlight the impacts of circadian dysfunction on aging and longevity and the reciprocal effects of aging on circadian function with examples from Drosophila to humans underscoring the highly conserved nature of these interactions. Additionally, we review the potential for using reinforcement of the circadian system to promote healthy aging and mitigate age-related pathologies. Advancements in medicine and public health have significantly increased human life span in the past century. With the demographics of countries worldwide shifting to an older population, there is a critical need to understand the factors that shape healthy aging. Drosophila melanogaster, as a model for aging and circadian interactions, has the capacity to facilitate the rapid advancement of research in this area and provide mechanistic insights for targeted investigations in mammals.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida
| | - Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida
| |
Collapse
|
10
|
Alzheimer's Disease and Sleep-Wake Disturbances: Amyloid, Astrocytes, and Animal Models. J Neurosci 2019; 38:2901-2910. [PMID: 29563238 DOI: 10.1523/jneurosci.1135-17.2017] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/21/2017] [Accepted: 10/18/2017] [Indexed: 01/24/2023] Open
Abstract
Sleep-wake abnormalities are common in patients with Alzheimer's disease, and can be a major reason for institutionalization. However, an emerging concept is that these sleep-wake disturbances are part of the causal pathway accelerating the neurodegenerative process. Recently, new findings have provided intriguing evidence for a positive feedback loop between sleep-wake dysfunction and β-amyloid (Aβ) aggregation. Studies in both humans and animal models have shown that extended periods of wakefulness increase Aβ levels and aggregation, and accumulation of Aβ causes fragmentation of sleep. This perspective is aimed at presenting evidence supporting causal links between sleep-wake dysfunction and aggregation of Aβ peptide in Alzheimer's disease, and explores the role of astrocytes, a specialized type of glial cell, in this context underlying Alzheimer's disease pathology. The utility of current animal models and the unexplored potential of alternative animal models for testing mechanisms involved in the reciprocal relationship between sleep disruption and Aβ are also discussed.Dual Perspectives Companion Paper: Microglia-Mediated Synapse Loss in Alzheimer's Disease by Lawrence Rajendran and Rosa Paolicelli.
Collapse
|
11
|
Higham JP, Malik BR, Buhl E, Dawson JM, Ogier AS, Lunnon K, Hodge JJL. Alzheimer's Disease Associated Genes Ankyrin and Tau Cause Shortened Lifespan and Memory Loss in Drosophila. Front Cell Neurosci 2019; 13:260. [PMID: 31244615 PMCID: PMC6581016 DOI: 10.3389/fncel.2019.00260] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by intracellular neurofibrillary tangles of hyperphosphorylated Tau, including the 0N4R isoform and accumulation of extracellular amyloid beta (Aβ) plaques. However, less than 5% of AD cases are familial, with many additional risk factors contributing to AD including aging, lifestyle, the environment and epigenetics. Recent epigenome-wide association studies (EWAS) of AD have identified a number of loci that are differentially methylated in the AD cortex. Indeed, hypermethylation and reduced expression of the Ankyrin 1 (ANK1) gene in AD has been reported in the cortex in numerous different post-mortem brain cohorts. Little is known about the normal function of ANK1 in the healthy brain, nor the role it may play in AD. We have generated Drosophila models to allow us to functionally characterize Drosophila Ank2, the ortholog of human ANK1 and to determine its interaction with human Tau and Aβ. We show expression of human Tau 0N4R or the oligomerizing Aβ 42 amino acid peptide caused shortened lifespan, degeneration, disrupted movement, memory loss, and decreased excitability of memory neurons with co-expression tending to make the pathology worse. We find that Drosophila with reduced neuronal Ank2 expression have shortened lifespan, reduced locomotion, reduced memory and reduced neuronal excitability similar to flies overexpressing either human Tau 0N4R or Aβ42. Therefore, we show that the mis-expression of Ank2 can drive disease relevant processes and phenocopy some features of AD. Therefore, we propose targeting human ANK1 may have therapeutic potential. This represents the first study to characterize an AD-relevant gene nominated from EWAS.
Collapse
Affiliation(s)
- James P. Higham
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Bilal R. Malik
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Edgar Buhl
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Jennifer M. Dawson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Anna S. Ogier
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Katie Lunnon
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - James J. L. Hodge
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
12
|
Czeczor JK, Genders AJ, Aston-Mourney K, Connor T, Hall LG, Hasebe K, Ellis M, De Jong KA, Henstridge DC, Meikle PJ, Febbraio MA, Walder K, McGee SL. APP deficiency results in resistance to obesity but impairs glucose tolerance upon high fat feeding. J Endocrinol 2018; 237:311-322. [PMID: 29674342 DOI: 10.1530/joe-18-0051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 04/19/2018] [Indexed: 12/22/2022]
Abstract
The amyloid precursor protein (APP) generates a number of peptides when processed through different cleavage mechanisms, including the amyloid beta peptide that is implicated in the development of Alzheimer's disease. It is well established that APP via its cleaved peptides regulates aspects of neuronal metabolism. Emerging evidence suggests that amyloidogenic processing of APP can lead to altered systemic metabolism, similar to that observed in metabolic disease states. In the present study, we investigated the effect of APP deficiency on obesity-induced alterations in systemic metabolism. Compared with WT littermates, APP-deficient mice were resistant to diet-induced obesity, which was linked to higher energy expenditure and lipid oxidation throughout the dark phase and was associated with increased spontaneous physical activity. Consistent with this lean phenotype, APP-deficient mice fed a high-fat diet (HFD) had normal insulin tolerance. However, despite normal insulin action, these mice were glucose intolerant, similar to WT mice fed a HFD. This was associated with reduced plasma insulin in the early phase of the glucose tolerance test. Analysis of the pancreas showed that APP was required to maintain normal islet and β-cell mass under high fat feeding conditions. These studies show that, in addition to regulating aspects of neuronal metabolism, APP is an important regulator of whole body energy expenditure and glucose homeostasis under high fat feeding conditions.
Collapse
Affiliation(s)
- Juliane K Czeczor
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Amanda J Genders
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Kathryn Aston-Mourney
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Timothy Connor
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Liam G Hall
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Kyoko Hasebe
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Megan Ellis
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Kirstie A De Jong
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | | | - Peter J Meikle
- Baker Heart and Diabetes InstituteMelbourne, Victoria, Australia
| | - Mark A Febbraio
- Division of Diabetes and MetabolismGarvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Ken Walder
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Sean L McGee
- Metabolic Research UnitSchool of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
- Baker Heart and Diabetes InstituteMelbourne, Victoria, Australia
| |
Collapse
|
13
|
O'Keefe L, Denton D. Using Drosophila Models of Amyloid Toxicity to Study Autophagy in the Pathogenesis of Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5195416. [PMID: 29888266 PMCID: PMC5985114 DOI: 10.1155/2018/5195416] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/20/2018] [Accepted: 04/10/2018] [Indexed: 12/24/2022]
Abstract
Autophagy is a conserved catabolic pathway that involves the engulfment of cytoplasmic components such as large protein aggregates and organelles that are delivered to the lysosome for degradation. This process is important in maintaining neuronal function and raises the possibility of a role for autophagy in neurodegenerative diseases. Alzheimer's disease (AD) is the most prevalent form of these diseases and is characterized by the accumulation of amyloid plaques in the brain which arise due to the misfolding and aggregation of toxic peptides, including amyloid beta (Aβ). There is substantial evidence from both AD patients and animal models that autophagy is dysregulated in this disease. However, it remains to be determined whether this is protective or pathogenic as there is evidence that autophagy can act to promote the degradation as well as function in the generation of toxic Aβ peptides. Understanding the molecular details of the extensive crosstalk that occurs between the autophagic and endolysosomal cellular pathways is essential for identifying the molecular details of amyloid toxicity. Drosophila models that express the toxic proteins that aggregate in AD have been generated and have been shown to recapitulate hallmarks of the disease. Here we focus on what is known about the role of autophagy in amyloid toxicity in AD from mammalian models and how Drosophila models can be used to further investigate AD pathogenesis.
Collapse
Affiliation(s)
- Louise O'Keefe
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, P.O. Box 11060, Adelaide, SA 5001, Australia
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| |
Collapse
|
14
|
Chauhan R, Chen KF, Kent BA, Crowther DC. Central and peripheral circadian clocks and their role in Alzheimer's disease. Dis Model Mech 2017; 10:1187-1199. [PMID: 28993311 PMCID: PMC5665458 DOI: 10.1242/dmm.030627] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Molecular and cellular oscillations constitute an internal clock that tracks the time of day and permits organisms to optimize their behaviour and metabolism to suit the daily demands they face. The workings of this internal clock become impaired with age. In this review, we discuss whether such age-related impairments in the circadian clock interact with age-related neurodegenerative disorders, such as Alzheimer's disease. Findings from mouse and fly models of Alzheimer's disease have accelerated our understanding of the interaction between neurodegeneration and circadian biology. These models show that neurodegeneration likely impairs circadian rhythms either by damaging the central clock or by blocking its communication with other brain areas and with peripheral tissues. The consequent sleep and metabolic deficits could enhance the susceptibility of the brain to further degenerative processes. Thus, circadian dysfunction might be both a cause and an effect of neurodegeneration. We also discuss the primary role of light in the entrainment of the central clock and describe important, alternative time signals, such as food, that play a role in entraining central and peripheral circadian clocks. Finally, we propose how these recent insights could inform efforts to develop novel therapeutic approaches to re-entrain arrhythmic individuals with neurodegenerative disease.
Collapse
Affiliation(s)
- Ruchi Chauhan
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Ko-Fan Chen
- Institute of Neurology, UCL, London, WC1N 3BG, UK
| | - Brianne A Kent
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Damian C Crowther
- Neuroscience, Innovative Medicines and Early Development, AstraZeneca, Granta Park, Cambridge, CB21 6GH, UK
| |
Collapse
|
15
|
Circadian Rhythm Neuropeptides in Drosophila: Signals for Normal Circadian Function and Circadian Neurodegenerative Disease. Int J Mol Sci 2017; 18:ijms18040886. [PMID: 28430154 PMCID: PMC5412466 DOI: 10.3390/ijms18040886] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023] Open
Abstract
Circadian rhythm is a ubiquitous phenomenon in many organisms ranging from prokaryotes to eukaryotes. During more than four decades, the intrinsic and exogenous regulations of circadian rhythm have been studied. This review summarizes the core endogenous oscillation in Drosophila and then focuses on the neuropeptides, neurotransmitters and hormones that mediate its outputs and integration in Drosophila and the links between several of these (pigment dispersing factor (PDF) and insulin-like peptides) and neurodegenerative disease. These signaling molecules convey important network connectivity and signaling information for normal circadian function, but PDF and insulin-like peptides can also convey signals that lead to apoptosis, enhanced neurodegeneration and cognitive decline in flies carrying circadian mutations or in a senescent state.
Collapse
|
16
|
Cedernaes J, Osorio RS, Varga AW, Kam K, Schiöth HB, Benedict C. Candidate mechanisms underlying the association between sleep-wake disruptions and Alzheimer's disease. Sleep Med Rev 2017; 31:102-111. [PMID: 26996255 PMCID: PMC4981560 DOI: 10.1016/j.smrv.2016.02.002] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 12/13/2022]
Abstract
During wakefulness, extracellular levels of metabolites in the brain increase. These include amyloid beta (Aβ), which contributes to the pathogenesis of Alzheimer's disease (AD). Counterbalancing their accumulation in the brain, sleep facilitates the removal of these metabolites from the extracellular space by convective flow of the interstitial fluid from the para-arterial to the para-venous space. However, when the sleep-wake cycle is disrupted (characterized by increased brain levels of the wake-promoting neuropeptide orexin and increased neural activity), the central nervous system (CNS) clearance of extracellular metabolites is diminished. Disruptions to the sleep-wake cycle have furthermore been linked to increased neuronal oxidative stress and impaired blood-brain barrier function - conditions that have also been proposed to play a role in the development and progression of AD. Notably, recent human and transgenic animal studies have demonstrated that AD-related pathophysiological processes that occur long before the clinical onset of AD, such as Aβ deposition in the brain, disrupt sleep and circadian rhythms. Collectively, as proposed in this review, these findings suggest the existence of a mechanistic interplay between AD pathogenesis and disrupted sleep-wake cycles, which is able to accelerate the development and progression of this disease.
Collapse
Affiliation(s)
| | - Ricardo S Osorio
- Center for Brain Health, NYU Langone Medical Center, New York, NY, USA.
| | - Andrew W Varga
- NYU Sleep Disorders Center, NYU Langone Medical Center, New York, NY, USA
| | - Korey Kam
- NYU Sleep Disorders Center, NYU Langone Medical Center, New York, NY, USA
| | | | | |
Collapse
|
17
|
Bertoldi K, Cechinel LR, Schallenberger B, Meireles L, Basso C, Lovatel GA, Bernardi L, Lamers ML, Siqueira IR. Aging process alters hippocampal and cortical secretase activities of Wistar rats. Behav Brain Res 2017; 317:374-381. [DOI: 10.1016/j.bbr.2016.09.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 09/26/2016] [Accepted: 09/30/2016] [Indexed: 11/30/2022]
|
18
|
Copenhaver PF, Ramaker JM. Neuronal migration during development and the amyloid precursor protein. CURRENT OPINION IN INSECT SCIENCE 2016; 18:1-10. [PMID: 27939704 PMCID: PMC5157842 DOI: 10.1016/j.cois.2016.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/06/2016] [Indexed: 06/06/2023]
Abstract
The Amyloid Precursor Protein (APP) is the source of amyloid peptides that accumulate in Alzheimer's disease. However, members of the APP family are strongly expressed in the developing nervous systems of invertebrates and vertebrates, where they regulate neuronal guidance, synaptic remodeling, and injury responses. In contrast to mammals, insects express only one APP ortholog (APPL), simplifying investigations into its normal functions. Recent studies have shown that APPL regulates neuronal migration in the developing insect nervous system, analogous to the roles ascribed to APP family proteins in the mammalian cortex. The comparative simplicity of insect systems offers new opportunities for deciphering the signaling mechanisms by which this enigmatic class of proteins contributes to the formation and function of the nervous system.
Collapse
Affiliation(s)
- Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Jenna M Ramaker
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA; Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
19
|
Song Q, Feng G, Huang Z, Chen X, Chen Z, Ping Y. Aberrant Axonal Arborization of PDF Neurons Induced by Aβ42-Mediated JNK Activation Underlies Sleep Disturbance in an Alzheimer's Model. Mol Neurobiol 2016; 54:6317-6328. [PMID: 27718103 DOI: 10.1007/s12035-016-0165-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022]
Abstract
Impaired sleep patterns are common symptoms of Alzheimer's disease (AD). Cellular mechanisms underlying sleep disturbance in AD remain largely unknown. Here, using a Drosophila Aβ42 AD model, we show that Aβ42 markedly decreases sleep in a large population, which is accompanied with postdevelopmental axonal arborization of wake-promoting pigment-dispersing factor (PDF) neurons. The arborization is mediated in part via JNK activation and can be reversed by decreasing JNK signaling activity. Axonal arborization and impaired sleep are correlated in Aβ42 and JNK kinase hemipterous mutant flies. Image reconstruction revealed that these aberrant fibers preferentially project to pars intercerebralis (PI), a fly brain region analogous to the mammalian hypothalamus. Moreover, PDF signaling in PI neurons was found to modulate sleep/wake activities, suggesting that excessive release of PDF by these aberrant fibers may lead to the impaired sleep in Aβ42 flies. Finally, inhibition of JNK activation in Aβ42 flies restores nighttime sleep loss, decreases Aβ42 accumulation, and attenuates neurodegeneration. These data provide a new mechanism by which sleep disturbance could be induced by Aβ42 burden, a key initiator of a complex pathogenic cascade in AD.
Collapse
Affiliation(s)
- Qian Song
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.,Shanghai Key Laboratory of Psychotic Disorders (No.13dz2260500), Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ge Feng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zehua Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.,School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Xiaoman Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.,School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhaohuan Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.,School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.,Institute of Systems Biomedicine, Collaborative Innovation Center of Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China. .,Shanghai Key Laboratory of Psychotic Disorders (No.13dz2260500), Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
20
|
Dissel S, Klose M, Donlea J, Cao L, English D, Winsky-Sommerer R, van Swinderen B, Shaw PJ. Enhanced sleep reverses memory deficits and underlying pathology in Drosophila models of Alzheimer's disease. Neurobiol Sleep Circadian Rhythms 2016; 2:15-26. [PMID: 29094110 PMCID: PMC5662006 DOI: 10.1016/j.nbscr.2016.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
To test the hypothesis that sleep can reverse cognitive impairment during Alzheimer's disease, we enhanced sleep in flies either co-expressing human amyloid precursor protein and Beta-secretase (APP:BACE), or in flies expressing human tau. The ubiquitous expression of APP:BACE or human tau disrupted sleep. The sleep deficits could be reversed and sleep could be enhanced when flies were administered the GABA-A agonist 4,5,6,7-tetrahydroisoxazolo-[5,4-c]pyridine-3-ol (THIP). Expressing APP:BACE disrupted both Short-term memory (STM) and Long-term memory (LTM) as assessed using Aversive Phototaxic Suppression (APS) and courtship conditioning. Flies expressing APP:BACE also showed reduced levels of the synaptic protein discs large (DLG). Enhancing sleep in memory-impaired APP:BACE flies fully restored both STM and LTM and restored DLG levels. Sleep also restored STM to flies expressing human tau. Using live-brain imaging of individual clock neurons expressing both tau and the cAMP sensor Epac1-camps, we found that tau disrupted cAMP signaling. Importantly, enhancing sleep in flies expressing human tau restored proper cAMP signaling. Thus, we demonstrate that sleep can be used as a therapeutic to reverse deficits that accrue during the expression of toxic peptides associated with Alzheimer's disease. THIP can be used to enhance sleep in two Drosophila models of Alzheimer's disease. Enhanced sleep reverses memory deficits in fly's expressing human APP:BACE and tau. Enhanced sleep restores cAMP levels in clock neurons expressing tau. Sleep can be used as a therapeutic to reverse Alzheimer's disease related deficits.
Collapse
Affiliation(s)
- Stephane Dissel
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Markus Klose
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Jeff Donlea
- Department of Neurobiology, University of California: Los Angeles Los Angeles, California, U.S.A
| | - Lijuan Cao
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Denis English
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Faculty of Health and Medical Sciences University of Surrey Guildford Surrey, GU2 7XH, United Kingdom
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane Qld 4072 Australia
| | - Paul J Shaw
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| |
Collapse
|
21
|
Cassar M, Kretzschmar D. Analysis of Amyloid Precursor Protein Function in Drosophila melanogaster. Front Mol Neurosci 2016; 9:61. [PMID: 27507933 PMCID: PMC4960247 DOI: 10.3389/fnmol.2016.00061] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/13/2016] [Indexed: 01/10/2023] Open
Abstract
The Amyloid precursor protein (APP) has mainly been investigated in connection with its role in Alzheimer’s Disease (AD) due to its cleavage resulting in the production of the Aβ peptides that accumulate in the plaques characteristic for this disease. However, APP is an evolutionary conserved protein that is not only found in humans but also in many other species, including Drosophila, suggesting an important physiological function. Besides Aβ, several other fragments are produced by the cleavage of APP; large secreted fragments derived from the N-terminus and a small intracellular C-terminal fragment. Although these fragments have received much less attention than Aβ, a picture about their function is finally emerging. In contrast to mammals, which express three APP family members, Drosophila expresses only one APP protein called APP-like or APPL. Therefore APPL functions can be studied in flies without the complication that other APP family members may have redundant functions. Flies lacking APPL are viable but show defects in neuronal outgrowth in the central and peripheral nervous system (PNS) in addition to synaptic changes. Furthermore, APPL has been connected with axonal transport functions. In the adult nervous system, APPL, and more specifically its secreted fragments, can protect neurons from degeneration. APPL cleavage also prevents glial death. Lastly, APPL was found to be involved in behavioral deficits and in regulating sleep/activity patterns. This review, will describe the role of APPL in neuronal development and maintenance and briefly touch on its emerging function in circadian rhythms while an accompanying review will focus on its role in learning and memory formation.
Collapse
Affiliation(s)
- Marlène Cassar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
22
|
Barekat A, Gonzalez A, Mauntz RE, Kotzebue RW, Molina B, El-Mecharrafie N, Conner CJ, Garza S, Melkani GC, Joiner WJ, Lipinski MM, Finley KD, Ratliff EP. Using Drosophila as an integrated model to study mild repetitive traumatic brain injury. Sci Rep 2016; 6:25252. [PMID: 27143646 PMCID: PMC4855207 DOI: 10.1038/srep25252] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/13/2016] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of morbidity and mortality worldwide. In addition, there has been a growing appreciation that even repetitive, milder forms of TBI (mTBI) can have long-term deleterious consequences to neural tissues. Hampering our understanding of genetic and environmental factors that influence the cellular and molecular responses to injury has been the limited availability of effective genetic model systems that could be used to identify the key genes and pathways that modulate both the acute and long-term responses to TBI. Here we report the development of a severe and mild-repetitive TBI model using Drosophila. Using this system, key features that are typically found in mammalian TBI models were also identified in flies, including the activation of inflammatory and autophagy responses, increased Tau phosphorylation and neuronal defects that impair sleep-related behaviors. This novel injury paradigm demonstrates the utility of Drosophila as an effective tool to validate genetic and environmental factors that influence the whole animal response to trauma and to identify prospective therapies needed for the treatment of TBI.
Collapse
Affiliation(s)
- Ayeh Barekat
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA.,Department of Biology, San Diego State University, San Diego, CA, USA
| | - Arysa Gonzalez
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - Ruth E Mauntz
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - Roxanne W Kotzebue
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA.,Department of Biology, San Diego State University, San Diego, CA, USA
| | - Brandon Molina
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA.,Department of Biology, San Diego State University, San Diego, CA, USA
| | - Nadja El-Mecharrafie
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA.,Department of Biology, San Diego State University, San Diego, CA, USA
| | | | - Shannon Garza
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Girish C Melkani
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - William J Joiner
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Marta M Lipinski
- Shock, Trauma, and Anesthesiology Research (STAR) Center; Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kim D Finley
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA.,Department of Biology, San Diego State University, San Diego, CA, USA.,Department of Chemistry, San Diego State University, San Diego, CA, USA
| | - Eric P Ratliff
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA.,Department of Biology, San Diego State University, San Diego, CA, USA.,Department of Chemistry, San Diego State University, San Diego, CA, USA
| |
Collapse
|