1
|
Drew VJ, Wang C, Kim T. Progressive sleep disturbance in various transgenic mouse models of Alzheimer's disease. Front Aging Neurosci 2023; 15:1119810. [PMID: 37273656 PMCID: PMC10235623 DOI: 10.3389/fnagi.2023.1119810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia. The relationship between AD and sleep dysfunction has received increased attention over the past decade. The use of genetically engineered mouse models with enhanced production of amyloid beta (Aβ) or hyperphosphorylated tau has played a critical role in the understanding of the pathophysiology of AD. However, their revelations regarding the progression of sleep impairment in AD have been highly dependent on the mouse model used and the specific techniques employed to examine sleep. Here, we discuss the sleep disturbances and general pathology of 15 mouse models of AD. Sleep disturbances covered in this review include changes to NREM and REM sleep duration, bout lengths, bout counts and power spectra. Our aim is to describe in detail the severity and chronology of sleep disturbances within individual mouse models of AD, as well as reveal broader trends of sleep deterioration that are shared among most models. This review also explores a variety of potential mechanisms relating Aβ accumulation and tau neurofibrillary tangles to the progressive deterioration of sleep observed in AD. Lastly, this review offers perspective on how study design might impact our current understanding of sleep disturbances in AD and provides strategies for future research.
Collapse
Affiliation(s)
- Victor J. Drew
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Chanung Wang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
2
|
Plucińska K, Mody N, Dekeryte R, Shearer K, Mcilroy GD, Delibegovic M, Platt B. High-fat diet exacerbates cognitive and metabolic abnormalities in neuronal BACE1 knock-in mice - partial prevention by Fenretinide. Nutr Neurosci 2022; 25:719-736. [PMID: 32862802 DOI: 10.1080/1028415x.2020.1806190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Objective: The β-site APP-cleaving enzyme 1 (BACE1) is a rate-limiting step in β-amyloid (Aβ) production in Alzheimer's disease (AD) brains, but recent evidence suggests that BACE1 is also involved in metabolic regulation. Here, we aimed to assess the effects of highfat diet (HFD) on metabolic and cognitive phenotypes in the diabetic BACE1 knock-in mice (PLB4) and WT controls; we additionally examined whether these phenotypes can be normalized with a synthetic retinoid (Fenretinide, Fen) targeting weight loss.Methods: Five-month old male WT and PLB4 mice were fed either (1) control chow diet, (2) 45%-saturated fat diet (HFD), (3) HFD with 0.04% Fen (HFD + Fen) or (4) control chow diet with 0.04% Fen (Fen) for 10 weeks. We assessed basic metabolic parameters, circadian rhythmicity, spatial habituation (Phenotyper) and working memory (Y-maze). Hypothalami, forebrain and liver tissues were assessed using Western blots, qPCR and ELISAs.Results: HFD feeding drastically worsened metabolism and induced early mortality (-40%) in otherwise viable PLB4 mice. This was ameliorated by Fen, despite no effects on glucose intolerance. In HFD-fed WT mice, Fen reduced weight gain, glucose intolerance and hepatic steatosis. The physiological changes induced in WT and PLB4 mice by HFD (+/-Fen) were accompanied by enhanced cerebral astrogliosis, elevated PTP1B, phopsho-eIF2α and altered hypothalamic transcription of Bace1, Pomc and Mc4r. Behaviourally, HFD feeding exacerbated spatial memory deficits in PLB4 mice, which was prevented by Fen and linked with increased full-length APP, normalized brain Aβ*56 oligomerization and astrogliosis.Conclusions: HFD induces early mortality and worsened cognition in the Alzheimer's-like BACE1 mice- partial prevention was achieved with Fenretinide, without improvements in glucose homeostasis.
Collapse
Affiliation(s)
- Kaja Plucińska
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- The Novo Nordisk Foundation Center for Basic Metabolic Research (CBMR), Integrative Physiology and Environmental Influences, University of Copenhagen, Copenhagen, Denmark
| | - Nimesh Mody
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Ruta Dekeryte
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Kirsty Shearer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - George D Mcilroy
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Mirela Delibegovic
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Bettina Platt
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
3
|
IL-6 and IL-1β upregulation and tau protein phosphorylation in response to chronic alcohol exposure in the mouse hippocampus. Neuroreport 2021; 32:851-857. [PMID: 34029285 DOI: 10.1097/wnr.0000000000001661] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alcoholism and alcohol abuse can lead to memory loss and cognitive dysfunction. The neuroinflammatory response plays an important role in the neurotoxic mechanism of chronic alcohol exposure. Additionally, the phosphorylation status of the tau protein is closely related to neurotoxicity and synaptic function. As inflammatory cytokines have been shown to regulate tau phosphorylation, in the present study, the aim was to determine whether cognitive impairment caused by chronic alcohol exposure is associated with neuroinflammation and tau hyperphosphorylation in the hippocampus. We established a chronic alcohol exposure model of C57BL/6J mice. The Y maze was used to assess the spatial recognition ability of mice, and ELISA was used to detect the levels of inflammatory cytokines IL-1β and IL-6 in the serum. Immunohistochemical and western blot assays were used to assess the expression levels of IL-1β and IL-6, as well as tau protein and its phosphorylation status in the hippocampus. We also analyzed the mRNA and protein expression of the synapse-associated proteins PSD95 and synaptophysin in the hippocampus. Our results showed that chronic alcohol exposure impaired the spatial recognition ability of mice upregulated the expression of IL-1β and IL-6 in the serum and hippocampus and increased the phosphorylation of tau protein in the hippocampus. In addition, chronic alcohol exposure downregulated PSD95 and synaptophysin protein levels. The present results indicate that hippocampal IL-1β, IL-6, and phosphorylated tau proteins may be involved in the neurotoxic mechanism of chronic alcohol exposure by mediating synaptic dysfunction.
Collapse
|
4
|
Li Y, Ding R, Ren X, Wen G, Dong Z, Yao H, Tan Y, Yu H, Wang X, Zhan X, Yao J, Lu Y, Zhang G, Wu X. Long-term ketamine administration causes Tau protein phosphorylation and Tau protein-dependent AMPA receptor reduction in the hippocampus of mice. Toxicol Lett 2019; 315:107-115. [PMID: 31470060 DOI: 10.1016/j.toxlet.2019.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/19/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022]
Abstract
As a recreational drug of abuse and an injectable anesthetic, ketamine has been shown to cause cognitive dysfunction and induce psychotic states. Although the specific mechanism is still unclear, it may be linked to synaptic receptors, including the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor. Recent evidence suggests that Tau protein phosphorylation and targeted delivery to the postsynaptic area is involved in maintaining neuronal plasticity, indicating that the neurotoxicity induced by ketamine may be related to the transfer of Tau protein after phosphorylation. In this study, we established a model of long-term (6 months) ketamine administration in wild-type (C57BL/6) and Tau knockout mice to investigate the effects of different doses of ketamine administration on Tau protein expression and phosphorylation in the mouse hippocampus. We also investigated changes in AMPA receptor expression in the synaptic membrane of wild-type and Tau knockout mice. Our results showed that long-term ketamine administration led to excessive Tau protein phosphorylation at Ser202/Thr205 and Ser396, but not at Ser199, Ser262 and Ser404. Most importantly, long-term ketamine administration decreased AMPA receptor levels in the hippocampal cell membrane in a Tau protein-dependent manner. Our results reveal the role of Tau protein phosphorylation in the mechanism of ketamine neurotoxicity, suggesting that the changes of membrane AMPA receptor and synaptic function induced by ketamine are mediated by abnormal phosphorylation of Tau protein at specific sites.
Collapse
Affiliation(s)
- Yanning Li
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China; Department of Forensic Medicine, School of Basic Medicine, Gannan Medical University, Ganzhou 341000, PR China
| | - Runtao Ding
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Zhibin Dong
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Hui Yao
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Yaqing Tan
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Hao Yu
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xiaolong Wang
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xiaoni Zhan
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, the Affiliated Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Guohua Zhang
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China.
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
5
|
Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, Ye S, Ye K, Wei D, Song Z, Chen D, Liu J. Fecal microbiota transplantation alleviated Alzheimer's disease-like pathogenesis in APP/PS1 transgenic mice. Transl Psychiatry 2019; 9:189. [PMID: 31383855 PMCID: PMC6683152 DOI: 10.1038/s41398-019-0525-3] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/22/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common dementia in the elderly. Treatment for AD is still a difficult task in clinic. AD is associated with abnormal gut microbiota. However, little is known about the role of fecal microbiota transplantation (FMT) in AD. Here, we evaluated the efficacy of FMT for the treatment of AD. We used an APPswe/PS1dE9 transgenic (Tg) mouse model. Cognitive deficits, brain deposits of amyloid-β (Aβ) and phosphorylation of tau, synaptic plasticity as well as neuroinflammation were assessed. Gut microbiota and its metabolites short-chain fatty acids (SCFAs) were analyzed by 16S rRNA sequencing and 1H nuclear magnetic resonance (NMR). Our results showed that FMT treatment could improve cognitive deficits and reduce the brain deposition of amyloid-β (Aβ) in APPswe/PS1dE9 transgenic (Tg) mice. These improvements were accompanied by decreased phosphorylation of tau protein and the levels of Aβ40 and Aβ42. We observed an increases in synaptic plasticity in the Tg mice, showing that postsynaptic density protein 95 (PSD-95) and synapsin I expression were increased after FMT. We also observed the decrease of COX-2 and CD11b levels in Tg mice after FMT. We also found that FMT treatment reversed the changes of gut microbiota and SCFAs. Thus, FMT may be a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Jing Sun
- 0000 0004 1764 2632grid.417384.dDepartment of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027 China
| | - Jingxuan Xu
- 0000 0004 1764 2632grid.417384.dDepartment of General Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027 China
| | - Yi Ling
- 0000 0004 1764 2632grid.417384.dDepartment of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027 China
| | - Fangyan Wang
- 0000 0001 0348 3990grid.268099.cDepartment of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Tianyu Gong
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Changwei Yang
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Shiqing Ye
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Keyue Ye
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Dianhui Wei
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Ziqing Song
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Danna Chen
- 0000 0001 0348 3990grid.268099.cDepartment of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Jiaming Liu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China. .,Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
6
|
Plucińska K, Crouch B, Yeap JM, Stoppelkamp S, Riedel G, Platt B. Histological and Behavioral Phenotypes of a Novel Mutated APP Knock-In Mouse. J Alzheimers Dis 2019; 65:165-180. [PMID: 30040726 DOI: 10.3233/jad-180336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene mutations within amyloid precursor protein (APP or AβPP) and/or presenilin 1 (PS1) genes are determinants of familial Alzheimer's disease (fAD) and remain fundamental for experimental models. Here, we generated a neuronal knock-in mouse (PLB2APP) with mutated human APPSwe/Lon and investigated histopathology and behavioral phenotypes. Additionally, PLB2APP mice were cross-bred with a presenilin (PS1A246E) line to assess the impact of this gene combination. Immunohistochemistry determined amyloid-β (Aβ) pathology, astrogliosis (via GFAP labelling), and neuronal densities in hippocampal and cortical brain regions. One-year old PLB2APP mice showed higher levels of intracellular Aβ in CA1, dentate gyrus, and cortical regions compared to PLBWT controls. Co-expression of PS1 reduced hippocampal but elevated cortical Aβ build-up. Amyloid plaques were sparse in aged PLB2APP mice, and co-expression of PS1 promoted plaque formation. Heightened GFAP expression followed the region-specific pattern of Aβ in PLB2APP and PLB2APP/PS1 mice. Behaviorally, habituation to a novel environment was delayed in 6-month-old PLB2APP mice, and overall home-cage activity was reduced in both lines at 6 and 12 months, particularly during the dark phase. Spatial learning in the water maze was impaired in PLB2APP mice independent of PS1 expression and associated with reduced spatial navigation strategies. Memory retrieval was compromised in PLB2APP mice only. Our data demonstrate that low expression of APP is sufficient to drive histopathological and cognitive changes in mice without overexpression or excessive plaque deposition. AD-like phenotypes were altered by co-expression of PS1, including a shift from hippocampal to cortical Aβ pathology, alongside reduced deficits in spatial learning.
Collapse
Affiliation(s)
- Kaja Plucińska
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Barry Crouch
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Jie M Yeap
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Sandra Stoppelkamp
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Bettina Platt
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
7
|
Naseri NN, Wang H, Guo J, Sharma M, Luo W. The complexity of tau in Alzheimer's disease. Neurosci Lett 2019; 705:183-194. [PMID: 31028844 PMCID: PMC7060758 DOI: 10.1016/j.neulet.2019.04.022] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is characterized by two major pathological lesions in the brain, amyloid plaques and neurofibrillary tangles (NFTs) composed mainly of amyloid-β (Aβ) peptides and hyperphosphorylated tau, respectively. Although accumulation of toxic Aβ species in the brain has been proposed as one of the important early events in AD, continued lack of success of clinical trials based on Aβ-targeting drugs has triggered the field to seek out alternative disease mechanisms and related therapeutic strategies. One of the new approaches is to uncover novel roles of pathological tau during disease progression. This review will primarily focus on recent advances in understanding the contributions of tau to AD.
Collapse
Affiliation(s)
- Nima N Naseri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| | - Hong Wang
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, USA
| | - Jennifer Guo
- The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
8
|
Bouter C, Bouter Y. 18F-FDG-PET in Mouse Models of Alzheimer's Disease. Front Med (Lausanne) 2019; 6:71. [PMID: 31058151 PMCID: PMC6482246 DOI: 10.3389/fmed.2019.00071] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/21/2019] [Indexed: 01/08/2023] Open
Abstract
Suitable animal models and in vivo biomarkers are essential for development and evaluation of new therapeutic strategies in Alzheimer's disease (AD). 18F-Fluorodeoxyglucose (18F-FDG)-positron-emission tomography (PET) is an imaging biomarker that allows the assessment of cerebral glucose metabolism in vivo. While 18F-FDG-PET/CT is an established tool in the evaluation of AD patients, its role in preclinical studies with AD mouse models remains unclear. Here, we want to review available studies on 18F-FDG-PET/CT in AD mouse models in order to evaluate the method and its impact in preclinical AD research. Only a limited number of studies using 18F-FDG-PET in AD mice were carried out so far showing contradictory findings in cerebral FDG uptake. Methodological differences as well as underlying pathological features of used mouse models seem to be accountable for those varying results. However, 18F-FDG-PET can be a valuable tool in longitudinal in vivo therapy monitoring with a lot of potential for future studies.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| |
Collapse
|
9
|
Sun J, Liu S, Ling Z, Wang F, Ling Y, Gong T, Fang N, Ye S, Si J, Liu J. Fructooligosaccharides Ameliorating Cognitive Deficits and Neurodegeneration in APP/PS1 Transgenic Mice through Modulating Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3006-3017. [PMID: 30816709 DOI: 10.1021/acs.jafc.8b07313] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Alzheimer's disease (AD) is closely related to gut microbial alteration. Prebiotic fructooligosaccharides (FOS) play major roles by regulating gut microbiota. The present study aimed to explore the effect and mechanism of FOS protection against AD via regulating gut microbiota. Male Apse/PSEN 1dE9 (APP/PS1) transgenic (Tg) mice were administrated with FOS for 6 weeks. Cognitive deficits and amyloid deposition were evaluated. The levels of synaptic plasticity markers including postsynaptic density protein 95 (PSD-95) and synapsin I, as well as phosphorylation of c-Jun N-terminal kinase (JNK), were determined. The intestinal microbial constituent was detected by 16S rRNA sequencing. Moreover, the levels of glucagon-like peptide-1 (GLP-1) in the gut and GLP-1 receptor (GLP-1R) in the brain were measured. The results indicated that FOS treatment ameliorated cognitive deficits and pathological changes in the Tg mice. FOS significantly upregulated the expression levels of synapsin I and PSD-95, as well as decreased phosphorylated level of JNK. The sequencing results showed that FOS reversed the altered microbial composition. Furthermore, FOS increased the level of GLP-1 and decreased the level of GLP-1R in the Tg mice. These findings indicated that FOS exerted beneficial effects against AD via regulating the gut microbiota-GLP-1/GLP-1R pathway.
Collapse
Affiliation(s)
- Jing Sun
- Department of Neurology , the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , Zhejiang 325027 , China
| | - Suzhi Liu
- Department of Neurology, The Affiliated Taizhou Hospital , Wenzhou Medical University , 150# Ximen Road , Linhai District, Taizhou 317000 , Zhejiang China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , Zhejiang 310003 , China
| | - Fangyan Wang
- Departments of Pathophysiology, School of Basic Medicine Science , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Yi Ling
- Department of Neurology , the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , Zhejiang 325027 , China
| | - Tianyu Gong
- Department of Preventive Medicine, School of Public Health and Management , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Na Fang
- Department of Preventive Medicine, School of Public Health and Management , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Shiqing Ye
- Department of Preventive Medicine, School of Public Health and Management , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Jue Si
- Department of Preventive Medicine, School of Public Health and Management , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Jiaming Liu
- Department of Neurology , the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , Zhejiang 325027 , China
- Department of Preventive Medicine, School of Public Health and Management , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| |
Collapse
|
10
|
Ueda A, Woods S, McElree I, O'Harrow TCDG, Inman C, Thenuwara S, Aftab M, Iyengar A. Two novel forms of ERG oscillation in Drosophila: age and activity dependence. J Neurogenet 2018; 32:118-126. [PMID: 29688104 DOI: 10.1080/01677063.2018.1461866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Over an animal's lifespan, neuronal circuits and systems often decline in an inherently heterogeneous fashion. To compare the age-dependent progression of changes in visual behavior with alterations in retinal physiology, we examined phototaxis and electroretinograms (ERGs) in a wild-type D. melanogaster strain (Canton-S) across their lifespan. In aged flies (beyond 50% median lifespan), we found a marked decline in phototaxis, while motor coordination was less disrupted, as indicated by relatively stronger negative geotaxis. These aged flies displayed substantially reduced ERG transient amplitudes while the receptor potentials (RP) remained largely intact. Using a repetitive light flash protocol, we serendipitously discovered two forms of activity-dependent oscillation in the ERG waveforms of young flies: 'light-off' and 'light-on' oscillations. After repeated 500 ms light flashes, light-off oscillations appeared during the ERG off-transients (frequency: 50-120 Hz, amplitude: ∼1 mV). Light-on oscillations (100-200 Hz, ∼0.3 mV) were induced by a series of 50 ms flashes, and were evident during the ERG on-transients. Both forms of oscillation were observed in other strains of D. melanogaster (Oregon-R, Berlin), additional Drosophila species (D. funerbris, D. euronotus, D. hydei, D. americana), and were evoked by a variety of light sources. Both light-off and light-on oscillations were distinct from previously described ERG oscillations in the visual mutant rosA in terms of location within the waveform and frequency. However, within rosA mutants, light-off oscillations, but not light-on oscillations could be recruited by the repetitive light flash protocol. Importantly though, we found that both forms of oscillation were rarely observed in aged flies. Although the physiological bases of these oscillations remain to be elucidated, they may provide important clues to age-related changes in neuronal excitability and synaptic transmission.
Collapse
Affiliation(s)
- Atsushi Ueda
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Scott Woods
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Ian McElree
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | | | - Casey Inman
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | | | - Muhammad Aftab
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Atulya Iyengar
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| |
Collapse
|
11
|
Dietrich K, Bouter Y, Müller M, Bayer TA. Synaptic Alterations in Mouse Models for Alzheimer Disease-A Special Focus on N-Truncated Abeta 4-42. Molecules 2018; 23:E718. [PMID: 29561816 PMCID: PMC6017701 DOI: 10.3390/molecules23040718] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 11/29/2022] Open
Abstract
This commentary reviews the role of the Alzheimer amyloid peptide Aβ on basal synaptic transmission, synaptic short-term plasticity, as well as short- and long-term potentiation in transgenic mice, with a special focus on N-terminal truncated Aβ4-42. Aβ4-42 is highly abundant in the brain of Alzheimer's disease (AD) patients. It demonstrates increased neurotoxicity compared to full length Aβ, suggesting an important role in the pathogenesis of AD. Transgenic Tg4-42 mice, a model for sporadic AD, express human Aβ4-42 in Cornu Ammonis (CA1) neurons, and develop age-dependent hippocampal neuron loss and neurological deficits. In contrast to other transgenic AD mouse models, the Tg4-42 model exhibits synaptic hyperexcitability, altered synaptic short-term plasticity with no alterations in short- and long-term potentiation. The outcomes of this study are discussed in comparison with controversial results from other AD mouse models.
Collapse
Affiliation(s)
- Katharina Dietrich
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, von-Siebold-Strasse 5, 37075 Göttingen, Germany.
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, von-Siebold-Strasse 5, 37075 Göttingen, Germany.
| | - Michael Müller
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Humboldtallee 23, 37073 Göttingen, Germany.
- Center for Physiology and Pathophysiology, Institute for Neuro- and Sense Physiology, University Medical Center (UMG), Georg-August-University, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, von-Siebold-Strasse 5, 37075 Göttingen, Germany.
| |
Collapse
|
12
|
Foster TC, Kyritsopoulos C, Kumar A. Central role for NMDA receptors in redox mediated impairment of synaptic function during aging and Alzheimer's disease. Behav Brain Res 2016; 322:223-232. [PMID: 27180169 DOI: 10.1016/j.bbr.2016.05.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/15/2016] [Accepted: 05/05/2016] [Indexed: 01/07/2023]
Abstract
Increased human longevity has magnified the negative impact that aging can have on cognitive integrity of older individuals experiencing some decline in cognitive function. Approximately 30% of the elderly will have cognitive problems that influence their independence. Impaired executive function and memory performance are observed in normal aging and yet can be an early sign of a progressive cognitive impairment of Alzheimer's disease (AD), the most common form of dementia. Brain regions that are vulnerable to aging exhibit the earliest pathology of AD. Senescent synaptic function is observed as a shift in Ca2+-dependent synaptic plasticity and similar mechanisms are thought to contribute to the early cognitive deficits associated with AD. In the case of aging, intracellular redox state mediates a shift in Ca2+ regulation including N-methyl-d-aspartate (NMDA) receptor hypofunction and increased Ca2+ release from intracellular stores to alter synaptic plasticity. AD can interact with these aging processes such that molecules linked to AD, β-amyloid (Aβ) and mutated presenilin 1 (PS1), can also degrade NMDA receptor function, promote Ca2+ release from intracellular stores, and may increase oxidative stress. Thus, age is one of the most important predictors of AD and brain aging likely contributes to the onset of AD. The focus of this review article is to provide an update on mechanisms that contribute to the senescent synapse and possible interactions with AD-related molecules, with special emphasis on regulation of NMDA receptors.
Collapse
Affiliation(s)
- T C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, United States of America.
| | - C Kyritsopoulos
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, United States of America
| | - A Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, United States of America.
| |
Collapse
|
13
|
Koss DJ, Robinson L, Drever BD, Plucińska K, Stoppelkamp S, Veselcic P, Riedel G, Platt B. Mutant Tau knock-in mice display frontotemporal dementia relevant behaviour and histopathology. Neurobiol Dis 2016; 91:105-23. [PMID: 26949217 DOI: 10.1016/j.nbd.2016.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/26/2016] [Accepted: 03/02/2016] [Indexed: 10/22/2022] Open
Abstract
Models of Tau pathology related to frontotemporal dementia (FTD) are essential to determine underlying neurodegenerative pathologies and resulting tauopathy relevant behavioural changes. However, existing models are often limited in their translational value due to Tau overexpression, and the frequent occurrence of motor deficits which prevent comprehensive behavioural assessments. In order to address these limitations, a forebrain-specific (CaMKIIα promoter), human mutated Tau (hTauP301L+R406W) knock-in mouse was generated out of the previously characterised PLB1Triple mouse, and named PLB2Tau. After confirmation of an additional hTau species (~60kDa) in forebrain samples, we identified age-dependent progressive Tau phosphorylation which coincided with the emergence of FTD relevant behavioural traits. In line with the non-cognitive symptomatology of FTD, PLB2Tau mice demonstrated early emerging (~6months) phenotypes of heightened anxiety in the elevated plus maze, depressive/apathetic behaviour in a sucrose preference test and generally reduced exploratory activity in the absence of motor impairments. Investigations of cognitive performance indicated prominent dysfunctions in semantic memory, as assessed by social transmission of food preference, and in behavioural flexibility during spatial reversal learning in a home cage corner-learning task. Spatial learning was only mildly affected and task-specific, with impairments at 12months of age in the corner learning but not in the water maze task. Electroencephalographic (EEG) investigations indicated a vigilance-stage specific loss of alpha power during wakefulness at both parietal and prefrontal recording sites, and site-specific EEG changes during non-rapid eye movement sleep (prefrontal) and rapid eye movement sleep (parietal). Further investigation of hippocampal electrophysiology conducted in slice preparations indicated a modest reduction in efficacy of synaptic transmission in the absence of altered synaptic plasticity. Together, our data demonstrate that the transgenic PLB2Tau mouse model presents with a striking behavioural and physiological face validity relevant for FTD, driven by the low level expression of mutant FTD hTau.
Collapse
Affiliation(s)
- David J Koss
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Lianne Robinson
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Benjamin D Drever
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Kaja Plucińska
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Sandra Stoppelkamp
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter Veselcic
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Gernot Riedel
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | - Bettina Platt
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
14
|
Koss DJ, Robinson L, Mietelska-Porowska A, Gasiorowska A, Sepčić K, Turk T, Jaspars M, Niewiadomska G, Scott RH, Platt B, Riedel G. Polymeric alkylpyridinium salts permit intracellular delivery of human Tau in rat hippocampal neurons: requirement of Tau phosphorylation for functional deficits. Cell Mol Life Sci 2015; 72:4613-32. [PMID: 26070304 PMCID: PMC11113860 DOI: 10.1007/s00018-015-1949-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/13/2015] [Accepted: 06/03/2015] [Indexed: 11/27/2022]
Abstract
Patients suffering from tauopathies including frontotemporal dementia (FTD) and Alzheimer's disease (AD) present with intra-neuronal aggregation of microtubule-associated protein Tau. During the disease process, Tau undergoes excessive phosphorylation, dissociates from microtubules and aggregates into insoluble neurofibrillary tangles (NFTs), accumulating in the soma. While many aspects of the disease pathology have been replicated in transgenic mouse models, a region-specific non-transgenic expression model is missing. Complementing existing models, we here report a novel region-specific approach to modelling Tau pathology. Local co-administration of the pore-former polymeric 1,3-alkylpyridinium salts (Poly-APS) extracted from marine sponges, and synthetic full-length 4R recombinant human Tau (hTau) was performed in vitro and in vivo. At low doses, Poly-APS was non-toxic and cultured cells exposed to Poly-APS (0.5 µg/ml) and hTau (1 µg/ml; ~22 µM) had normal input resistance, resting-state membrane potentials and Ca(2+) transients induced either by glutamate or KCl, as did cells exposed to a low concentration of the phosphatase inhibitor Okadaic acid (OA; 1 nM, 24 h). Combined hTau loading and phosphatase inhibition resulted in a collapse of the membrane potential, suppressed excitation and diminished glutamate and KCl-stimulated Ca(2+) transients. Stereotaxic infusions of Poly-APS (0.005 µg/ml) and hTau (1 µg/ml) bilaterally into the dorsal hippocampus at multiple sites resulted in hTau loading of neurons in rats. A separate cohort received an additional 7-day minipump infusion of OA (1.2 nM) intrahippocampally. When tested 2 weeks after surgery, rats treated with Poly-APS+hTau+OA presented with subtle learning deficits, but were also impaired in cognitive flexibility and recall. Hippocampal plasticity recorded from slices ex vivo was diminished in Poly-APS+hTau+OA subjects, but not in other treatment groups. Histological sections confirmed the intracellular accumulation of hTau in CA1 pyramidal cells and along their processes; phosphorylated Tau was present only within somata. This study demonstrates that cognitive, physiological and pathological symptoms reminiscent of tauopathies can be induced following non-mutant hTau delivery into CA1 in rats, but functional consequences hinge on increased Tau phosphorylation. Collectively, these data validate a novel model of locally infused recombinant hTau protein as an inducer of Tau pathology in the hippocampus of normal rats; future studies will provide insights into the pathological spread and maturation of Tau pathology.
Collapse
Affiliation(s)
- Dave J Koss
- School of Medical Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK
| | - Lianne Robinson
- School of Medical Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK
- Behavioural Neuroscience Core Facility, Division of Neuroscience, University of Dundee, Dundee, UK
| | | | - Anna Gasiorowska
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- Mossakowski Medical Research Centre, Warsaw, Poland
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tom Turk
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Marcel Jaspars
- Department of Chemistry, Marine Biodiscovery Centre, University of Aberdeen, Aberdeen, UK
| | - Grazyna Niewiadomska
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Roderick H Scott
- School of Medical Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK
| | - Bettina Platt
- School of Medical Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK
| | - Gernot Riedel
- School of Medical Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK.
| |
Collapse
|
15
|
Jyoti A, Plano A, Riedel G, Platt B. Progressive age-related changes in sleep and EEG profiles in the PLB1Triple mouse model of Alzheimer’s disease. Neurobiol Aging 2015; 36:2768-84. [DOI: 10.1016/j.neurobiolaging.2015.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 05/05/2015] [Accepted: 07/02/2015] [Indexed: 12/01/2022]
|
16
|
Melis V, Zabke C, Stamer K, Magbagbeolu M, Schwab K, Marschall P, Veh RW, Bachmann S, Deiana S, Moreau PH, Davidson K, Harrington KA, Rickard JE, Horsley D, Garman R, Mazurkiewicz M, Niewiadomska G, Wischik CM, Harrington CR, Riedel G, Theuring F. Different pathways of molecular pathophysiology underlie cognitive and motor tauopathy phenotypes in transgenic models for Alzheimer's disease and frontotemporal lobar degeneration. Cell Mol Life Sci 2015; 72:2199-222. [PMID: 25523019 PMCID: PMC4427622 DOI: 10.1007/s00018-014-1804-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/22/2014] [Accepted: 12/08/2014] [Indexed: 11/04/2022]
Abstract
A poorly understood feature of the tauopathies is their very different clinical presentations. The frontotemporal lobar degeneration (FTLD) spectrum is dominated by motor and emotional/psychiatric abnormalities, whereas cognitive and memory deficits are prominent in the early stages of Alzheimer's disease (AD). We report two novel mouse models overexpressing different human tau protein constructs. One is a full-length tau carrying a double mutation [P301S/G335D; line 66 (L66)] and the second is a truncated 3-repeat tau fragment which constitutes the bulk of the PHF core in AD corresponding to residues 296-390 fused with a signal sequence targeting it to the endoplasmic reticulum membrane (line 1; L1). L66 has abundant tau pathology widely distributed throughout the brain, with particularly high counts of affected neurons in hippocampus and entorhinal cortex. The pathology is neuroanatomically static and declines with age. Behaviourally, the model is devoid of a higher cognitive phenotype but presents with sensorimotor impairments and motor learning phenotypes. L1 displays a much weaker histopathological phenotype, but shows evidence of neuroanatomical spread and amplification with age that resembles the Braak staging of AD. Behaviourally, the model has minimal motor deficits but shows severe cognitive impairments affecting particularly the rodent equivalent of episodic memory which progresses with advancing age. In both models, tau aggregation can be dissociated from abnormal phosphorylation. The two models make possible the demonstration of two distinct but nevertheless convergent pathways of tau molecular pathogenesis. L1 appears to be useful for modelling the cognitive impairment of AD, whereas L66 appears to be more useful for modelling the motor features of the FTLD spectrum. Differences in clinical presentation of AD-like and FTLD syndromes are therefore likely to be inherent to the respective underlying tauopathy, and are not dependent on presence or absence of concomitant APP pathology.
Collapse
Affiliation(s)
- V. Melis
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - C. Zabke
- CCR/Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Germany
| | - K. Stamer
- CCR/Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Germany
| | - M. Magbagbeolu
- CCR/Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Germany
| | - K. Schwab
- CCR/Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Germany
| | - P. Marschall
- CCR/Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Germany
| | - R. W. Veh
- Charité-Universitätsmedizin Berlin, Institute of Anatomy, Berlin, Germany
| | - S. Bachmann
- Charité-Universitätsmedizin Berlin, Institute of Anatomy, Berlin, Germany
| | - S. Deiana
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - P.-H. Moreau
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - K. Davidson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - K. A. Harrington
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - J. E. Rickard
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - D. Horsley
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - R. Garman
- Consultants in Veterinary Pathology Inc., Murrysville, PA USA
| | | | | | - C. M. Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - C. R. Harrington
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, Scotland, UK
| | - G. Riedel
- School of Medical Sciences, University of Aberdeen, Foresterhill, AB25 2ZD Scotland, UK
| | - F. Theuring
- CCR/Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Germany
| |
Collapse
|
17
|
Abstract
Key neuropathological hallmarks of Alzheimer's disease (AD) are elevated levels of amyloid β-peptide (Aβ) species generated via amyloid precursor protein (APP) endoproteolysis and cleavage by the rate-limiting β-site enzyme 1 (BACE1). Because rodents do not develop amyloid pathologies, we here investigated whether AD-like endophenotypes can be created in mice by expression of human bace1. To avoid pitfalls of existing models, we introduced hbace1 via knock-in under the control of the CaMKII α promoter into the safe HPRT locus. We report amyloidogenic processing of murine APP in the hBACE1 mice (termed PLB4), resulting in the formation of toxic APP metabolites that accumulate intra- and extraneuronally in hippocampus and cortex. Pronounced accumulation of Aβ*56 and Aβ hexamers in the absence of plaque deposition was detected in brain tissue from symptomatic PLB4 mice. Heightened levels of inflammation (gliosis) also appeared in several AD-related brain regions (dentate gyrus, hippocampal area CA1, piriform and parietal cortices) at 6 and 12 months of age. Behaviorally, deficits in habituation to a novel environment and semantic-like memory (social transmission of food preference) were detected from 3 to 4 months of age. Impairments in spatial learning strategies in long-term reference (water maze) and working memory (Y-maze) tasks presented at 6 months, and were distinct from reductions in locomotor activity and anxiety. Overall, our data indicate for the first time that targeted, subtle forebrain-specific expression through single gene knock-in of hBACE1 is sufficient to generate AD-relevant cognitive impairments amid corresponding histopathologies, confirming human BACE as the key parameter in amyloid pathogenesis.
Collapse
|
18
|
Ryan D, Koss D, Porcu E, Woodcock H, Robinson L, Platt B, Riedel G. Spatial learning impairments in PLB1Triple knock-in Alzheimer mice are task-specific and age-dependent. Cell Mol Life Sci 2013; 70:2603-19. [PMID: 23535719 PMCID: PMC11113905 DOI: 10.1007/s00018-013-1314-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 02/27/2013] [Accepted: 02/28/2013] [Indexed: 12/21/2022]
Abstract
We recently generated an advanced mouse model of Alzheimer's disease (AD) by targeted knock-in of single-copy mutated human amyloid precursor-protein (APP) and tau genes, crossed with a non-symptomatic presenilin (PS1A246E) over-expressing mouse line. These PLB1Triple mice presented with age-dependent and AD-relevant phenotypes. Homozygous PLB1Triple mice aged 4-12 months were assessed here in a battery of spatial learning tasks: Exp.1 radial-arm water maze (spatial reference and working memory) Exp.2 open-field water maze (spatial reference memory); Exp.3 home cage observation system with spatial learning (IntelliCage); Exp.4 spontaneous object recognition (SOR; novel object and spatial object shift). A separate test with high-expression transgenic APP mice matching the design of experiment 1 was also performed. Spatial deficits in PLB1Triple mice were confirmed at 12, but not 4 months in both water maze tasks. PSAPP mice, by contrast, presented with severe yet non-progressive spatial learning deficits already at 4 months. During tests of spatial learning in SOR and IntelliCage, PLB1Triple mice neither acquired the location of the water-rewarded corner, nor recognize novel or spatially shifted objects at 4 months, indicating these protocols to be more sensitive than the water maze. Collectively and in line with AD symptomatology, PLB1Triple mice present with a graded and progressive age-dependent loss of spatial memory that can be revealed by the use of a battery of tasks. With the emergence of subtle deficits progressively increasing in severity, PLB1Triple mice may offer a more patho-physiologically relevant model of dementia than aggressive expression models.
Collapse
Affiliation(s)
- D. Ryan
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - D. Koss
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - E. Porcu
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - H. Woodcock
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - L. Robinson
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - B. Platt
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - G. Riedel
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| |
Collapse
|