1
|
O'Connell A, Quinlan L, Kwakowsky A. β-amyloid's neurotoxic mechanisms as defined by in vitro microelectrode arrays: a review. Pharmacol Res 2024; 209:107436. [PMID: 39369863 DOI: 10.1016/j.phrs.2024.107436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Alzheimer's disease is characterized by the aggregation of β-amyloid, a pathological feature believed to drive the neuronal loss and cognitive decline commonly seen in the disease. Given the growing prevalence of this progressive neurodegenerative disease, understanding the exact mechanisms underlying this process has become a top priority. Microelectrode arrays are commonly used for chronic, non-invasive recording of both spontaneous and evoked neuronal activity from diverse in vitro disease models and to evaluate therapeutic or toxic compounds. To date, microelectrode arrays have been used to investigate β-amyloids' toxic effects, β-amyloids role in specific pathological features and to assess pharmacological approaches to treat Alzheimer's disease. The versatility of microelectrode arrays means these studies use a variety of methods and investigate different disease models and brain regions. This review provides an overview of these studies, highlighting their disparities and presenting the status of the current literature. Despite methodological differences, the current literature indicates that β-amyloid has an inhibitory effect on synaptic plasticity and induces network connectivity disruptions. β-amyloid's effect on spontaneous neuronal activity appears more complex. Overall, the literature corroborates the theory that β-amyloid induces neurotoxicity, having a progressive deleterious effect on neuronal signaling and plasticity. These studies also confirm that microelectrode arrays are valuable tools for investigating β-amyloid pathology from a functional perspective, helping to bridge the gap between cellular and network pathology and disease symptoms. The use of microelectrode arrays provides a functional insight into Alzheimer's disease pathology which will aid in the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Aoife O'Connell
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland
| | - Leo Quinlan
- Physiology, School of Medicine, Regenerative Medicine Institute, University of Galway, Ireland
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland.
| |
Collapse
|
2
|
Ando K, Küçükali F, Doeraene E, Nagaraj S, Antonelli EM, Thazin Htut M, Yilmaz Z, Kosa AC, Lopez-Guitierrez L, Quintanilla-Sánchez C, Aydin E, Ramos AR, Mansour S, Turbant S, Schurmans S, Sleegers K, Erneux C, Brion JP, Leroy K. Alteration of gene expression and protein solubility of the PI 5-phosphatase SHIP2 are correlated with Alzheimer's disease pathology progression. Acta Neuropathol 2024; 147:94. [PMID: 38833073 PMCID: PMC11150309 DOI: 10.1007/s00401-024-02745-7] [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: 04/15/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
A recent large genome-wide association study has identified EGFR (encoding the epidermal growth factor EGFR) as a new genetic risk factor for late-onset AD. SHIP2, encoded by INPPL1, is taking part in the signalling and interactome of several growth factor receptors, such as the EGFR. While INPPL1 has been identified as one of the most significant genes whose RNA expression correlates with cognitive decline, the potential alteration of SHIP2 expression and localization during the progression of AD remains largely unknown. Here we report that gene expression of both EGFR and INPPL1 was upregulated in AD brains. SHIP2 immunoreactivity was predominantly detected in plaque-associated astrocytes and dystrophic neurites and its increase was correlated with amyloid load in the brain of human AD and of 5xFAD transgenic mouse model of AD. While mRNA of INPPL1 was increased in AD, SHIP2 protein undergoes a significant solubility change being depleted from the soluble fraction of AD brain homogenates and co-enriched with EGFR in the insoluble fraction. Using FRET-based flow cytometry biosensor assay for tau-tau interaction, overexpression of SHIP2 significantly increased the FRET signal while siRNA-mediated downexpression of SHIP2 significantly decreased FRET signal. Genetic association analyses suggest that some variants in INPPL1 locus are associated with the level of CSF pTau. Our data support the hypothesis that SHIP2 is an intermediate key player of EGFR and AD pathology linking amyloid and tau pathologies in human AD.
Collapse
Affiliation(s)
- Kunie Ando
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium.
| | - Fahri Küçükali
- Complex Genetics of Alzheimer's Disease Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Emilie Doeraene
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Siranjeevi Nagaraj
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Eugenia Maria Antonelli
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - May Thazin Htut
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute (UNI), 808 Route de Lennik, 1070, Brussels, Belgium
| | - Andreea-Claudia Kosa
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Lidia Lopez-Guitierrez
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Carolina Quintanilla-Sánchez
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Emmanuel Aydin
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Ana Raquel Ramos
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Salwa Mansour
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute (UNI), 808 Route de Lennik, 1070, Brussels, Belgium
| | - Sabrina Turbant
- Biobanque Neuro-CEB, Hôpital de la Pitié-Salpétrière, Paris, France
- Plateforme de Ressources Biologiques (PRB), Hôpital de La Pitié-Salpêtrière, AP-HP, Paris, France
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA Research Centre, University of Liège, Liège, Belgium
| | - Kristel Sleegers
- Complex Genetics of Alzheimer's Disease Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christophe Erneux
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Jean-Pierre Brion
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Karelle Leroy
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium.
| |
Collapse
|
3
|
Luan J, Guo N, Hu F, Gou X, Xu L. Aged AβPPswe/PS1ΔE9 mice as a useful animal model for studying the link between immunological senescence and diseases. Immunol Lett 2024; 266:106842. [PMID: 38355057 DOI: 10.1016/j.imlet.2024.106842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 02/16/2024]
Abstract
The APPswe/PS1ΔE9 mouse is a double transgenic murine model that harbors two transgenes for Alzheimer's Disease (AD)-related mutant proteins. We previously discovered that this double transgenic animal had a premature immunosenescence phenotype. However, it is unclear how this phenotype progresses to a later stage. This study aimed to elucidate the changes in systemic characteristics aside from those associated with AD between elderly APPswe/PS1ΔE9 mice and littermate control wild-type mice. Tumors in all organs were considerably more frequent in AD mice aged 24 months than in the control wild-type mice. In addition, the survival rate of aged AD mice was considerably lower than that of wild-type control mice. Further, we discovered that the phenotypic difference was mainly caused by severe immunological aging, as evidenced by a high proportion of exhausted T lymphocytes in AD mice compared to wild-type mice of the same age. Based on our findings, the harm produced by normal aging is not as severe as immunological senescence. Addressing immunological aging, as opposed to anti-aging alone, may be a more crucial target for a long life free of cancer.
Collapse
Affiliation(s)
- Jing Luan
- Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Na Guo
- Institution of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Fengrui Hu
- Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China.
| | - Lixian Xu
- Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
4
|
Jiang L, Roberts R, Wong M, Zhang L, Webber CJ, Libera J, Wang Z, Kilci A, Jenkins M, Ortiz AR, Dorrian L, Sun J, Sun G, Rashad S, Kornbrek C, Daley SA, Dedon PC, Nguyen B, Xia W, Saito T, Saido TC, Wolozin B. β-amyloid accumulation enhances microtubule associated protein tau pathology in an APP NL-G-F/MAPT P301S mouse model of Alzheimer's disease. Front Neurosci 2024; 18:1372297. [PMID: 38572146 PMCID: PMC10987964 DOI: 10.3389/fnins.2024.1372297] [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: 01/17/2024] [Accepted: 03/01/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction The study of the pathophysiology study of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular -amyloid (A) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. Methods The humanized APPNL-G-F knock-in mouse line was crossed to the PS19 MAPTP301S, over-expression mouse line to create the dual APPNL-G-F/PS19 MAPTP301S line. The resulting pathologies were characterized by immunochemical methods and PCR. Results We now report on a double transgenic APPNL-G-F/PS19 MAPTP301S mouse that at 6 months of age exhibits robust A plaque accumulation, intense MAPT pathology, strong inflammation and extensive neurodegeneration. The presence of A pathology potentiated the other major pathologies, including MAPT pathology, inflammation and neurodegeneration. MAPT pathology neither changed levels of amyloid precursor protein nor potentiated A accumulation. Interestingly, study of immunofluorescence in cleared brains indicates that microglial inflammation was generally stronger in the hippocampus, dentate gyrus and entorhinal cortex, which are regions with predominant MAPT pathology. The APPNL-G-F/MAPTP301S mouse model also showed strong accumulation of N6-methyladenosine (m6A), which was recently shown to be elevated in the AD brain. m6A primarily accumulated in neuronal soma, but also co-localized with a subset of astrocytes and microglia. The accumulation of m6A corresponded with increases in METTL3 and decreases in ALKBH5, which are enzymes that add or remove m6A from mRNA, respectively. Discussion Our understanding of the pathophysiology of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular -amyloid (A) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. The APPNL-G-F/MAPTP301S mouse recapitulates many features of AD pathology beginning at 6 months of aging, and thus represents a useful new mouse model for the field.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Rebecca Roberts
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Melissa Wong
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Lushuang Zhang
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Chelsea Joy Webber
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Jenna Libera
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Zihan Wang
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Alper Kilci
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Matthew Jenkins
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Alejandro Rondón Ortiz
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Luke Dorrian
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Jingjing Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore, Singapore
| | - Guangxin Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sherif Rashad
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | | | - Sarah Anne Daley
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Peter C. Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore, Singapore
| | - Brian Nguyen
- LifeCanvas Technologies, Cambridge, MA, United States
| | - Weiming Xia
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Benjamin Wolozin
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| |
Collapse
|
5
|
Marshall J, Huynh K, Lancaster G, Ng J, Collins J, Pernes G, Liang A, Featherby T, Mellet N, Drew B, Calkin A, King A, Meikle P, Febbraio M, Adlard P, Henstridge D. Behavioral, metabolic, and lipidomic characterization of the 5xFADxTg30 mouse model of Alzheimer's disease. iScience 2024; 27:108800. [PMID: 38292430 PMCID: PMC10826307 DOI: 10.1016/j.isci.2024.108800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is associated with both extracellular amyloid-β (Aβ) plaques and intracellular tau-containing neurofibrillary tangles (NFT). We characterized the behavioral, metabolic and lipidomic phenotype of the 5xFADxTg30 mouse model which contains overexpression of both Aβ and tau. Our results independently reproduce several phenotypic traits described previously for this model, while providing additional characterization. This model develops many aspects associated with AD including frailty, decreased survival, initiation of aspects of cognitive decline and alterations to specific lipid classes and molecular lipid species in the plasma and brain. Notably, some sex-specific differences exist in this model and motor impairment with aging in this model does compromise the utility of the model for some movement-based behavioral assessments of cognitive function. These findings provide a reference for individuals interested in using this model to understand the pathology associated with elevated Aβ and tau or for testing potential therapeutics for the treatment of AD.
Collapse
Affiliation(s)
- J.P.S. Marshall
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - K. Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - G.I. Lancaster
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - J. Ng
- School of Health Sciences, The University of Tasmania, Launceston, TAS, Australia
| | - J.M. Collins
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - G. Pernes
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - A. Liang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - T. Featherby
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - N.A. Mellet
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - B.G. Drew
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - A.C. Calkin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - A.E. King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - P.J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - M.A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia
| | - P.A. Adlard
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - D.C. Henstridge
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Health Sciences, The University of Tasmania, Launceston, TAS, Australia
| |
Collapse
|
6
|
Malik N, Miah MU, Galgani A, McAleese K, Walker L, LeBeau FE, Attems J, Outeiro TF, Thomas A, Koss DJ. Regional AT-8 reactive tau species correlate with intracellular Aβ levels in cases of low AD neuropathologic change. Acta Neuropathol 2024; 147:40. [PMID: 38353753 PMCID: PMC10866780 DOI: 10.1007/s00401-024-02691-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024]
Abstract
The amyloid cascade hypothesis states that Aβ aggregates induce pathological changes in tau, leading to neurofibrillary tangles (NFTs) and cell death. A caveat with this hypothesis is the spatio-temporal divide between plaques and NFTs. This has been addressed by the inclusion of soluble Aβ and tau species in the revised amyloid cascade hypothesis. Nevertheless, despite the potential for non-plaque Aβ to contribute to tau pathology, few studies have examined relative correlative strengths between total Aβ, plaque Aβ and intracellular Aβ with tau pathology within a single tissue cohort. Employing frozen and fixed frontal cortex grey and white matter tissue from non-AD controls (Con; n = 39) and Alzheimer's disease (AD) cases (n = 21), biochemical and immunohistochemical (IHC) measures of Aβ and AT-8 phosphorylated tau were assessed. Biochemical native-state dot blots from crude tissue lysates demonstrated robust correlations between total Aβ and AT-8 tau, when considered as a combined cohort (Con and AD) and when as Con and AD cases, separately. In contrast, no associations between Aβ plaques and AT-8 were reported when using IHC measurements in either Con or AD cases. However, when intracellular Aβ was measured via the Aβ specific antibody MOAB-2, a correlative relationship with AT-8 tau was reported in non-AD controls but not in AD cases. Collectively the data suggests that accumulating intracellular Aβ may influence AT-8 pathology, early in AD-related neuropathological change. Despite the lower levels of phospho-tau and Aβ in controls, the robust correlative relationships observed suggest a physiological association of Aβ production and tau phosphorylation, which may be modified during disease. This study is supportive of a revised amyloid cascade hypothesis and demonstrates regional associative relationships between tau pathology and intracellular Aβ, but not extracellular Aβ plaques.
Collapse
Affiliation(s)
- Nauman Malik
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Mohi-Uddin Miah
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Alessandro Galgani
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Kirsty McAleese
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Lauren Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Fiona E LeBeau
- Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Johannes Attems
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Tiago F Outeiro
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alan Thomas
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - David J Koss
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK.
| |
Collapse
|
7
|
Jiang L, Roberts R, Wong M, Zhang L, Webber CJ, Kilci A, Jenkins M, Sun J, Sun G, Rashad S, Dedon PC, Daley SA, Xia W, Ortiz AR, Dorrian L, Saito T, Saido TC, Wolozin B. Accumulation of m 6A exhibits stronger correlation with MAPT than β-amyloid pathology in an APP NL-G-F /MAPT P301S mouse model of Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-2745852. [PMID: 37292629 PMCID: PMC10246280 DOI: 10.21203/rs.3.rs-2745852/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The study for the pathophysiology study of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular β-amyloid (Aβ) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. We now report on a double transgenic APPNL-G-F MAPTP301S mouse that at 6 months of age exhibits robust Aβ plaque accumulation, intense MAPT pathology, strong inflammation and extensive neurodegeneration. The presence of Aβ pathology potentiated the other major pathologies, including MAPT pathology, inflammation and neurodegeneration. However, MAPT pathology neither changed levels of amyloid precursor protein nor potentiated Aβ accumulation. The APPNL-G-F/MAPTP301S mouse model also showed strong accumulation of N6-methyladenosine (m6A), which was recently shown to be elevated in the AD brain. M6A primarily accumulated in neuronal soma, but also co-localized with a subset of astrocytes and microglia. The accumulation of m6A corresponded with increases in METTL3 and decreases in ALKBH5, which are enzymes that add or remove m6A from mRNA, respectively. Thus, the APPNL-G-F/MAPTP301S mouse recapitulates many features of AD pathology beginning at 6 months of aging.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Rebecca Roberts
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Melissa Wong
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Lushuang Zhang
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Chelsea Joy Webber
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Alper Kilci
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Matthew Jenkins
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Jingjing Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore 138602, Singapore
| | - Guangxin Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sherif Rashad
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Peter C Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Sarah Anne Daley
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, 01730, USA
| | - Weiming Xia
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, 01730, USA
| | - Alejandro Rondón Ortiz
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Luke Dorrian
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, 351-0198,Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, 351-0198,Japan
| | - Benjamin Wolozin
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Neurology, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA USA
| |
Collapse
|
8
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
9
|
Jiang L, Roberts R, Wong M, Zhang L, Webber CJ, Kilci A, Jenkins M, Sun G, Rashad S, Sun J, Dedon PC, Daley SA, Xia W, Ortiz AR, Dorrian L, Saito T, Saido TC, Wolozin B. Accumulation of m 6A exhibits stronger correlation with MAPT than β-amyloid pathology in an APP NL-G-F /MAPT P301S mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534515. [PMID: 37034774 PMCID: PMC10081259 DOI: 10.1101/2023.03.28.534515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The study for the pathophysiology study of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular β-amyloid (Aβ) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. We now report on a double transgenic APPNL-G-F MAPTP301S mouse that at 6 months of age exhibits robust Aβ plaque accumulation, intense MAPT pathology, strong inflammation and extensive neurodegeneration. The presence of Aβ pathology potentiated the other major pathologies, including MAPT pathology, inflammation and neurodegeneration. However, MAPT pathology neither changed levels of amyloid precursor protein nor potentiated Aβ accumulation. The APPNL-G-F/MAPTP301S mouse model also showed strong accumulation of N6-methyladenosine (m6A), which was recently shown to be elevated in the AD brain. M6A primarily accumulated in neuronal soma, but also co-localized with a subset of astrocytes and microglia. The accumulation of m6A corresponded with increases in METTL3 and decreases in ALKBH5, which are enzymes that add or remove m6A from mRNA, respectively. Thus, the APPNL-G-F/MAPTP301S mouse recapitulates many features of AD pathology beginning at 6 months of aging.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Rebecca Roberts
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Melissa Wong
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Lushuang Zhang
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Chelsea Joy Webber
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Alper Kilci
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Matthew Jenkins
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Guangxin Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sherif Rashad
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Jingjing Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore 138602, Singapore
| | - Peter C Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore 138602, Singapore
| | - Sarah Anne Daley
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, 01730, USA
| | - Weiming Xia
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, 01730, USA
| | - Alejandro Rondón Ortiz
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Luke Dorrian
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, 351-0198, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, 351-0198, Japan
| | - Benjamin Wolozin
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, 02118
- Department of Neurology, Chobanian and Avedesian School of Medicine, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA USA
| |
Collapse
|
10
|
Gorina YV, Vlasova OL, Bolshakova AV, Salmina AB. Alzheimer’s Disease: a Search for the Best Experimental Models to Decode Cellular and Molecular Mechanisms of Its Development. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
11
|
Arora S, Kanekiyo T, Singh J. Functionalized nanoparticles for brain targeted BDNF gene therapy to rescue Alzheimer's disease pathology in transgenic mouse model. Int J Biol Macromol 2022; 208:901-911. [PMID: 35378156 DOI: 10.1016/j.ijbiomac.2022.03.203] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is actively produced and utilized in cortical circuits throughout life to sustain neuronal function and synaptic plasticity. In animal models of Alzheimer's Disease (AD), highly invasive BDNF gene therapy using viral vectors has successfully shown enhanced synaptic protein expression, proliferation of neurons and attenuation of amyloidogenic processes. However, to eliminate virus-related safety issues and invasive procedures, our present study has explored brain-targeted lipid-based nanoparticles that can deliver plasmid encoding BDNF to brain in a safe and efficient manner. Efficacy of these nanoparticles was tested in early (6-months) and advanced stage (9-months) transgenic APP/PS1 AD mice. Liposomes were surface-functionalized with brain targeting ligand, mannose, and cell-penetrating peptides (rabies virus-derived peptide or penetratin). These bifunctionalized nanoparticles enhanced BDNF expression by ~2 times and resulted in >40% (p < 0.05) reduction in toxic amyloid-beta peptides in 6- and 9-months old APP/PS1 mice brains compared to their age-matched untreated controls. Plaque load was reduced ~7 and ~3 times (p < 0.05), respectively, whereas synaptic proteins, synaptophysin and PSD-95, were found to be increased >90% (p < 0.05) in both age groups of transgenic mice treated with bifunctionalized nanoparticles. No untoward adverse effects were observed throughout treatment, suggesting a safe and effective strategy to rescue AD pathology.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA.
| |
Collapse
|
12
|
Yoshida S, Hasegawa T. Deciphering the prion-like behavior of pathogenic protein aggregates in neurodegenerative diseases. Neurochem Int 2022; 155:105307. [PMID: 35181393 DOI: 10.1016/j.neuint.2022.105307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are hitherto classified based on their core clinical features, the anatomical distribution of neurodegeneration, and the cell populations mainly affected. On the other hand, the wealth of neuropathological, genetic, molecular and biochemical studies have identified the existence of distinct insoluble protein aggregates in the affected brain regions. These findings have spread the use of a collective term, proteinopathy, for neurodegenerative disorders with particular type of structurally altered protein accumulation. Particularly, a recent breakthrough in this field came with the discovery that these protein aggregates can transfer from one cell to another, thereby converting normal proteins to potentially toxic, misfolded species in a prion-like manner. In this review, we focus specifically on the molecular and cellular basis that underlies the seeding activity and transcellular spreading phenomenon of neurodegeneration-related protein aggregates, and discuss how these events contribute to the disease progression.
Collapse
Affiliation(s)
- Shun Yoshida
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan; Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa, Yamagata, 992-1202, Japan
| | - Takafumi Hasegawa
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan.
| |
Collapse
|
13
|
Ginsenoside Rd Attenuates Tau Phosphorylation in Olfactory Bulb, Spinal Cord, and Telencephalon by Regulating Glycogen Synthase Kinase 3 β and Cyclin-Dependent Kinase 5. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2021:4485957. [PMID: 34987593 PMCID: PMC8720614 DOI: 10.1155/2021/4485957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Ginseng is a plant of the family Acanthopanaceae. It has been used for thousands of years in China. It is known as the king of hundred herbs. It was recorded first in Shennong Baicao Jing. It has been found that ginsenoside Rd is a neuroprotective agent. This article aims to explore the protective roles of ginsenoside Rd in Alzheimer's disease. Rd, a Chinese herb, may be a promising treatment drug for Alzheimer's disease (AD) and is also reported to be related to several pathological changes, including the deposition of Aβ and tau hyperphosphorylation in AD as it decreases the deposition of tau hyperphosphorylation in APP transgenic mice. METHODS In this study, APP transgenic mice were pretreated with 10 mg/kg Rd for six months, and the effect of Rd on neuropathological deficits in the olfactory bulb, spinal cord, and telencephalon of APP transgenic mice was investigated. The phosphorylation levels of tau (S199/202, S396, S404, and Tau5) and the activities of the proteins glycogen synthase kinase 3β (Tyr216) and cyclin-dependent kinase 5 (P25/P35) were measured. RESULTS The pretreatment of Rd effectively decreased the production and deposition of hyperphosphorylated tau (S199/202, S396, and S404) protein by depressing the expression of glycogen synthase kinase 3β (GSK-3β/Tyr216) and cyclin-dependent kinase 5 (CDK5/P25). CONCLUSION These findings suggest that ginsenoside Rd could improve the pathological changes of AD in the olfactory bulb, spinal cord, and telencephalon, which further demonstrated the potential therapeutic effect of Rd in early AD.
Collapse
|
14
|
Cuadrado-Tejedor M, Pérez-González M, Alfaro-Ruiz R, Badesso S, Sucunza D, Espelosin M, Ursúa S, Lachen-Montes M, Fernández-Irigoyen J, Santamaria E, Luján R, García-Osta A. Amyloid-Driven Tau Accumulation on Mitochondria Potentially Leads to Cognitive Deterioration in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222111950. [PMID: 34769380 PMCID: PMC8584544 DOI: 10.3390/ijms222111950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 11/18/2022] Open
Abstract
Despite the well-accepted role of the two main neuropathological markers (β-amyloid and tau) in the progression of Alzheimer’s disease, the interaction and specific contribution of each of them is not fully elucidated. To address this question, in the present study, an adeno-associated virus (AAV9) carrying the mutant P301L form of human tau, was injected into the dorsal hippocampi of APP/PS1 transgenic mice or wild type mice (WT). Three months after injections, memory tasks, biochemical and immunohistochemical analysis were performed. We found that the overexpression of hTauP301L accelerates memory deficits in APP/PS1 mice, but it did not affect memory function of WT mice. Likewise, biochemical assays showed that only in the case of APP/PS1-hTauP301L injected mice, an important accumulation of tau was observed in the insoluble urea fraction. Similarly, electron microscopy images revealed that numerous clusters of tau immunoparticles appear at the dendrites of APP/PS1 injected mice and not in WT animals, suggesting that the presence of amyloid is necessary to induce tau aggregation. Interestingly, these tau immunoparticles accumulate in dendritic mitochondria in the APP/PS1 mice, whereas most of mitochondria in WT injected mice remain free of tau immunoparticles. Taken together, it seems that amyloid induces tau aggregation and accumulation in the dendritic mitochondria and subsequently may alter synapse function, thus, contributing to accelerate cognitive decline in APP/PS1 mice.
Collapse
Affiliation(s)
- Mar Cuadrado-Tejedor
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
- Correspondence: (M.C.-T.); (A.G.-O.)
| | - Marta Pérez-González
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Rocío Alfaro-Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, 02008 Albacete, Spain; (R.A.-R.); (R.L.)
| | - Sara Badesso
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Diego Sucunza
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
| | - María Espelosin
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
| | - Susana Ursúa
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
| | - Mercedes Lachen-Montes
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (M.L.-M.); (J.F.-I.); (E.S.)
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (M.L.-M.); (J.F.-I.); (E.S.)
| | - Enrique Santamaria
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (M.L.-M.); (J.F.-I.); (E.S.)
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, 02008 Albacete, Spain; (R.A.-R.); (R.L.)
| | - Ana García-Osta
- Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, IdiSNA, 31008 Pamplona, Spain; (M.P.-G.); (S.B.); (M.E.); (S.U.)
- Correspondence: (M.C.-T.); (A.G.-O.)
| |
Collapse
|
15
|
Wang X, Liu EJ, Liu Q, Li SH, Li T, Zhou QZ, Liu YC, Zhang H, Wang JZ. Tau Acetylation in Entorhinal Cortex Induces its Chronic Hippocampal Propagation and Cognitive Deficits in Mice. J Alzheimers Dis 2021; 77:241-255. [PMID: 32804150 DOI: 10.3233/jad-200529] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Increased tau acetylation at K174, K274, K280, and K281 has been observed in the brains of Alzheimer's disease (AD) patients or in transgenic mice, but the role of acetylation in tau propagation is elusive. OBJECTIVE To study the effect of tau acetylation in entorhinal cortex on tau transmission and learning and memory. METHODS Stereotactic brain injection, behavioral test, electrophysiological recording, immunohistochemistry, and immunofluorescence were used. RESULTS We constructed the hyperacetylation mimics of tau (AAV-Tau-4Q), the non-acetylation tau mutant (AAV-Tau-4R), and the wild-type tau (AAV-Tau-WT). By overexpressing these different tau proteins in the entorhinal cortex (EC) of 2-month-old mice, we found that overexpressing Tau-4Q in EC for 3 or 6 months (to 5 or 8 months of age) neither induces tau propagation to dentate gyrus (DG) nor glial activation in DG, nor spatial memory deficit. However, overexpressing Tau-WT and Tau-4Q in EC for 13.5 months (15.5 months of age) at 2 months promoted tau propagation respectively to granulosa and hilus of DG with glial activation, synaptic dysfunction, and memory deficit, while overexpressing Tau-4R abolished tau propagation with improved cellular pathologies and cognitive functions. Furthermore, overexpressing Tau-4Q in unilateral DG of 2-month-old mice for 8 weeks also promoted its contralateral transmission with glial activation, and mice with tau (Tau-WT, Tau-4Q, and Tau-4R) overexpression in DG showed cognitive deficits compared with the empty vector controls. CONCLUSION Tau acetylation induces a time-dependent propagation from EC to DG, and only hippocampus but not EC tau accumulation induces cognitive deficits.
Collapse
Affiliation(s)
- Xin Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - En-Jie Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Hong Li
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu-Zhi Zhou
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Chao Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
16
|
Cacabelos R, Carrera I, Martínez-Iglesias O, Cacabelos N, Naidoo V. What is the gold standard model for Alzheimer's disease drug discovery and development? Expert Opin Drug Discov 2021; 16:1415-1440. [PMID: 34330186 DOI: 10.1080/17460441.2021.1960502] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Alzheimer's disease models (ADMs) are currently used for drug development (DD). More than 20,000 molecules were screened for AD treatment over decades, with only one drug (Aducanumab)FDA-approved over the past 18 years. A revision of pathogenic concepts and ADMs are needed.Areas covered: The authors discuss herein preclinical models including: (i) in vitro models (cell lines, primary neuron cell cultures, iPSC-derived brain cells), (ii) ex vivo models, and (iii) in vivo models (artificial, transgenic, non-transgenic and induced).Expert opinion: The following types of ADMs have been reported: Mouse models (45.08%), Rat models (15.04%), Non-human Primate models (0.76%), Rabbit models (0.46%), Cat models (0.53%), Pig models (0.30%), Guinea pig models (0.15%), Octodon degu models (0.02%), Dog models (0.54%), Drosophila melanogaster models (1.79%), Zebrafish models (0.50%), Caenorhabditis elegans (1.21%), Cell culture models (3.31%), Cholinergic models (8.26%), Neurotoxic models (6.79%), Neuroinflammation models (6.92%), Neurovascular models (7.88%), and Microbiome models (0.45%).No single ADM faithfully reproduces all the pathogenic events in the human AD phenotype spectrum. ADMs should be different for (i) pathogenic studies vs basic DD, and (ii) preventive interventions vs symptomatic treatments. There cannot be an ideal ADM for DD, because AD is a spectrum of syndromes. DD can integrate pathogenic, mechanistic, metabolic, transporter and pleiotropic genes in a multisystem model.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Departments of Genomic Medicine, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, Spain
| | - Iván Carrera
- Health Biotechnology, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, Spain
| | - Olaia Martínez-Iglesias
- Medical Epigenetics, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, Spain
| | - Natalia Cacabelos
- Medical Documentation, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, Spain
| | - Vinogran Naidoo
- Basic Neuroscience, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, Spain
| |
Collapse
|
17
|
Zampar S, Wirths O. Characterization of a Mouse Model of Alzheimer's Disease Expressing Aβ4-42 and Human Mutant Tau. Int J Mol Sci 2021; 22:ijms22105191. [PMID: 34069029 PMCID: PMC8156793 DOI: 10.3390/ijms22105191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 01/04/2023] Open
Abstract
The relationship between the two most prominent neuropathological hallmarks of Alzheimer’s Disease (AD), extracellular amyloid-β (Aβ) deposits and intracellular accumulation of hyperphosphorylated tau in neurofibrillary tangles (NFT), remains at present not fully understood. A large body of evidence places Aβ upstream in the cascade of pathological events, triggering NFTs formation and the subsequent neuron loss. Extracellular Aβ deposits were indeed causative of an increased tau phosphorylation and accumulation in several transgenic models but the contribution of soluble Aβ peptides is still controversial. Among the different Aβ variants, the N-terminally truncated peptide Aβ4–42 is among the most abundant. To understand whether soluble Aβ4–42 peptides impact the onset or extent of tau pathology, we have crossed the homozygous Tg4–42 mouse model of AD, exclusively expressing Aβ4–42 peptides, with the PS19 (P301S) tau transgenic model. Behavioral assessment showed that the resulting double-transgenic line presented a partial worsening of motor performance and spatial memory deficits in the aged group. While an increased loss of distal CA1 pyramidal neurons was detected in young mice, no significant alterations in hippocampal tau phosphorylation were observed in immunohistochemical analyses.
Collapse
|
18
|
Lee SH, Meilandt WJ, Xie L, Gandham VD, Ngu H, Barck KH, Rezzonico MG, Imperio J, Lalehzadeh G, Huntley MA, Stark KL, Foreman O, Carano RA, Friedman BA, Sheng M, Easton A, Bohlen CJ, Hansen DV. Trem2 restrains the enhancement of tau accumulation and neurodegeneration by β-amyloid pathology. Neuron 2021; 109:1283-1301.e6. [DOI: 10.1016/j.neuron.2021.02.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/07/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
|
19
|
Spatial memory deficiency early in 6xTg Alzheimer's disease mouse model. Sci Rep 2021; 11:1334. [PMID: 33446720 PMCID: PMC7809274 DOI: 10.1038/s41598-020-79344-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/08/2020] [Indexed: 01/27/2023] Open
Abstract
Alzheimer’s disease (AD) is mainly characterized by the deposition of extracellular amyloid plaques and intracellular accumulation of neurofibrillary tangles (NFTs). While the recent 5xFAD AD mouse model exhibits many AD-related phenotypes and a relatively early and aggressive amyloid β production, it does not show NFTs. Here, we developed and evaluated a novel AD mouse model (6xTg-AD, 6xTg) by crossbreeding 5xFAD mice with mice expressing mutant (P301L) tau protein (MAPT). Through behavioral and histopathological tests, we analyzed cognitive changes and neuropathology in 6xTg mice compared to their respective parental strains according to age. Spatial memory deficits occurred in 6xTg mice at 2 months of age, earlier than they occurred in 5xFAD mice. Histopathological data revealed aggressive Aβ42 and p-tau accumulation in 6xTg mice. Microglial activation occurred in the cortex and hippocampus of 6xTg mice beginning at 2 months. In 6xTg model mice, the synaptic loss was observed in the cortex from 4 months of age and in the hippocampus from 6 months of age, and neuronal loss appeared in the cortex from 4 months of age and in the hippocampus 6 months of age, earlier than it is observed in the 5xFAD and JNPL3 models. These results showed that each pathological symptom appeared much faster than in their parental animal models. In conclusion, these novel 6xTg-AD mice might be an advanced animal model for studying AD, representing a promising approach to developing effective therapy.
Collapse
|
20
|
Laversenne V, Nazeeruddin S, Källstig EC, Colin P, Voize C, Schneider BL. Anti-Aβ antibodies bound to neuritic plaques enhance microglia activity and mitigate tau pathology. Acta Neuropathol Commun 2020; 8:198. [PMID: 33225991 PMCID: PMC7681991 DOI: 10.1186/s40478-020-01069-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/27/2020] [Indexed: 11/10/2022] Open
Abstract
The brain pathology of Alzheimer's disease (AD) is characterized by the misfolding and aggregation of both the amyloid beta (Aβ) peptide and hyperphosphorylated forms of the tau protein. Initial Aβ deposition is considered to trigger a sequence of deleterious events contributing to tau pathology, neuroinflammation and ultimately causing the loss of synapses and neurons. To assess the effect of anti-Aβ immunization in this context, we generated a mouse model by overexpressing the human tau protein in the hippocampus of 5xFAD mice. Aβ plaque deposition combined with human tau overexpression leads to an array of pathological manifestations including the formation of tau-positive dystrophic neurites and accumulation of hyperphosphorylated tau at the level of neuritic plaques. Remarkably, the presence of human tau reduces microglial clustering in proximity to the Aβ plaques, which may affect the barrier role of microglia. In this mouse model, continuous administration of anti-Aβ antibodies enhances the clustering of microglial cells even in the presence of tau. Anti-Aβ immunization increases plaque compaction, reduces the spread of tau in the hippocampal formation and prevents the formation of tau-positive dystrophic neurites. However, the treatment does not significantly reduce tau-induced neurodegeneration in the dentate gyrus. These results highlight that anti-Aβ immunization is able to enhance microglial activity around neuritic plaques, mitigating part of the tau-induced pathological manifestations.
Collapse
|
21
|
Neuron Loss in Alzheimer's Disease: Translation in Transgenic Mouse Models. Int J Mol Sci 2020; 21:ijms21218144. [PMID: 33143374 PMCID: PMC7663280 DOI: 10.3390/ijms21218144] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Transgenic mouse models represent an essential tool for the exploration of Alzheimer’s disease (AD) pathological mechanisms and the development of novel treatments, which at present provide only symptomatic and transient effects. While a variety of mouse models successfully reflects the main neuropathological hallmarks of AD, such as extracellular amyloid-β (Aβ) deposits, intracellular accumulation of Tau protein, the development of micro- and astrogliosis, as well as behavioral deficits, substantial neuron loss, as a key feature of the disease, seems to be more difficult to achieve. In this review, we summarize information on classic and more recent transgenic mouse models for AD, focusing in particular on loss of pyramidal, inter-, and cholinergic neurons. Although the cause of neuron loss in AD is still a matter of scientific debate, it seems to be linked to intraneuronal Aβ accumulation in several transgenic mouse models, especially in pyramidal neurons.
Collapse
|
22
|
Houben S, de Fisenne MA, Ando K, Vanden Dries V, Poncelet L, Yilmaz Z, Mansour S, De Decker R, Brion JP, Leroy K. Intravenous Injection of PHF-Tau Proteins From Alzheimer Brain Exacerbates Neuroinflammation, Amyloid Beta, and Tau Pathologies in 5XFAD Transgenic Mice. Front Mol Neurosci 2020; 13:106. [PMID: 32765217 PMCID: PMC7381181 DOI: 10.3389/fnmol.2020.00106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/20/2020] [Indexed: 02/01/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation in the brain of intraneuronal aggregates of abnormally and hyperphosphorylated tau proteins and of extracellular deposits of amyloid-β surrounded by dystrophic neurites. Numerous experimental models have shown that tau pathology develops in the brain after intracerebral injection of brain homogenates or pathological tau [paired helical filaments (PHF)-tau)] from AD brains. Further investigations are however necessary to identify or exclude potential extracerebral routes of tau pathology transmission, e.g., through the intravascular route. In this study, we have analyzed the effect of intravenous injection of PHF-tau proteins from AD brains on the formation of tau and amyloid pathologies in the brain of wild-type (WT) mice and of 5XFAD mice (an amyloid model). We observed that 5XFAD mice with a disrupted blood-brain barrier showed increased plaque-associated astrogliosis, microgliosis, and increased deposits of Aβ40 and Aβ42 after intravenous injection of PHF-tau proteins. In addition, an increased phosphotau immunoreactivity was observed in plaque-associated dystrophic neurites. These results suggest that blood products contaminated by PHF-tau proteins could potentially induce an exacerbation of neuroinflammation and AD pathologies.
Collapse
Affiliation(s)
- Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie-Ange de Fisenne
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Virginie Vanden Dries
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Luc Poncelet
- Laboratory of Anatomy, Biomechanics and Organogenesis, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Salwa Mansour
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Robert De Decker
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
23
|
Thierry M, Boluda S, Delatour B, Marty S, Seilhean D, Potier MC, Duyckaerts C. Human subiculo-fornico-mamillary system in Alzheimer's disease: Tau seeding by the pillar of the fornix. Acta Neuropathol 2020; 139:443-461. [PMID: 31822997 DOI: 10.1007/s00401-019-02108-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/29/2019] [Accepted: 11/30/2019] [Indexed: 02/07/2023]
Abstract
In Alzheimer's disease (AD), Tau and Aβ aggregates involve sequentially connected regions, sometimes distantly separated. These alterations were studied in the pillar of the fornix (PoF), an axonal tract, to analyse the role of axons in their propagation. The PoF axons mainly originate from the subicular neurons and project to the mamillary body. Forty-seven post-mortem cases at various Braak stages (Tau) and Thal phases (Aβ) were analysed by immunohistochemistry. The distribution of the lesions showed that the subiculum was affected before the mamillary body, but neither Tau aggregation nor Aβ deposition was consistently first. The subiculum and the mamillary body contained Gallyas positive neurofibrillary tangles, immunolabelled by AT8, TG3, PHF1, Alz50 and C3 Tau antibodies. In the PoF, only thin and fragmented threads were observed, exclusively in the cases with neurofibrillary tangles in the subiculum. The threads were made of Gallyas negative, AT8 and TG3 positive Tau. They were intra-axonal and devoid of paired helical filaments at electron microscopy. We tested PoF homogenates containing Tau AT8 positive axons in a Tau P301S biosensor HEK cell line and found a seeding activity. There was no Aβ immunoreactivity detected in the PoF. We could follow microcryodissected AT8 positive axons entering the mamillary body; contacts between Tau positive endings and Aβ positive diffuse or focal deposits were observed in CLARITY-cleared mamillary body. In conclusion, we show that non-fibrillary, hyperphosphorylated Tau is transported by the axons of the PoF from the subiculum to the mamillary body and has a seeding activity. Either Tau aggregation or Aβ accumulation may occur first in this system: this inconstant order is incompatible with a cause-and-effects relationship. However, both pathologies were correlated and intimately associated, indicating an interaction of the two processes, once initiated.
Collapse
Affiliation(s)
- Manon Thierry
- Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
- Laboratoire de Neuropathologie Raymond Escourolle, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, 47, Blvd de l'Hôpital, 75651, Paris Cedex 13, France
| | - Susana Boluda
- Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
- Laboratoire de Neuropathologie Raymond Escourolle, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, 47, Blvd de l'Hôpital, 75651, Paris Cedex 13, France
| | - Benoît Delatour
- Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
| | - Serge Marty
- Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
| | - Danielle Seilhean
- Laboratoire de Neuropathologie Raymond Escourolle, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, 47, Blvd de l'Hôpital, 75651, Paris Cedex 13, France
| | - Marie-Claude Potier
- Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
| | - Charles Duyckaerts
- Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France.
- Laboratoire de Neuropathologie Raymond Escourolle, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, 47, Blvd de l'Hôpital, 75651, Paris Cedex 13, France.
| |
Collapse
|
24
|
McAllister BB, Lacoursiere SG, Sutherland RJ, Mohajerani MH. Intracerebral seeding of amyloid-β and tau pathology in mice: Factors underlying prion-like spreading and comparisons with α-synuclein. Neurosci Biobehav Rev 2020; 112:1-27. [PMID: 31996301 DOI: 10.1016/j.neubiorev.2020.01.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is characterized neuropathologically by progressive neurodegeneration and by the presence of amyloid plaques and neurofibrillary tangles. These plaques and tangles are composed, respectively, of amyloid-beta (Aβ) and tau proteins. While long recognized as hallmarks of AD, it remains unclear what causes the formation of these insoluble deposits. One theory holds that prion-like templated misfolding of Aβ and tau induces these proteins to form pathological aggregates, and propagation of this misfolding causes the stereotyped progression of pathology commonly seen in AD. Supporting this theory, numerous studies have been conducted in which aggregated Aβ, tau, or α-synuclein is injected intracerebrally into pathology-free host animals, resulting in robust formation of pathology. Here, we review this literature, focusing on in vivo intracerebral seeding of Aβ and tau in mice. We compare the results of these experiments to what is known about the seeding and spread of α-synuclein pathology, and we discuss how this research informs our understanding of the factors underlying the onset, progression, and outcomes of proteinaceous pathologies.
Collapse
Affiliation(s)
- Brendan B McAllister
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - Sean G Lacoursiere
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - Robert J Sutherland
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada.
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada.
| |
Collapse
|
25
|
DeVos SL, Corjuc BT, Commins C, Dujardin S, Bannon RN, Corjuc D, Moore BD, Bennett RE, Jorfi M, Gonzales JA, Dooley PM, Roe AD, Pitstick R, Irimia D, Frosch MP, Carlson GA, Hyman BT. Tau reduction in the presence of amyloid-β prevents tau pathology and neuronal death in vivo. Brain 2019; 141:2194-2212. [PMID: 29733334 DOI: 10.1093/brain/awy117] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/05/2018] [Indexed: 11/14/2022] Open
Abstract
Several studies have now supported the use of a tau lowering agent as a possible therapy in the treatment of tauopathy disorders, including Alzheimer's disease. In human Alzheimer's disease, however, concurrent amyloid-β deposition appears to synergize and accelerate tau pathological changes. Thus far, tau reduction strategies that have been tested in vivo have been examined in the setting of tau pathology without confounding amyloid-β deposition. To determine whether reducing total human tau expression in a transgenic model where there is concurrent amyloid-β plaque formation can still reduce tau pathology and protect against neuronal loss, we have taken advantage of the regulatable tau transgene in APP/PS1 × rTg4510 mice. These mice develop both neurofibrillary tangles as well as amyloid-β plaques throughout the cortex and hippocampus. By suppressing human tau expression for 6 months in the APP/PS1 × rTg4510 mice using doxycycline, AT8 tau pathology, bioactivity, and astrogliosis were reduced, though importantly to a lesser extent than lowering tau in the rTg4510 alone mice. Based on non-denaturing gels and proteinase K digestions, the remaining tau aggregates in the presence of amyloid-β exhibit a longer-lived aggregate conformation. Nonetheless, lowering the expression of the human tau transgene was sufficient to equally ameliorate thioflavin-S positive tangles and prevent neuronal loss equally well in both the APP/PS1 × rTg4510 mice and the rTg4510 cohort. Together, these results suggest that, although amyloid-β stabilizes tau aggregates, lowering total tau levels is still an effective strategy for the treatment of tau pathology and neuronal loss even in the presence of amyloid-β deposition.
Collapse
Affiliation(s)
- Sarah L DeVos
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Bianca T Corjuc
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Caitlin Commins
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Simon Dujardin
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Riley N Bannon
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Diana Corjuc
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Benjamin D Moore
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Rachel E Bennett
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Mehdi Jorfi
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Jose A Gonzales
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Patrick M Dooley
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Allyson D Roe
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Rose Pitstick
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Daniel Irimia
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Matthew P Frosch
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA.,C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - George A Carlson
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Bradley T Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
26
|
Estrada LD, Ahumada P, Cabrera D, Arab JP. Liver Dysfunction as a Novel Player in Alzheimer's Progression: Looking Outside the Brain. Front Aging Neurosci 2019; 11:174. [PMID: 31379558 PMCID: PMC6650779 DOI: 10.3389/fnagi.2019.00174] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/25/2019] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) afflicts an estimated 20 million people worldwide and is the fourth-leading cause of death in the developed world. The most common cause of dementia in older individuals, AD is characterized by neuropathologies including synaptic and neuronal degeneration, amyloid plaques, and neurofibrillary tangles (NTFs). Amyloid plaques are primarily composed of amyloid-beta peptide (Aβ), which accumulates in the brains of patients with AD. Further, small aggregates termed Aβ oligomers are implicated in the synaptic loss and neuronal degeneration underlying early cognitive impairments. Whether Aβ accumulates in part because of dysregulated clearance from the brain remains unclear. The flow of substances (e.g., nutrients, drugs, toxins) in and out of the brain is mediated by the blood-brain-barrier (BBB). The BBB exhibits impairment in AD patients and animal models. The effect of BBB impairment on Aβ, and whether BBB function is affected by non-neurological pathologies that impair peripheral clearance requires further investigation. In particular, impaired peripheral clearance is a feature of nonalcoholic fatty liver disease (NAFLD), a spectrum of liver disorders characterized by accumulation of fat in the liver accompanied by varying degrees of inflammation and hepatocyte injury. NAFLD has reached epidemic proportions, with an estimated prevalence between 20% and 30% of the general population. This chronic condition may influence AD pathogenesis. This review article summarizes the current state of the literature linking NAFLD and AD, highlighting the role of the major Aβ efflux and clearance protein, the LRP-1 receptor, which is abundantly expressed in liver, brain, and vasculature.
Collapse
Affiliation(s)
- Lisbell D. Estrada
- Bionanotechnology Laboratory, Integrative Center for Applied Biology and Chemistry (CIBQA), Department of Chemical & Biological Sciences, Universidad Bernardo O’Higgins, Santiago, Chile
| | - Pablo Ahumada
- Bionanotechnology Laboratory, Integrative Center for Applied Biology and Chemistry (CIBQA), Department of Chemical & Biological Sciences, Universidad Bernardo O’Higgins, Santiago, Chile
| | - Daniel Cabrera
- Bionanotechnology Laboratory, Integrative Center for Applied Biology and Chemistry (CIBQA), Department of Chemical & Biological Sciences, Universidad Bernardo O’Higgins, Santiago, Chile
- Laboratório de Hepatologia Experimental, Gastroenterology Department, Facultad de Medicina, Centro de Envejecimiento y Regeneración (CARE Chile-UC), P. Universidad Catolica de Chile, Santiago, Chile
| | - Juan P. Arab
- Laboratório de Hepatologia Experimental, Gastroenterology Department, Facultad de Medicina, Centro de Envejecimiento y Regeneración (CARE Chile-UC), P. Universidad Catolica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Poncelet L, Ando K, Vergara C, Mansour S, Suain V, Yilmaz Z, Reygel A, Gilissen E, Brion JP, Leroy K. A 4R tauopathy develops without amyloid deposits in aged cat brains. Neurobiol Aging 2019; 81:200-212. [PMID: 31306814 DOI: 10.1016/j.neurobiolaging.2019.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/13/2019] [Accepted: 05/30/2019] [Indexed: 11/26/2022]
Abstract
Human tauopathies are neurodegenerative diseases with accumulation of abnormally phosphorylated and aggregated tau proteins forming neurofibrillary tangles. We investigated the development of tau pathology in aged cat brains as a model of neurofibrillary tangle formation occurring spontaneously during aging. In 4 of 6 cats aged between 18 and 21 years, we found a somatodendritic accumulation of phosphorylated and aggregated tau in neurons and oligodendrocytes. Two of these 4 cats had no amyloid immunoreactivity. These tau inclusions were mainly composed of 4R tau isoforms and straight filaments and colocalized with the active form of the glycogen synthase kinase-3 (GSK3). Cat brains with a tau pathology showed a significant cortical atrophy and neuronal loss. We demonstrate in this study the presence of a tau pathology in aged cat brains that develop independently of amyloid deposits. The colocalization of the active form of the GSK3 with tau inclusions as observed in human tauopathies suggests that this kinase could be responsible for the abnormal tau phosphorylation observed in aged cat brains, representing a mechanism of tau pathology development shared between a naturally occurring tauopathy in aged cats and human tauopathies.
Collapse
Affiliation(s)
- Luc Poncelet
- Laboratory of Anatomy, Biomechanics and Organogenesis, ULB neuroscience institute, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Cristina Vergara
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Salwa Mansour
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Valérie Suain
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Alain Reygel
- Royal Museum for Central Africa, Vertebrate Unit, Tervuren, Belgium
| | - Emmanuel Gilissen
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium; Royal Museum for Central Africa, BIOCOL Unit, Tervuren, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, ULB Neuroscience Institute, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium.
| |
Collapse
|
28
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 543] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
29
|
Vergara C, Houben S, Suain V, Yilmaz Z, De Decker R, Vanden Dries V, Boom A, Mansour S, Leroy K, Ando K, Brion JP. Amyloid-β pathology enhances pathological fibrillary tau seeding induced by Alzheimer PHF in vivo. Acta Neuropathol 2019; 137:397-412. [PMID: 30599077 DOI: 10.1007/s00401-018-1953-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022]
Abstract
Neuropathological analysis in Alzheimer's disease (AD) and experimental evidence in transgenic models overexpressing frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17) mutant tau suggest that amyloid-β pathology enhances the development of tau pathology. In this work, we analyzed this interaction independently of the overexpression of an FTDP-17 mutant tau, by analyzing tau pathology in wild-type (WT), 5xFAD, APP-/- and tau-/- mice after stereotaxic injection in the somatosensory cortex of short-length native human AD-PHF. Gallyas and phosphotau-positive tau inclusions developed in WT, 5xFAD, and APP-/- but not in tau-/- mice. Ultrastructural analysis demonstrated their intracellular localization and that they were composed of straight filaments. These seeded tau inclusions were composed only of endogenous murine tau exhibiting a tau antigenic profile similar to tau aggregates in AD. Insoluble tau level was higher and ipsilateral anteroposterior and contralateral cortical spreading of tau inclusions was more important in AD-PHF-injected 5xFAD mice than in WT mice. The formation of large plaque-associated dystrophic neurites positive for oligomeric and phosphotau was observed in 5xFAD mice injected with AD-PHF but never in control-injected or in non-injected 5xFAD mice. An increased level of the p25 activator of CDK5 kinase was found in AD-PHF-injected 5xFAD mice. These data demonstrate in vivo that the presence of Aβ pathology enhances experimentally induced tau seeding of endogenous, wild-type tau expressed at physiological level, and demonstrate the fibrillar nature of heterotopically seeded endogenous tau. These observations further support the hypothesis that Aβ enhances tau pathology development in AD through increased pathological tau spreading.
Collapse
Affiliation(s)
- Cristina Vergara
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Valérie Suain
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Robert De Decker
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Virginie Vanden Dries
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Alain Boom
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Salwa Mansour
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, 808, route de Lennik, Bldg GE, 1070, Brussels, Belgium.
| |
Collapse
|
30
|
Mudher A, Colin M, Dujardin S, Medina M, Dewachter I, Alavi Naini SM, Mandelkow EM, Mandelkow E, Buée L, Goedert M, Brion JP. What is the evidence that tau pathology spreads through prion-like propagation? Acta Neuropathol Commun 2017; 5:99. [PMID: 29258615 PMCID: PMC5735872 DOI: 10.1186/s40478-017-0488-7] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022] Open
Abstract
Emerging experimental evidence suggests that the spread of tau pathology in the brain in Tauopathies reflects the propagation of abnormal tau species along neuroanatomically connected brain areas. This propagation could occur through a "prion-like" mechanism involving transfer of abnormal tau seeds from a "donor cell" to a "recipient cell" and recruitment of normal tau in the latter to generate new tau seeds. This review critically appraises the evidence that the spread of tau pathology occurs via such a "prion-like" mechanism and proposes a number of recommendations for directing future research. Recommendations for definitions of frequently used terms in the tau field are presented in an attempt to clarify and standardize interpretation of research findings. Molecular and cellular factors affecting tau aggregation are briefly reviewed, as are potential contributions of physiological and pathological post-translational modifications of tau. Additionally, the experimental evidence for tau seeding and "prion-like" propagation of tau aggregation that has emerged from cellular assays and in vivo models is discussed. Propagation of tau pathology using "prion-like" mechanisms is expected to incorporate several steps including cellular uptake, templated seeding, secretion and intercellular transfer through synaptic and non-synaptic pathways. The experimental findings supporting each of these steps are reviewed. The clinical validity of these experimental findings is then debated by considering the supportive or contradictory findings from patient samples. Further, the role of physiological tau release in this scenario is examined because emerging data shows that tau is secreted but the physiological function (if any) of this secretion in the context of propagation of pathological tau seeds is unclear. Bona fide prions exhibit specific properties, including transmission from cell to cell, tissue to tissue and organism to organism. The propagation of tau pathology has so far not been shown to exhibit all of these steps and how this influences the debate of whether or not abnormal tau species can propagate in a "prion-like" manner is discussed. The exact nature of tau seeds responsible for propagation of tau pathology in human tauopathies remains controversial; it might be tightly linked to the existence of tau strains stably propagating peculiar patterns of neuropathological lesions, corresponding to the different patterns seen in human tauopathies. That this is a property shared by all seed-competent tau conformers is not yet firmly established. Further investigation is also required to clarify the relationship between propagation of tau aggregates and tau-induced toxicity. Genetic variants identified as risks factors for tauopathies might play a role in propagation of tau pathology, but many more studies are needed to document this. The contribution of selective vulnerability of neuronal populations, as an alternative to prion-like mechanisms to explain spreading of tau pathology needs to be clarified. Learning from the prion field will be helpful to enhance our understanding of propagation of tau pathology. Finally, development of better models is expected to answer some of these key questions and allow for the testing of propagation-centred therapies.
Collapse
Affiliation(s)
- Amrit Mudher
- University of Southampton, Biological Sciences, Faculty of Natural and Environmental Sciences, SO17 1BJ, Southampton, UK.
| | - Morvane Colin
- Univ. Lille, Inserm, CHU-Lille, UMR-S 1172, LabEx DISTALZ, 59000, Lille, France
| | - Simon Dujardin
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Miguel Medina
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Ilse Dewachter
- Dementia Research Group, BioMedical Research Institute, Hasselt University, 3500, Hasselt, Belgium
| | - Seyedeh Maryam Alavi Naini
- Institut de Biologie Paris Seine-Laboratoire Neuroscience Paris Seine INSERM UMRS 1130, CNRS UMR 8246, UPMC UM 118 Université Pierre et Marie Curie, Paris, France
| | - Eva-Maria Mandelkow
- DZNE (German Ctr. Neurodegen. Diseases), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
- DESY, Hamburg, Germany
| | - Eckhard Mandelkow
- DZNE (German Ctr. Neurodegen. Diseases), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
- DESY, Hamburg, Germany
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, UMR-S 1172, LabEx DISTALZ, 59000, Lille, France
| | - Michel Goedert
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CB2 0QH, Cambridge, UK
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology Université Libre de Bruxelles, Faculty of Medicine, ULB Neuroscience Institute (UNI) 808, route de Lennik 1070, Brussels, Belgium.
| |
Collapse
|
31
|
Axonal and myelinic pathology in 5xFAD Alzheimer's mouse spinal cord. PLoS One 2017; 12:e0188218. [PMID: 29176903 PMCID: PMC5703477 DOI: 10.1371/journal.pone.0188218] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/02/2017] [Indexed: 01/14/2023] Open
Abstract
As an extension of the brain, the spinal cord has unique properties which could allow us to gain a better understanding of CNS pathology. The brain and cord share the same cellular components, yet the latter is simpler in cytoarchitecture and connectivity. In Alzheimer's research, virtually all focus is on brain pathology, however it has been shown that transgenic Alzheimer's mouse models accumulate beta amyloid plaques in spinal cord, suggesting that the cord possesses the same molecular machinery and conditions for plaque formation. Here we report a spatial-temporal map of plaque load in 5xFAD mouse spinal cord. We found that plaques started to appear at 11 weeks, then exhibited a time dependent increase and differential distribution along the cord. More plaques were found in cervical than other spinal levels at all time points examined. Despite heavy plaque load at 6 months, the number of cervical motor neurons in 5xFAD mice is comparable to wild type littermates. On detailed microscopic examination, fine beta amyloid-containing and beta sheet-rich thread-like structures were found in the peri-axonal space of many axons. Importantly, these novel structures appear before any plaque deposits are visible in young mice spinal cord and they co-localize with axonal swellings at later stages, suggesting that these thread-like structures might represent the initial stages of plaque formation, and could play a role in axonal damage. Additionally, we were able to demonstrate increasing myelinopathy in aged 5xFAD mouse spinal cord using the lipid probe Nile Red with high resolution. Collectively, we found significant amyloid pathology in grey and white matter of the 5xFAD mouse spinal cord which indicates that this structure maybe a useful platform to study mechanisms of Alzheimer's pathology and disease progression.
Collapse
|
32
|
Bennett RE, DeVos SL, Dujardin S, Corjuc B, Gor R, Gonzalez J, Roe AD, Frosch MP, Pitstick R, Carlson GA, Hyman BT. Enhanced Tau Aggregation in the Presence of Amyloid β. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1601-1612. [PMID: 28500862 DOI: 10.1016/j.ajpath.2017.03.011] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/01/2017] [Accepted: 03/22/2017] [Indexed: 01/12/2023]
Abstract
Amyloid plaques and neurofibrillary tangles co-occur in Alzheimer disease, but with different topological and temporal patterns. Whether these two lesions are independent or pathobiologically related is uncertain. For example, amyloid deposition in the neocortex precedes the spread of tau neurofibrillary tangles from the limbic areas to the cortex. We examined the aggregation properties of tau isolated from human cases with early tau pathology (Braak II) with and without plaques. Using a well-established HEK cell biosensor assay, we show that tau from cases with plaques has an enhanced ability to induce tau aggregates compared to tau from cases without plaques. To further explore this effect, we combined mice carrying the APP/PS1 transgene array that develop plaques with rTg4510 mice carrying the P301L mutant human tau transgene that develop extensive tau pathology with age. The resulting APP/PS1-rTg4510 mice had a threefold increase in tau seeding activity over the rTg4510 strain, without change in tau production or extracellular release. Surprisingly, this effect was observed before overt amyloid deposition. The enhancement of tau aggregation was also apparent by an increase in histological measures of tau pathology in young APP/PS1-rTg4510 mice and an increase in high-molecular-weight tau. Overall, these data provide evidence that amyloid β acts to enhance tau pathology by increasing the formation of tau species capable of seeding new aggregates.
Collapse
Affiliation(s)
- Rachel E Bennett
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Sarah L DeVos
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Simon Dujardin
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Bianca Corjuc
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Rucha Gor
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Jose Gonzalez
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Allyson D Roe
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Matthew P Frosch
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Bradley T Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| |
Collapse
|
33
|
Hüttenrauch M, Walter S, Kaufmann M, Weggen S, Wirths O. Limited Effects of Prolonged Environmental Enrichment on the Pathology of 5XFAD Mice. Mol Neurobiol 2016; 54:6542-6555. [DOI: 10.1007/s12035-016-0167-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
|
34
|
Audouard E, Houben S, Masaracchia C, Yilmaz Z, Suain V, Authelet M, De Decker R, Buée L, Boom A, Leroy K, Ando K, Brion JP. High-Molecular-Weight Paired Helical Filaments from Alzheimer Brain Induces Seeding of Wild-Type Mouse Tau into an Argyrophilic 4R Tau Pathology in Vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2709-22. [PMID: 27497324 DOI: 10.1016/j.ajpath.2016.06.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/14/2016] [Accepted: 06/06/2016] [Indexed: 12/14/2022]
Abstract
In Alzheimer disease, the development of tau pathology follows neuroanatomically connected pathways, suggesting that abnormal tau species might recruit normal tau by passage from cell to cell. Herein, we analyzed the effect of stereotaxic brain injection of human Alzheimer high-molecular-weight paired helical filaments (PHFs) in the dentate gyrus of wild-type and mutant tau THY-Tau22 mice. After 3 months of incubation, wild-type and THY-Tau22 mice developed an atrophy of the dentate gyrus and a tau pathology characterized by Gallyas and tau-positive grain-like inclusions into granule cells that extended in the hippocampal hilus and eventually away into the alveus, and the fimbria. Gallyas-positive neuropil threads and oligodendroglial coiled bodies were also observed. These tau inclusions were composed only of mouse tau, and were immunoreactive with antibodies to 4R tau, phosphotau, misfolded tau, ubiquitin, and p62. Although local hyperphosphorylation of tau was increased in the dentate gyrus in THY-Tau22 mice, the development of neurofibrillary tangles made of mutant human tau was not accelerated in the hippocampus, indicating that wild-type human PHFs were inefficient in seeding tau aggregates made of G272V/P301S mutant human tau. Our results indicate thus that injection of human wild-type Alzheimer PHF seeded aggregation of wild-type murine tau into an argyrophilic 4R tau pathology, and constitutes an interesting model independent of expression of a mutant tau protein.
Collapse
Affiliation(s)
- Emilie Audouard
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Caterina Masaracchia
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Valérie Suain
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Michèle Authelet
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Robert De Decker
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Luc Buée
- INSERM, U837, Université de Lille 2, Lille, France
| | - Alain Boom
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium.
| |
Collapse
|
35
|
Endres K, Reinhardt S, Geladaris A, Knies J, Grimm M, Hartmann T, Schmitt U. Transnasal delivery of human A-beta peptides elicits impaired learning and memory performance in wild type mice. BMC Neurosci 2016; 17:44. [PMID: 27377996 PMCID: PMC4932715 DOI: 10.1186/s12868-016-0280-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/24/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Murine models of Alzheimer's disease (AD) are mainly based on overexpression of pathologic amyloid precursor protein and/or presenilins. Those genes resemble underlying cause of early onset type of AD while about 99 % of all human cases are to be characterized as sporadic, late onset. Appropriate animal models for this type of AD are still missing. We here investigated, if transnasal delivery of A-beta 42 peptides might serve to mimic pathological effects in mice. RESULTS A-beta 42 peptides, used for the behavioral study, showed the expected dose-dependent toxicity in neur oblastoma cell line SH-SY5Y and were able to form higher molecular weight species in vitro. Upon delivery into nostrils of wild type mice, protein bands that might represent aggregation products of the exogenously applied human A-beta 42 were only observed in total brain homogenates from mice pre-treated with mannitol. By using TAMRA-labeled A-beta 42 peptides we demonstrated, that transport throughout the brain was achieved already 1 h after administration. FVB/N mice treated with A-beta 42 for 3 days were significantly impaired in the cue-retention condition of the fear conditioning task as compared to controls whereas A-beta-treated C57B6/J mice were impaired in the context condition. In the Morris water maze test, these mice also displayed a delayed learning performance, indicated by significantly longer time to find the platform. Those deficits were also seen for memory performance in the probe trial as measured by number of crossings of the former platform position and time spent in the goal quadrant. CONCLUSIONS Existing AD mouse models are of genetic origin and need prolonged housing time before onset of pathology. Our short-term treatment induced learning and memory deficits via exogenous application of A-beta peptides comparable to those observed for the transgenic animals. With the transnasal A-beta 42 treatment we present an approach to investigate purely A-beta related changes suitable as a model for symptoms of Alzheimer's dementia (AD). Resulting behavioral deficits were indicative for familial type of Alzheimer's disease as well as for the late onset variant.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany.
| | - Sven Reinhardt
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Anastasia Geladaris
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Julia Knies
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Marcus Grimm
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany.,Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Tobias Hartmann
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany.,Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Ulrich Schmitt
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| |
Collapse
|
36
|
Walker LC, Schelle J, Jucker M. The Prion-Like Properties of Amyloid-β Assemblies: Implications for Alzheimer's Disease. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a024398. [PMID: 27270558 DOI: 10.1101/cshperspect.a024398] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Since the discovery that prion diseases can be transmitted to experimental animals by inoculation with afflicted brain matter, researchers have speculated that the brains of patients suffering from other neurodegenerative diseases might also harbor causative agents with transmissible properties. Foremost among these disorders is Alzheimer's disease (AD), the most common cause of dementia in the elderly. A growing body of research supports the concept that the pathogenesis of AD is initiated and sustained by the endogenous, seeded misfolding and aggregation of the protein fragment amyloid-β (Aβ). At the molecular level, this mechanism of nucleated protein self-assembly is virtually identical to that of prions consisting of the prion protein (PrP). The formation, propagation, and spread of Aβ seeds within the brain can thus be considered a fundamental feature of AD pathogenesis.
Collapse
Affiliation(s)
- Lary C Walker
- Yerkes National Primate Research Center and Department of Neurology, Emory University, Atlanta, Georgia 30322
| | - Juliane Schelle
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), D-72076 Tübingen, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), D-72076 Tübingen, Germany
| |
Collapse
|
37
|
Chen W, Abud EA, Yeung ST, Lakatos A, Nassi T, Wang J, Blum D, Buée L, Poon WW, Blurton-Jones M. Increased tauopathy drives microglia-mediated clearance of beta-amyloid. Acta Neuropathol Commun 2016; 4:63. [PMID: 27339073 PMCID: PMC4918195 DOI: 10.1186/s40478-016-0336-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/13/2016] [Indexed: 12/23/2022] Open
Abstract
Alzheimer disease is characterized by the accumulation of β-amyloid (Aβ) plaques and tau-laden neurofibrillary tangles. Emerging studies suggest that in neurodegenerative diseases, aggregation of one protein species can promote other proteinopathies and that inflammation plays an important role in this process. To study the interplay between Aβ deposition, tau pathology, and microgliosis, we established a new AD transgenic mouse model by crossing 5xfAD mice with Thy-Tau22 transgenic mice. The resulting 'T5x' mice exhibit a greater than three-fold increase in misfolded and hyperphosphorylated tau and further substantiates the hypothesis that Aβ accelerates tau pathology. Surprisingly, T5x mice exhibit a 40-50 % reduction in Aβ plaque load and insoluble Aβ species when compared with aged-matched 5xfAD littermates. T5x mice exhibit significant changes in cytokine production, an almost doubling of microglial number, and a dramatic shift in microglia activation state. Furthermore, T5x microglia exhibit increased phagocytic capacity that enhances the clearance of insoluble Aβ and decreasing plaque load. Therefore, our results suggest that strategies to increase the phagocytic ability of microglia can be employed to reduce Aβ and that tau-induced changes in microglial activation state can promote the clearance of Aβ.
Collapse
|
38
|
Vasconcelos B, Stancu IC, Buist A, Bird M, Wang P, Vanoosthuyse A, Van Kolen K, Verheyen A, Kienlen-Campard P, Octave JN, Baatsen P, Moechars D, Dewachter I. Heterotypic seeding of Tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of Tau-pathology in vivo. Acta Neuropathol 2016; 131:549-69. [PMID: 26739002 PMCID: PMC4789256 DOI: 10.1007/s00401-015-1525-x] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/13/2015] [Accepted: 12/13/2015] [Indexed: 11/27/2022]
Abstract
Genetic, clinical, histopathological and biomarker data strongly support Beta-amyloid (Aβ) induced spreading of Tau-pathology beyond entorhinal cortex (EC), as a crucial process in conversion from preclinical cognitively normal to Alzheimer‘s Disease (AD), while the underlying mechanism remains unclear. In vivo preclinical models have reproducibly recapitulated Aβ-induced Tau-pathology. Tau pathology was thereby also induced by aggregated Aβ, in functionally connected brain areas, reminiscent of a prion-like seeding process. In this work we demonstrate, that pre-aggregated Aβ can directly induce Tau fibrillization by cross-seeding, in a cell-free assay, comparable to that demonstrated before for alpha-synuclein and Tau. We furthermore demonstrate, in a well-characterized cellular Tau-aggregation assay that Aβ-seeds cross-seeded Tau-pathology and strongly catalyzed pre-existing Tau-aggregation, reminiscent of the pathogenetic process in AD. Finally, we demonstrate that heterotypic seeded Tau by pre-aggregated Aβ provides efficient seeds for induction and propagation of Tau-pathology in vivo. Prion-like, heterotypic seeding of Tau fibrillization by Aβ, providing potent seeds for propagating Tau pathology in vivo, as demonstrated here, provides a compelling molecular mechanism for Aβ-induced propagation of Tau-pathology, beyond regions with pre-existing Tau-pathology (entorhinal cortex/locus coeruleus). Cross-seeding along functional connections could thereby resolve the initial spatial dissociation between amyloid- and Tau-pathology, and preferential propagation of Tau-pathology in regions with pre-existing ‘silent’ Tau-pathology, by conversion of a ‘silent’ Tau pathology to a ‘spreading’ Tau-pathology, observed in AD.
Collapse
Affiliation(s)
- Bruno Vasconcelos
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Ilie-Cosmin Stancu
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Arjan Buist
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| | - Matthew Bird
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Peng Wang
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Alexandre Vanoosthuyse
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Kristof Van Kolen
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| | - An Verheyen
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| | - Pascal Kienlen-Campard
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Jean-Noël Octave
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium
| | - Peter Baatsen
- VIB11 vzw Center for the Biology of Disease, KU Leuven, 3000, Leuven, Belgium
| | - Diederik Moechars
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| | - Ilse Dewachter
- Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200, Brussels, Belgium.
| |
Collapse
|
39
|
Eisele YS, Duyckaerts C. Propagation of Aß pathology: hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies. Acta Neuropathol 2016; 131:5-25. [PMID: 26715565 DOI: 10.1007/s00401-015-1516-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022]
Abstract
In brains of patients with Alzheimer's disease (AD), Aβ peptides accumulate in parenchyma and, almost invariably, also in the vascular walls. Although Aβ aggregation is, by definition, present in AD, its impact is only incompletely understood. It occurs in a stereotypical spatiotemporal distribution within neuronal networks in the course of the disease. This suggests a role for synaptic connections in propagating Aβ pathology, and possibly of axonal transport in an antero- or retrograde way-although, there is also evidence for passive, extracellular diffusion. Striking, in AD, is the conjunction of tau and Aβ pathology. Tau pathology in the cell body of neurons precedes Aβ deposition in their synaptic endings in several circuits such as the entorhino-dentate, cortico-striatal or subiculo-mammillary connections. However, genetic evidence suggests that Aβ accumulation is the first step in AD pathogenesis. To model the complexity and consequences of Aβ aggregation in vivo, various transgenic (tg) rodents have been generated. In rodents tg for the human Aβ precursor protein, focal injections of preformed Aβ aggregates can induce Aβ deposits in the vicinity of the injection site, and over time in more distant regions of the brain. This suggests that Aβ shares with α-synuclein, tau and other proteins the property to misfold and aggregate homotypic molecules. We propose to group those proteins under the term "propagons". Propagons may lack the infectivity of prions. We review findings from neuropathological examinations of human brains in different stages of AD and from studies in rodent models of Aβ aggregation and discuss putative mechanisms underlying the initiation and spread of Aβ pathology.
Collapse
Affiliation(s)
- Yvonne S Eisele
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Charles Duyckaerts
- Laboratoire de Neuropathologie Raymond-Escourolle, Hopital de la Pitie-Salpetriere, 47, boulevard de l'Hopital, 75651, Paris Cedex 13, France.
- ICM, equipe Alzheimer-Prion, 47, boulevard de l'Hopital, 750713, Paris, France.
| |
Collapse
|
40
|
Talwar P, Sinha J, Grover S, Rawat C, Kushwaha S, Agarwal R, Taneja V, Kukreti R. Dissecting Complex and Multifactorial Nature of Alzheimer's Disease Pathogenesis: a Clinical, Genomic, and Systems Biology Perspective. Mol Neurobiol 2015; 53:4833-64. [PMID: 26351077 DOI: 10.1007/s12035-015-9390-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/11/2015] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by loss of memory and other cognitive functions. AD can be classified into familial AD (FAD) and sporadic AD (SAD) based on heritability and into early onset AD (EOAD) and late onset AD (LOAD) based on age of onset. LOAD cases are more prevalent with genetically complex architecture. In spite of significant research focused on understanding the etiological mechanisms, search for diagnostic biomarker(s) and disease-modifying therapy is still on. In this article, we aim to comprehensively review AD literature on established etiological mechanisms including role of beta-amyloid and apolipoprotein E (APOE) along with promising newer etiological factors such as epigenetic modifications that have been associated with AD suggesting its multifactorial nature. As genomic studies have recently played a significant role in elucidating AD pathophysiology, a systematic review of findings from genome-wide linkage (GWL), genome-wide association (GWA), genome-wide expression (GWE), and epigenome-wide association studies (EWAS) was conducted. The availability of multi-dimensional genomic data has further coincided with the advent of computational and network biology approaches in recent years. Our review highlights the importance of integrative approaches involving genomics and systems biology perspective in elucidating AD pathophysiology. The promising newer approaches may provide reliable means of early and more specific diagnosis and help identify therapeutic interventions for LOAD.
Collapse
Affiliation(s)
- Puneet Talwar
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India.,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Juhi Sinha
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Sandeep Grover
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India.,Department of Paediatrics, Division of Pneumonology-Immunology, Charité University Medical Centre, Berlin, Germany
| | - Chitra Rawat
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India.,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Suman Kushwaha
- Institute of Human Behaviour and Allied Sciences (IHBAS), Delhi, India
| | - Rachna Agarwal
- Institute of Human Behaviour and Allied Sciences (IHBAS), Delhi, India
| | - Vibha Taneja
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India. .,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India.
| |
Collapse
|
41
|
Kanno T, Tsuchiya A, Tanaka A, Nishizaki T. Combination of PKCε Activation and PTP1B Inhibition Effectively Suppresses Aβ-Induced GSK-3β Activation and Tau Phosphorylation. Mol Neurobiol 2015; 53:4787-97. [DOI: 10.1007/s12035-015-9405-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/18/2015] [Indexed: 11/30/2022]
|
42
|
Activation of peroxisome proliferator-activated receptor α stimulates ADAM10-mediated proteolysis of APP. Proc Natl Acad Sci U S A 2015; 112:8445-50. [PMID: 26080426 DOI: 10.1073/pnas.1504890112] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Amyloid precursor protein (APP) derivative β-amyloid (Aβ) plays an important role in the pathogenesis of Alzheimer's disease (AD). Sequential proteolysis of APP by β-secretase and γ-secretase generates Aβ. Conversely, the α-secretase "a disintegrin and metalloproteinase" 10 (ADAM10) cleaves APP within the eventual Aβ sequence and precludes Aβ generation. Therefore, up-regulation of ADAM10 represents a plausible therapeutic strategy to combat overproduction of neurotoxic Aβ. Peroxisome proliferator-activated receptor α (PPARα) is a transcription factor that regulates genes involved in fatty acid metabolism. Here, we determined that the Adam10 promoter harbors PPAR response elements; that knockdown of PPARα, but not PPARβ or PPARγ, decreases the expression of Adam10; and that lentiviral overexpression of PPARα restored ADAM10 expression in Ppara(-/-) neurons. Gemfibrozil, an agonist of PPARα, induced the recruitment of PPARα:retinoid x receptor α, but not PPARγ coactivator 1α (PGC1α), to the Adam10 promoter in wild-type mouse hippocampal neurons and shifted APP processing toward the α-secretase, as determined by augmented soluble APPα and decreased Aβ production. Accordingly, Ppara(-/-) mice displayed elevated SDS-stable, endogenous Aβ and Aβ1-42 relative to wild-type littermates, whereas 5XFAD mice null for PPARα (5X/α(-/-)) exhibited greater cerebral Aβ load relative to 5XFAD littermates. These results identify PPARα as an important factor regulating neuronal ADAM10 expression and, thus, α-secretase proteolysis of APP.
Collapse
|
43
|
Stancu IC, Vasconcelos B, Ris L, Wang P, Villers A, Peeraer E, Buist A, Terwel D, Baatsen P, Oyelami T, Pierrot N, Casteels C, Bormans G, Kienlen-Campard P, Octave JN, Moechars D, Dewachter I. Templated misfolding of Tau by prion-like seeding along neuronal connections impairs neuronal network function and associated behavioral outcomes in Tau transgenic mice. Acta Neuropathol 2015; 129:875-94. [PMID: 25862635 PMCID: PMC4436846 DOI: 10.1007/s00401-015-1413-4] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
Prion-like seeding and propagation of Tau-pathology have been demonstrated experimentally and may underlie the stereotyped progression of neurodegenerative Tauopathies. However, the involvement of templated misfolding of Tau in neuronal network dysfunction and behavioral outcomes remains to be explored in detail. Here we analyzed the repercussions of prion-like spreading of Tau-pathology via neuronal connections on neuronal network function in TauP301S transgenic mice. Spontaneous and GABA(A)R-antagonist-induced neuronal network activity were affected following templated Tau-misfolding using synthetic preformed Tau fibrils in cultured primary neurons. Electrophysiological analysis in organotypic hippocampal slices of Tau transgenic mice demonstrated impaired synaptic transmission and impaired long-term potentiation following Tau-seed induced Tau-aggregation. Intracerebral injection of Tau-seeds in TauP301S mice, caused prion-like spreading of Tau-pathology through functionally connected neuroanatomical pathways. Electrophysiological analysis revealed impaired synaptic plasticity in hippocampal CA1 region 6 months after Tau-seeding in entorhinal cortex (EC). Furthermore, templated Tau aggregation impaired cognitive function, measured in the object recognition test 6 months post-seeding. In contrast, Tau-seeding in basal ganglia and subsequent spreading through functionally connected neuronal networks involved in motor control, resulted in motoric deficits reflected in clasping and impaired inverted grid hanging, not significantly affected following Tau-seeding in EC. Immunostaining, biochemical and electron microscopic analysis in the different models suggested early pathological forms of Tau, including Tau-oligomers, rather than fully mature neurofibrillary tangles (NFTs) as culprits of neuronal dysfunction. We here demonstrate for the first time using in vitro, ex vivo and in vivo models, that prion-like spreading of Tau-misfolding by Tau seeds, along unique neuronal connections, causes neuronal network dysfunction and associated behavioral dysfunction. Our data highlight the potential relevance of this mechanism in the symptomatic progression in Tauopathies. We furthermore demonstrate that the initial site of Tau-seeding thereby determines the behavioral outcome, potentially underlying the observed heterogeneity in (familial) Tauopathies, including in TauP301 mutants.
Collapse
Affiliation(s)
- Ilie-Cosmin Stancu
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Bruno Vasconcelos
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Laurence Ris
- />Department of Neurosciences, University of Mons, 7000 Mons, Belgium
| | - Peng Wang
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Agnès Villers
- />Department of Neurosciences, University of Mons, 7000 Mons, Belgium
| | - Eve Peeraer
- />Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Arjan Buist
- />Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Dick Terwel
- />reMYND nv, Gaston Geenslaan 1, 3001 Leuven, Belgium
| | - Peter Baatsen
- />VIB11 vzw Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium
| | - Tutu Oyelami
- />Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Nathalie Pierrot
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Cindy Casteels
- />MoSAIC-Molecular Small Animal Imaging Centre, KU Leuven, 3000 Leuven, Belgium
| | - Guy Bormans
- />MoSAIC-Molecular Small Animal Imaging Centre, KU Leuven, 3000 Leuven, Belgium
| | - Pascal Kienlen-Campard
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Jean-Nöel Octave
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Diederik Moechars
- />Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Ilse Dewachter
- />Alzheimer Dementia Group, Institute of Neuroscience, Catholic University of Louvain, 1200 Brussels, Belgium
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Alzheimer's disease is a complex multifactorial age-related neurodegenerative disorder. Current transgenic animal models do not fully recapitulate human Alzheimer's disease at the molecular, cellular and behavioural levels. This review aims to address the clinical relevance of using 'physiologically' aged rats, dogs and Octodon degus, as more representative 'natural' ecologically valid models to elucidate mechanistic aspects of Alzheimer's disease, and for the development of therapeutic agents to attenuate age-related cognitive decline. RECENT FINDINGS Aged rats, dogs and O. degus decline cognitively and ultimately develop Alzheimer's disease-like symptoms in response to the natural ageing process. Aged rats provide a tractable and popular model to examine the neurobiological basis underlying cognitive decline with age, but they do not develop Alzheimer's disease pathology. Progressive accumulation of abnormal amyloid-beta in extracellular plaques and surrounding cerebral vasculature is a common feature in human Alzheimer's disease, aged canine model and most nonhuman primates. Interestingly, the O. degus develops amyloid-beta deposits, neurofibrillary tangles containing hyperphosphorylated tau protein, altered cholinergic transmission and cognitive deficits analogous to those observed in Alzheimer's disease. Natural animal models better represent the full pathophysiology of Alzheimer's disease and are not only a viable alternative to transgenic models, but also are arguably the preferable model. SUMMARY 'Natural' models are useful to elucidate the neurobiological basis of Alzheimer's disease and develop effective therapeutic strategies that can be translated into human clinical trials.
Collapse
|
45
|
Malm T, Mariani M, Donovan LJ, Neilson L, Landreth GE. Activation of the nuclear receptor PPARδ is neuroprotective in a transgenic mouse model of Alzheimer's disease through inhibition of inflammation. J Neuroinflammation 2015; 12:7. [PMID: 25592770 PMCID: PMC4310027 DOI: 10.1186/s12974-014-0229-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/21/2014] [Indexed: 12/23/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a multifactorial disorder associated with the accumulation of soluble forms of beta-amyloid (Aβ) and its subsequent deposition into plaques. One of the major contributors to neuronal death is chronic and uncontrolled inflammatory activation of microglial cells around the plaques and their secretion of neurotoxic molecules. A shift in microglial activation towards a phagocytic phenotype has been proposed to confer benefit in models of AD. Peroxisome proliferator activator receptor δ (PPARδ) is a transcription factor with potent anti-inflammatory activation properties and PPARδ agonism leads to reduction in brain Aβ levels in 5XFAD mice. This study was carried out to elucidate the involvement of microglial activation in the PPARδ-mediated reduction of Aβ burden and subsequent outcome to neuronal survival in a 5XFAD mouse model of AD. Methods 5XFAD mice were orally treated with the PPARδ agonist GW0742 for 2 weeks. The brain Aβ load, glial activation, and neuronal survival were assessed by immunohistochemistry and quantitative PCR. In addition, the ability of GW0742 to prevent direct neuronal death as well as inflammation-induced neuron death was analyzed in vitro. Results Our results show for the first time that a short treatment period of 5XFAD mice was effective in reducing the parenchymal Aβ load without affecting the levels of intraneuronal Aβ. This was concomitant with a decrease in overall microglial activation and reduction in proinflammatory mediators. Instead, microglial immunoreactivity around Aβ deposits was increased. Importantly, the reduction in the proinflammatory milieu elicited by GW0742 treatment resulted in attenuation of neuronal loss in vivo in the subiculum of 5XFAD mice. In addition, whereas GW0742 failed to protect primary neurons against glutamate-induced cell death, it prevented inflammation-induced neuronal death in microglia-neuron co-cultures in vitro. Conclusions This study demonstrates that GW0742 treatment has a prominent anti-inflammatory effect in 5XFAD mice and suggests that PPARδ agonists may have therapeutic utility in treating AD.
Collapse
Affiliation(s)
- Tarja Malm
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| | - Monica Mariani
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Lauren J Donovan
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Lee Neilson
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Gary E Landreth
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
46
|
Stover KR, Campbell MA, Van Winssen CM, Brown RE. Analysis of motor function in 6-month-old male and female 3xTg-AD mice. Behav Brain Res 2014; 281:16-23. [PMID: 25486177 DOI: 10.1016/j.bbr.2014.11.046] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 11/24/2014] [Accepted: 11/29/2014] [Indexed: 12/19/2022]
Abstract
The 3xTg-AD mouse has high validity as a model of Alzheimer's disease (AD) because it develops both amyloid beta plaques and neurofibrillary tangles. Human patients with AD typically develop motor deficits, which worsen as the disease progresses, but 3xTg-AD mice have been reported to show enhanced motor abilities. We investigated the motor behaviour phenotype of male and female 3xTg-AD and B6129SF2 wildtype mice on a battery of motor behaviours at 6 months of age. Compared to wildtype mice, the 3xTg-AD mice had enhanced motor performance on the Rotarod, but worse performance on the grid suspension task. In gait analysis 3xTg-AD mice had a longer stride length and made more foot slips on the balance beam than wildtype mice. There was no overall difference in voluntary wheel-running activity between genotypes, but there was a disruption in circadian activity rhythm in 3xTg-AD mice. In some motor tasks, such as the Rotarod and balance beam, females appeared to perform better than males, but this sex differences was accounted for by differences in body weight. Our results indicate that while the 3xTg-AD mice show enhanced performance on the Rotarod, they have poorer performance on other motor behaviour tasks, indicating that their motor behaviour phenotype is more complex than previously reported. The presence of the P301L transgene may explain the enhancement of Rotarod performance but the poorer performance on other motor behaviour tasks may be due to other transgenes.
Collapse
Affiliation(s)
- Kurt R Stover
- Department of Psychology and Neuroscience, Dalhousie University, PO Box 1500, Halifax, NS B3H 4R2, Canada
| | - Mackenzie A Campbell
- Department of Psychology and Neuroscience, Dalhousie University, PO Box 1500, Halifax, NS B3H 4R2, Canada
| | - Christine M Van Winssen
- Department of Psychology and Neuroscience, Dalhousie University, PO Box 1500, Halifax, NS B3H 4R2, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, PO Box 1500, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
47
|
Stancu IC, Vasconcelos B, Terwel D, Dewachter I. Models of β-amyloid induced Tau-pathology: the long and "folded" road to understand the mechanism. Mol Neurodegener 2014; 9:51. [PMID: 25407337 PMCID: PMC4255655 DOI: 10.1186/1750-1326-9-51] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 10/14/2014] [Indexed: 02/28/2023] Open
Abstract
The amyloid cascade hypothesis has been the prevailing hypothesis in Alzheimer’s Disease research, although the final and most wanted proof i.e. fully successful anti-amyloid clinical trials in patients, is still lacking. This may require a better in depth understanding of the cascade. Particularly, the exact toxic forms of Aβ and Tau, the molecular link between them and their respective contributions to the disease process need to be identified in detail. Although the lack of final proof has raised substantial criticism on the hypothesis per se, accumulating experimental evidence in in vitro models, in vivo models and from biomarkers analysis in patients supports the amyloid cascade and particularly Aβ-induced Tau-pathology, which is the focus of this review. We here discuss available models that recapitulate Aβ-induced Tau-pathology and review some potential underlying mechanisms. The availability and diversity of these models that mimic the amyloid cascade partially or more complete, provide tools to study remaining questions, which are crucial for development of therapeutic strategies for Alzheimer’s Disease.
Collapse
Affiliation(s)
| | | | | | - Ilse Dewachter
- Catholic University of Louvain, Institute of Neuroscience, Alzheimer Dementia, Av, E, Mounier 53, Av, Hippocrate 54, B-1200 Brussels, Belgium.
| |
Collapse
|
48
|
Stancu IC, Ris L, Vasconcelos B, Marinangeli C, Goeminne L, Laporte V, Haylani LE, Couturier J, Schakman O, Gailly P, Pierrot N, Kienlen-Campard P, Octave JN, Dewachter I. Tauopathy contributes to synaptic and cognitive deficits in a murine model for Alzheimer's disease. FASEB J 2014; 28:2620-31. [PMID: 24604080 DOI: 10.1096/fj.13-246702] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tau alterations are now considered an executor of neuronal demise and cognitive dysfunction in Alzheimer's disease (AD). Mouse models combining amyloidosis and tauopathy and their parental counterparts are important tools to further investigate the interplay of abnormal amyloid-β (Aβ) and Tau species in pathogenesis, synaptic and neuronal dysfunction, and cognitive decline. Here, we crossed APP/PS1 mice with 5 early-onset familial AD mutations (5xFAD) and TauP301S (PS19) transgenic mice, denoted F(+)/T(+) mice, and phenotypically compared them to their respective parental strains, denoted F(+)/T(-) and F(-)/T(+) respectively, as controls. We found dramatically aggravated tauopathy (~10-fold) in F(+)/T(+) mice compared to the parental F(-)/T(+) mice. In contrast, amyloidosis was unaltered compared to the parental F(+)/T(-) mice. Tauopathy was invariably and very robustly aggravated in hippocampal and cortical brain regions. Most important, F(+)/T(+) displayed aggravated cognitive deficits in a hippocampus-dependent spatial navigation task, compared to the parental F(+)/T(-) strain, while parental F(-)/T(+) mice did not display cognitive impairment. Basal synaptic transmission was impaired in F(+)/T(+) mice compared to nontransgenic mice and the parental strains (≥40%). Finally, F(+)/T(+) mice displayed a significant hippocampal atrophy (~20%) compared to nontransgenic mice, in contrast to the parental strains. Our data indicate for the first time that pathological Aβ species (or APP/PS1) induced changes in Tau contribute to cognitive deficits correlating with synaptic deficits and hippocampal atrophy in an AD model. Our data lend support to the amyloid cascade hypothesis with a role of pathological Aβ species as initiator and pathological Tau species as executor.
Collapse
Affiliation(s)
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Mons, Belgium
| | | | | | | | | | | | | | - Olivier Schakman
- Department of Cell Physiology, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium; and
| | - Philippe Gailly
- Department of Cell Physiology, Institute of Neuroscience, Catholic University of Louvain, Brussels, Belgium; and
| | | | | | | | | |
Collapse
|