1
|
Abdelmoaty MM, Yeapuri P, Machhi J, Lu Y, Namminga KL, Kadry R, Lu E, Bhattarai S, Mosley RL, Gendelman HE. Immune senescence in aged APP/PS1 mice. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:317-330. [PMID: 38023614 PMCID: PMC10659760 DOI: 10.1515/nipt-2023-0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023]
Abstract
Objectives To evaluate the linkage between age and deficits in innate and adaptive immunity which heralds both Alzheimer's disease (AD) onset and progression. The pathobiological events which underlie and tie these outcomes remain not fully understood. Methods To investigate age-dependent immunity in AD, we evaluated innate and adaptive immunity in coordinate studies of regulatory T cell (Treg) function, T cell frequencies, and microglial integrity. These were assessed in blood, peripheral lymphoid tissues, and the hippocampus of transgenic (Tg) amyloid precursor protein/presenilin 1 (APP/PS1) against non-Tg mice. Additionally, immune arrays of hippocampal tissue were performed at 4, 6, 12, and 20 months of age. Results APP/PS1 mice showed progressive impairment of Treg immunosuppressive function with age. There was partial restoration of Treg function in 20-month-old mice. Ingenuity pathway analyses of hippocampal tissues were enriched in inflammatory, oxidative, and cellular activation pathways that paralleled advancing age and AD-pathobiology. Operative genes in those pathways included, but were not limited to triggering receptor on myeloid cells 1 (TREM1), T helper type 1 (Th1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. Interleukin-17 (IL-17), nitric oxide, acute phase, and T cell receptor signaling pathways were also perturbed. Significant inflammation was observed at 6- and 12-months. However, at 20-months, age associated partial restoration of Treg function reduced inflammatory phenotype. Conclusions Impaired Treg function, inflammation and oxidative stress were associated with AD pathology. Age associated partial restoration of Treg function in old mice reduced the hippocampal inflammatory phenotype. Restoring Treg suppressive function can be a therapeutic modality for AD.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
2
|
Latina V, De Introna M, Caligiuri C, Loviglio A, Florio R, La Regina F, Pignataro A, Ammassari-Teule M, Calissano P, Amadoro G. Immunotherapy with Cleavage-Specific 12A12mAb Reduces the Tau Cleavage in Visual Cortex and Improves Visuo-Spatial Recognition Memory in Tg2576 AD Mouse Model. Pharmaceutics 2023; 15:pharmaceutics15020509. [PMID: 36839831 PMCID: PMC9965010 DOI: 10.3390/pharmaceutics15020509] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Tau-targeted immunotherapy is a promising approach for treatment of Alzheimer's disease (AD). Beyond cognitive decline, AD features visual deficits consistent with the manifestation of Amyloid β-protein (Aβ) plaques and neurofibrillary tangles (NFT) in the eyes and higher visual centers, both in animal models and affected subjects. We reported that 12A12-a monoclonal cleavage-specific antibody (mAb) which in vivo neutralizes the neurotoxic, N-terminal 20-22 kDa tau fragment(s)-significantly reduces the retinal accumulation in Tg(HuAPP695Swe)2576 mice of both tau and APP/Aβ pathologies correlated with local inflammation and synaptic deterioration. Here, we report the occurrence of N-terminal tau cleavage in the primary visual cortex (V1 area) and the beneficial effect of 12A12mAb treatment on phenotype-associated visuo-spatial deficits in this AD animal model. We found out that non-invasive administration of 12 A12mAb markedly reduced the pathological accumulation of both truncated tau and Aβ in the V1 area, correlated to significant improvement in visual recognition memory performance along with local increase in two direct readouts of cortical synaptic plasticity, including the dendritic spine density and the expression level of activity-regulated cytoskeleton protein Arc/Arg3.1. Translation of these findings to clinical therapeutic interventions could offer an innovative tau-directed opportunity to delay or halt the visual impairments occurring during AD progression.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Margherita De Introna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Chiara Caligiuri
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Alessia Loviglio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Rita Florio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Martine Ammassari-Teule
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Via Ercole Ramarini 32, 00015 Rome, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-06-49255252
| |
Collapse
|
3
|
Mice in translational neuroscience: What R we doing? Prog Neurobiol 2022; 217:102330. [PMID: 35872220 DOI: 10.1016/j.pneurobio.2022.102330] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 12/28/2022]
Abstract
Animal models play a pivotal role in translational neuroscience but recurrent problems in data collection, analyses, and interpretation, lack of biomarkers, and a tendency to over-reliance on mice have marred neuroscience progress, leading to one of the highest attrition rates in drug translation. Global initiatives to improve reproducibility and model selection are being implemented. Notwithstanding, mice are still the preferred animal species to model human brain disorders even when the translation has been shown to be limited. Non-human primates are better positioned to provide relevant translational information because of their higher brain complexity and homology to humans. Among others, lack of resources and formal training, strict legislation, and ethical issues may impede broad access to large animals. We propose that instead of increasingly restrictive legislation, more resources for training, education, husbandry, and data sharing are urgently needed. The creation of multidisciplinary teams, in which veterinarians need to play a key role, would be critical to improve translational efficiency. Furthermore, it is not usually acknowledged by researchers and regulators the value of comparative studies in lower species, that are instrumental in toxicology, target identification, and mechanistic studies. Overall, we highlight here the need for a conceptual shift in neuroscience research and policies to reach the patients.
Collapse
|
4
|
Fernandes A, Caldeira C, Cunha C, Ferreiro E, Vaz AR, Brites D. Differences in Immune-Related Genes Underlie Temporal and Regional Pathological Progression in 3xTg-AD Mice. Cells 2022; 11:cells11010137. [PMID: 35011699 PMCID: PMC8750089 DOI: 10.3390/cells11010137] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 01/27/2023] Open
Abstract
The prevalence of Alzheimer’s disease (AD), the most common cause of age-associated dementia, is estimated to increase over the next decades. Evidence suggests neuro-immune signaling deregulation and risk genes beyond the amyloid-β (Aβ) deposition in AD pathology. We examined the temporal profile of inflammatory mediators and microglia deactivation/activation in the brain cortex and hippocampus of 3xTg-AD mice at 3- and 9-month-old. We found upregulated APP processing, decreased expression of CD11b, CX3CR1, MFG-E8, TNF-α, IL-1β, MHC-II and C/EBP-α and increased miR-146a in both brain regions in 3-month-old 3xTG-AD mice, suggestive of a restrictive regulation. Enhanced TNF-α, IL-1β, IL-6, iNOS, SOCS1 and Arginase 1 were only present in the hippocampus of 9-month-old animals, though elevation of HMGB1 and reduction of miR-146a and miR-124 were common features in the hippocampus and cortex regions. miR-155 increased early in the cortex and later in both regions, supporting its potential as a biomarker. Candidate downregulated target genes by cortical miR-155 included Foxo3, Runx2 and CEBPβ at 3 months and Foxo3, Runx2 and Socs1 at 9 months, which are implicated in cell survival, but also in Aβ pathology and microglia/astrocyte dysfunction. Data provide new insights across AD state trajectory, with divergent microglia phenotypes and inflammatory-associated features, and identify critical targets for drug discovery and combinatorial therapies.
Collapse
Affiliation(s)
- Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Correspondence: (A.F.); (D.B.); Tel.: +351-217946450 (D.B.)
| | - Cláudia Caldeira
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
| | - Carolina Cunha
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
- Bruno Silva-Santos Lab, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Elisabete Ferreiro
- MitoXT-Mitochondrial Toxicologu and Experimental Therapeutics Laboratory, CNC-Center for Neuroscience and Cell Biology, Universidade de Coimbra, 3004-516 Coimbra, Portugal;
- III-Institute for Interdisciplinary Research (IIIUC), Universidade de Coimbra, 3004-516 Coimbra, Portugal
| | - Ana Rita Vaz
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
- Correspondence: (A.F.); (D.B.); Tel.: +351-217946450 (D.B.)
| |
Collapse
|
5
|
Bassil R, Shields K, Granger K, Zein I, Ng S, Chih B. Improved modeling of human AD with an automated culturing platform for iPSC neurons, astrocytes and microglia. Nat Commun 2021; 12:5220. [PMID: 34471104 PMCID: PMC8410795 DOI: 10.1038/s41467-021-25344-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/27/2021] [Indexed: 11/17/2022] Open
Abstract
Advancement in human induced pluripotent stem cell (iPSC) neuron and microglial differentiation protocols allow for disease modeling using physiologically relevant cells. However, iPSC differentiation and culturing protocols have posed challenges to maintaining consistency. Here, we generated an automated, consistent, and long-term culturing platform of human iPSC neurons, astrocytes, and microglia. Using this platform we generated a iPSC AD model using human derived cells, which showed signs of Aβ plaques, dystrophic neurites around plaques, synapse loss, dendrite retraction, axon fragmentation, phospho-Tau induction, and neuronal cell death in one model. We showed that the human iPSC microglia internalized and compacted Aβ to generate and surround the plaques, thereby conferring some neuroprotection. We investigated the mechanism of action of anti-Aβ antibodies protection and found that they protected neurons from these pathologies and were most effective before pTau induction. Taken together, these results suggest that this model can facilitate target discovery and drug development efforts. Human induced pluripotent stem cell (iPSC) cells have been used to model disease in specific cell types. Here, the authors develop an automated long-term culturing platform of human iPSC neurons, astrocytes, and microglia and use it to model some cellular aspects of Alzheimer’s disease.
Collapse
Affiliation(s)
- Reina Bassil
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA.,Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA, USA
| | - Kenneth Shields
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Kevin Granger
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Ivan Zein
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Shirley Ng
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Ben Chih
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA. .,Department of Neuroscience, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
6
|
Vitale P, Salgueiro-Pereira AR, Lupascu CA, Willem M, Migliore R, Migliore M, Marie H. Analysis of Age-Dependent Alterations in Excitability Properties of CA1 Pyramidal Neurons in an APPPS1 Model of Alzheimer's Disease. Front Aging Neurosci 2021; 13:668948. [PMID: 34177555 PMCID: PMC8230571 DOI: 10.3389/fnagi.2021.668948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/14/2021] [Indexed: 11/29/2022] Open
Abstract
Age-dependent accumulation of amyloid-β, provoking increasing brain amyloidopathy, triggers abnormal patterns of neuron activity and circuit synchronization in Alzheimer’s disease (AD) as observed in human AD patients and AD mouse models. Recent studies on AD mouse models, mimicking this age-dependent amyloidopathy, identified alterations in CA1 neuron excitability. However, these models generally also overexpress mutated amyloid precursor protein (APP) and presenilin 1 (PS1) and there is a lack of a clear correlation of neuronal excitability alterations with progressive amyloidopathy. The active development of computational models of AD points out the need of collecting such experimental data to build a reliable disease model exhibiting AD-like disease progression. We therefore used the feature extraction tool of the Human Brain Project (HBP) Brain Simulation Platform to systematically analyze the excitability profile of CA1 pyramidal neuron in the APPPS1 mouse model. We identified specific features of neuron excitability that best correlate either with over-expression of mutated APP and PS1 or increasing Aβ amyloidopathy. Notably, we report strong alterations in membrane time constant and action potential width and weak alterations in firing behavior. Also, using a CA1 pyramidal neuron model, we evidence amyloidopathy-dependent alterations in Ih. Finally, cluster analysis of these recordings showed that we could reliably assign a trace to its correct group, opening the door to a more refined, less variable analysis of AD-affected neurons. This inter-disciplinary analysis, bringing together experimentalists and modelers, helps to further unravel the neuronal mechanisms most affected by AD and to build a biologically plausible computational model of the AD brain.
Collapse
Affiliation(s)
- Paola Vitale
- Institute of Biophysics, National Research Council, Palermo, Italy
| | | | | | - Michael Willem
- Biomedical Center, Ludwig Maximilian University of Munich, Munich, Germany
| | - Rosanna Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Hélène Marie
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| |
Collapse
|
7
|
Abstract
Transformation of astrocytes into reactive states is considered one of the major pathological hallmarks of prion and other neurodegenerative diseases. Recent years witnessed a growing appreciation of the view that reactive astrocytes are intimately involved in chronic neurodegeneration; however, little is known about their role in disease pathogenesis. The current article reviews the progress of the last few years and critically discusses controversial questions of whether reactive astrocytes associated with prion diseases are neurotoxic or neuroprotective and whether bidirectional A1–A2 model is applicable for describing polarization of astrocytes. Moreover, other important topics, including reversibility of a transition to a reactive state, along with the role of microglia and other stimuli in triggering astrocyte activation are reviewed. Defining the role of reactive astrocytes in pathogenesis of neurodegenerative diseases will open unrealized opportunities for developing new therapeutic approaches against prion and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ilia V Baskakov
- Department of Anatomy and Neurobiology, And Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
The degree of astrocyte activation is predictive of the incubation time to prion disease. Acta Neuropathol Commun 2021; 9:87. [PMID: 33980286 PMCID: PMC8114720 DOI: 10.1186/s40478-021-01192-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022] Open
Abstract
In neurodegenerative diseases including Alzheimer’s, Parkinson’s and prion diseases, astrocytes acquire disease-associated reactive phenotypes. With growing appreciation of their role in chronic neurodegeneration, the questions whether astrocytes lose their ability to perform homeostatic functions in the reactive states and whether the reactive phenotypes are neurotoxic or neuroprotective remain unsettled. The current work examined region-specific changes in expression of genes, which report on astrocyte physiological functions and their reactive states, in C57Black/6J mice challenged with four prion strains via two inoculation routes. Unexpectedly, strong reverse correlation between the incubation time to the diseases and the degree of astrocyte activation along with disturbance in functional pathways was observed. The animal groups with the most severe astrocyte response and degree of activation showed the most rapid disease progression. The degree of activation tightly intertwined with the global transformation of the homeostatic state, characterized by disturbances in multiple gene sets responsible for normal physiological functions producing a neurotoxic, reactive phenotype as a net result. The neurotoxic reactive phenotype exhibited a universal gene signature regardless of the prion strain. The current work suggests that the degree of astrocyte activation along with the disturbance in their physiological pathways contribute to the faster progression of disease and perhaps even drive prion pathogenesis.
Collapse
|
9
|
Kushwaha R, Sinha A, Makarava N, Molesworth K, Baskakov IV. Non-cell autonomous astrocyte-mediated neuronal toxicity in prion diseases. Acta Neuropathol Commun 2021; 9:22. [PMID: 33546775 PMCID: PMC7866439 DOI: 10.1186/s40478-021-01123-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
Under normal conditions, astrocytes perform a number of important physiological functions centered around neuronal support and synapse maintenance. In neurodegenerative diseases including Alzheimer’s, Parkinson’s and prion diseases, astrocytes acquire reactive phenotypes, which are sustained throughout the disease progression. It is not known whether in the reactive states associated with prion diseases, astrocytes lose their ability to perform physiological functions and whether the reactive states are neurotoxic or, on the contrary, neuroprotective. The current work addresses these questions by testing the effects of reactive astrocytes isolated from prion-infected C57BL/6J mice on primary neuronal cultures. We found that astrocytes isolated at the clinical stage of the disease exhibited reactive, pro-inflammatory phenotype, which also showed downregulation of genes involved in neurogenic and synaptogenic functions. In astrocyte-neuron co-cultures, astrocytes from prion-infected animals impaired neuronal growth, dendritic spine development and synapse maturation. Toward examining the role of factors secreted by reactive astrocytes, astrocyte-conditioned media was found to have detrimental effects on neuronal viability and synaptogenic functions via impairing synapse integrity, and by reducing spine size and density. Reactive microglia isolated from prion-infected animals were found to induce phenotypic changes in primary astrocytes reminiscent to those observed in prion-infected mice. In particular, astrocytes cultured with reactive microglia-conditioned media displayed hypertrophic morphology and a downregulation of genes involved in neurogenic and synaptogenic functions. In summary, the current study provided experimental support toward the non-cell autonomous mechanisms behind neurotoxicity in prion diseases and demonstrated that the astrocyte reactive phenotype associated with prion diseases is synaptotoxic.
Collapse
|
10
|
Forsell L, Vos EN, Jayaraman K, Edman A, Hussaini SA. BrainWiki-A Wiki-Style, User Driven, Comparative Brain Anatomy Tool. Front Neuroanat 2020; 14:548172. [PMID: 33192339 PMCID: PMC7604473 DOI: 10.3389/fnana.2020.548172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/23/2020] [Indexed: 11/13/2022] Open
Abstract
The mouse is the most important animal model within neuroscientific research, a position strengthened by the wide-spread use of transgenic mouse models. Discoveries in animals are followed by corroboration in humans, and the interchange between these fields of research is essential to our understanding of the human brain. With the advent of advanced technologies such as single-cell transcriptomics, epigenetic profiling and diffusion MRI, many prominent research institutes and collaborations have emerged, aiming to construct complete human or mouse brain atlases with data on gene expression, connectivity and cell types. These initiatives are indispensable resources, but frequently require extensive, time-consuming development, and rely on updates by the provider. They often come in the shape of applications which require practice or prior technical know-how. Importantly, none of them place the human and the mouse brain next to each other to allow for immediate comparison. We present BrainWiki, a user-friendly, web-based atlas that links the human and the mouse brain together, side-by-side. The platform gives the user a simple overview of brain anatomy along with published articles relating to each brain region that allows the user to delve deeper into the current state of research concerning circuitry, brain functions and pathology. The website relies on interactivity and supports user contributions resulting in a dynamic website that evolves at the pace of neuroscience. It is designed to allow for constant updates and new features in the future which will contain data such as gene expression and neuronal cell types.
Collapse
Affiliation(s)
- Linda Forsell
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, United States.,Karolinska Institutet, Stockholm, Sweden
| | | | - Keerthi Jayaraman
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, United States
| | - Axel Edman
- Karolinska Institutet, Stockholm, Sweden
| | - S Abid Hussaini
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
11
|
Stazi M, Wirths O. Chronic Memantine Treatment Ameliorates Behavioral Deficits, Neuron Loss, and Impaired Neurogenesis in a Model of Alzheimer's Disease. Mol Neurobiol 2020; 58:204-216. [PMID: 32914393 PMCID: PMC7695672 DOI: 10.1007/s12035-020-02120-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/05/2020] [Indexed: 02/06/2023]
Abstract
Memantine, a non-competitive NMDA receptor antagonist possessing neuroprotective properties, belongs to the small group of drugs which have been approved for the treatment of Alzheimer's disease (AD). While several preclinical studies employing different transgenic AD mouse models have described beneficial effects with regard to rescued behavioral deficits or reduced amyloid plaque pathology, it is largely unknown whether memantine might have beneficial effects on neurodegeneration. In the current study, we assessed whether memantine treatment has an impact on hippocampal neuron loss and associated behavioral deficits in the Tg4-42 mouse model of AD. We demonstrate that a chronic oral memantine treatment for 4 months diminishes hippocampal CA1 neuron loss and rescues learning and memory performance in different behavioral paradigms, such as Morris water maze or a novel object recognition task. Cognitive benefits of chronic memantine treatment were accompanied by an amelioration of impaired adult hippocampal neurogenesis. Taken together, our results demonstrate that memantine successfully counteracts pathological alterations in a preclinical mouse model of AD.
Collapse
Affiliation(s)
- Martina Stazi
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August University, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August University, Von-Siebold-Str. 5, 37075, Göttingen, Germany.
| |
Collapse
|
12
|
Region-specific glial homeostatic signature in prion diseases is replaced by a uniform neuroinflammation signature, common for brain regions and prion strains with different cell tropism. Neurobiol Dis 2020; 137:104783. [PMID: 32001329 DOI: 10.1016/j.nbd.2020.104783] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 02/08/2023] Open
Abstract
Chronic neuroinflammation is recognized as a major neuropathological hallmark in a broad spectrum of neurodegenerative diseases including Alzheimer's, Parkinson's, Frontal Temporal Dementia, Amyotrophic Lateral Sclerosis, and prion diseases. Both microglia and astrocytes exhibit region-specific homeostatic transcriptional identities, which under chronic neurodegeneration, transform into reactive phenotypes in a region- and disease-specific manner. Little is known about region-specific identity of glia in prion diseases. The current study was designed to determine whether the region-specific homeostatic signature of glia changes with the progression of prion diseases, and whether these changes occur in a region-dependent or universal manner. Also of interest was whether different prion strains give rise to different reactive phenotypes. To answer these questions, we analyzed gene expression in the thalamus, cortex, hypothalamus and hippocampus of mice infected with 22L and ME7 prion strains using a Nanostring Neuroinflammation panel at the subclinical, early clinical and advanced stages of the disease. We found that at the preclinical stage of the disease, the region-specific homeostatic identities were preserved. However, with the appearance of clinical signs, the region-specific signatures were partially lost and replaced with a neuroinflammation signature. While the same sets of genes were activated by both prion strains, the timing of neuroinflammation and the degree of activation in different brain regions was strain-specific. Changes in astrocyte function scored at the top of the activated pathways. Moreover, clustering analysis suggested that the astrocyte function pathway responded to prion infection prior to the Activated Microglia or Neuron and Neurotransmission pathways. The current work established neuroinflammation gene expression signature associated with prion diseases. Our results illustrate that with the disease progression, the region-specific homeostatic transcriptome signatures are replaced by the region-independent neuroinflammation signature, which is common for prion strains with different cell tropism. The prion-associated neuroinflammation signature identified in the current study overlapped only partially with the microglia degenerative phenotype and the disease-associated microglia phenotype reported for animal models of other neurodegenerative diseases.
Collapse
|
13
|
Essayan-Perez S, Zhou B, Nabet AM, Wernig M, Huang YWA. Modeling Alzheimer's disease with human iPS cells: advancements, lessons, and applications. Neurobiol Dis 2019; 130:104503. [PMID: 31202913 PMCID: PMC6689423 DOI: 10.1016/j.nbd.2019.104503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/24/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
One in three people will develop Alzheimer's disease (AD) or another dementia and, despite intense research efforts, treatment options remain inadequate. Understanding the mechanisms of AD pathogenesis remains our principal hurdle to developing effective therapeutics to tackle this looming medical crisis. In light of recent discoveries from whole-genome sequencing and technical advances in humanized models, studying disease risk genes with induced human neural cells presents unprecedented advantages. Here, we first review the current knowledge of the proposed mechanisms underlying AD and focus on modern genetic insights to inform future studies. To highlight the utility of human pluripotent stem cell-based innovations, we then present an update on efforts in recapitulating the pathophysiology by induced neuronal, non-neuronal and a collection of brain cell types, departing from the neuron-centric convention. Lastly, we examine the translational potentials of such approaches, and provide our perspectives on the promise they offer to deepen our understanding of AD pathogenesis and to accelerate the development of intervention strategies for patients and risk carriers.
Collapse
Affiliation(s)
- Sofia Essayan-Perez
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Bo Zhou
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America; Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Amber M Nabet
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Yu-Wen Alvin Huang
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America.
| |
Collapse
|
14
|
Seifirad S, Haghpanah V. Inappropriate modeling of chronic and complex disorders: How to reconsider the approach in the context of predictive, preventive and personalized medicine, and translational medicine. EPMA J 2019; 10:195-209. [PMID: 31462938 PMCID: PMC6695463 DOI: 10.1007/s13167-019-00176-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/02/2019] [Indexed: 12/14/2022]
Abstract
Preclinical investigations such as animal modeling make the basis of clinical investigations and subsequently patient care. Predictive, preventive, and personalized medicine (PPPM) not only highlights a patient-tailored approach by choosing the right medication, the right dose at the right time point but it as well essentially requires early identification, by the means of complex and state-of-the-art technologies of unmanifested pathological processes in an individual, in order to deliver targeted prevention early enough to reverse manifestation of a pathology. Such an approach can be achieved by taking into account clinical, pathological, environmental, and psychosocial characteristics of the patients or an individual who has a suboptimal health condition. Inappropriate modeling of chronic and complex disorders, in this context, may diminish the predictive potential and slow down the development of PPPM and consequently modern healthcare. Therefore, it is the common goal of PPPM and translational medicine to find the solution for the problem we present in our review. Both, translational medicine and PPPM in parallel, essentially need accurate surrogates for misleading animal models. This study was therefore undertaken to provide shreds of evidence against the validity of animal models. Limitations of current animal models and drug development strategies based on animal modeling have been systematically discussed. Finally, a variety of potential surrogates have been suggested to change the unfavorable situation in medical research and consequently in healthcare.
Collapse
Affiliation(s)
- Soroush Seifirad
- PERFUSE Study Group, Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Mango D, Saidi A, Cisale GY, Feligioni M, Corbo M, Nisticò R. Targeting Synaptic Plasticity in Experimental Models of Alzheimer's Disease. Front Pharmacol 2019; 10:778. [PMID: 31379566 PMCID: PMC6646937 DOI: 10.3389/fphar.2019.00778] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/17/2019] [Indexed: 12/28/2022] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) of hippocampal synaptic transmission represent the principal experimental models underlying learning and memory. Alterations of synaptic plasticity are observed in several neurodegenerative disorders, including Alzheimer’s disease (AD). Indeed, synaptic dysfunction is an early event in AD, making it an attractive therapeutic target for pharmaceutical intervention. To date, intensive investigations have characterized hippocampal synaptic transmission, LTP, and LTD in in vitro and in murine models of AD. In this review, we describe the synaptic alterations across the main AD models generated so far. We then examine the clinical perspective of LTP/LTD studies and discuss the limitations of non-clinical models and how to improve their predictive validity in the drug discovery process.
Collapse
Affiliation(s)
- Dalila Mango
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - Amira Saidi
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - Giusy Ylenia Cisale
- Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
| | - Marco Feligioni
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy.,Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Robert Nisticò
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy.,School of Pharmacy, Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
16
|
De Rossi P, Andrew RJ, Musial TF, Buggia‐Prevot V, Xu G, Ponnusamy M, Ly H, Krause SV, Rice RC, de l’Estoile V, Valin T, Salem S, Despa F, Borchelt DR, Bindokas VP, Nicholson DA, Thinakaran G. Aberrant accrual of BIN1 near Alzheimer's disease amyloid deposits in transgenic models. Brain Pathol 2018; 29:485-501. [PMID: 30506549 PMCID: PMC6542723 DOI: 10.1111/bpa.12687] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/20/2018] [Indexed: 01/11/2023] Open
Abstract
Bridging integrator 1 (BIN1) is the most significant late-onset Alzheimer's disease (AD) susceptibility locus identified via genome-wide association studies. BIN1 is an adaptor protein that regulates membrane dynamics in the context of endocytosis and membrane remodeling. An increase in BIN1 expression and changes in the relative levels of alternatively spliced BIN1 isoforms have been reported in the brains of patients with AD. BIN1 can bind to Tau, and an increase in BIN1 expression correlates with Tau pathology. In contrast, the loss of BIN1 expression in cultured cells elevates Aβ production and Tau propagation by insfluencing endocytosis and recycling. Here, we show that BIN1 accumulates adjacent to amyloid deposits in vivo. We found an increase in insoluble BIN1 and a striking accrual of BIN1 within and near amyloid deposits in the brains of multiple transgenic models of AD. The peri-deposit aberrant BIN1 localization was conspicuously different from the accumulation of APP and BACE1 within dystrophic neurites. Although BIN1 is highly expressed in mature oligodendrocytes, BIN1 association with amyloid deposits occurred in the absence of the accretion of other oligodendrocyte or myelin proteins. Finally, super-resolution microscopy and immunogold electron microscopy analyses highlight the presence of BIN1 in proximity to amyloid fibrils at the edges of amyloid deposits. These results reveal the aberrant accumulation of BIN1 is a feature associated with AD amyloid pathology. Our findings suggest a potential role for BIN1 in extracellular Aβ deposition in vivo that is distinct from its well-characterized function as an adaptor protein in endocytosis and membrane remodeling.
Collapse
Affiliation(s)
- Pierre De Rossi
- Department of NeurobiologyThe University of ChicagoChicagoIL
| | | | - Timothy F. Musial
- Department of Neurological SciencesRush University Medical CenterChicagoIL
| | | | - Guilian Xu
- Center for Translational Research in Neurodegenerative DiseaseUniversity of FloridaGainesvilleFL
| | | | - Han Ly
- Departments of Pharmacology and Nutritional Sciences, and Neurology, College of MedicineUniversity of KentuckyLexingtonKY
| | - Sofia V. Krause
- Department of NeurobiologyThe University of ChicagoChicagoIL
| | - Richard C. Rice
- Department of NeurobiologyThe University of ChicagoChicagoIL
| | | | - Tess Valin
- Department of NeurobiologyThe University of ChicagoChicagoIL
| | - Someya Salem
- Department of NeurobiologyThe University of ChicagoChicagoIL
| | - Florin Despa
- Departments of Pharmacology and Nutritional Sciences, and Neurology, College of MedicineUniversity of KentuckyLexingtonKY
| | - David R. Borchelt
- Center for Translational Research in Neurodegenerative DiseaseUniversity of FloridaGainesvilleFL
| | - Vytas P. Bindokas
- Integrated Light Microscopy FacilityThe University of ChicagoChicagoIL
| | | | - Gopal Thinakaran
- Department of NeurobiologyThe University of ChicagoChicagoIL,Departments of Neurology, and PathologyThe University of Chicago, The University of ChicagoChicagoIL
| |
Collapse
|
17
|
Dai SJ, Zhang JY, Bao YT, Zhou XJ, Lin LN, Fu YB, Zhang YJ, Li CY, Yang YX. Intracerebroventricular injection of Aβ1-42 combined with two-vessel occlusion accelerate Alzheimer’s disease development in rats. Pathol Res Pract 2018; 214:1583-1595. [DOI: 10.1016/j.prp.2018.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/02/2018] [Accepted: 07/22/2018] [Indexed: 10/28/2022]
|
18
|
Galloway CR, Ravipati K, Singh S, Lebois EP, Cohen RM, Levey AI, Manns JR. Hippocampal place cell dysfunction and the effects of muscarinic M 1 receptor agonism in a rat model of Alzheimer's disease. Hippocampus 2018; 28:568-585. [PMID: 29742799 DOI: 10.1002/hipo.22961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 04/02/2018] [Accepted: 05/06/2018] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that disproportionately impacts memory and the hippocampus. However, it is unclear how AD pathology influences the activity of surviving neurons in the hippocampus to contribute to the memory symptoms in AD. One well-understood connection between spatial memory and neuronal activity in healthy brains is the activity of place cells, neurons in the hippocampus that fire preferentially in a specific location of a given environment (the place field of the place cell). In the present study, place cells were recorded from the hippocampus in a recently-developed rat model of AD (Tg-F344 AD) at an age (12-20 months) at which the AD rats showed marked spatial memory deficits. Place cells in the CA2 and CA3 pyramidal regions of the hippocampus in AD rats showed sharply reduced spatial fidelity relative to wild-type (WT) rats. In contrast, spiking activity of place cells recorded in region CA1 in AD rats showed good spatial fidelity that was similar to CA1 place cells in WT rats. Oral administration of the M1 muscarinic acetylcholine receptor agonist VU0364572 impacted place cell firing rates in CA1 and CA2/3 hippocampal regions, but did not improve the spatial fidelity of CA2/3 hippocampal place cells in AD rats. The results indicated that, to the extent the spatial memory impairment in AD rats was attributable to hippocampal dysfunction, the memory impairment was more attributable to dysfunction in hippocampal regions CA2 and CA3 rather than CA1.
Collapse
Affiliation(s)
| | - Kaushik Ravipati
- Neuroscience and Behavioral Biology Program, Emory University, Atlanta, Georgia
| | - Suyashi Singh
- Neuroscience and Behavioral Biology Program, Emory University, Atlanta, Georgia
| | - Evan P Lebois
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Robert M Cohen
- Department of Psychiatry, Emory University, Atlanta, Georgia
| | - Allan I Levey
- Department of Neurology, Emory University, Atlanta, Georgia
| | - Joseph R Manns
- Department of Psychology, Emory University, Atlanta, Georgia
| |
Collapse
|
19
|
Watts JC, Prusiner SB. β-Amyloid Prions and the Pathobiology of Alzheimer's Disease. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a023507. [PMID: 28193770 DOI: 10.1101/cshperspect.a023507] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in humans and will pose a considerable challenge to healthcare systems in the coming years. Aggregation of the β-amyloid (Aβ) peptide within the brain is thought to be an initiating event in AD pathogenesis. Many recent studies in transgenic mice have provided evidence that Aβ aggregates become self-propagating during disease, leading to a cascade of protein aggregation in the brain, which may underlie the progressive nature of AD. The ability to self-propagate and the existence of distinct "strains" reveals that Aβ aggregates exhibit many properties indistinguishable from those of prions composed of PrPSc proteins. Here, we review the evidence that Aβ can become a prion during disease and discuss how Aβ prions may be important for understanding the pathobiology of AD.
Collapse
Affiliation(s)
- Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Biochemistry, University of Toronto, Toronto, Ontario M5T 2S8, Canada
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Departments of Neurology and of Biochemistry and Biophysics, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
20
|
Paspalas CD, Carlyle BC, Leslie S, Preuss TM, Crimins JL, Huttner AJ, van Dyck CH, Rosene DL, Nairn AC, Arnsten AFT. The aged rhesus macaque manifests Braak stage III/IV Alzheimer's-like pathology. Alzheimers Dement 2018; 14:680-691. [PMID: 29241829 PMCID: PMC6178089 DOI: 10.1016/j.jalz.2017.11.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/18/2017] [Accepted: 11/07/2017] [Indexed: 01/10/2023]
Abstract
INTRODUCTION An animal model of late-onset Alzheimer's disease is needed to research what causes degeneration in the absence of dominant genetic insults and why the association cortex is particularly vulnerable to degeneration. METHODS We studied the progression of tau and amyloid cortical pathology in the aging rhesus macaque using immunoelectron microscopy and biochemical assays. RESULTS Aging macaques exhibited the same qualitative pattern and sequence of tau and amyloid cortical pathology as humans, reaching Braak stage III/IV. Pathology began in the young-adult entorhinal cortex with protein kinase A-phosphorylation of tau, progressing to fibrillation with paired helical filaments and mature tangles in oldest animals. Tau pathology in the dorsolateral prefrontal cortex paralleled but lagged behind the entorhinal cortex, not afflicting the primary visual cortex. DISCUSSION The aging rhesus macaque provides the long-sought animal model for exploring the etiology of late-onset Alzheimer's disease and for testing preventive strategies.
Collapse
Affiliation(s)
| | - Becky C Carlyle
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Shannon Leslie
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Todd M Preuss
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Center, Emory University, Atlanta, GA, USA
| | - Johanna L Crimins
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Anita J Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Christopher H van Dyck
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Douglas L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
21
|
Ruiz-Riquelme A, Lau HHC, Stuart E, Goczi AN, Wang Z, Schmitt-Ulms G, Watts JC. Prion-like propagation of β-amyloid aggregates in the absence of APP overexpression. Acta Neuropathol Commun 2018; 6:26. [PMID: 29615128 PMCID: PMC5883524 DOI: 10.1186/s40478-018-0529-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/21/2018] [Indexed: 11/10/2022] Open
Abstract
The amyloid cascade hypothesis posits that the initiating event in Alzheimer's disease (AD) is the aggregation and deposition of the β-amyloid (Aβ) peptide, which is a proteolytic cleavage product of the amyloid precursor protein (APP). Mounting evidence suggests that the formation and spread of prion-like Aβ aggregates during AD may contribute to disease progression. Inoculation of transgenic mice that overexpress APP with pre-formed Aβ aggregates results in the prion-like induction of cerebral Aβ deposition. To determine whether Aβ deposition can also be induced when physiological APP levels are present in the brain, we inoculated AppNL-F mice, a knock-in model of AD that avoids potential artifacts associated with APP overexpression, with Aβ aggregates derived from the brains of AD patients or transgenic mice. In all cases, induced Aβ deposition was apparent in the corpus callosum, olfactory bulb, and meningeal blood vessels of inoculated mice at 130-150 days post-inoculation, whereas uninoculated and buffer-inoculated animals exhibited minimal or no Aβ deposits at these ages. Interestingly, despite being predominantly composed of protease-resistant Aβ42 aggregates, the induced parenchymal Aβ deposits were largely diffuse and were unreactive to an amyloid-binding dye. These results demonstrate that APP overexpression is not a prerequisite for the prion-like induction of cerebral Aβ deposition. Accordingly, spreading of Aβ deposition may contribute to disease progression in AD patients.
Collapse
|
22
|
Cheng F, Fransson LÅ, Mani K. Common traffic routes for imported spermine and endosomal glypican-1-derived heparan sulfate in fibroblasts. Exp Cell Res 2018; 364:133-142. [PMID: 29408503 DOI: 10.1016/j.yexcr.2018.01.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 11/26/2022]
Abstract
Import of the polyamine spermine from the extracellular environment depends on the presence of cell surface heparan sulfate proteoglycans, such as glypican-1. This proteoglycan is internalized by endocytosis, releases its heparan sulfate chains in endosomes by a nitric oxide-, copper- and amyloid precursor protein-dependent mechanism, then penetrates the membrane and is transported to the nucleus and then to autophagosomes. This process is spontaneous or induced by ascorbate depending on the growth-state of the cell. Here, we have explored possible connections between the heparan sulfate traffic route and spermine uptake and delivery in wild-type and Tg2576 mouse fibroblasts. Cells were examined by deconvolution immunofluorescence microscopy. The antibodies used were specific for spermine, glypican-1-derived heparan sulfate, Rab7, nucleolin and a marker for autophagosomes. Endogenous immunostainable spermine was primarily associated with autophagosomes. When spermine synthesis was inhibited, imported spermine appeared in Rab7-positive endosomes. When ascorbate was added, heparan sulfate and spermine were transported to the nucleus where they colocalized with nucleolin. Spermine also appeared in autophagosomes. In a pulse-chase experiment, heparan sulfate and spermine were first arrested in late endosomes by actinomycin D treatment. During the chase, when arrest was abolished, heparan sulfate and spermine were both transported to the nucleus and targeted nucleolin. In amyloid precursor protein-/--fibroblasts, ascorbate failed to induce release of heparan sulfate and spermine remained in the endosomes. We propose that cell surface glypican-1 carries spermine to the endosomes and that the released heparan sulfate carries spermine across the membrane into the cytosol and then to the nucleus.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Biomedical CenterA13, SE-221 84 Lund, Sweden
| | - Lars-Åke Fransson
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Biomedical CenterA13, SE-221 84 Lund, Sweden
| | - Katrin Mani
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Biomedical CenterA13, SE-221 84 Lund, Sweden.
| |
Collapse
|
23
|
Cytochrome b561, copper, β-cleaved amyloid precursor protein and niemann-pick C1 protein are involved in ascorbate-induced release and membrane penetration of heparan sulfate from endosomal S-nitrosylated glypican-1. Exp Cell Res 2017; 360:171-179. [DOI: 10.1016/j.yexcr.2017.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 11/21/2022]
|
24
|
Dugger BN, Perl DP, Carlson GA. Neurodegenerative Disease Transmission and Transgenesis in Mice. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a023549. [PMID: 28193724 DOI: 10.1101/cshperspect.a023549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Although the discovery of the prion protein (PrP) resulted from its co-purification with scrapie infectivity in Syrian hamsters, work with genetically defined and genetically modified mice proved crucial for understanding the fundamental processes involved not only in prion diseases caused by PrP misfolding, aggregation, and spread but also in other, much more common, neurodegenerative brain diseases. In this review, we focus on methodological and conceptual approaches used to study scrapie and related PrP misfolding diseases in mice and how these approaches have advanced our understanding of related disorders including Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Brittany N Dugger
- Institute for Neurodegenerative Diseases, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Daniel P Perl
- F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - George A Carlson
- Institute for Neurodegenerative Diseases, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158.,McLaughlin Research Institute of Biomedical Sciences, Great Falls, Montana 59405
| |
Collapse
|
25
|
Rorabaugh JM, Chalermpalanupap T, Botz-Zapp CA, Fu VM, Lembeck NA, Cohen RM, Weinshenker D. Chemogenetic locus coeruleus activation restores reversal learning in a rat model of Alzheimer's disease. Brain 2017; 140:3023-3038. [PMID: 29053824 PMCID: PMC5841201 DOI: 10.1093/brain/awx232] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 01/17/2023] Open
Abstract
See Grinberg and Heinsen (doi:10.1093/brain/awx261) for a scientific commentary on this article. Clinical evidence suggests that aberrant tau accumulation in the locus coeruleus and noradrenergic dysfunction may be a critical early step in Alzheimer’s disease progression. Yet, an accurate preclinical model of these phenotypes that includes early pretangle tau accrual in the locus coeruleus, loss of locus coeruleus innervation and deficits locus coeruleus/norepinephrine modulated behaviours, does not exist, hampering the identification of underlying mechanisms and the development of locus coeruleus-based therapies. Here, a transgenic rat (TgF344-AD) expressing disease-causing mutant amyloid precursor protein (APPsw) and presenilin-1 (PS1ΔE9) was characterized for histological and behavioural signs of locus coeruleus dysfunction reminiscent of mild cognitive impairment/early Alzheimer’s disease. In TgF344-AD rats, hyperphosphorylated tau was detected in the locus coeruleus prior to accrual in the medial entorhinal cortex or hippocampus, and tau pathology in the locus coeruleus was negatively correlated with noradrenergic innervation in the medial entorhinal cortex. Likewise, TgF344-AD rats displayed progressive loss of hippocampal norepinephrine levels and locus coeruleus fibres in the medial entorhinal cortex and dentate gyrus, with no frank noradrenergic cell body loss. Cultured mouse locus coeruleus neurons expressing hyperphosphorylation-prone mutant human tau had shorter neurites than control neurons, but similar cell viability, suggesting a causal link between pretangle tau accrual and altered locus coeruleus fibre morphology. TgF344-AD rats had impaired reversal learning in the Morris water maze compared to their wild-type littermates, which was rescued by chemogenetic locus coeruleus activation via designer receptors exclusively activated by designer drugs (DREADDs). Our results indicate that TgF344-AD rats uniquely meet several key criteria for a suitable model of locus coeruleus pathology and dysfunction early in Alzheimer’s disease progression, and suggest that a substantial window of opportunity for locus coeruleus/ norepinephrine-based therapeutics exists.
Collapse
Affiliation(s)
- Jacki M Rorabaugh
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA 30322, USA
| | | | - Christian A Botz-Zapp
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Vanessa M Fu
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Natalie A Lembeck
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Robert M Cohen
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta GA 30322, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA 30322, USA
| |
Collapse
|
26
|
Chalermpalanupap T, Weinshenker D, Rorabaugh JM. Down but Not Out: The Consequences of Pretangle Tau in the Locus Coeruleus. Neural Plast 2017; 2017:7829507. [PMID: 29038736 PMCID: PMC5605916 DOI: 10.1155/2017/7829507] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/20/2017] [Accepted: 07/20/2017] [Indexed: 12/12/2022] Open
Abstract
Degeneration of locus coeruleus (LC) is an underappreciated hallmark of Alzheimer's disease (AD). The LC is the main source of norepinephrine (NE) in the forebrain, and its degeneration is highly correlated with cognitive impairment and amyloid-beta (Aβ) and tangle pathology. Hyperphosphorylated tau in the LC is among the first detectable AD-like neuropathology in the brain, and while the LC/NE system impacts multiple aspects of AD (e.g., cognition, neuropathology, and neuroinflammation), the functional consequences of hyperphosphorylated tau accrual on LC neurons are not known. Recent evidence suggests that LC neurons accumulate aberrant tau species for decades before frank LC cell body degeneration occurs in AD, suggesting that a therapeutic window exists. In this review, we combine the literature on how pathogenic tau affects forebrain neurons with the known properties and degeneration patterns of LC neurons to synthesize hypotheses on hyperphosphorylated tau-induced dysfunction of LC neurons and the prion-like spread of pretangle tau from the LC to the forebrain. We also propose novel experiments using both in vitro and in vivo models to address the many questions surrounding the impact of hyperphosphorylated tau on LC neurons in AD and its role in disease progression.
Collapse
Affiliation(s)
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jacki M. Rorabaugh
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
27
|
Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ. EFAD transgenic mice as a human APOE relevant preclinical model of Alzheimer's disease. J Lipid Res 2017; 58:1733-1755. [PMID: 28389477 PMCID: PMC5580905 DOI: 10.1194/jlr.r076315] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/06/2017] [Indexed: 01/12/2023] Open
Abstract
Identified in 1993, APOE4 is the greatest genetic risk factor for sporadic Alzheimer's disease (AD), increasing risk up to 15-fold compared with APOE3, with APOE2 decreasing AD risk. However, the functional effects of APOE4 on AD pathology remain unclear and, in some cases, controversial. In vivo progress to understand how the human (h)-APOE genotypes affect AD pathology has been limited by the lack of a tractable familial AD-transgenic (FAD-Tg) mouse model expressing h-APOE rather than mouse (m)-APOE. The disparity between m- and h-apoE is relevant for virtually every AD-relevant pathway, including amyloid-β (Aβ) deposition and clearance, neuroinflammation, tau pathology, neural plasticity and cerebrovascular deficits. EFAD mice were designed as a temporally useful preclinical FAD-Tg-mouse model expressing the h-APOE genotypes for identifying mechanisms underlying APOE-modulated symptoms of AD pathology. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research. Here we review insights gleaned from the EFAD mice and summarize future directions.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Evangelina Avila-Munoz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Nicole Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612.
| |
Collapse
|
28
|
Arber C, Lovejoy C, Wray S. Stem cell models of Alzheimer's disease: progress and challenges. ALZHEIMERS RESEARCH & THERAPY 2017; 9:42. [PMID: 28610595 PMCID: PMC5470327 DOI: 10.1186/s13195-017-0268-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/22/2017] [Indexed: 02/08/2023]
Abstract
A major challenge to our understanding of the molecular mechanisms of Alzheimer’s disease (AD) has been the lack of physiologically relevant in vitro models which capture the precise patient genome, in the cell type of interest, with physiological expression levels of the gene(s) of interest. Induced pluripotent stem cell (iPSC) technology, together with advances in 2D and 3D neuronal differentiation, offers a unique opportunity to overcome this challenge and generate a limitless supply of human neurons for in vitro studies. iPSC-neuron models have been widely employed to model AD and we discuss in this review the progress that has been made to date using patient-derived neurons to recapitulate key aspects of AD pathology and how these models have contributed to a deeper understanding of AD molecular mechanisms, as well as addressing the key challenges posed by using this technology and what progress is being made to overcome these. Finally, we highlight future directions for the use of iPSC-neurons in AD research and highlight the potential value of this technology to neurodegenerative research in the coming years.
Collapse
Affiliation(s)
- Charles Arber
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Christopher Lovejoy
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Selina Wray
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.
| |
Collapse
|
29
|
Hallmarks of Alzheimer's Disease in Stem-Cell-Derived Human Neurons Transplanted into Mouse Brain. Neuron 2017; 93:1066-1081.e8. [PMID: 28238547 DOI: 10.1016/j.neuron.2017.02.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 12/12/2016] [Accepted: 01/31/2017] [Indexed: 01/11/2023]
Abstract
Human pluripotent stem cells (PSCs) provide a unique entry to study species-specific aspects of human disorders such as Alzheimer's disease (AD). However, in vitro culture of neurons deprives them of their natural environment. Here we transplanted human PSC-derived cortical neuronal precursors into the brain of a murine AD model. Human neurons differentiate and integrate into the brain, express 3R/4R Tau splice forms, show abnormal phosphorylation and conformational Tau changes, and undergo neurodegeneration. Remarkably, cell death was dissociated from tangle formation in this natural 3D model of AD. Using genome-wide expression analysis, we observed upregulation of genes involved in myelination and downregulation of genes related to memory and cognition, synaptic transmission, and neuron projection. This novel chimeric model for AD displays human-specific pathological features and allows the analysis of different genetic backgrounds and mutations during the course of the disease.
Collapse
|
30
|
Astrocytic transporters in Alzheimer's disease. Biochem J 2017; 474:333-355. [DOI: 10.1042/bcj20160505] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/16/2016] [Accepted: 11/29/2016] [Indexed: 12/26/2022]
Abstract
Astrocytes play a fundamental role in maintaining the health and function of the central nervous system. Increasing evidence indicates that astrocytes undergo both cellular and molecular changes at an early stage in neurological diseases, including Alzheimer's disease (AD). These changes may reflect a change from a neuroprotective to a neurotoxic phenotype. Given the lack of current disease-modifying therapies for AD, astrocytes have become an interesting and viable target for therapeutic intervention. The astrocyte transport system covers a diverse array of proteins involved in metabolic support, neurotransmission and synaptic architecture. Therefore, specific targeting of individual transporter families has the potential to suppress neurodegeneration, a characteristic hallmark of AD. A small number of the 400 transporter superfamilies are expressed in astrocytes, with evidence highlighting a fraction of these are implicated in AD. Here, we review the current evidence for six astrocytic transporter subfamilies involved in AD, as reported in both animal and human studies. This review confirms that astrocytes are indeed a viable target, highlights the complexities of studying astrocytes and provides future directives to exploit the potential of astrocytes in tackling AD.
Collapse
|
31
|
Kim D, Baik SH, Kang S, Cho SW, Bae J, Cha MY, Sailor MJ, Mook-Jung I, Ahn KH. Close Correlation of Monoamine Oxidase Activity with Progress of Alzheimer's Disease in Mice, Observed by in Vivo Two-Photon Imaging. ACS CENTRAL SCIENCE 2016; 2:967-975. [PMID: 28058286 PMCID: PMC5200925 DOI: 10.1021/acscentsci.6b00309] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Indexed: 05/26/2023]
Abstract
Monoamine oxidases (MAOs) play an important role in Alzheimer's disease (AD) pathology. We report in vivo comonitoring of MAO activity and amyloid-β (Aβ) plaques dependent on the aging of live mice with AD, using a two-photon fluorescence probe. The probe under the catalytic action of MAO produces a dipolar fluorophore that senses Aβ plaques, a general AD biomarker, enabling us to comonitor the enzyme activity and the progress of AD indicated by Aβ plaques. The results show that the progress of AD has a close correlation with MAO activity, which can be categorized into three stages: slow initiation stage up to three months, an aggressive stage, and a saturation stage from nine months. Histological analysis also reveals elevation of MAO activity around Aβ plaques in aged mice. The close correlation between the MAO activity and AD progress observed by in vivo monitoring for the first time prompts us to investigate the enzyme as a potential biomarker of AD.
Collapse
Affiliation(s)
- Dokyoung Kim
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Sung Hoon Baik
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Seokjo Kang
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Seo Won Cho
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
| | - Juryang Bae
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
| | - Moon-Yong Cha
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Michael J. Sailor
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Inhee Mook-Jung
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Kyo Han Ahn
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
| |
Collapse
|
32
|
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
|
33
|
Rosen RF, Tomidokoro Y, Farberg AS, Dooyema J, Ciliax B, Preuss TM, Neubert TA, Ghiso JA, LeVine H, Walker LC. Comparative pathobiology of β-amyloid and the unique susceptibility of humans to Alzheimer's disease. Neurobiol Aging 2016; 44:185-196. [PMID: 27318146 DOI: 10.1016/j.neurobiolaging.2016.04.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 10/21/2022]
Abstract
The misfolding and accumulation of the protein fragment β-amyloid (Aβ) is an early and essential event in the pathogenesis of Alzheimer's disease (AD). Despite close biological similarities among primates, humans appear to be uniquely susceptible to the profound neurodegeneration and dementia that characterize AD, even though nonhuman primates deposit copious Aβ in senile plaques and cerebral amyloid-β angiopathy as they grow old. Because the amino acid sequence of Aβ is identical in all primates studied to date, we asked whether differences in the properties of aggregated Aβ might underlie the vulnerability of humans and the resistance of other primates to AD. In a comparison of aged squirrel monkeys (Saimiri sciureus) and humans with AD, immunochemical and mass spectrometric analyses indicate that the populations of Aβ fragments are largely similar in the 2 species. In addition, Aβ-rich brain extracts from the brains of aged squirrel monkeys and AD patients similarly seed the deposition of Aβ in a transgenic mouse model. However, the epitope exposure of aggregated Aβ differs in sodium dodecyl sulfate-stable oligomeric Aβ from the 2 species. In addition, the high-affinity binding of (3)H Pittsburgh Compound B to Aβ is significantly diminished in tissue extracts from squirrel monkeys compared with AD patients. These findings support the hypothesis that differences in the pathobiology of aggregated Aβ among primates are linked to post-translational attributes of the misfolded protein, such as molecular conformation and/or the involvement of species-specific cofactors.
Collapse
Affiliation(s)
- Rebecca F Rosen
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
| | | | - Aaron S Farberg
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Jeromy Dooyema
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Brian Ciliax
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd M Preuss
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas A Neubert
- Department of Biochemistry and Molecular Pharmacology, Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, USA
| | - Jorge A Ghiso
- Department of Pathology, NYU School of Medicine, New York, NY, USA; Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| | - Harry LeVine
- Department of Molecular & Cellular Biochemistry, Center on Aging, Center for Structural Biology, University of Kentucky, Lexington, KY, USA
| | - Lary C Walker
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
34
|
Luo J, Lee SH, VandeVrede L, Qin Z, Ben Aissa M, Larson J, Teich AF, Arancio O, D'Souza Y, Elharram A, Koster K, Tai LM, LaDu MJ, Bennett BM, Thatcher GRJ. A multifunctional therapeutic approach to disease modification in multiple familial mouse models and a novel sporadic model of Alzheimer's disease. Mol Neurodegener 2016; 11:35. [PMID: 27129593 PMCID: PMC4850651 DOI: 10.1186/s13024-016-0103-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 04/22/2016] [Indexed: 12/20/2022] Open
Abstract
Background Clinical failures singularly targeting amyloid-β pathology indicate a critical need for alternative Alzheimer’s disease (AD) therapeutic strategies. The mixed pathology reported in a large population of AD patients demands a multifunctional drug approach. Since activation of cAMP response element binding protein (CREB) plays a crucial role in synaptic strengthening and memory formation, we retooled a clinical drug with known neuroprotective and anti-inflammatory activity to activate CREB, and validated this novel multifunctional drug, NMZ, in 4 different mouse models of AD. Results NMZ was tested in three mouse models of familial AD and one model of sporadic AD. In 3 × Tg hippocampal slices, NMZ restored LTP. In vivo, memory was improved with NMZ in all animal models with robust cognitive deficits. NMZ treatment lowered neurotoxic forms of Aβ in both APP/PS1 and 3 × Tg transgenic mice while also restoring neuronal plasticity biomarkers in the 3 × Tg mice. In EFAD mice, incorporation of the major genetic AD risk factor, hAPOE4, did not mute the beneficial drug effects. In a novel sporadic mouse model that manifests AD-like pathology caused by accelerated oxidative stress in the absence of any familial AD mutation, oral administration of NMZ attenuated hallmark AD pathology and restored biomarkers of synaptic and neuronal function. Conclusions The multifunctional approach, embodied by NMZ, was successful in mouse models of AD incorporating Aβ pathology (APP/PS1), tau pathology (3xTg), and APOE4, the major human genetic risk factor for AD (EFAD). The efficacy observed in a novel model of sporadic AD (Aldh2−/−) demonstrates that the therapeutic approach is not limited to rare, familial AD genetic mutations. The multifunctional drug, NMZ, was not designed directly to target Aβ and tau pathology; however, the attenuation of this hallmark pathology suggests the approach to be a highly promising, disease-modifying strategy for AD and mixed pathology dementia.
Collapse
Affiliation(s)
- Jia Luo
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Sue H Lee
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Lawren VandeVrede
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Zhihui Qin
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Manel Ben Aissa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA.,Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - John Larson
- Department of Psychiatry, Neuropsychiatric Institute, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew F Teich
- Department of Pathology, The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ottavio Arancio
- Department of Pathology, The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Yohan D'Souza
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Ahmed Elharram
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Kevin Koster
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Brian M Bennett
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
35
|
Müller-Schiffmann A, Herring A, Abdel-Hafiz L, Chepkova AN, Schäble S, Wedel D, Horn AHC, Sticht H, de Souza Silva MA, Gottmann K, Sergeeva OA, Huston JP, Keyvani K, Korth C. Amyloid-β dimers in the absence of plaque pathology impair learning and synaptic plasticity. Brain 2015; 139:509-25. [DOI: 10.1093/brain/awv355] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/17/2015] [Indexed: 11/12/2022] Open
Abstract
Abstract
Despite amyloid plaques, consisting of insoluble, aggregated amyloid-β peptides, being a defining feature of Alzheimer’s disease, their significance has been challenged due to controversial findings regarding the correlation of cognitive impairment in Alzheimer’s disease with plaque load. The amyloid cascade hypothesis defines soluble amyloid-β oligomers, consisting of multiple amyloid-β monomers, as precursors of insoluble amyloid-β plaques. Dissecting the biological effects of single amyloid-β oligomers, for example of amyloid-β dimers, an abundant amyloid-β oligomer associated with clinical progression of Alzheimer’s disease, has been difficult due to the inability to control the kinetics of amyloid-β multimerization. For investigating the biological effects of amyloid-β dimers, we stabilized amyloid-β dimers by an intermolecular disulphide bridge via a cysteine mutation in the amyloid-β peptide (Aβ-S8C) of the amyloid precursor protein. This construct was expressed as a recombinant protein in cells and in a novel transgenic mouse, termed tgDimer mouse. This mouse formed constant levels of highly synaptotoxic soluble amyloid-β dimers, but not monomers, amyloid-β plaques or insoluble amyloid-β during its lifespan. Accordingly, neither signs of neuroinflammation, tau hyperphosphorylation or cell death were observed. Nevertheless, these tgDimer mice did exhibit deficits in hippocampal long-term potentiation and age-related impairments in learning and memory, similar to what was observed in classical Alzheimer’s disease mouse models. Although the amyloid-β dimers were unable to initiate the formation of insoluble amyloid-β aggregates in tgDimer mice, after crossbreeding tgDimer mice with the CRND8 mouse, an amyloid-β plaque generating mouse model, Aβ-S8C dimers were sequestered into amyloid-β plaques, suggesting that amyloid-β plaques incorporate neurotoxic amyloid-β dimers that by themselves are unable to self-assemble. Our results suggest that within the fine interplay between different amyloid-β species, amyloid-β dimer neurotoxic signalling, in the absence of amyloid-β plaque pathology, may be involved in causing early deficits in synaptic plasticity, learning and memory that accompany Alzheimer’s disease.
10.1093/brain/awv355_video_abstract awv355_video_abstract
Collapse
Affiliation(s)
| | - Arne Herring
- 2 Institute of Neuropathology, University of Duisburg-Essen, Germany
| | - Laila Abdel-Hafiz
- 3 Centre for Behavioural Neuroscience, Heinrich Heine University, Düsseldorf, Germany
| | - Aisa N. Chepkova
- 4 Institute for Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Sandra Schäble
- 3 Centre for Behavioural Neuroscience, Heinrich Heine University, Düsseldorf, Germany
- *Present address: Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University, Düsseldorf, Germany
| | - Diana Wedel
- 1 Department Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Anselm H. C. Horn
- 5 Institute for Biochemistry, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- 5 Institute for Biochemistry, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Kurt Gottmann
- 4 Institute for Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Olga A. Sergeeva
- 4 Institute for Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Joseph P. Huston
- 3 Centre for Behavioural Neuroscience, Heinrich Heine University, Düsseldorf, Germany
| | - Kathy Keyvani
- 2 Institute of Neuropathology, University of Duisburg-Essen, Germany
| | - Carsten Korth
- 1 Department Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
36
|
Cavaleri F. Review of Amyotrophic Lateral Sclerosis, Parkinson’s and Alzheimer’s diseases helps further define pathology of the novel paradigm for Alzheimer’s with heavy metals as primary disease cause. Med Hypotheses 2015; 85:779-90. [DOI: 10.1016/j.mehy.2015.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/25/2015] [Accepted: 10/11/2015] [Indexed: 01/07/2023]
|
37
|
Cañete T, Blázquez G, Tobeña A, Giménez-Llort L, Fernández-Teruel A. Cognitive and emotional alterations in young Alzheimer's disease (3xTgAD) mice: effects of neonatal handling stimulation and sexual dimorphism. Behav Brain Res 2014; 281:156-71. [PMID: 25446741 DOI: 10.1016/j.bbr.2014.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/30/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
Abstract
Alzheimer disease is the most common neurodegenerative disorder and cause of senile dementia. It is characterized by an accelerated memory loss, and alterations of mood, reason, judgment and language. The main neuropathological hallmarks of the disorder are β-amyloid (βA) plaques and neurofibrillary Tau tangles. The triple transgenic 3xTgAD mouse model develops βA and Tau pathologies in a progressive manner which mimicks the pattern that takes place in the human brain with AD, and showing cognitive alterations characteristic of the disease. The present study intended to examine whether 3xTgAD mice of both sexes present cognitive, emotional and other behavioral alterations at the early age of 4 months, an age in which only some intraneuronal amyloid accumulation is found. Neonatal handling (H) is an early-life treatment known to produce profound and long-lasting behavioral and neurobiological effects in rodents, as well as improvements in cognitive functions. Therefore, we also aimed at evaluating the effects of H on the behavioral/cognitive profile of 4-month-old male and female 3xTgAD mice. The results indicate that, (1) 3xTgAD mice present spatial learning/memory deficits and emotional alterations already at the early age of 4 months, (2) there exists sexual dimorphism effects on several behavioral variables at this age, (3) neonatal handling exerts a preventive effect on some cognitive (spatial learning) and emotional alterations appearing in 3xTgAD mice already at early ages, and 4) H treatment appears to produce stronger positive effects in females than in males in several spatial learning measures and in the open field test.
Collapse
Affiliation(s)
- T Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| | - G Blázquez
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - A Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - L Giménez-Llort
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - A Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
38
|
Liu L, Luo S, Zeng L, Wang W, Yuan L, Jian X. Degenerative alterations in noradrenergic neurons of the locus coeruleus in Alzheimer's disease. Neural Regen Res 2014; 8:2249-55. [PMID: 25206534 PMCID: PMC4146034 DOI: 10.3969/j.issn.1673-5374.2013.24.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 06/12/2013] [Indexed: 11/18/2022] Open
Abstract
Mice carrying mutant amyloid-β precursor protein and presenilin-1 genes (APP/PS1 double transgenic mice) have frequently been used in studies of Alzheimer's disease; however, such studies have focused mainly on hippocampal and cortical changes. The severity of Alzheimer's disease is known to correlate with the amount of amyloid-β protein deposition and the number of dead neurons in the locus coeruleus. In the present study, we assigned APP/PS1 double transgenic mice to two groups according to age: young mice (5–6 months old) and aged mice (16–17 months old). Age-matched wild-type mice were used as controls. Immunohistochemistry for tyrosine hydroxylase (a marker of catecholaminergic neurons in the locus coeruleus) revealed that APP/PS1 mice had 23% fewer cells in the locus coeruleus compared with aged wild-type mice. APP/PS1 mice also had increased numbers of cell bodies of neurons positive for tyrosine hydroxylase, but fewer tyrosine hydroxylase-positive fibers, which were also short, thick and broken. Quantitative analysis using unbiased stereology showed a significant age-related increase in the mean volume of tyrosine droxylase-positive neurons in aged APP/PS1 mice compared with young APP/PS1 mice. Moreover, the mean volume of tyrosine hydroxylase-positive neurons was positively correlated with the total volume of the locus coeruleus. These findings indicate that noradrenergic neurons and fibers in the locus coeruleus are predisposed to degenerative alterations in APP/PS1 double transgenic mice.
Collapse
Affiliation(s)
- Lihua Liu
- Department of Histology & Embryology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China ; Department of Nursing, Medical College of Hunan Normal University, Changsha 10013, Hunan Province, China
| | - Saiping Luo
- Third Department of Surgery, Agricultural Division Four Hospital, Xinjiang Production and Construction Corps, Yili 835000, Xinjiang Uygur Autonomous Region, China
| | - Leping Zeng
- Department of Anatomy & Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Weihong Wang
- Department of Nursing, Medical College of Hunan Normal University, Changsha 10013, Hunan Province, China
| | - Liming Yuan
- Department of Anatomy, Medical College of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Xiaohong Jian
- Department of Anatomy, Medical College of Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|
39
|
Joffre C, Nadjar A, Lebbadi M, Calon F, Laye S. n-3 LCPUFA improves cognition: the young, the old and the sick. Prostaglandins Leukot Essent Fatty Acids 2014; 91:1-20. [PMID: 24908517 DOI: 10.1016/j.plefa.2014.05.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 01/01/2023]
Abstract
Due to the implication of docosahexaenoic acid (DHA) in neurogenesis, synaptogenesis, neurite outgrowth and to its high incorporation into the brain, this n-3 long chain polyunsaturated fatty acid (LCPUFA) is considered as crucial in the development and maintenance of the learning memory performance throughout life. In the present chapter we aimed at reviewing data investigating the relation between DHA and cognition during the perinatal period, young adult- and adulthood and neurodegenerative diseases such as Alzheimer disease (AD). In Humans, dietary DHA supplementation from the perinatal period to adulthood does not reveal a clear and consistent memory improvement whereas it is the case in animal studies. The positive effects observed in animal models may have been enhanced by using n-3 PUFA deficient animal models as controls. In animal models of AD, a general consensus on the beneficial effects of n-3 LCPUFA in attenuating cognitive impairment was established. These studies make DHA a potential suitable micronutrient for the maintenance of cognitive performance at all periods of life.
Collapse
Affiliation(s)
- C Joffre
- Université Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France.
| | - A Nadjar
- Université Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France.
| | - M Lebbadi
- Centre de Recherche du CHUL, Axe Neurosciences, T2-05, 2705, Boulevard Laurier, Québec, QC, Canada G1V 4G2.
| | - F Calon
- Centre de Recherche du CHUL, Axe Neurosciences, T2-05, 2705, Boulevard Laurier, Québec, QC, Canada G1V 4G2.
| | - S Laye
- Université Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, F-33000 Bordeaux, France.
| |
Collapse
|
40
|
Watts JC, Prusiner SB. Mouse models for studying the formation and propagation of prions. J Biol Chem 2014; 289:19841-9. [PMID: 24860095 DOI: 10.1074/jbc.r114.550707] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prions are self-propagating protein conformers that cause a variety of neurodegenerative disorders in humans and animals. Mouse models have played key roles in deciphering the biology of prions and in assessing candidate therapeutics. The development of transgenic mice that form prions spontaneously in the brain has advanced our understanding of sporadic and genetic prion diseases. Furthermore, the realization that many proteins can become prions has necessitated the development of mouse models for assessing the potential transmissibility of common neurodegenerative diseases. As the universe of prion diseases continues to expand, mouse models will remain crucial for interrogating these devastating illnesses.
Collapse
Affiliation(s)
- Joel C Watts
- From the Institute for Neurodegenerative Diseases and the Department of Neurology, University of California, San Francisco, California 94143
| | - Stanley B Prusiner
- From the Institute for Neurodegenerative Diseases and the Department of Neurology, University of California, San Francisco, California 94143
| |
Collapse
|
41
|
Blázquez G, Cañete T, Tobeña A, Giménez-Llort L, Fernández-Teruel A. Cognitive and emotional profiles of aged Alzheimer's disease (3×TgAD) mice: effects of environmental enrichment and sexual dimorphism. Behav Brain Res 2014; 268:185-201. [PMID: 24746486 DOI: 10.1016/j.bbr.2014.04.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/05/2014] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease is a neurodegenerative disorder associated with age which represents the most common cause of dementia. It is characterized by an accelerated memory loss compared to normal aging, and deterioration of other cognitive abilities that interfere with mood, reason, judgment and language. The main neuropathological hallmarks of the disorder are β-amyloid (βA) plaques and neurofibrillary Tau tangles. Triple transgenic 3×TgAD mouse model develops βA and Tau pathologies in a progressive manner, with a specific temporal and anatomic profile mimicking the pattern that takes place in the human brain with AD, and showing cognitive alterations characteristic of the disease. Environmental enrichment treatment in mice induces behavioral and emotional reactivity changes, including cognitive improvements in some AD-related transgenic mice. The present work intended to characterize the behavioral profile of 3×TgAD mice at advanced stages of neuropathological development (12 and 15 months of age) and to investigate whether environmental enrichment administered during adulthood was able to modify some of their behavioral and cognitive alterations. Results show that, at advanced stages of the disease 3×TgAD mice show deficits of spatial learning acquisition, as well as short-term and working memory deficits, while displaying increased levels of anxiety/fearfulness and normal sensorimotor functions. 3×TgAD mice also show sexual dimorphism, as reflected by increased cognitive deficits in females and increased levels of novelty-induced behavioral inhibition in males. Environmental enrichment exerts some slight positive effects, by mainly improving the initial acquisition of the spatial learning and working memory in 12-month-old 3×TgAD mice. Such effects vary depending on the gender.
Collapse
Affiliation(s)
- Gloria Blázquez
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, Autonomous University of Barcelona, 08193 Bellaterra, Spain.
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, Autonomous University of Barcelona, 08193 Bellaterra, Spain.
| | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, Autonomous University of Barcelona, 08193 Bellaterra, Spain.
| | - Lydia Giménez-Llort
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, Autonomous University of Barcelona, 08193 Bellaterra, Spain.
| | - Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, Autonomous University of Barcelona, 08193 Bellaterra, Spain.
| |
Collapse
|
42
|
Kerrigan TL, Brown JT, Randall AD. Characterization of altered intrinsic excitability in hippocampal CA1 pyramidal cells of the Aβ-overproducing PDAPP mouse. Neuropharmacology 2014; 79:515-24. [PMID: 24055500 PMCID: PMC3989024 DOI: 10.1016/j.neuropharm.2013.09.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/21/2013] [Accepted: 09/04/2013] [Indexed: 11/27/2022]
Abstract
Transgenic mice that accumulate Aβ peptides in the CNS are commonly used to interrogate functional consequences of Alzheimer's disease-associated amyloidopathy. In addition to changes to synaptic function, there is also growing evidence that changes to intrinsic excitability of neurones can arise in these models of amyloidopathy. Furthermore, some of these alterations to intrinsic properties may occur relatively early within the age-related progression of experimental amyloidopathy. Here we report a detailed comparison between the intrinsic excitability properties of hippocampal CA1 pyramidal neurones in wild-type (WT) and PDAPP mice. The latter is a well-established model of Aβ accumulation which expresses human APP harbouring the Indiana (V717F) mutation. At the age employed in this study (9-10 months) CNS Abeta was elevated in PDAPP mice but significant plaque pathology was absent. PDAPP mice exhibited no differences in subthreshold intrinsic properties including resting potential, input resistance, membrane time constant and sag. When CA1 cells of PDAPP mice were given depolarizing stimuli of various amplitudes they initially fired at a higher frequency than WT cells. Commensurate with this, PDAPP cells exhibited a larger fast afterdepolarizing potential. PDAPP mice had narrower spikes but action potential threshold, rate of rise and peak were not different. Thus not all changes seen in our previous studies of amyloidopathy models were present in PDAPP mice; however, narrower spikes, larger ADPs and the propensity to fire at higher frequencies were consistent with our prior work and thus may represent robust, cross-model, indices of amyloidopathy. This article is part of a Special Issue entitled 'Neurodevelopment Disorder'.
Collapse
Affiliation(s)
- T L Kerrigan
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - J T Brown
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, UK; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, The Hatherly Building, Exeter EX4 4PS, UK
| | - A D Randall
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol BS8 1TD, UK; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, The Hatherly Building, Exeter EX4 4PS, UK.
| |
Collapse
|
43
|
Aβ-induced Golgi fragmentation in Alzheimer's disease enhances Aβ production. Proc Natl Acad Sci U S A 2014; 111:E1230-9. [PMID: 24639524 DOI: 10.1073/pnas.1320192111] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Golgi fragmentation occurs in neurons of patients with Alzheimer's disease (AD), but the underlying molecular mechanism causing the defects and the subsequent effects on disease development remain unknown. In this study, we examined the Golgi structure in APPswe/PS1E9 transgenic mouse and tissue culture models. Our results show that accumulation of amyloid beta peptides (Aβ) leads to Golgi fragmentation. Further biochemistry and cell biology studies revealed that Golgi fragmentation in AD is caused by phosphorylation of Golgi structural proteins, such as GRASP65, which is induced by Aβ-triggered cyclin-dependent kinase-5 activation. Significantly, both inhibition of cyclin-dependent kinase-5 and expression of nonphosphorylatable GRASP65 mutants rescued the Golgi structure and reduced Aβ secretion by elevating α-cleavage of the amyloid precursor protein. Our study demonstrates a molecular mechanism for Golgi fragmentation and its effects on amyloid precursor protein trafficking and processing in AD, suggesting Golgi as a potential drug target for AD treatment.
Collapse
|
44
|
Mate I, Cruces J, Vida C, Sanfeliu C, Manassra R, Giménez-Llort L, De la Fuente M. [Premature immunosenescence in triple-transgenic mice for Alzheimer's disease]. Rev Esp Geriatr Gerontol 2013; 49:15-9. [PMID: 24315805 DOI: 10.1016/j.regg.2013.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/23/2013] [Accepted: 04/26/2013] [Indexed: 11/30/2022]
Abstract
INTRODUCTION A deterioration of the neuroimmunoendocrine network has been observed in Alzheimer's disease (AD). However, the peripheral immune response has hardly been investigated in this pathology. Since some immune function parameters have been established as good markers of the rate of ageing, and can predict longevity, the aim of the present work was to study some of these functions in splenic leucocytes in transgenic mice for AD of different ages. MATERIAL AND METHODS Young female (4 ± 1 months), adult (9 ± 1 months), and mature (12 ± 1 months) triple-transgenic mice for AD (3 xTgAD) and non-transgenic (NTg) control mice of the same ages were used. The chemotaxis, the anti-tumour activity of « natural killer » (NK) cells and the lymphoproliferative response in the presence of the mitogens concanavalin A and lipopolysaccharide, functions that decrease with age, were determined in splenic leucocytes. In addition, the differences in lifespan between 3 xTgAD and NTg were studied in parallel using other animals, until their death through natural causes. RESULTS In 3 xTgAD, with respect to NTg, chemotaxis decreased at all ages studied, whereas in lymphoproliferative response this reduction was shown at 4 months and 9 months. NK activity was diminished only in young 3 xTgAD with respect to NTg. The 3 xTgAD showed a shorter lifespan than the NTg control group. CONCLUSIONS The 3 xTgAD mice show a premature immunosenescence, which could explain their early mortality. The determination of these immune functions at peripheral level could serve as a marker of the progression of the Alzheimer's disease.
Collapse
Affiliation(s)
- Ianire Mate
- Departamento de Fisiología, Facultad de Biología, Universidad Complutense de Madrid, Madrid, España
| | - Julia Cruces
- Departamento de Fisiología, Facultad de Biología, Universidad Complutense de Madrid, Madrid, España
| | - Carmen Vida
- Departamento de Fisiología, Facultad de Biología, Universidad Complutense de Madrid, Madrid, España
| | - Coral Sanfeliu
- Instituto de Investigación Biomédica de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, España
| | - Rashed Manassra
- Departamento de Fisiología, Facultad de Biología, Universidad Complutense de Madrid, Madrid, España
| | - Lydia Giménez-Llort
- Instituto de Neurociencia, Universidad Autónoma de Barcelona, Bellaterra, Barcelona, España
| | - Mónica De la Fuente
- Departamento de Fisiología, Facultad de Biología, Universidad Complutense de Madrid, Madrid, España.
| |
Collapse
|
45
|
Glennon EBC, Whitehouse IJ, Miners JS, Kehoe PG, Love S, Kellett KAB, Hooper NM. BIN1 is decreased in sporadic but not familial Alzheimer's disease or in aging. PLoS One 2013; 8:e78806. [PMID: 24205320 PMCID: PMC3804620 DOI: 10.1371/journal.pone.0078806] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/22/2013] [Indexed: 01/31/2023] Open
Abstract
Bridging integrator 1 (BIN1) has been implicated in sporadic Alzheimer's disease (AD) by a number of genome wide association studies (GWAS) in a variety of populations. Here we measured BIN1 in frontal cortex samples from 24 sporadic AD and 24 age-matched non-dementia brains and correlated the expression of this protein with markers of AD. BIN1 was reduced by 87% (p=0.007) in sporadic AD compared to non-dementia controls, but BIN1 in sporadic AD did not correlate with soluble Aβ (r(s)=-0.084, p=0.698), insoluble Aβ (r(s)=0.237, p=0.269), Aβ plaque load (r(s)=0.063, p=0.771) or phospho-tau load (r(s)=-0.160, p=0.489). In contrast to our findings in sporadic AD, BIN1 was unchanged in the hippocampus from 6 cases of familial AD compared to 6 age-matched controls (p=0.488). BIN1 declined with age in a cohort of non-dementia control cases between 25 and 88 years but the correlation was not significant (rs=-0.449, p=0.081). Although BIN1 is known to have a role in endocytosis, and the processing of the amyloid precursor protein (APP) to form amyloid-β (Aβ) peptides is dependent on endocytosis, knockdown of BIN1 by targeted siRNA or the overexpression of BIN1 in a human neuroblastoma cell line (SH-SY5Y) had no effect on APP processing. These data suggest that the alteration in BIN1 is involved in the pathogenesis of sporadic, but not familial AD and is not a consequence of AD neurodegeneration or the ageing process, a finding in keeping with the numerous GWAS that implicate BIN1 in sporadic AD. However, the mechanism of its contribution remains to be established.
Collapse
Affiliation(s)
- Elizabeth B. C. Glennon
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Isobel J. Whitehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - J. Scott Miners
- Dementia Research Group, School of Clinical Sciences, Institute of Clinical Neurosciences, University of Bristol, Bristol, United Kingdom
| | - Patrick G. Kehoe
- Dementia Research Group, School of Clinical Sciences, Institute of Clinical Neurosciences, University of Bristol, Bristol, United Kingdom
| | - Seth Love
- Dementia Research Group, School of Clinical Sciences, Institute of Clinical Neurosciences, University of Bristol, Bristol, United Kingdom
| | - Katherine A. B. Kellett
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nigel M. Hooper
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
46
|
Camboni M, Wang CM, Miranda C, Yoon JH, Xu R, Zygmunt D, Kaspar BK, Martin PT. Active and passive immunization strategies based on the SDPM1 peptide demonstrate pre-clinical efficacy in the APPswePSEN1dE9 mouse model for Alzheimer's disease. Neurobiol Dis 2013; 62:31-43. [PMID: 24021662 DOI: 10.1016/j.nbd.2013.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/18/2013] [Accepted: 09/01/2013] [Indexed: 12/31/2022] Open
Abstract
Recent clinical and pre-clinical studies suggest that both active and passive immunization strategies targeting Aβ amyloid may have clinical benefit in Alzheimer's disease. Here, we demonstrate that vaccination of APPswePSEN1dE9 mice with SDPM1, an engineered non-native Aβ amyloid-specific binding peptide, lowers brain Aβ amyloid plaque burden and brain Aβ1-40 and Aβ1-42 peptide levels, improves cognitive learning and memory in Morris water maze tests and increases the expression of synaptic brain proteins. This was the case in young mice immunized prior to development of significant brain amyloid burden, and in older mice, where brain amyloid was already present. Active immunization was optimized using ALUM as an adjuvant to stimulate production of anti-SDPM1 and anti-Aβ amyloid antibodies. Intracerebral injection of P4D6, an SDPM1 peptide-mimotope antibody, also lowered brain amyloid plaque burden in APPswePSEN1dE9 mice. Additionally, P4D6 inhibited Aβ amyloid-mediated toxicity in cultured neuronal cells. The protein sequence of the variable domain within the P4D6 heavy chain was found to mimic a multimer of the SDPM1 peptide motif. These data demonstrate the efficacy of active and passive vaccine strategies to target Aβ amyloid oligomers using an engineered peptide-mimotope strategy.
Collapse
Affiliation(s)
- Marybeth Camboni
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Chiou-Miin Wang
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Carlos Miranda
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Jung Hae Yoon
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Rui Xu
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Deborah Zygmunt
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Brian K Kaspar
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA; Department of Pediatrics, The Ohio State University, USA; Department of Neuroscience, The Ohio State University, USA
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA; Department of Pediatrics, The Ohio State University, USA; Department of Physiology and Cell Biology, The Ohio State University, USA.
| |
Collapse
|
47
|
Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med 2013; 2:a006247. [PMID: 22762014 DOI: 10.1101/cshperspect.a006247] [Citation(s) in RCA: 541] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tau represents the subunit protein of one of the major hallmarks of Alzheimer disease (AD), the neurofibrillary tangles, and is therefore of major interest as an indicator of disease mechanisms. Many of the unusual properties of Tau can be explained by its nature as a natively unfolded protein. Examples are the large number of structural conformations and biochemical modifications (phosphorylation, proteolysis, glycosylation, and others), the multitude of interaction partners (mainly microtubules, but also other cytoskeletal proteins, kinases, and phosphatases, motor proteins, chaperones, and membrane proteins). The pathological aggregation of Tau is counterintuitive, given its high solubility, but can be rationalized by short hydrophobic motifs forming β structures. The aggregation of Tau is toxic in cell and animal models, but can be reversed by suppressing expression or by aggregation inhibitors. This review summarizes some of the structural, biochemical, and cell biological properties of Tau and Tau fibers. Further aspects of Tau as a diagnostic marker and therapeutic target, its involvement in other Tau-based diseases, and its histopathology are covered by other chapters in this volume.
Collapse
Affiliation(s)
- Eva-Maria Mandelkow
- Max-Planck Unit for Structural Molecular Biology, c/o DESY, 22607 Hamburg, Germany; DZNE, German Center for Neurodegenerative Diseases, and CAESAR Research Center, 53175 Bonn, Germany.
| | | |
Collapse
|
48
|
Kaushal A, Wani WY, Anand R, Gill KD. Spontaneous and induced nontransgenic animal models of AD: modeling AD using combinatorial approach. Am J Alzheimers Dis Other Demen 2013; 28:318-26. [PMID: 23687185 PMCID: PMC10852793 DOI: 10.1177/1533317513488914] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative and dementing disorder, is characterized by extracellular amyloid deposition, intracellular neurofibrillary tangle formation, and neuronal loss. We are still behind in AD research in terms of knowledge regarding understanding its pathophysiology and designing therapeutics because of the lack of an accurate animal model for AD. A complete animal model of AD should imitate all the cognitive, behavioral, and neuropathological features of the disease. Partial models are currently in use, which only mimic specific and not all of the components of AD pathology. Currently the transgenic animals are the popular models for AD research, but different genetic backgrounds of these transgenic animals remain a major confounding factor. This review attempts to summarize the current literature on nontransgenic animal models of AD and to highlight the potential of exploiting spontaneous and induced animal models for neuropathological, neurochemical, neurobehavioral, and neuroprotective studies of AD.
Collapse
Affiliation(s)
- Alka Kaushal
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Willayat Yousuf Wani
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - R. Anand
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Kiran Dip Gill
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
49
|
Canugovi C, Misiak M, Ferrarelli LK, Croteau DL, Bohr VA. The role of DNA repair in brain related disease pathology. DNA Repair (Amst) 2013; 12:578-87. [PMID: 23721970 DOI: 10.1016/j.dnarep.2013.04.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Oxidative DNA damage is implicated in brain aging, neurodegeneration and neurological diseases. Damage can be created by normal cellular metabolism, which accumulates with age, or by acute cellular stress conditions which create bursts of oxidative damage. Brain cells have a particularly high basal level of metabolic activity and use distinct oxidative damage repair mechanisms to remove oxidative damage from DNA and dNTP pools. Accumulation of this damage in the background of a functional DNA repair response is associated with normal aging, but defective repair in brain cells can contribute to neurological dysfunction. Emerging research strongly associates three common neurodegenerative conditions, Alzheimer's, Parkinson's and stroke, with defects in the ability to repair chronic or acute oxidative damage in neurons. This review explores the current knowledge of the role of oxidative damage repair in preserving brain function and highlights the emerging models and methods being used to advance our knowledge of the pathology of neurodegenerative disease.
Collapse
Affiliation(s)
- Chandrika Canugovi
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
50
|
Lithner CU, Lacor PN, Zhao WQ, Mustafiz T, Klein WL, Sweatt JD, Hernandez CM. Disruption of neocortical histone H3 homeostasis by soluble Aβ: implications for Alzheimer's disease. Neurobiol Aging 2013; 34:2081-90. [PMID: 23582659 DOI: 10.1016/j.neurobiolaging.2012.12.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 12/28/2012] [Indexed: 01/08/2023]
Abstract
Amyloid-β peptide (Aβ) fragment misfolding may play a crucial role in the progression of Alzheimer's disease (AD) pathophysiology as well as epigenetic mechanisms at the DNA and histone level. We hypothesized that histone H3 homeostasis is disrupted in association with the appearance of soluble Aβ at an early stage in AD progression. We identified, localized, and compared histone H3 modifications in multiple model systems (neural-like SH-SY5Y, primary neurons, Tg2576 mice, and AD neocortex), and narrowed our focus to investigate 3 key motifs associated with regulating transcriptional activation and inhibition: acetylated lysine 14, phosphorylated serine 10 and dimethylated lysine 9. Our results in vitro and in vivo indicate that multimeric soluble Aβ may be a potent signaling molecule indirectly modulating the transcriptional activity of DNA by modulating histone H3 homeostasis. These findings reveal potential loci of transcriptional disruption relevant to AD. Identifying genes that undergo significant epigenetic alterations in response to Aβ could aid in the understanding of the pathogenesis of AD, as well as suggesting possible new treatment strategies.
Collapse
Affiliation(s)
- Christina Unger Lithner
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|