101
|
Long X, Tao Y, Chen XC, Deng B, Cai J, Zhang SJ. Getting Lost: Place Cells and Grid Cells in Rodent Models of Alzheimer's Disease. Neurosci Bull 2021; 37:894-897. [PMID: 33811610 DOI: 10.1007/s12264-021-00670-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/26/2020] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xiaoyang Long
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuan Tao
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xi-Chan Chen
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Bin Deng
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jing Cai
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Sheng-Jia Zhang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
102
|
Leng K, Li E, Eser R, Piergies A, Sit R, Tan M, Neff N, Li SH, Rodriguez RD, Suemoto CK, Leite REP, Ehrenberg AJ, Pasqualucci CA, Seeley WW, Spina S, Heinsen H, Grinberg LT, Kampmann M. Molecular characterization of selectively vulnerable neurons in Alzheimer's disease. Nat Neurosci 2021; 24:276-287. [PMID: 33432193 PMCID: PMC7854528 DOI: 10.1038/s41593-020-00764-7] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/20/2020] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is characterized by the selective vulnerability of specific neuronal populations, the molecular signatures of which are largely unknown. To identify and characterize selectively vulnerable neuronal populations, we used single-nucleus RNA sequencing to profile the caudal entorhinal cortex and the superior frontal gyrus-brain regions where neurofibrillary inclusions and neuronal loss occur early and late in AD, respectively-from postmortem brains spanning the progression of AD-type tau neurofibrillary pathology. We identified RORB as a marker of selectively vulnerable excitatory neurons in the entorhinal cortex and subsequently validated their depletion and selective susceptibility to neurofibrillary inclusions during disease progression using quantitative neuropathological methods. We also discovered an astrocyte subpopulation, likely representing reactive astrocytes, characterized by decreased expression of genes involved in homeostatic functions. Our characterization of selectively vulnerable neurons in AD paves the way for future mechanistic studies of selective vulnerability and potential therapeutic strategies for enhancing neuronal resilience.
Collapse
Affiliation(s)
- Kun Leng
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
| | - Emmy Li
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Rana Eser
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Antonia Piergies
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Rene Sit
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Norma Neff
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Song Hua Li
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Roberta Diehl Rodriguez
- Department of Neurology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
| | - Claudia Kimie Suemoto
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
- Division of Geriatrics, Department of Clinical Medicine, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
| | | | - Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos A Pasqualucci
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
| | - William W Seeley
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Helmut Heinsen
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
- Department of Psychiatry, University of Würzburg, Würzburg, Germany
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil.
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Martin Kampmann
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
103
|
Spatial memory deficiency early in 6xTg Alzheimer's disease mouse model. Sci Rep 2021; 11:1334. [PMID: 33446720 PMCID: PMC7809274 DOI: 10.1038/s41598-020-79344-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/08/2020] [Indexed: 01/27/2023] Open
Abstract
Alzheimer’s disease (AD) is mainly characterized by the deposition of extracellular amyloid plaques and intracellular accumulation of neurofibrillary tangles (NFTs). While the recent 5xFAD AD mouse model exhibits many AD-related phenotypes and a relatively early and aggressive amyloid β production, it does not show NFTs. Here, we developed and evaluated a novel AD mouse model (6xTg-AD, 6xTg) by crossbreeding 5xFAD mice with mice expressing mutant (P301L) tau protein (MAPT). Through behavioral and histopathological tests, we analyzed cognitive changes and neuropathology in 6xTg mice compared to their respective parental strains according to age. Spatial memory deficits occurred in 6xTg mice at 2 months of age, earlier than they occurred in 5xFAD mice. Histopathological data revealed aggressive Aβ42 and p-tau accumulation in 6xTg mice. Microglial activation occurred in the cortex and hippocampus of 6xTg mice beginning at 2 months. In 6xTg model mice, the synaptic loss was observed in the cortex from 4 months of age and in the hippocampus from 6 months of age, and neuronal loss appeared in the cortex from 4 months of age and in the hippocampus 6 months of age, earlier than it is observed in the 5xFAD and JNPL3 models. These results showed that each pathological symptom appeared much faster than in their parental animal models. In conclusion, these novel 6xTg-AD mice might be an advanced animal model for studying AD, representing a promising approach to developing effective therapy.
Collapse
|
104
|
Toniolo S, Sen A, Husain M. Modulation of Brain Hyperexcitability: Potential New Therapeutic Approaches in Alzheimer's Disease. Int J Mol Sci 2020; 21:E9318. [PMID: 33297460 PMCID: PMC7730926 DOI: 10.3390/ijms21239318] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
People with Alzheimer's disease (AD) have significantly higher rates of subclinical and overt epileptiform activity. In animal models, oligomeric Aβ amyloid is able to induce neuronal hyperexcitability even in the early phases of the disease. Such aberrant activity subsequently leads to downstream accumulation of toxic proteins, and ultimately to further neurodegeneration and neuronal silencing mediated by concomitant tau accumulation. Several neurotransmitters participate in the initial hyperexcitable state, with increased synaptic glutamatergic tone and decreased GABAergic inhibition. These changes appear to activate excitotoxic pathways and, ultimately, cause reduced long-term potentiation, increased long-term depression, and increased GABAergic inhibitory remodelling at the network level. Brain hyperexcitability has therefore been identified as a potential target for therapeutic interventions aimed at enhancing cognition, and, possibly, disease modification in the longer term. Clinical trials are ongoing to evaluate the potential efficacy in targeting hyperexcitability in AD, with levetiracetam showing some encouraging effects. Newer compounds and techniques, such as gene editing via viral vectors or brain stimulation, also show promise. Diagnostic challenges include identifying best biomarkers for measuring sub-clinical epileptiform discharges. Determining the timing of any intervention is critical and future trials will need to carefully stratify participants with respect to the phase of disease pathology.
Collapse
Affiliation(s)
- Sofia Toniolo
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department Clinical Neurosciences, John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Masud Husain
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| |
Collapse
|
105
|
Fan YG, Pang ZQ, Wu TY, Zhang YH, Xuan WQ, Wang Z, Yu X, Li YC, Guo C, Wang ZY. Vitamin D deficiency exacerbates Alzheimer-like pathologies by reducing antioxidant capacity. Free Radic Biol Med 2020; 161:139-149. [PMID: 33068737 DOI: 10.1016/j.freeradbiomed.2020.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/05/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
Vitamin D (VD) deficiency is prevalent among aging people and Alzheimer's disease (AD) patients. However, the roles of VD deficiency in the pathology of AD remain largely unexplored. In this study, APP/PS1 mice were fed a VD-deficient diet for 13 weeks to evaluate the effects of VD deficiency on the learning and memory functions and the neuropathological characteristics of the mice. Our study revealed that VD deficiency accelerated cognitive impairment in the APP/PS1 mice. Mechanistic studies revealed that VD deficiency promoted glial activation and increased inflammatory factor secretion. Furthermore, VD deficiency increased the production and deposition of Aβ by elevating the expression levels of amyloid precursor protein (APP) and β-site APP cleavage enzyme 1 (BACE1). In addition, VD deficiency increased the phosphorylation of Tau at Thr181, Thr205 and Ser396 by increasing the activities of cyclin-dependent kinase 5 (CDK5) and glycogen synthase kinase 3α/β (GSK3α/β) and promoted synaptic dystrophy and neuronal loss. All these effects of VD deficiency may be ascribed to enhanced oxidative stress via the downregulation of superoxide dismutase 1 (SOD1), glutathione peroxidase 4 (GPx4) and cystine/glutamate exchanger (xCT). Taken together, our data suggest that VD deficiency exacerbates Alzheimer-like pathologies via promoting inflammatory stress, increasing Aβ production and elevating Tau phosphorylation by decreasing antioxidant capacity in the brains of APP/PS1 mice. Hence, rescuing the VD status of AD patients should be taken into consideration during the treatment of AD.
Collapse
Affiliation(s)
- Yong-Gang Fan
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Zhong-Qiu Pang
- College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Ting-Yao Wu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yan-Hui Zhang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Wen-Qiang Xuan
- College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Zhuo Wang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Xin Yu
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Yan-Chun Li
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China.
| | - Zhan-You Wang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
106
|
Ridler T, Witton J, Phillips KG, Randall AD, Brown JT. Impaired speed encoding and grid cell periodicity in a mouse model of tauopathy. eLife 2020; 9:e59045. [PMID: 33242304 PMCID: PMC7690954 DOI: 10.7554/elife.59045] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Dementia is associated with severe spatial memory deficits which arise from dysfunction in hippocampal and parahippocampal circuits. For spatially sensitive neurons, such as grid cells, to faithfully represent the environment these circuits require precise encoding of direction and velocity information. Here, we have probed the firing rate coding properties of neurons in medial entorhinal cortex (MEC) in a mouse model of tauopathy. We find that grid cell firing patterns are largely absent in rTg4510 mice, while head-direction tuning remains largely intact. Conversely, neural representation of running speed information was significantly disturbed, with smaller proportions of MEC cells having firing rates correlated with locomotion in rTg4510 mice. Additionally, the power of local field potential oscillations in the theta and gamma frequency bands, which in wild-type mice are tightly linked to running speed, was invariant in rTg4510 mice during locomotion. These deficits in locomotor speed encoding likely severely impact path integration systems in dementia.
Collapse
Affiliation(s)
- Thomas Ridler
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Hatherly LaboratoriesExeterUnited Kingdom
| | - Jonathan Witton
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Hatherly LaboratoriesExeterUnited Kingdom
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUnited Kingdom
| | - Keith G Phillips
- Lilly United Kingdom Erl Wood Manor WindleshamSurreyUnited Kingdom
| | - Andrew D Randall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Hatherly LaboratoriesExeterUnited Kingdom
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUnited Kingdom
| | - Jonathan T Brown
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Hatherly LaboratoriesExeterUnited Kingdom
| |
Collapse
|
107
|
Salvadores N, Gerónimo-Olvera C, Court FA. Axonal Degeneration in AD: The Contribution of Aβ and Tau. Front Aging Neurosci 2020; 12:581767. [PMID: 33192476 PMCID: PMC7593241 DOI: 10.3389/fnagi.2020.581767] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) represents the most common age-related neurodegenerative disorder, affecting around 35 million people worldwide. Despite enormous efforts dedicated to AD research over decades, there is still no cure for the disease. Misfolding and accumulation of Aβ and tau proteins in the brain constitute a defining signature of AD neuropathology, and mounting evidence has documented a link between aggregation of these proteins and neuronal dysfunction. In this context, progressive axonal degeneration has been associated with early stages of AD and linked to Aβ and tau accumulation. As the axonal degeneration mechanism has been starting to be unveiled, it constitutes a promising target for neuroprotection in AD. A comprehensive understanding of the mechanism of axonal destruction in neurodegenerative conditions is therefore critical for the development of new therapies aimed to prevent axonal loss before irreversible neuronal death occurs in AD. Here, we review current evidence of the involvement of Aβ and tau pathologies in the activation of signaling cascades that can promote axonal demise.
Collapse
Affiliation(s)
- Natalia Salvadores
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Cristian Gerónimo-Olvera
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, United States
| |
Collapse
|
108
|
Fung CW, Guo J, Fu H, Figueroa HY, Konofagou EE, Duff KE. Atrophy associated with tau pathology precedes overt cell death in a mouse model of progressive tauopathy. SCIENCE ADVANCES 2020; 6:6/42/eabc8098. [PMID: 33067235 PMCID: PMC7567584 DOI: 10.1126/sciadv.abc8098] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/02/2020] [Indexed: 06/11/2023]
Abstract
Tau pathology in Alzheimer's disease (AD) first develops in the entorhinal cortex (EC), then spreads to the hippocampus, followed by the neocortex. Overall, tau pathology correlates well with neurodegeneration and cell loss, but the spatial and temporal association between tau pathology and overt volume loss (atrophy) associated with structural changes or cell loss is unclear. Using in vivo magnetic resonance imaging (MRI) with tensor-based morphometry (TBM), we mapped the spatiotemporal pattern of structural changes in a mouse model of AD-like progressive tauopathy. A novel, coregistered in vivo MRI atlas was then applied to identify regions in the medial temporal lobe that had a significant volume reduction. Our study shows that in a mouse model of tauopathy spread, the propagation of tau pathology from the EC to the hippocampus is associated with TBM-related atrophy, but atrophy in the dentate gyrus and subiculum precedes overt cell loss.
Collapse
Affiliation(s)
- Christine W Fung
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, 500 W 120th Street, New York, NY 10025, USA
| | - Jia Guo
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
- Zuckerman Institute, Columbia University, 3227 Broadway, New York, NY 10027, USA
| | - Hongjun Fu
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH 43210, USA
| | - Helen Y Figueroa
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, 500 W 120th Street, New York, NY 10025, USA
| | - Karen E Duff
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA
- UK Dementia Research Institute at University College London, London, UK
| |
Collapse
|
109
|
Coughlan G, Puthusseryppady V, Lowry E, Gillings R, Spiers H, Minihane AM, Hornberger M. Test-retest reliability of spatial navigation in adults at-risk of Alzheimer's disease. PLoS One 2020; 15:e0239077. [PMID: 32960930 PMCID: PMC7508365 DOI: 10.1371/journal.pone.0239077] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/28/2020] [Indexed: 12/16/2022] Open
Abstract
The Virtual Supermarket Task (VST) and Sea Hero Quest detect high-genetic-risk Alzheimer`s disease (AD). We aimed to determine their test-retest reliability in a preclinical AD population. Over two time points, separated by an 18-month period, 59 cognitively healthy individuals underwent a neuropsychological and spatial navigation assessment. At baseline, participants were classified as low-genetic-risk of AD or high-genetic-risk of AD. We calculated two-way mixed effects intraclass correlation coefficients (ICC) for task parameters and used repeated measures ANOVAS to determine whether genetic risk or sex contributed to test-retest variability. The egocentric parameter of the VST measure showed the highest test-retest reliability (ICC = .72), followed by the SHQ distance travelled parameter (ICC = .50). Post hoc longitudinal analysis showed that boundary-based navigation predicts worsening episodic memory concerns in high-risk (F = 5.01, P = 0.03), but in not low-risk, AD candidates. The VST and the Sea Hero Quest produced parameters with acceptable test-retest reliability. Further research in larger sample sizes is desirable.
Collapse
Affiliation(s)
- Gillian Coughlan
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | | | - Ellen Lowry
- Department of Psychology, University of East Anglia, Norwich, United Kingdom
| | - Rachel Gillings
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Hugo Spiers
- Department of Experimental Psychology, Institute of Behavioural Neuroscience, University College London, London, United Kingdom
| | | | - Michael Hornberger
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
110
|
Li L, Venkataraman L, Chen S, Fu H. Function of WFS1 and WFS2 in the Central Nervous System: Implications for Wolfram Syndrome and Alzheimer's disease. Neurosci Biobehav Rev 2020; 118:775-783. [PMID: 32949681 DOI: 10.1016/j.neubiorev.2020.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
L.P. Li, L. Venkataraman, S. Chen, and H.J. Fu. Function of WFS1 and WFS2 in the Central Nervous System: Implications for Wolfram Syndrome and Alzheimer's Disease. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Wolfram syndrome (WS) is a rare monogenetic spectrum disorder characterized by insulin-dependent juvenile-onset diabetes mellitus, diabetes insipidus, optic nerve atrophy, hearing loss, progressive neurodegeneration, and a wide spectrum of psychiatric manifestations. Most WS patients belong to Wolfram Syndrome type 1 (WS1) caused by mutations in the Wolfram Syndrome 1 (WFS1/Wolframin) gene, while a small fraction of patients belongs to Wolfram Syndrome type 2 (WS2) caused by pathogenic variants in the CDGSH Iron Sulfur Domain 2 (CISD2/WFS2) gene. Although currently there is no treatment for this life-threatening disease, the molecular mechanisms underlying the pathogenesis of WS have been proposed. Interestingly, Alzheimer's disease (AD), an age-dependent neurodegenerative disease, shares some common mechanisms with WS. In this review, we focus on the function of WFS1 and WFS2 in the central nervous system as well as their implications in WS and AD. We also propose three future directions for elucidating the role of WFS1 and WFS2 in WS and AD.
Collapse
Affiliation(s)
- Liangping Li
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Lalitha Venkataraman
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Shuo Chen
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
111
|
Rodriguez GA, Barrett GM, Duff KE, Hussaini SA. Chemogenetic attenuation of neuronal activity in the entorhinal cortex reduces Aβ and tau pathology in the hippocampus. PLoS Biol 2020; 18:e3000851. [PMID: 32822389 PMCID: PMC7467290 DOI: 10.1371/journal.pbio.3000851] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/02/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
High levels of the amyloid-beta (Aβ) peptide have been shown to disrupt neuronal function and induce hyperexcitability, but it is unclear what effects Aβ-associated hyperexcitability may have on tauopathy pathogenesis or propagation in vivo. Using a novel transgenic mouse line to model the impact of human APP (hAPP)/Aβ accumulation on tauopathy in the entorhinal cortex–hippocampal (EC-HIPP) network, we demonstrate that hAPP overexpression aggravates EC-Tau aggregation and accelerates pathological tau spread into the hippocampus. In vivo recordings revealed a strong role for hAPP/Aβ, but not tau, in the emergence of EC neuronal hyperactivity and impaired theta rhythmicity. Chronic chemogenetic attenuation of EC neuronal hyperactivity led to reduced hAPP/Aβ accumulation and reduced pathological tau spread into downstream hippocampus. These data strongly support the hypothesis that in Alzheimer’s disease (AD), Aβ-associated hyperactivity accelerates the progression of pathological tau along vulnerable neuronal circuits, and demonstrates the utility of chronic, neuromodulatory approaches in ameliorating AD pathology in vivo. A novel, triple transgenic mouse model of Alzheimer's disease reveals that amyloid beta-associated neuronal hyperactivity and network dysfunction accelerates the spread of pathological tau from the entorhinal cortex into the hippocampus. Chronic attenuation of neuronal activity using chemogenetics reduces this effect, supporting a role for neuronal hyperactivity in Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Gustavo A. Rodriguez
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Geoffrey M. Barrett
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Karen E. Duff
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- UK Dementia Research Institute at University College London, London, United Kingdom
- * E-mail: (SAH); (KED)
| | - S. Abid Hussaini
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- * E-mail: (SAH); (KED)
| |
Collapse
|
112
|
Morrone CD, Bazzigaluppi P, Beckett TL, Hill ME, Koletar MM, Stefanovic B, McLaurin J. Regional differences in Alzheimer's disease pathology confound behavioural rescue after amyloid-β attenuation. Brain 2020; 143:359-373. [PMID: 31782760 PMCID: PMC6935751 DOI: 10.1093/brain/awz371] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Failure of Alzheimer’s disease clinical trials to improve or stabilize cognition has led to the need for a better understanding of the driving forces behind cognitive decline in the presence of active disease processes. To dissect contributions of individual pathologies to cognitive function, we used the TgF344-AD rat model, which recapitulates the salient hallmarks of Alzheimer’s disease pathology observed in patient populations (amyloid, tau inclusions, frank neuronal loss, and cognitive deficits). scyllo-Inositol treatment attenuated amyloid-β peptide in disease-bearing TgF344-AD rats, which rescued pattern separation in the novel object recognition task and executive function in the reversal learning phase of the Barnes maze. Interestingly, neither activities of daily living in the burrowing task nor spatial memory in the Barnes maze were rescued by attenuating amyloid-β peptide. To understand the pathological correlates leading to behavioural rescue, we examined the neuropathology and in vivo electrophysiological signature of the hippocampus. Amyloid-β peptide attenuation reduced hippocampal tau pathology and rescued adult hippocampal neurogenesis and neuronal function, via improvements in cross-frequency coupling between theta and gamma bands. To investigate mechanisms underlying the persistence of spatial memory deficits, we next examined neuropathology in the entorhinal cortex, a region whose input to the hippocampus is required for spatial memory. Reduction of amyloid-β peptide in the entorhinal cortex had no effect on entorhinal tau pathology or entorhinal-hippocampal neuronal network dysfunction, as measured by an impairment in hippocampal response to entorhinal stimulation. Thus, rescue or not of cognitive function is dependent on regional differences of amyloid-β, tau and neuronal network dysfunction, demonstrating the importance of staging disease in patients prior to enrolment in clinical trials. These results further emphasize the need for combination therapeutic approaches across disease progression.
Collapse
Affiliation(s)
- Christopher D Morrone
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, 1 King's College Cir, Toronto, ON, Canada
| | - Paolo Bazzigaluppi
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Tina L Beckett
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Mary E Hill
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Margaret M Koletar
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Medical Biophysics, 101 College St Suite 15-701, Toronto, ON, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, 1 King's College Cir, Toronto, ON, Canada
| |
Collapse
|
113
|
Harris SS, Wolf F, De Strooper B, Busche MA. Tipping the Scales: Peptide-Dependent Dysregulation of Neural Circuit Dynamics in Alzheimer’s Disease. Neuron 2020; 107:417-435. [DOI: 10.1016/j.neuron.2020.06.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/24/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
|
114
|
Bierbrauer A, Kunz L, Gomes CA, Luhmann M, Deuker L, Getzmann S, Wascher E, Gajewski PD, Hengstler JG, Fernandez-Alvarez M, Atienza M, Cammisuli DM, Bonatti F, Pruneti C, Percesepe A, Bellaali Y, Hanseeuw B, Strange BA, Cantero JL, Axmacher N. Unmasking selective path integration deficits in Alzheimer's disease risk carriers. SCIENCE ADVANCES 2020; 6:eaba1394. [PMID: 32923622 PMCID: PMC7455192 DOI: 10.1126/sciadv.aba1394] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 07/15/2020] [Indexed: 05/11/2023]
Abstract
Alzheimer's disease (AD) manifests with progressive memory loss and spatial disorientation. Neuropathological studies suggest early AD pathology in the entorhinal cortex (EC) of young adults at genetic risk for AD (APOE ε4-carriers). Because the EC harbors grid cells, a likely neural substrate of path integration (PI), we examined PI performance in APOE ε4-carriers during a virtual navigation task. We report a selective impairment in APOE ε4-carriers specifically when recruitment of compensatory navigational strategies via supportive spatial cues was disabled. A separate fMRI study revealed that PI performance was associated with the strength of entorhinal grid-like representations when no compensatory strategies were available, suggesting grid cell dysfunction as a mechanistic explanation for PI deficits in APOE ε4-carriers. Furthermore, posterior cingulate/retrosplenial cortex was involved in the recruitment of compensatory navigational strategies via supportive spatial cues. Our results provide evidence for selective PI deficits in AD risk carriers, decades before potential disease onset.
Collapse
Affiliation(s)
- Anne Bierbrauer
- Department of Neuropsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
- Corresponding author. (A.B.); (L.K.); (N.A.)
| | - Lukas Kunz
- Epilepsy Center, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106 Freiburg im Breisgau, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
- Corresponding author. (A.B.); (L.K.); (N.A.)
| | - Carlos A. Gomes
- Department of Neuropsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Maike Luhmann
- Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Lorena Deuker
- Department of Neuropsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Stephan Getzmann
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Edmund Wascher
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Patrick D. Gajewski
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Marina Fernandez-Alvarez
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Network Center for Biomedical Research in Neurodegenerative Disease (CIBERNED), Seville, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Network Center for Biomedical Research in Neurodegenerative Disease (CIBERNED), Seville, Spain
| | - Davide M. Cammisuli
- Department of Medicine and Surgery, Laboratory of Clinical Psychology, Clinical Psychophysiology and Clinical Neuropsychology, University of Parma, Parma, Italy
| | - Francesco Bonatti
- Department of Medicine and Surgery, Medical Genetics, University of Parma, Parma, Italy
| | - Carlo Pruneti
- Department of Medicine and Surgery, Laboratory of Clinical Psychology, Clinical Psychophysiology and Clinical Neuropsychology, University of Parma, Parma, Italy
| | - Antonio Percesepe
- Department of Medicine and Surgery, Medical Genetics, University of Parma, Parma, Italy
| | - Youssef Bellaali
- Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Hanseeuw
- Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bryan A. Strange
- Department of Neuroimaging, Alzheimer’s Disease Research Centre, Reina Sofia–CIEN Foundation, Madrid, Spain
- Laboratory for Clinical Neuroscience, Centre for Biomedical Technology, Universidad Politecnica de Madrid, Madrid, Spain
| | - Jose L. Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Network Center for Biomedical Research in Neurodegenerative Disease (CIBERNED), Seville, Spain
| | - Nikolai Axmacher
- Department of Neuropsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Xinjiekouwai Street 19, Beijing 100875, China
- Corresponding author. (A.B.); (L.K.); (N.A.)
| |
Collapse
|
115
|
Disrupted Place Cell Remapping and Impaired Grid Cells in a Knockin Model of Alzheimer's Disease. Neuron 2020; 107:1095-1112.e6. [PMID: 32697942 DOI: 10.1016/j.neuron.2020.06.023] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/13/2020] [Accepted: 06/22/2020] [Indexed: 11/24/2022]
Abstract
Patients with Alzheimer's disease (AD) suffer from spatial memory impairment and wandering behavior, but the brain circuit mechanisms causing such symptoms remain largely unclear. In healthy brains, spatially tuned hippocampal place cells and entorhinal grid cells exhibit distinct spike patterns in different environments, a circuit function called "remapping." We tested remapping in amyloid precursor protein knockin (APP-KI) mice with impaired spatial memory. CA1 neurons, including place cells, showed disrupted remapping, although their spatial tuning was only mildly diminished. Medial entorhinal cortex (MEC) neurons severely lost their spatial tuning and grid cells were almost absent. Fast gamma oscillatory coupling between the MEC and CA1 was also impaired. Mild disruption of MEC grid cells emerged in younger APP-KI mice, although the spatial memory and CA1 remapping of the animals remained intact. These results point to remapping impairment in the hippocampus, possibly linked to grid cell disruption, as circuit mechanisms underlying spatial memory impairment in AD.
Collapse
|
116
|
Bertan F, Wischhof L, Sosulina L, Mittag M, Dalügge D, Fornarelli A, Gardoni F, Marcello E, Di Luca M, Fuhrmann M, Remy S, Bano D, Nicotera P. Loss of Ryanodine Receptor 2 impairs neuronal activity-dependent remodeling of dendritic spines and triggers compensatory neuronal hyperexcitability. Cell Death Differ 2020; 27:3354-3373. [PMID: 32641776 PMCID: PMC7853040 DOI: 10.1038/s41418-020-0584-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/15/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
Dendritic spines are postsynaptic domains that shape structural and functional properties of neurons. Upon neuronal activity, Ca2+ transients trigger signaling cascades that determine the plastic remodeling of dendritic spines, which modulate learning and memory. Here, we study in mice the role of the intracellular Ca2+ channel Ryanodine Receptor 2 (RyR2) in synaptic plasticity and memory formation. We demonstrate that loss of RyR2 in pyramidal neurons of the hippocampus impairs maintenance and activity-evoked structural plasticity of dendritic spines during memory acquisition. Furthermore, post-developmental deletion of RyR2 causes loss of excitatory synapses, dendritic sparsification, overcompensatory excitability, network hyperactivity and disruption of spatially tuned place cells. Altogether, our data underpin RyR2 as a link between spine remodeling, circuitry dysfunction and memory acquisition, which closely resemble pathological mechanisms observed in neurodegenerative disorders.
Collapse
Affiliation(s)
- Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Manuel Mittag
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dennis Dalügge
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Martin Fuhrmann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stefan Remy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | |
Collapse
|
117
|
Xu Y, Zhao M, Han Y, Zhang H. GABAergic Inhibitory Interneuron Deficits in Alzheimer's Disease: Implications for Treatment. Front Neurosci 2020; 14:660. [PMID: 32714136 PMCID: PMC7344222 DOI: 10.3389/fnins.2020.00660] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized clinically by severe cognitive deficits and pathologically by amyloid plaques, neuronal loss, and neurofibrillary tangles. Abnormal amyloid β-protein (Aβ) deposition in the brain is often thought of as a major initiating factor in AD neuropathology. However, gamma-aminobutyric acid (GABA) inhibitory interneurons are resistant to Aβ deposition, and Aβ decreases synaptic glutamatergic transmission to decrease neural network activity. Furthermore, there is now evidence suggesting that neural network activity is aberrantly increased in AD patients and animal models due to functional deficits in and decreased activity of GABA inhibitory interneurons, contributing to cognitive deficits. Here we describe the roles played by excitatory neurons and GABA inhibitory interneurons in Aβ-induced cognitive deficits and how altered GABA interneurons regulate AD neuropathology. We also comprehensively review recent studies on how GABA interneurons and GABA receptors can be exploited for therapeutic benefit. GABA interneurons are an emerging therapeutic target in AD, with further clinical trials urgently warranted.
Collapse
Affiliation(s)
- Yilan Xu
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Manna Zhao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Yuying Han
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
118
|
Functional connectivity between the entorhinal and posterior cingulate cortices underpins navigation discrepancies in at-risk Alzheimer's disease. Neurobiol Aging 2020; 90:110-118. [DOI: 10.1016/j.neurobiolaging.2020.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 01/29/2023]
|
119
|
Jiang Y, Zhang Y, Su L. MiR-539-5p Decreases amyloid β-protein production, hyperphosphorylation of Tau and Memory Impairment by Regulating PI3K/Akt/GSK-3β Pathways in APP/PS1 Double Transgenic Mice. Neurotox Res 2020; 38:524-535. [PMID: 32415525 DOI: 10.1007/s12640-020-00217-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
The production of amyloid β (Aβ) and tau hyperphosphorylation have been identified as key processes in Alzheimer's disease (AD) pathogenesis. MiR-539-5p has been found to be abnormally expressed in brain tissue; however, the functional role of miR-539-5p in the pathogenesis of AD remains unclear. In our study, we found that the expression of miR-539-5p was significantly downregulated in humans and mice with AD and was negatively correlated with expression of APP, caveolin 1, and GSK-3β. Moreover, upregulation of miR-539-5p inhibited Aβ accumulation, tau phosphorylation, oxidative stress, and apoptosis and improved memory ability in AD mice. Furthermore, by using bioinformatics tool and dual-luciferase reporter assay, APP, Caveolin 1, and GSK-3β were confirmed as direct targets of miR-539-5p. In addition, the PI3K/AKT/GSK-3β signaling pathway can be regulated by miR-539-5p. In conclusion, this study provided a novel insight into the pathologic mechanism of AD by identifying that miR-539-5p plays a neuroprotective role in AD.
Collapse
Affiliation(s)
- Yushu Jiang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou City, 450000, Henan Province, China
| | - Yuan Zhang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The Central Laboratory, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, No.3002 Sungang West Road, Futian District, Shenzhen City, 518035, Guangdong Province, China.
| | - Li Su
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen City, 518055, Guangdong Province, China
| |
Collapse
|
120
|
Liu B, Kou J, Li F, Huo D, Xu J, Zhou X, Meng D, Ghulam M, Artyom B, Gao X, Ma N, Han D. Lemon essential oil ameliorates age-associated cognitive dysfunction via modulating hippocampal synaptic density and inhibiting acetylcholinesterase. Aging (Albany NY) 2020; 12:8622-8639. [PMID: 32392535 PMCID: PMC7244039 DOI: 10.18632/aging.103179] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
The lemon essential oil (LEO), extracted from the fruit of lemon, has been used to treat multiple pathological diseases, such as diabetes, inflammation, cardiovascular diseases, depression and hepatobiliary dysfunction. The study was designed to study the effects of LEO on cognitive dysfunction induced by Alzheimer’s disease (AD). We used APP/PS1 double transgene (APP/PS1) AD mice in the experiment; these mice exhibit significant deficits in synaptic density and hippocampal-dependent spatial related memory. The effects of LEO on learning and memory were examined using the Morris Water Maze (MWM) test, Novel object recognition test, and correlative indicators, including a neurotransmitter (acetylcholinesterase, AChE), a nerve growth factor (brain-derived neurotrophic factor, BDNF), a postsynaptic marker (PSD95), and presynaptic markers (synapsin-1, and synaptophysin), in APP/PS1 mice. Histopathology was performed to estimate the effects of LEO on AD mice. A significantly lowered brain AChE depression in APP/PS1 and wild-type C57BL/6L (WT) mice. PSD95/ Synaptophysin, the index of synaptic density, was noticeably improved in histopathologic changes. Hence, it can be summarized that memory-enhancing activity might be associated with a reduction in the AChE levels and is elevated by BDNF, PSD95, and synaptophysin through enhancing synaptic plasticity.
Collapse
Affiliation(s)
- Bonan Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Fuyan Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Da Huo
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Jiaran Xu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Xiaoxi Zhou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Dehao Meng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Murtaza Ghulam
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Bobkov Artyom
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin 150081, China.,Translational Medicine Center of Northern China, Harbin 150081, China.,Heilongjiang Provincial key Laboratory of Genetically Modified Model Animal, Harbin Medical University, Ministry of Education, Harbin 150081, China.,China Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Ning Ma
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin 150081, China.,Translational Medicine Center of Northern China, Harbin 150081, China
| | - Dong Han
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin 150081, China.,Translational Medicine Center of Northern China, Harbin 150081, China
| |
Collapse
|
121
|
Koller EJ, Gonzalez De La Cruz E, Machula T, Ibanez KR, Lin WL, Williams T, Riffe CJ, Ryu D, Strang KH, Liu X, Janus C, Golde TE, Dickson D, Giasson BI, Chakrabarty P. Combining P301L and S320F tau variants produces a novel accelerated model of tauopathy. Hum Mol Genet 2020; 28:3255-3269. [PMID: 31261380 DOI: 10.1093/hmg/ddz151] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/22/2019] [Accepted: 06/21/2019] [Indexed: 01/14/2023] Open
Abstract
Understanding the biological functions of tau variants can illuminate differential etiologies of Alzheimer's disease (AD) and primary tauopathies. Though the end-stage neuropathological attributes of AD and primary tauopathies are similar, the etiology and behavioral outcomes of these diseases follow unique and divergent trajectories. To study the divergent physiological properties of tau variants on a uniform immunogenetic background, we created somatic transgenesis CNS models of tauopathy utilizing neonatal delivery of adeno-associated viruses expressing wild-type (WT) or mutant tau in non-transgenic mice. We selected four different tau variants-WT tau associated with AD, P301L mutant tau associated with frontotemporal dementia (FTD), S320F mutant tau associated with Pick's disease and a combinatorial approach using P301L/S320F mutant tau. CNS-targeted expression of WT and P301L mutant tau results in robust tau hyperphosphorylation without tangle pathology, gradually developing age-progressive memory deficits. In contrast, the S320F variant, especially in combination with P301L, produces an AD-type tangle pathology, focal neuroinflammation and memory impairment on an accelerated time scale. Using the doubly mutated P301L/S320F tau variant, we demonstrate that combining different mutations can have an additive effect on neuropathologies and associated co-morbidities, possibly hinting at involvement of unique functional pathways. Importantly, we also show that overexpression of wild-type tau as well as an FTD-associated tau variant can lead to cognitive deficits even in the absence of tangles. Together, our data highlights the synergistic neuropathologies and associated cognitive and synaptic alterations of the combinatorial tau variant leading to a robust model of tauopathy.
Collapse
Affiliation(s)
- Emily J Koller
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Elsa Gonzalez De La Cruz
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Timothy Machula
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Kristen R Ibanez
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Wen-Lang Lin
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, USA
| | - Tosha Williams
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Cara J Riffe
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Daniel Ryu
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Kevin H Strang
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Xuefei Liu
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Christopher Janus
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Todd E Golde
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Dennis Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, USA
| | - Benoit I Giasson
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
122
|
Vimal SK, Zuo H, Wang Z, Wang H, Long Z, Bhattacharyya S. Self-Therapeutic Nanoparticle That Alters Tau Protein and Ameliorates Tauopathy Toward a Functional Nanomedicine to Tackle Alzheimer's. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906861. [PMID: 32191383 DOI: 10.1002/smll.201906861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/30/2020] [Accepted: 02/13/2020] [Indexed: 06/10/2023]
Abstract
Tauopathy is a complex disorder associated at the junction of several other pathologies. Intrinsically disordered tau protein remains therapeutically challenging due to its undruggable nature and is a possible reason for monumental failure of several tau-based therapies. Herein, nanogold remodeled tau is reported as a pseudo-nanochaperon and shows therapeutic benefit by passive targeting in transgenic tau P301L mutant mice. Treatment with nanogold polyethylene glycol (Au-PEG) conjugate moderately improves the learning ability of the tau P301L mice that corroborates with diminished phosphorylated tau burden. Circulating total tau level that acts in a prion fashion is significantly reduced upon Au-PEG treatment. Similarly, a high level of tau is found in macaque monkey serum and Au-PEG inhibits amyloidosis of Alzheimer's patients and primate's serum samples ex vivo. Addtionally, brain MRI of an old aged macaque monkey shows the decrease of grey matter, which correlates with mutual loss of grey matter upon progressive dementia as reported. Au-PEG tunes tau and other circulating pro-dementia factors that are present in human AD serum, by remodeling the protein and repairing aberrant proteostasis. Alteration of proteotoxic tau function by nanogold as a kinetic stablizer holds translational potential to combat socially challenging dementia.
Collapse
Affiliation(s)
- Sunil Kumar Vimal
- Department of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| | - Hua Zuo
- Department of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| | - Zhengwu Wang
- Sichuan Hengshu Bio-tech Co.LTD, Tanqiao Group, Xinlian Village, Xijie Town, Yibin City, 644600, P. R. China
| | - Hongrun Wang
- Sichuan Hengshu Bio-tech Co.LTD, Tanqiao Group, Xinlian Village, Xijie Town, Yibin City, 644600, P. R. China
| | - Zhiliang Long
- Sleep and Neuroimaging Center, Faculty of Psychology, Southwest University, Chongqing, 400715, P. R. China
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
123
|
Schöberl F, Pradhan C, Irving S, Buerger K, Xiong G, Kugler G, Kohlbecher S, Engmann J, Werner P, Brendel M, Schneider E, Perneczky R, Jahn K, la Fougère C, Bartenstein P, Brandt T, Dieterich M, Zwergal A. Real-space navigation testing differentiates between amyloid-positive and -negative aMCI. Neurology 2020; 94:e861-e873. [PMID: 31896617 DOI: 10.1212/wnl.0000000000008758] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/05/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To distinguish between patients with amyloid-positive (A+) and -negative (A-) amnestic mild cognitive impairment (aMCI) by simultaneously investigating navigation performance, visual exploration behavior, and brain activations during a real-space navigation paradigm. METHODS Twenty-one patients with aMCI were grouped into A+ (n = 11) and A- cases by amyloid-PET imaging and amyloid CSF levels and compared to 15 healthy controls. Neuropsychological deficits were quantified by use of the Consortium to Establish a Registry for Alzheimer's Disease-plus cognitive battery. All participants performed a navigation task in which they had to find items in a realistic spatial environment and had to apply egocentric and allocentric route planning strategies. 18F-fluorodeoxyglucose was injected at the start to detect navigation-induced brain activations. Subjects wore a gaze-controlled, head-fixed camera that recorded their visual exploration behavior. RESULTS A+ patients performed worse during egocentric and allocentric navigation compared to A- patients and controls (p < 0.001). Both aMCI subgroups used fewer shortcuts, moved more slowly, and stayed longer at crossings. Word-list learning, figural learning, and Trail-Making tests did not differ in the A+ and A- subgroups. A+ patients showed a reduced activation of the right hippocampus, retrosplenial, and parietal cortex during navigation compared to A- patients (p < 0.005). CONCLUSIONS A+ patients with aMCI perform worse than A- patients with aMCI in egocentric and allocentric route planning because of a more widespread impairment of their cerebral navigation network. Navigation testing in real space is a promising approach to identify patients with aMCI with underlying Alzheimer pathology.
Collapse
Affiliation(s)
- Florian Schöberl
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Cauchy Pradhan
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Stephanie Irving
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Katharina Buerger
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Guoming Xiong
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Günter Kugler
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Stefan Kohlbecher
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Julia Engmann
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Philipp Werner
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Matthias Brendel
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Erich Schneider
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Robert Perneczky
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Klaus Jahn
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Christian la Fougère
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Peter Bartenstein
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Thomas Brandt
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Marianne Dieterich
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany
| | - Andreas Zwergal
- From the Department of Neurology (F.S., J.E., P.W., A.Z., M.D.), University Hospital, German Center for Vertigo and Balance Disorders (F.S., C.P., S.I., G.X., G.K., S.K., E.S., K.J., C.l.F., P.B., T.B., M.D., A.Z.), DSGZ, Institute for Stroke and Dementia Research (K.B.), ISD, University Hospital, Department of Nuclear Medicine (G.X., M.B., P.B.), Department of Psychiatry (R.P.), and Clinical Neurosciences (T.B.), Ludwig Maximilian University of Munich; German Center for Neurodegenerative Diseases (K.B., R.P., M.D.), DZNE, Munich; Institute for Medical Technology (E.S.), Brandenburg University of Technology Cottbus-Senftenberg; Munich Cluster of Systems Neurology (R.P., P.B., M.D.), SyNergy, Germany; Ageing Epidemiology Research Unit (R.P.), School of Public Health, Imperial College, London, UK; Neurological Hospital (K.J.), Schön Klinik Bad Aibling; and Department of Nuclear Medicine (C.l.F.), Eberhard Karl University of Tübingen, Germany.
| |
Collapse
|
124
|
Ye J, Yin Y, Liu H, Fang L, Tao X, Wei L, Zuo Y, Yin Y, Ke D, Wang J. Tau inhibits PKA by nuclear proteasome-dependent PKAR2α elevation with suppressed CREB/GluA1 phosphorylation. Aging Cell 2020; 19:e13055. [PMID: 31668016 PMCID: PMC6974714 DOI: 10.1111/acel.13055] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 07/28/2019] [Accepted: 10/05/2019] [Indexed: 01/03/2023] Open
Abstract
Intraneuronal accumulation of wild-type tau plays a key role in Alzheimer's disease, while the mechanisms underlying tauopathy and memory impairment remain unclear. Here, we report that overexpressing full-length wild-type human tau (hTau) in mouse hippocampus induces learning and memory deficits with remarkably reduced levels of multiple synapse- and memory-associated proteins. Overexpressing hTau inhibits the activity of protein kinase A (PKA) and decreases the phosphorylation level of cAMP-response element binding protein (CREB), GluA1, and TrkB with reduced BDNF mRNA and protein levels both in vitro and in vivo. Simultaneously, overexpressing hTau increased PKAR2α (an inhibitory subunit of PKA) in nuclear fraction and inactivated proteasome activity. With an increased association of PKAR2α with PA28γ (a nuclear proteasome activator), the formation of PA28γ-20S proteasome complex remarkably decreased in the nuclear fraction, followed by a reduced interaction of PKAR2α with 20S proteasome. Both downregulating PKAR2α by shRNA and upregulating proteasome by expressing PA28γ rescued hTau-induced PKA inhibition and CREB dephosphorylation, and upregulating PKA improved hTau-induced cognitive deficits in mice. Together, these data reveal that intracellular tau accumulation induces synapse and memory impairments by inhibiting PKA/CREB/BDNF/TrkB and PKA/GluA1 signaling, and deficit of PA28γ-20S proteasome complex formation contributes to PKAR2α elevation and PKA inhibition.
Collapse
Affiliation(s)
- Jinwang Ye
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Ministry of Education of China for Neurological Disorders Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yaling Yin
- Department of Physiology and Neurobiology School of Basic Medical Sciences Xinxiang Medical University Xinxiang China
| | - Huanhuan Liu
- School of Pharmacy Xinxiang Medical University Xinxiang China
| | - Lin Fang
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Ministry of Education of China for Neurological Disorders Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiaoqing Tao
- Department of Physiology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Linyu Wei
- Department of Physiology and Neurobiology School of Basic Medical Sciences Xinxiang Medical University Xinxiang China
| | - Yue Zuo
- School of Pharmacy Xinxiang Medical University Xinxiang China
| | - Ying Yin
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Ministry of Education of China for Neurological Disorders Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Dan Ke
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Ministry of Education of China for Neurological Disorders Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jian‐Zhi Wang
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Ministry of Education of China for Neurological Disorders Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Department of Physiology and Neurobiology School of Basic Medical Sciences Xinxiang Medical University Xinxiang China
- Co‐innovation Center of Neurodegeneration Nantong University Nantong China
| |
Collapse
|
125
|
Pickett EK, Herrmann AG, McQueen J, Abt K, Dando O, Tulloch J, Jain P, Dunnett S, Sohrabi S, Fjeldstad MP, Calkin W, Murison L, Jackson RJ, Tzioras M, Stevenson A, d'Orange M, Hooley M, Davies C, Colom-Cadena M, Anton-Fernandez A, King D, Oren I, Rose J, McKenzie CA, Allison E, Smith C, Hardt O, Henstridge CM, Hardingham GE, Spires-Jones TL. Amyloid Beta and Tau Cooperate to Cause Reversible Behavioral and Transcriptional Deficits in a Model of Alzheimer's Disease. Cell Rep 2019; 29:3592-3604.e5. [PMID: 31825838 PMCID: PMC6915767 DOI: 10.1016/j.celrep.2019.11.044] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 08/16/2019] [Accepted: 11/11/2019] [Indexed: 02/08/2023] Open
Abstract
A key knowledge gap blocking development of effective therapeutics for Alzheimer's disease (AD) is the lack of understanding of how amyloid beta (Aβ) peptide and pathological forms of the tau protein cooperate in causing disease phenotypes. Within a mouse tau-deficient background, we probed the molecular, cellular, and behavioral disruption triggered by the influence of wild-type human tau on human Aβ-induced pathology. We find that Aβ and tau work cooperatively to cause a hyperactivity behavioral phenotype and to cause downregulation of transcription of genes involved in synaptic function. In both our mouse model and human postmortem tissue, we observe accumulation of pathological tau in synapses, supporting the potential importance of synaptic tau. Importantly, tau reduction in the mice initiated after behavioral deficits emerge corrects behavioral deficits, reduces synaptic tau levels, and substantially reverses transcriptional perturbations, suggesting that lowering synaptic tau levels may be beneficial in AD.
Collapse
Affiliation(s)
- Eleanor K Pickett
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Abigail G Herrmann
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Jamie McQueen
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Kimberly Abt
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Owen Dando
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Jane Tulloch
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Pooja Jain
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Sophie Dunnett
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Sadaf Sohrabi
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Maria P Fjeldstad
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Will Calkin
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Leo Murison
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Rosemary J Jackson
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Makis Tzioras
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Anna Stevenson
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Marie d'Orange
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Monique Hooley
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Caitlin Davies
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Marti Colom-Cadena
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Alejandro Anton-Fernandez
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Declan King
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Iris Oren
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Jamie Rose
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Chris-Anne McKenzie
- Centre for Clinical Brain Sciences and Sudden Death Brain Bank, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Elizabeth Allison
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences and Sudden Death Brain Bank, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Oliver Hardt
- McGill University Department of Psychology, Montreal QC H3A 1B1, Canada; The University of Edinburgh Simons Initiative for the Developing Brain, George Square, Edinburgh EH8 9JZ, UK
| | - Christopher M Henstridge
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Giles E Hardingham
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK
| | - Tara L Spires-Jones
- The University of Edinburgh Centre for Discovery Brain Sciences, 1 George Square, Edinburgh EH8 9JZ, UK; UK Dementia Research Institute at Edinburgh, George Square, Edinburgh EH8 9JZ, UK.
| |
Collapse
|
126
|
Memory retrieval modulates spatial tuning of single neurons in the human entorhinal cortex. Nat Neurosci 2019; 22:2078-2086. [PMID: 31712776 PMCID: PMC6897360 DOI: 10.1038/s41593-019-0523-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 09/23/2019] [Indexed: 01/18/2023]
Abstract
The medial temporal lobe is critical for both spatial navigation and memory. Although single neurons in the medial temporal lobe activate to represent locations in the environment during navigation, how this spatial tuning relates to memory for events involving those locations remains unclear. We examined memory-related changes in spatial tuning by recording single-neuron activity from neurosurgical patients performing a virtual-reality object-location memory task. We identified 'memory-trace cells' with activity that was spatially tuned to the retrieved location of the specific object that participants were cued to remember. Memory-trace cells in the entorhinal cortex, in particular, encoded discriminable representations of different memories through a memory-specific rate code. These findings indicate that single neurons in the human entorhinal cortex change their spatial tuning to target relevant memories for retrieval.
Collapse
|
127
|
Visual versus Verbal Working Memory in Statistically Determined Patients with Mild Cognitive Impairment: On behalf of the Consortium for Clinical and Epidemiological Neuropsychological Data Analysis (CENDA). J Int Neuropsychol Soc 2019; 25:1001-1010. [PMID: 31543085 DOI: 10.1017/s1355617719000808] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Previous research in mild cognitive impairment (MCI) suggests that visual episodic memory impairment may emerge before analogous verbal episodic memory impairment. The current study examined working memory (WM) test performance in MCI to assess whether patients present with greater visual versus verbal WM impairment. WM performance was also assessed in relation to hippocampal occupancy (HO), a ratio of hippocampal volume to ventricular dilation adjusted for demographic variables and intracranial volume. METHODS Jak et al. (2009) (The American Journal of Geriatric Psychiatry, 17, 368-375) and Edmonds, Delano-Wood, Galasko, Salmon, & Bondi (2015) (Journal of Alzheimer's Disease, 47(1), 231-242) criteria classify patients into four groups: little to no cognitive impairment (non-MCI); subtle cognitive impairment (SCI); amnestic MCI (aMCI); and a combined mixed/dysexecutive MCI (mixed/dys MCI). WM was assessed using co-normed Wechsler Adult Intelligence Scale-IV (WAIS-IV) Digit Span Backwards and Wechsler Memory Scale-IV (WMS-IV) Symbol Span Z-scores. RESULTS Between-group analyses found worse WMS-IV Symbol Span and WAIS-IV Digit Span Backwards performance for mixed/dys MCI compared to non-MCI patients. Within-group analyses found no differences for non-MCI patients; however, all other groups scored lower on WMS-IV Symbol Span than WAIS-IV Digit Span Backwards. Regression analysis with HO as the dependent variable was statistically significant for WMS-IV Symbol Span performance. WAIS-IV Digit Span Backwards performance failed to reach statistical significance. CONCLUSIONS Worse WMS-IV Symbol Span performance was observed in patient groups with measurable neuropsychological impairment and better WMS-IV Symbol Span performance was associated with higher HO ratios. These results suggest that visual WM may be particularly sensitive to emergent illness compared to analogous verbal WM tests.
Collapse
|
128
|
Grøntvedt GR, Schröder TN, Sando SB, White L, Bråthen G, Doeller CF. Alzheimer's disease. Curr Biol 2019; 28:R645-R649. [PMID: 29870699 DOI: 10.1016/j.cub.2018.04.080] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The German psychiatrist and neuropathologist Alois Alzheimer was fascinated by the symptoms of Auguste D., a 50-year-old woman admitted to the Frankfurt Psychiatric Hospital in 1901 who suffered from memory disturbances, paranoia and progressive confusion. After her death and autopsy, Alzheimer described histological alterations in her brain that later came to be known as amyloid plaques and neurofibrillary tangles (Figure 1). The case report was published in a psychiatric textbook some years later, and this peculiar and (at the time) seemingly rare illness was later named Alzheimer's disease.
Collapse
Affiliation(s)
- Gøril Rolfseng Grøntvedt
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Tobias Navarro Schröder
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sigrid Botne Sando
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Linda White
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Geir Bråthen
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Christian F Doeller
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
129
|
Spatial navigation deficits - overlooked cognitive marker for preclinical Alzheimer disease? Nat Rev Neurol 2019; 14:496-506. [PMID: 29980763 DOI: 10.1038/s41582-018-0031-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Detection of incipient Alzheimer disease (AD) pathophysiology is critical to identify preclinical individuals and target potentially disease-modifying therapies towards them. Current neuroimaging and biomarker research is strongly focused in this direction, with the aim of establishing AD fingerprints to identify individuals at high risk of developing this disease. By contrast, cognitive fingerprints for incipient AD are virtually non-existent as diagnostics and outcomes measures are still focused on episodic memory deficits as the gold standard for AD, despite their low sensitivity and specificity for identifying at-risk individuals. This Review highlights a novel feature of cognitive evaluation for incipient AD by focusing on spatial navigation and orientation deficits, which are increasingly shown to be present in at-risk individuals. Importantly, the navigation system in the brain overlaps substantially with the regions affected by AD in both animal models and humans. Notably, spatial navigation has fewer verbal, cultural and educational biases than current cognitive tests and could enable a more uniform, global approach towards cognitive fingerprints of AD and better cognitive treatment outcome measures in future multicentre trials. The current Review appraises the available evidence for spatial navigation and/or orientation deficits in preclinical, prodromal and confirmed AD and identifies research gaps and future research priorities.
Collapse
|
130
|
Abstract
Animal models are indispensable tools for Alzheimer disease (AD) research. Over the course of more than two decades, an increasing number of complementary rodent models has been generated. These models have facilitated testing hypotheses about the aetiology and progression of AD, dissecting the associated pathomechanisms and validating therapeutic interventions, thereby providing guidance for the design of human clinical trials. However, the lack of success in translating rodent data into therapeutic outcomes may challenge the validity of the current models. This Review critically evaluates the genetic and non-genetic strategies used in AD modelling, discussing their strengths and limitations, as well as new opportunities for the development of better models for the disease.
Collapse
|
131
|
Wang Y, Xu X, Wang R. The place cell activity is information-efficient constrained by energy. Neural Netw 2019; 116:110-118. [DOI: 10.1016/j.neunet.2019.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/15/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
|
132
|
Yan T, Ding F, Zhao Y. Integrated identification of key genes and pathways in Alzheimer's disease via comprehensive bioinformatical analyses. Hereditas 2019; 156:25. [PMID: 31346329 PMCID: PMC6636172 DOI: 10.1186/s41065-019-0101-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/09/2019] [Indexed: 12/23/2022] Open
Abstract
Background Alzheimer's disease (AD) is known to be caused by multiple factors, meanwhile the pathogenic mechanism and development of AD associate closely with genetic factors. Existing understanding of the molecular mechanisms underlying AD remains incomplete. Methods Gene expression data (GSE48350) derived from post-modern brain was extracted from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were derived from hippocampus and entorhinal cortex regions between AD patients and healthy controls and detected via Morpheus. Functional enrichment analyses, including Gene Ontology (GO) and pathway analyses of DEGs, were performed via Cytoscape and followed by the construction of protein-protein interaction (PPI) network. Hub proteins were screened using the criteria: nodes degree≥10 (for hippocampus tissues) and ≥ 8 (for entorhinal cortex tissues). Molecular Complex Detection (MCODE) was used to filtrate the important clusters. University of California Santa Cruz (UCSC) and the database of RNA-binding protein specificities (RBPDB) were employed to identify the RNA-binding proteins of the long non-coding RNA (lncRNA). Results 251 & 74 genes were identified as DEGs, which consisted of 56 & 16 up-regulated genes and 195 & 58 down-regulated genes in hippocampus and entorhinal cortex, respectively. Biological analyses demonstrated that the biological processes and pathways related to memory, transmembrane transport, synaptic transmission, neuron survival, drug metabolism, ion homeostasis and signal transduction were enriched in these genes. 11 genes were identified as hub genes in hippocampus and entorhinal cortex, and 3 hub genes were identified as the novel candidates involved in the pathology of AD. Furthermore, 3 lncRNAs were filtrated, whose binding proteins were closely associated with AD. Conclusions Through GO enrichment analyses, pathway analyses and PPI analyses, we showed a comprehensive interpretation of the pathogenesis of AD at a systematic biology level, and 3 novel candidate genes and 3 lncRNAs were identified as novel and potential candidates participating in the pathology of AD. The results of this study could supply integrated insights for understanding the pathogenic mechanism underlying AD and potential novel therapeutic targets.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Bioengineering, Harbin Institute of Technology, Weihai, 264209 Shandong China
| | - Feng Ding
- Department of Bioengineering, Harbin Institute of Technology, Weihai, 264209 Shandong China
| | - Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai, 264209 Shandong China
| |
Collapse
|
133
|
Karakatsani ME, Kugelman T, Ji R, Murillo M, Wang S, Niimi Y, Small SA, Duff KE, Konofagou EE. Unilateral Focused Ultrasound-Induced Blood-Brain Barrier Opening Reduces Phosphorylated Tau from The rTg4510 Mouse Model. Am J Cancer Res 2019; 9:5396-5411. [PMID: 31410223 PMCID: PMC6691580 DOI: 10.7150/thno.28717] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 06/21/2019] [Indexed: 11/21/2022] Open
Abstract
The neuropathological hallmarks of Alzheimer's disease include amyloid plaques and neurofibrillary tangles. Tau pathology correlates well with impaired neuronal activity and dementia. Focused ultrasound coupled with systemic administration of microbubbles has previously been shown to open the blood-brain barrier and induce an immune response, which, in an amyloid AD mouse model, resulted in the reduction of the amyloid brain load. Methods: In this study, we investigated the effect of focused ultrasound at the early stages of tau pathology (pre-tangle) in the rTg4510 mouse model. Results: Reduction of phosphorylated tau from the hippocampal formation processes, and particularly the pyramidal CA1 neurons, was noted in the ultrasound-treated brains without an associated increase in the phosphorylated tau-affected cell somas, typically associated with disease progression. Attenuation of the pathology was found to correlate well with the ultrasound-initiated immune response without compromising neuronal integrity. Unilateral ultrasound application resulted in a bilateral effect indicating a broader reduction of the phosphorylated tau. Conclusion: Findings presented herein reinforce the premise of ultrasound in reducing tau pathology and thus curbing the progression of Alzheimer's disease.
Collapse
|
134
|
Naseri NN, Wang H, Guo J, Sharma M, Luo W. The complexity of tau in Alzheimer's disease. Neurosci Lett 2019; 705:183-194. [PMID: 31028844 PMCID: PMC7060758 DOI: 10.1016/j.neulet.2019.04.022] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is characterized by two major pathological lesions in the brain, amyloid plaques and neurofibrillary tangles (NFTs) composed mainly of amyloid-β (Aβ) peptides and hyperphosphorylated tau, respectively. Although accumulation of toxic Aβ species in the brain has been proposed as one of the important early events in AD, continued lack of success of clinical trials based on Aβ-targeting drugs has triggered the field to seek out alternative disease mechanisms and related therapeutic strategies. One of the new approaches is to uncover novel roles of pathological tau during disease progression. This review will primarily focus on recent advances in understanding the contributions of tau to AD.
Collapse
Affiliation(s)
- Nima N Naseri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| | - Hong Wang
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, USA
| | - Jennifer Guo
- The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
135
|
Frere S, Slutsky I. Alzheimer's Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2019; 97:32-58. [PMID: 29301104 DOI: 10.1016/j.neuron.2017.11.028] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) starts from pure cognitive impairments and gradually progresses into degeneration of specific brain circuits. Although numerous factors initiating AD have been extensively studied, the common principles underlying the transition from cognitive deficits to neuronal loss remain unknown. Here we describe an evolutionarily conserved, integrated homeostatic network (IHN) that enables functional stability of central neural circuits and safeguards from neurodegeneration. We identify the critical modules comprising the IHN and propose a central role of neural firing in controlling the complex homeostatic network at different spatial scales. We hypothesize that firing instability and impaired synaptic plasticity at early AD stages trigger a vicious cycle, leading to dysregulation of the whole IHN. According to this hypothesis, the IHN collapse represents the major driving force of the transition from early memory impairments to neurodegeneration. Understanding the core elements of homeostatic control machinery, the reciprocal connections between distinct IHN modules, and the role of firing homeostasis in this hierarchy has important implications for physiology and should offer novel conceptual approaches for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
136
|
Kunz L, Maidenbaum S, Chen D, Wang L, Jacobs J, Axmacher N. Mesoscopic Neural Representations in Spatial Navigation. Trends Cogn Sci 2019; 23:615-630. [PMID: 31130396 PMCID: PMC6601347 DOI: 10.1016/j.tics.2019.04.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 01/21/2023]
Abstract
Recent evidence suggests that mesoscopic neural oscillations measured via intracranial electroencephalography exhibit spatial representations, which were previously only observed at the micro- and macroscopic level of brain organization. Specifically, theta (and gamma) oscillations correlate with movement, speed, distance, specific locations, and goal proximity to boundaries. In entorhinal cortex (EC), they exhibit hexadirectional modulation, which is putatively linked to grid cell activity. Understanding this mesoscopic neural code is crucial because information represented by oscillatory power and phase may complement the information content at other levels of brain organization. Mesoscopic neural oscillations help bridge the gap between single-neuron and macroscopic brain signals of spatial navigation and may provide a mechanistic basis for novel biomarkers and therapeutic targets to treat diseases causing spatial disorientation.
Collapse
Affiliation(s)
- Lukas Kunz
- Epilepsy Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.
| | - Shachar Maidenbaum
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
| | - Dong Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China
| | - Liang Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| | - Joshua Jacobs
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
| | - Nikolai Axmacher
- Department of Neuropsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
137
|
Howett D, Castegnaro A, Krzywicka K, Hagman J, Marchment D, Henson R, Rio M, King JA, Burgess N, Chan D. Differentiation of mild cognitive impairment using an entorhinal cortex-based test of virtual reality navigation. Brain 2019; 142:1751-1766. [PMID: 31121601 PMCID: PMC6536917 DOI: 10.1093/brain/awz116] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/15/2019] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
The entorhinal cortex is one of the first regions to exhibit neurodegeneration in Alzheimer's disease, and as such identification of entorhinal cortex dysfunction may aid detection of the disease in its earliest stages. Extensive evidence demonstrates that the entorhinal cortex is critically implicated in navigation underpinned by the firing of spatially modulated neurons. This study tested the hypothesis that entorhinal-based navigation is impaired in pre-dementia Alzheimer's disease. Forty-five patients with mild cognitive impairment (26 with CSF Alzheimer's disease biomarker data: 12 biomarker-positive and 14 biomarker-negative) and 41 healthy control participants undertook an immersive virtual reality path integration test, as a measure of entorhinal-based navigation. Behavioural performance was correlated with MRI measures of entorhinal cortex volume, and the classification accuracy of the path integration task was compared with a battery of cognitive tests considered sensitive and specific for early Alzheimer's disease. Biomarker-positive patients exhibited larger errors in the navigation task than biomarker-negative patients, whose performance did not significantly differ from controls participants. Path-integration performance correlated with Alzheimer's disease molecular pathology, with levels of CSF amyloid-β and total tau contributing independently to distance error. Path integration errors were negatively correlated with the volumes of the total entorhinal cortex and of its posteromedial subdivision. The path integration task demonstrated higher diagnostic sensitivity and specificity for differentiating biomarker positive versus negative patients (area under the curve = 0.90) than was achieved by the best of the cognitive tests (area under the curve = 0.57). This study demonstrates that an entorhinal cortex-based virtual reality navigation task can differentiate patients with mild cognitive impairment at low and high risk of developing dementia, with classification accuracy superior to reference cognitive tests considered to be highly sensitive to early Alzheimer's disease. This study provides evidence that navigation tasks may aid early diagnosis of Alzheimer's disease, and the basis of this in animal cellular and behavioural studies provides the opportunity to answer the unmet need for translatable outcome measures for comparing treatment effect across preclinical and clinical trial phases of future anti-Alzheimer's drugs.
Collapse
Affiliation(s)
- David Howett
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andrea Castegnaro
- Institute of Cognitive Neuroscience, University College London, London, UK
- Department of Electrical Engineering, University College London, London, UK
| | - Katarzyna Krzywicka
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Johanna Hagman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Deepti Marchment
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Richard Henson
- MRC Cognition and Brain Sciences Unit, and Department of Psychiatry, University of Cambridge, UK
| | - Miguel Rio
- Department of Electrical Engineering, University College London, London, UK
| | - John A King
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Neil Burgess
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Dennis Chan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
138
|
Bellmund JLS, Gärdenfors P, Moser EI, Doeller CF. Navigating cognition: Spatial codes for human thinking. Science 2019; 362:362/6415/eaat6766. [PMID: 30409861 DOI: 10.1126/science.aat6766] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The hippocampal formation has long been suggested to underlie both memory formation and spatial navigation. We discuss how neural mechanisms identified in spatial navigation research operate across information domains to support a wide spectrum of cognitive functions. In our framework, place and grid cell population codes provide a representational format to map variable dimensions of cognitive spaces. This highly dynamic mapping system enables rapid reorganization of codes through remapping between orthogonal representations across behavioral contexts, yielding a multitude of stable cognitive spaces at different resolutions and hierarchical levels. Action sequences result in trajectories through cognitive space, which can be simulated via sequential coding in the hippocampus. In this way, the spatial representational format of the hippocampal formation has the capacity to support flexible cognition and behavior.
Collapse
Affiliation(s)
- Jacob L S Bellmund
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, The Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, NTNU, Norwegian University of Science and Technology, Trondheim, Norway. .,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands.,Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Peter Gärdenfors
- Department of Philosophy and Cognitive Science, Lund University, Lund, Sweden.,Centre for Artificial Intelligence, University of Technology Sydney, Sydney, Australia
| | - Edvard I Moser
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, The Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christian F Doeller
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, The Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, NTNU, Norwegian University of Science and Technology, Trondheim, Norway. .,Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
139
|
Arzy S, Schacter DL. Self-Agency and Self-Ownership in Cognitive Mapping. Trends Cogn Sci 2019; 23:476-487. [PMID: 31064702 DOI: 10.1016/j.tics.2019.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/13/2019] [Accepted: 04/03/2019] [Indexed: 01/24/2023]
Abstract
The concepts of agency of one's actions and ownership of one's experience have proved useful in relating body representations to bodily consciousness. Here we apply these concepts to cognitive maps. Agency is defined as 'the sense that I am the one who is generating the experience represented on a cognitive map', while ownership is defined as 'the sense that I am the one who is undergoing an experience, represented on a cognitive map'. The roles of agency and ownership are examined with respect to the transformation between egocentric and allocentric representations and the underlying neurocognitive and computational mechanisms; and within the neuropsychiatric domain, including Alzheimer's disease (AD) and other memory-related disorders, in which the senses of agency and ownership may be disrupted.
Collapse
Affiliation(s)
- Shahar Arzy
- Department of Psychology, Harvard University, Cambridge, MA, USA; Neuropsychiatry Laboratory, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel.
| | | |
Collapse
|
140
|
Crouch B, Yeap JM, Pais B, Riedel G, Platt B. Of mice and motion: Behavioural-EEG phenotyping of Alzheimer’s disease mouse models. J Neurosci Methods 2019; 319:89-98. [DOI: 10.1016/j.jneumeth.2018.06.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/14/2018] [Accepted: 06/28/2018] [Indexed: 01/22/2023]
|
141
|
Hammond TR, Marsh SE, Stevens B. Immune Signaling in Neurodegeneration. Immunity 2019; 50:955-974. [PMID: 30995509 PMCID: PMC6822103 DOI: 10.1016/j.immuni.2019.03.016] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-β, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.
Collapse
Affiliation(s)
- Timothy R Hammond
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
142
|
Stimmell AC, Baglietto-Vargas D, Moseley SC, Lapointe V, Thompson LM, LaFerla FM, McNaughton BL, Wilber AA. Impaired Spatial Reorientation in the 3xTg-AD Mouse Model of Alzheimer's Disease. Sci Rep 2019; 9:1311. [PMID: 30718609 PMCID: PMC6361963 DOI: 10.1038/s41598-018-37151-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023] Open
Abstract
In early Alzheimer's disease (AD) spatial navigation is impaired; however, the precise cause of this impairment is unclear. Recent evidence suggests that getting lost is one of the first impairments to emerge in AD. It is possible that getting lost represents a failure to use distal cues to get oriented in space. Therefore, we set out to look for impaired use of distal cues for spatial orientation in a mouse model of amyloidosis (3xTg-AD). To do this, we trained mice to shuttle to the end of a track and back to an enclosed start box to receive a water reward. Then, mice were trained to stop in an unmarked reward zone to receive a brain stimulation reward. The time required to remain in the zone for a reward was increased across training, and the track was positioned in a random start location for each trial. We found that 6-month female, but not 3-month female, 6-month male, or 12-month male, 3xTg-AD mice were impaired. 6-month male and female mice had only intracellular pathology and male mice had less pathology, particularly in the dorsal hippocampus. Thus, AD may cause spatial disorientation as a result of impaired use of landmarks.
Collapse
Affiliation(s)
- Alina C Stimmell
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA.
| | | | - Shawn C Moseley
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Valérie Lapointe
- Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Lauren M Thompson
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Frank M LaFerla
- Neurobiology and Behavior, University of California Irvine, Irvine, California, USA
| | - Bruce L McNaughton
- Neurobiology and Behavior, University of California Irvine, Irvine, California, USA
- Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Aaron A Wilber
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA.
| |
Collapse
|
143
|
Martini F, Rosa SG, Klann IP, Fulco BCW, Carvalho FB, Rahmeier FL, Fernandes MC, Nogueira CW. A multifunctional compound ebselen reverses memory impairment, apoptosis and oxidative stress in a mouse model of sporadic Alzheimer's disease. J Psychiatr Res 2019; 109:107-117. [PMID: 30521994 DOI: 10.1016/j.jpsychires.2018.11.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/24/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023]
Abstract
Alzheimer 's disease (AD) is characterized by progressive cognitive decline including memory impairment, cortical dysfunction, and neuropsychiatric disturbances. The drug discovery to treat AD consists to develop compounds able to act in multiple molecular targets involved in the pathogenesis of the disease and the repositioning of old drugs for new application. This way, the intracerebroventricular (icv) injection of streptozotocin (STZ) has been used as a metabolic model of sporadic AD. The aim of the present study was to investigate whether ebselen (1-10 mg/kg), a multifunctional selenoorganic compound, ameliorates memory impairment, hippocampal oxidative stress, apoptosis and cell proliferation in a mouse model of sporadic AD induced by icv STZ (3 mg/kg, 1 μl/min). The administration of ebselen (10 mg/kg, i.p.) reversed memory impairment and hippocampal oxidative stress, by increasing the activities of antioxidant enzymes and the level of a non-enzymatic antioxidant defense, in Swiss mice administered with icv STZ. The anti-apoptotic property of ebselen was demonstrated by its effectiveness against the increase in the ratios of Bax/Bcl-2, cleaved PARP/PARP and the cleaved caspase-3 levels in the hippocampus of icv STZ mice. Although ebselen reversed memory impairment, it was ineffective against the reduction in the number of BrdU positive cells induced by icv STZ. In conclusion, the multifunctional selenoorganic compound ebselen was effective to reverse memory impairment, hippocampal oxidative stress and apoptosis in a mouse model of sporadic AD induced by icv STZ.
Collapse
Affiliation(s)
- Franciele Martini
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, CEP 97105-900, Santa Maria, Rio Grande do Sul, Brazil
| | - Suzan Gonçalves Rosa
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, CEP 97105-900, Santa Maria, Rio Grande do Sul, Brazil
| | - Isabella Pregardier Klann
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, CEP 97105-900, Santa Maria, Rio Grande do Sul, Brazil
| | - Bruna Cruz Weber Fulco
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, CEP 97105-900, Santa Maria, Rio Grande do Sul, Brazil
| | - Fabiano Barbosa Carvalho
- Laboratório de Patologia da Fundação, Universidade Federal de Ciências da Saúde de Porto Alegre, CEP 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Francine Luciano Rahmeier
- Laboratório de Patologia da Fundação, Universidade Federal de Ciências da Saúde de Porto Alegre, CEP 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marilda Cruz Fernandes
- Laboratório de Patologia da Fundação, Universidade Federal de Ciências da Saúde de Porto Alegre, CEP 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristina Wayne Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, CEP 97105-900, Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
144
|
Zhao S, Zhang L, Yang C, Li Z, Rong S. Procyanidins and Alzheimer’s Disease. Mol Neurobiol 2019; 56:5556-5567. [DOI: 10.1007/s12035-019-1469-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
|
145
|
Presynaptic Pathophysiology Encoded in Different Domains of Tau - Hyper-Versus Hypoexcitability? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:97-103. [PMID: 32096031 DOI: 10.1007/978-981-32-9358-8_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mutations in MAPT (Tau) have been implicated in several types of tauopathy, but the pathways leading to neurodegeneration have remained elusive and are heterogeneous. Here we describe the effects of two mutations, both linked to AD or FTD, that are located in different domains of Tau and show different pathways of toxicity. The deletion mutation ΔK280 lies in the repeat domain and strongly increases β-structure and hence aggregation, whereas the mutation A152T lies in the N-terminal projection domain, has little effect on aggregation but instead on signalling. Both mutations cause presynaptic dysfunction, but in opposite ways, leading to hypoexcitability/hypoactivity vs. hyperexcitability/excitotoxicity, respectively. In organotypic slices these abnormal states can be reversed by drugs, e.g. Tau aggregation inhibitors or modulators of glutamate uptake. This information could contribute to the understanding of "normal" Tau biology and possible therapeutical strategies.
Collapse
|
146
|
Nanowired delivery of cerebrolysin with neprilysin and p-Tau antibodies induces superior neuroprotection in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2019; 245:145-200. [DOI: 10.1016/bs.pbr.2019.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
147
|
Progressive release of mesoporous nano-selenium delivery system for the multi-channel synergistic treatment of Alzheimer's disease. Biomaterials 2018; 197:417-431. [PMID: 30638753 DOI: 10.1016/j.biomaterials.2018.12.027] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/20/2018] [Accepted: 12/23/2018] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with a complex pathogenesis. Controlled release, target ability, and multi-channel synergistic treatment are key factors associated with the success of AD drugs. Herein, we report a novel mesoporous nano-selenium (MSe) release delivery system (MSe-Res/Fc-β-CD/Bor) based on the borneol (Bor) target, β-cyclodextrin nanovalves (Fc-β-CD) with loaded resveratrol (Res). Previous experiments have shown that MSe-Res/Fc-β-CD/Bor first releases Bor by interacting with blood or intracellular esterases, allowing the nanosystem to pass through the blood-brain barrier (BBB). Subsequently, the Fc-β-CD is opened by the redox (H2O2) response to the release of Res at the lesion site. We demonstrated that MSe-Res/Fc-β-CD/Bor inhibited aggregation of β-amyloid proteins (Aβ), mitigated oxidative stress, and suppressed tau hyperphosphorylation, while protecting nerve cells and successfully improving memory impairment in APP/PS1 mice. Interestingly, compared with rivastigmine (Riv) positive drugs alone, the MSe/Fc-β-CD/Bor loaded with Riv had a better pharmacokinetic index. These results indicate that MSe-Res/Fc-β-CD/Bor could be a prospective drug for treating AD.
Collapse
|
148
|
Fu H, Possenti A, Freer R, Nakano Y, Hernandez Villegas NC, Tang M, Cauhy PVM, Lassus BA, Chen S, Fowler SL, Figueroa HY, Huey ED, Johnson GVW, Vendruscolo M, Duff KE. A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Nat Neurosci 2018; 22:47-56. [PMID: 30559469 PMCID: PMC6330709 DOI: 10.1038/s41593-018-0298-7] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/23/2018] [Indexed: 01/04/2023]
Abstract
Excitatory neurons are preferentially impaired in early Alzheimer's disease but the pathways contributing to their relative vulnerability remain largely unknown. Here we report that pathological tau accumulation takes place predominantly in excitatory neurons compared to inhibitory neurons, not only in the entorhinal cortex, a brain region affected in early Alzheimer's disease, but also in areas affected later by the disease. By analyzing RNA transcripts from single-nucleus RNA datasets, we identified a specific tau homeostasis signature of genes differentially expressed in excitatory compared to inhibitory neurons. One of the genes, BCL2-associated athanogene 3 (BAG3), a facilitator of autophagy, was identified as a hub, or master regulator, gene. We verified that reducing BAG3 levels in primary neurons exacerbated pathological tau accumulation, whereas BAG3 overexpression attenuated it. These results define a tau homeostasis signature that underlies the cellular and regional vulnerability of excitatory neurons to tau pathology.
Collapse
Affiliation(s)
- Hongjun Fu
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA.
| | - Andrea Possenti
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Rosie Freer
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Yoshikazu Nakano
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | | | - Maoping Tang
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Paula V M Cauhy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA.,Federal University of Uberlândia, Uberlândia, Brazil
| | - Benjamin A Lassus
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Shuo Chen
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Stephanie L Fowler
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Helen Y Figueroa
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Edward D Huey
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA.,Departments of Psychiatry and Neurology, Columbia University, New York, NY, USA
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Karen E Duff
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
149
|
Progressive impairment of directional and spatially precise trajectories by TgF344-Alzheimer's disease rats in the Morris Water Task. Sci Rep 2018; 8:16153. [PMID: 30385825 PMCID: PMC6212523 DOI: 10.1038/s41598-018-34368-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Spatial navigation is impaired in early stages of Alzheimer’s disease, and may be a defining behavioral marker of preclinical AD. A new rat model (TgF344-AD) of AD overcomes many limitations of other rodent models, though spatial navigation has not been comprehensively assessed. Using the hidden and cued platform variants of the Morris water task, a longitudinal assessment of spatial navigation was conducted on TgF344-AD (n = 16) and Fischer 344 (n = 12) male and female rats at three age ranges: 4 to 5 months, 7 to 8, and 10 to 11 months of age. TgF344-AD rats exhibited largely intact navigation at 4–5 months, with deficits in the hidden platform task emerging at 7–8 months and becoming significantly pronounced at 10–11 months of age. In general, TgF344-AD rats displayed less accurate swim trajectories to the platform and searched a wider area around the platform region compared to wildtype rats. Impaired navigation occurred in the absence of deficits in acquiring the procedural task demands or navigation to the cued platform location. Together, the results indicate that TgF344-AD rats exhibit comparable navigational deficits to those found in individuals with preclinical-AD.
Collapse
|
150
|
Nie B, Wu D, Liang S, Liu H, Sun X, Li P, Huang Q, Zhang T, Feng T, Ye S, Zhang Z, Shan B. A stereotaxic MRI template set of mouse brain with fine sub-anatomical delineations: Application to MEMRI studies of 5XFAD mice. Magn Reson Imaging 2018; 57:83-94. [PMID: 30359719 DOI: 10.1016/j.mri.2018.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/16/2018] [Accepted: 10/18/2018] [Indexed: 01/22/2023]
Abstract
PURPOSE Manganese-enhanced magnetic resonance imaging (MEMRI) can help us trace the active neurons and neuronal pathway in transgenic mouse AD model. 5XFAD has been widespread accepted as a valuable model system for studying brain dysfunction progresses in the courses of AD. To further understand the development of AD at early stages, an effective and objective data analysis platform for MEMRI studies should be constructed. MATERIALS AND METHODS A set of stereotaxic templates of mouse brain in Paxinos and Franklin space, "the Institute of High Energy Physics Mouse Template", or IMT for short, was constructed by iteratively registration and averaging. An atlas image was reconstructed from the Paxinos and Franklin atlas figures and each sub-anatomical segmentation was assigning a unique integer. An analysis SPM plug-in toolbox was further created, that automates and standardizes the time-consuming processes of brain extraction, tissue segmentation, and statistical analysis for MEMRI scans. RESULTS The IMT comprised a T2WI template image, a MEMRI template image, intracranial tissue segmentations, and accompany with a digital mouse brain atlas image, in which 707 sub-anatomical brain regions are delineated. Data analyses were performed on groups of developing 5XFAD mice to demonstrate the usage of IMT, and the results shows that abnormal neuronal activity occurs at early stage in 5XFAD mice. CONCLUSION We have constructed a stereotaxic template set of mouse brain named IMT with fine delineations of sub-anatomical structures, which is compatible with SPM. It will give a widely range of researchers a standardized coordinate system for localization of any mouse brain related data.
Collapse
Affiliation(s)
- Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| | - Di Wu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shengxiang Liang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China; College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Physical Science and Technology College, Zhengzhou University, Zhengzhou 450001, China
| | - Hua Liu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi Sun
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China; Physical Science and Technology College, Zhengzhou University, Zhengzhou 450001, China
| | - Panlong Li
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China; Physical Science and Technology College, Zhengzhou University, Zhengzhou 450001, China
| | - Qi Huang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianhao Zhang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Feng
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China; Physical Science and Technology College, Zhengzhou University, Zhengzhou 450001, China
| | - Songtao Ye
- College of Information Engineering, Xiangtan University, Xiangtan 411105, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Baoci Shan
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China; School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China.
| |
Collapse
|