1
|
Capilla-López MD, Deprada A, Andrade-Talavera Y, Martínez-Gallego I, Coatl-Cuaya H, Sotillo P, Rodríguez-Alvarez J, Rodríguez-Moreno A, Parra-Damas A, Saura CA. Synaptic vulnerability to amyloid-β and tau pathologies differentially disrupts emotional and memory neural circuits. Mol Psychiatry 2025; 30:2966-2979. [PMID: 39885298 PMCID: PMC12185336 DOI: 10.1038/s41380-025-02901-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/22/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is characterized by memory loss and neuropsychiatric symptoms associated with cerebral amyloid-β (Aβ) and tau pathologies, but whether and how these factors differentially disrupt neural circuits remains unclear. Here, we investigated the vulnerability of memory and emotional circuits to Aβ and tau pathologies in mice expressing mutant human amyloid precursor protein (APP), Tau or both APP/Tau in excitatory neurons. APP/Tau mice develop age- and sex-dependent Aβ and phosphorylated tau pathologies, the latter exacerbated at early stages, in vulnerable brain regions. Early memory deficits were associated with hippocampal tau pathology in Tau and APP/Tau mice, whereas anxiety and fear appeared linked to intracellular Aβ in the basolateral amygdala (BLA) of APP and APP/Tau mice. Transcriptome hippocampal profiling revealed gene changes affecting myelination and RNA processing in Tau mice, and inflammation and synaptic-related pathways in APP/Tau mice at 6 months. At 9 months, we detected common and region-specific changes in astrocytic, microglia and 63 AD-associated genes in the hippocampus and BLA of APP/Tau mice. Spatial learning deficits were associated with synaptic tau accumulation and synapse disruption in the hippocampus of Tau and APP/Tau mice, whereas emotional disturbances were linked to Aβ pathology but not synaptic tau in the BLA. Interestingly, Aβ and tau exhibited synergistic detrimental effects in long-term potentiation (LTP) in the hippocampus but they counteract with each other to mitigate LTP impairments in the amygdala. These findings indicate that Aβ and tau pathologies cause region-specific effects and synergize to induce synaptic dysfunction and immune responses, contributing to the differing vulnerability of memory and emotional neural circuits in AD.
Collapse
Grants
- A2022047S BrightFocus Foundation (BrightFocus)
- 2021 SGR00142 Departament d'Innovació, Universitats i Empresa, Generalitat de Catalunya (Department of Innovation, Education and Enterprise, Government of Catalonia)
- GA. 101061171 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 Marie Skłodowska-Curie Actions (H2020 Excellent Science - Marie Skłodowska-Curie Actions)
- Departament d'Innovació, Universitats i Empresa, Generalitat de Catalunya (Department of Innovation, Education and Enterprise, Government of Catalonia)
- Ministerio de Ciencia, Innovación y Universidades from Spain (PID2019-106615RB-I00, PID2022-137668OB-I00 and PDC2021-121350-I00), Generalitat de Catalunya (2021 SGR00142) and BrightFocus Foundation (A2022047S)
- BES-2017-082072 predoctoral fellowship from Ministerio de Ciencia e Innovación (Spain).
- FPU 18/02486 fellowship from Ministerio de Universidades (Spain).
- FPU 17/04283 fellowship from Ministerio de Universidades (Spain).
- Ministerio de Ciencia, Innovación y Universidades (SAF2017-89271-R, PID2020-11751ORB-I00,PID2023-151925OB-I00) and CIBERNED (CB06/05/0042)
- Juan de la Cierva (IJC2019-042468-I) and Ramón y Cajal programs grants from Ministerio de Ciencia, Innovación y Universidades(Spain)
- PREP2022-000069 predoctoral fellowship from Ministerio de Ciencia, Innovación y Universidades (Spain)
Collapse
Affiliation(s)
- Maria Dolores Capilla-López
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Angel Deprada
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Irene Martínez-Gallego
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Sevilla, Spain
| | - Heriberto Coatl-Cuaya
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Sevilla, Spain
| | - Paula Sotillo
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José Rodríguez-Alvarez
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonio Rodríguez-Moreno
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Sevilla, Spain
| | - Arnaldo Parra-Damas
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
2
|
Srivastava I, Goikolea J, Ayberk Kaya T, Latorre-Leal M, Eroli F, Pereira Iglesias M, Álvarez-Jiménez L, Arroyo-García LE, Shimozawa M, Nilsson P, Fisahn A, Lindskog M, Maioli S, Loera-Valencia R. Reactive Astrocytes with Reduced Function of Glutamate Transporters in the AppNL-G-F Knock-in Mice. ACS Chem Neurosci 2025. [PMID: 40421586 DOI: 10.1021/acschemneuro.4c00714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Alzheimer's disease (AD) is associated with synaptic and memory dysfunction. One of the hallmarks of AD is reactive astrogliosis, with reactive astrocytes surrounding amyloid plaques in the brain. Astrocytes have also been shown to be actively involved in disease progression, nevertheless, mechanistic information about their role in synaptic transmission during AD pathology is lacking. Astrocytes maintain synaptic transmission by taking up extracellular glutamate during synaptic activity through astrocytic glutamate transporter GLT-1, but its function has been difficult to measure in real-time in AD pathology. Here, we used an App knock-in AD model (AppNL-G-F) carrying the Swedish, Arctic and Beyreuther mutations associated with AD and exhibiting AD-like Aβ plaque deposition and memory impairment. Using immunohistochemistry, patch-clamp of astrocytes, and Western blot from tissue and FACS isolated synaptosomes, we found that AppNL-G-F mice at 6-8 months of age have astrocytes with clearly altered morphology compared to wild-type (WT). Moreover, astrocyte glutamate clearance function in AppNL-G-F mice, measured as electrophysiological recordings of glutamate transporter currents, was severely impaired compared to WT animals. The reduction of glutamate uptake by astrocytes cannot be explained by GLT-1 protein levels, which were unchanged in synaptosomes and hippocampus of AppNL-G-F mice. Our data suggest that astrocytic glutamate transporters are affected by excess Aβ42 in the brain contributing to synaptic dysfunction in the hippocampus. This data contributes to the notion of restoring astrocyte synaptic function as a potential therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Ipsit Srivastava
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Julen Goikolea
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Tamer Ayberk Kaya
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - María Latorre-Leal
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Francesca Eroli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Marta Pereira Iglesias
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Laura Álvarez-Jiménez
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Luis Enrique Arroyo-García
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Makoto Shimozawa
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - André Fisahn
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Maria Lindskog
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Silvia Maioli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Raúl Loera-Valencia
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Chihuahua, Av. H. Colegio Militar 4700, Nombre de Dios, 31150 Chihuahua, Chih. Mexico
| |
Collapse
|
3
|
Li J, Tanzillo A, Pizzirusso G, Caccavano A, Chittajallu R, Sohn M, Abebe D, Zhang Y, Pelkey K, Dale RK, McBain CJ, Petros TJ. Reducing methylation of histone 3.3 lysine 4 in the medial ganglionic eminence and hypothalamus recapitulates neurodevelopmental disorder phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.02.651761. [PMID: 40568105 PMCID: PMC12190312 DOI: 10.1101/2025.05.02.651761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
Methylation of lysine 4 on histone H3 (H3K4) is enriched on active promoters and enhancers and correlates with gene activation. Disruption of H3K4 methylation is associated with numerous neurodevelopmental diseases (NDDs) that display intellectual disability and abnormal body growth. Here, we perturb H3K4 methylation in the medial ganglionic eminence (MGE) and the hypothalamus, two brain regions associated with these disease phenotypes. These mutant mice have fewer forebrain interneurons, deficient network rhythmogenesis, and increased spontaneous seizures and seizure susceptibility. Mutant mice are significantly smaller than control littermates, but they eventually became obese due to striking changes in the genetic and cellular hypothalamus environment in these mice. Perturbation of H3K4 methylation in these cells produces deficits in numerous NDD-associated behaviors, with a bias for more severe phenotypes in female mice. Single cell sequencing reveals transcriptional changes in the embryonic and adult brain that underlie many of these phenotypes. In sum, our findings highlight the critical role of H3K4 methylation in regulating survival and cell-specific gene regulatory mechanisms in forebrain GABAergic and hypothalamic cells during neurodevelopment to control network excitability and body size homoeostasis.
Collapse
|
4
|
Morrissey ZD, Kumar P, Phan TX, Maienschein-Cline M, Leow A, Lazarov O. Neurogenesis drives hippocampal formation-wide spatial transcription alterations in health and Alzheimer's disease. FRONTIERS IN DEMENTIA 2025; 4:1546433. [PMID: 40309339 PMCID: PMC12041076 DOI: 10.3389/frdem.2025.1546433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025]
Abstract
The mechanism by which neurogenesis regulates the profile of neurons and glia in the hippocampal formation is not known. Further, the effect of neurogenesis on neuronal vulnerability characterizing the entorhinal cortex in Alzheimer's disease (AD) is unknown. Here, we used in situ sequencing to investigate the spatial transcription profile of neurons and glia in the hippocampal circuitry in wild-type mice and in familial AD (FAD) mice expressing varying levels of neurogenesis. This approach revealed that in addition to the dentate gyrus, neurogenesis modulates the cellular profile in the entorhinal cortex and CA regions of the hippocampus. Notably, enhancing neurogenesis in FAD mice led to partial restoration of neuronal and cellular profile in these brain areas, resembling the profile of their wild-type counterparts. This approach provides a platform for the examination of the cellular dynamics in the hippocampal formation in health and in AD.
Collapse
Affiliation(s)
- Zachery D. Morrissey
- Graduate Program in Neuroscience, University of Illinois Chicago, Chicago, IL, United States
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL, United States
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, United States
| | - Pavan Kumar
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, United States
| | - Trongha X. Phan
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, United States
| | | | - Alex Leow
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL, United States
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
Gonzalez MM, Magondu B, Rowan MJM, Forest CR. Regional susceptibility of PV interneurons in an hAPP-KI mouse model of Alzheimer's disease pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646485. [PMID: 40236235 PMCID: PMC11996532 DOI: 10.1101/2025.04.01.646485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Early-stage Alzheimer's pathology correlates with disrupted neuronal excitability, which can drive network and cognitive dysfunction even prior to neurodegeneration. However, the emergence and extent of these changes may vary by brain region and cell types situated in those regions. Here we aimed to investigate the effects of AD pathology on different neuron subtypes in both the entorhinal cortex, a region with enhanced pathology in early AD, and the primary visual cortex, a relatively unaffected region in early-stage AD. We designed and employed a semi-automated patch clamp electrophysiology apparatus to record from fast-spiking parvalbumin interneurons and excitatory neurons in these regions, recording from over 150 cells in young adult APP-KI mice. In entorhinal cortex, amyloid overproduction resulted in PV interneuron hypoexcitability, whereas excitatory neurons were concurrently hyperexcitable. Conversely, neurons of either subclass were largely unaffected in the visual cortex. Together, these findings suggest that fast-spiking parvalbumin interneurons in the entorhinal cortex, but not in the visual cortex, play an integral role in AD progression.
Collapse
|
6
|
Tong J, Gao J, Qi Y, Gao Z, Wang Q, Liu Y, Yuan T, Ren M, Yang G, Li Z, Li J, Sun H, Zhao X, Leung YY, Mu Y, Xu J, Lu C, Peng S, Ge L. GABA AR-PPT1 palmitoylation homeostasis controls synaptic transmission and circuitry oscillation. Transl Psychiatry 2024; 14:488. [PMID: 39695089 DOI: 10.1038/s41398-024-03206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
The infantile neuronal ceroid lipofuscinosis, also called CLN1 disease, is a fatal neurodegenerative disease caused by mutations in the CLN1 gene encoding palmitoyl protein thioesterase 1 (PPT1). Identifying the depalmitoylation substrates of PPT1 is crucial for understanding CLN1 disease. In this study, we found that GABAAR, the critical synaptic protein essential for inhibitory neurotransmission, is a substrate of PPT1. PPT1 depalmitoylates GABAAR α1 subunit at Cystein-260, while binding to Cystein-165 and -179. Mutations of PPT1 or its GABAAR α1 subunit binding site enhanced inhibitory synaptic transmission and strengthened oscillations powers but disrupted phase coupling in CA1 region and impaired learning and memory in 1- to 2-months-old PPT1-deficient and Gabra1em1 mice. Our study highlights the critical role of PPT1 in maintaining GABAAR palmitoylation homeostasis and reveals a previously unknown molecular pathway in CLN1 diseases induced by PPT1 mutations.
Collapse
Affiliation(s)
- Jia Tong
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, He'nan, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jingjing Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yawei Qi
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Ziyan Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Qianqian Wang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Yang Liu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Tiangang Yuan
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Minglong Ren
- Institute of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Guixia Yang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Zhaoyue Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jin Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Hongyuan Sun
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Xing Zhao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Yeung-Yeung Leung
- Division of Brain Sciences, Imperial College Faculty of Medicine, Du Cane Road, London, UK
| | - Yonghui Mu
- Basic Medical College, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jiamin Xu
- Institute of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Chengbiao Lu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
- He'nan International Joint Laboratory for Non-invasive Neural Modulation, Department of Physiology and Pathology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, He'nan, China.
| | - Shiyong Peng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
| | - Lihao Ge
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, He'nan, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
| |
Collapse
|
7
|
Brown EB, Lloyd E, Riley R, Panahidizjikan Z, Martin-Peña A, McFarlane S, Dahanukar A, Keene AC. Aging is associated with a modality-specific decline in taste. iScience 2024; 27:110919. [PMID: 39381735 PMCID: PMC11460507 DOI: 10.1016/j.isci.2024.110919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/27/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
Deficits in chemosensory processing are associated with healthy aging, as well as numerous neurodegenerative disorders, including Alzheimer's disease (AD). The fruit fly, Drosophila melanogaster, is a powerful model for studying chemosensation, aging, and aging-related pathologies, yet the effects of aging and neurodegeneration on taste function remain largely unexplored. Aging impaired response to sugars, but not medium-chain fatty acids that are sensed by a shared population of neurons. Selective expression of the human amyloid beta (Aβ) peptide phenocopied the effects of aging. Functional imaging of gustatory axon terminals revealed reduced response to sugar, but not fatty acids. Axonal innervation of the fly taste center was largely intact in aged flies; however, axonal innervation was reduced upon expression of Aβ. A comparison of transcript expression within the sugar-sensing taste neurons revealed age-related changes in 66 genes. Together, these findings suggest that different mechanisms underly taste deficits in aged and AD model flies.
Collapse
Affiliation(s)
- Elizabeth B. Brown
- Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA
- Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Evan Lloyd
- Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA
- Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Rose Riley
- Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Zohre Panahidizjikan
- Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Alfonso Martin-Peña
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Samuel McFarlane
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Anupama Dahanukar
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
8
|
Adam L, Kumar R, Arroyo‐Garcia LE, Molenkamp WH, Nowak JS, Klute H, Farzadfard A, Alkenayeh R, Nielsen J, Biverstål H, Otzen DE, Johansson J, Abelein A. Specific inhibition of α-synuclein oligomer generation and toxicity by the chaperone domain Bri2 BRICHOS. Protein Sci 2024; 33:e5091. [PMID: 38980078 PMCID: PMC11232276 DOI: 10.1002/pro.5091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 07/10/2024]
Abstract
Protein misfolding and aggregation are involved in several neurodegenerative disorders, such as α-synuclein (αSyn) implicated in Parkinson's disease, where new therapeutic approaches remain essential to combat these devastating diseases. Elucidating the microscopic nucleation mechanisms has opened new opportunities to develop therapeutics against toxic mechanisms and species. Here, we show that naturally occurring molecular chaperones, represented by the anti-amyloid Bri2 BRICHOS domain, can be used to target αSyn-associated nucleation processes and structural species related to neurotoxicity. Our findings revealed that BRICHOS predominantly suppresses the formation of new nucleation units on the fibrils surface (secondary nucleation), decreasing the oligomer generation rate. Further, BRICHOS directly binds to oligomeric αSyn species and effectively diminishes αSyn fibril-related toxicity. Hence, our studies show that molecular chaperones can be utilized as tools to target molecular processes and structural species related to αSyn neurotoxicity and have the potential as protein-based treatments against neurodegenerative disorders.
Collapse
Affiliation(s)
- Laurène Adam
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Rakesh Kumar
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Luis Enrique Arroyo‐Garcia
- Department of Neurobiology, Care Sciences and Society, Division of NeurogeriatricsKarolinska InstitutetSolnaSweden
| | | | - Jan Stanislaw Nowak
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhus CDenmark
| | - Hannah Klute
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Azad Farzadfard
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhus CDenmark
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Rami Alkenayeh
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Janni Nielsen
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhus CDenmark
| | - Henrik Biverstål
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhus CDenmark
| | - Jan Johansson
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Axel Abelein
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| |
Collapse
|
9
|
Pizzirusso G, Preka E, Goikolea J, Aguilar-Ruiz C, Rodriguez-Rodriguez P, Vazquez-Cabrera G, Laterza S, Latorre-Leal M, Eroli F, Blomgren K, Maioli S, Nilsson P, Fragkopoulou A, Fisahn A, Arroyo-García LE. Dynamic microglia alterations associate with hippocampal network impairments: A turning point in amyloid pathology progression. Brain Behav Immun 2024; 119:286-300. [PMID: 38608739 DOI: 10.1016/j.bbi.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/12/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Alzheimer's disease is a progressive neurological disorder causing memory loss and cognitive decline. The underlying causes of cognitive deterioration and neurodegeneration remain unclear, leading to a lack of effective strategies to prevent dementia. Recent evidence highlights the role of neuroinflammation, particularly involving microglia, in Alzheimer's disease onset and progression. Characterizing the initial phase of Alzheimer's disease can lead to the discovery of new biomarkers and therapeutic targets, facilitating timely interventions for effective treatments. We used the AppNL-G-F knock-in mouse model, which resembles the amyloid pathology and neuroinflammatory characteristics of Alzheimer's disease, to investigate the transition from a pre-plaque to an early plaque stage with a combined functional and molecular approach. Our experiments show a progressive decrease in the power of cognition-relevant hippocampal gamma oscillations during the early stage of amyloid pathology, together with a modification of fast-spiking interneuron intrinsic properties and postsynaptic input. Consistently, transcriptomic analyses revealed that these effects are accompanied by changes in synaptic function-associated pathways. Concurrently, homeostasis- and inflammatory-related microglia signature genes were downregulated. Moreover, we found a decrease in Iba1-positive microglia in the hippocampus that correlates with plaque aggregation and neuronal dysfunction. Collectively, these findings support the hypothesis that microglia play a protective role during the early stages of amyloid pathology by preventing plaque aggregation, supporting neuronal homeostasis, and overall preserving the oscillatory network's functionality. These results suggest that the early alteration of microglia dynamics could be a pivotal event in the progression of Alzheimer's disease, potentially triggering plaque deposition, impairment of fast-spiking interneurons, and the breakdown of the oscillatory circuitry in the hippocampus.
Collapse
Affiliation(s)
- Giusy Pizzirusso
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Sweden
| | - Efthalia Preka
- Department of Women's and Children's Health, Karolinska Institutet, Sweden
| | - Julen Goikolea
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Celia Aguilar-Ruiz
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Patricia Rodriguez-Rodriguez
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | | | - Simona Laterza
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Maria Latorre-Leal
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Francesca Eroli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Sweden; Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | | | - André Fisahn
- Department of Women's and Children's Health, Karolinska Institutet, Sweden.
| | - Luis Enrique Arroyo-García
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Sweden.
| |
Collapse
|
10
|
Portal B, Södergren M, Parés i Borrell T, Giraud R, Metzendorf NG, Hultqvist G, Nilsson P, Lindskog M. Early Astrocytic Dysfunction Is Associated with Mistuned Synapses as well as Anxiety and Depressive-Like Behavior in the AppNL-F Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 100:1017-1037. [PMID: 38995780 PMCID: PMC11307019 DOI: 10.3233/jad-231461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 07/14/2024]
Abstract
Background Alzheimer's disease (AD) is the most common neurodegenerative disease. Unfortunately, efficient and affordable treatments are still lacking for this neurodegenerative disorder, it is therefore urgent to identify new pharmacological targets. Astrocytes are playing a crucial role in the tuning of synaptic transmission and several studies have pointed out severe astrocyte reactivity in AD. Reactive astrocytes show altered physiology and function, suggesting they could have a role in the early pathophysiology of AD. Objective We aimed to characterize early synaptic impairments in the AppNL-F knock-in mouse model of AD, especially to understand the contribution of astrocytes to early brain dysfunctions. Methods The AppNL-F mouse model carries two disease-causing mutations inserted in the amyloid precursor protein gene. This strain does not start to develop amyloid-β plaques until 9 months of age. Thanks to electrophysiology, we investigated synaptic function, at both neuronal and astrocytic levels, in 6-month-old animals and correlate the synaptic activity with emotional behavior. Results Electrophysiological recordings in the hippocampus revealed an overall synaptic mistuning at a pre-plaque stage of the pathology, associated to an intact social memory but a stronger depressive-like behavior. Astrocytes displayed a reactive-like morphology and a higher tonic GABA current compared to control mice. Interestingly, we here show that the synaptic impairments in hippocampal slices are partially corrected by a pre-treatment with the monoamine oxidase B blocker deprenyl or the fast-acting antidepressant ketamine (5 mg/kg). Conclusions We propose that reactive astrocytes can induce synaptic mistuning early in AD, before plaques deposition, and that these changes are associated with emotional symptoms.
Collapse
Affiliation(s)
- Benjamin Portal
- Department for Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Moa Södergren
- Department for Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Romain Giraud
- Department for Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Nicole G. Metzendorf
- Department of Pharmacy, Division of Protein Drug Design, Uppsala University, Uppsala, Sweden
| | - Greta Hultqvist
- Department of Pharmacy, Division of Protein Drug Design, Uppsala University, Uppsala, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Maria Lindskog
- Department for Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Miao J, Liu X, Lan J. 40 Hz Electroacupuncture relieves the memory dysfunction of 5xFAD mice by regulating neuronal electrical activity. Brain Res 2023; 1821:148576. [PMID: 37714422 DOI: 10.1016/j.brainres.2023.148576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
In this investigation, we probed the impacts of 40 Hz Electroacupuncture (EA) on the cognitive function and brain activity in 5xFAD mice. Three groups of mice were constituted: the Model group of 5xFAD mice, the Wild Type (WT) group of littermate controls, and the EA group of 5xFAD mice subjected to EA treatment. Behavioral tests were conducted to evaluate memory function and anxiety levels, while the presence of Aβ plaques were detected via immunostaining, and neuronal activity was measured using multichannel recordings. Our results indicated that EA therapy enhanced memory function and anxiety-like behavior in 5xFAD mice, as well as diminishing the abundance and dimensions of Aβ plaques in the hippocampus and mPFC regions. Notably, the suppression of astrocyte activation was observed, which was potentially associated with alterations in gamma oscillation. Furthermore, the synaptic transmission of neurons was amplified, suggesting a possible modulation in neural activity. These findings indicate that 40 Hz EA could influence cognitive performance and potentially affect neuronal activity in 5xFAD mice, while the direct connection between EA and neuronal electrical activity regulation requires further exploration. The potential frequency-specific effects of EA on protective mechanisms in the brain was not addressed in this study and thus presents a direction for future research.
Collapse
Affiliation(s)
- Jifei Miao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital. Shenzhen, China
| | - Xiaoming Liu
- Shenzhen Bao'an Traditional Chinese Medicine Hospital. Shenzhen, China
| | - Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital. Shenzhen, China.
| |
Collapse
|
12
|
Chen KS, Noureldein MH, Rigan DM, Hayes JM, Savelieff MG, Feldman EL. Regional interneuron transcriptional changes reveal pathologic markers of disease progression in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565165. [PMID: 37961679 PMCID: PMC10635060 DOI: 10.1101/2023.11.01.565165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and leading cause of dementia, characterized by neuronal and synapse loss, amyloid-β and tau protein aggregates, and a multifactorial pathology involving neuroinflammation, vascular dysfunction, and disrupted metabolism. Additionally, there is growing evidence of imbalance between neuronal excitation and inhibition in the AD brain secondary to dysfunction of parvalbumin (PV)- and somatostatin (SST)-positive interneurons, which differentially modulate neuronal activity. Importantly, impaired interneuron activity in AD may occur upstream of amyloid-β pathology rendering it a potential therapeutic target. To determine the underlying pathologic processes involved in interneuron dysfunction, we spatially profiled the brain transcriptome of the 5XFAD AD mouse model versus controls, across four brain regions, dentate gyrus, hippocampal CA1 and CA3, and cortex, at early-stage (12 weeks-of-age) and late-stage (30 weeks-of-age) disease. Global comparison of differentially expressed genes (DEGs) followed by enrichment analysis of 5XFAD versus control highlighted various biological pathways related to RNA and protein processing, transport, and clearance in early-stage disease and neurodegeneration pathways at late-stage disease. Early-stage DEGs examination found shared, e.g ., RNA and protein biology, and distinct, e.g ., N-glycan biosynthesis, pathways enriched in PV-versus somatostatin SST-positive interneurons and in excitatory neurons, which expressed neurodegenerative and axon- and synapse-related pathways. At late-stage disease, PV-positive interneurons featured cancer and cancer signaling pathways along with neuronal and synapse pathways, whereas SST-positive interneurons showcased glycan biosynthesis and various infection pathways. Late-state excitatory neurons were primarily characterized by neurodegenerative pathways. These fine-grained transcriptomic profiles for PV- and SST-positive interneurons in a time- and spatial-dependent manner offer new insight into potential AD pathophysiology and therapeutic targets.
Collapse
|
13
|
Naia L, Shimozawa M, Bereczki E, Li X, Liu J, Jiang R, Giraud R, Leal NS, Pinho CM, Berger E, Falk VL, Dentoni G, Ankarcrona M, Nilsson P. Mitochondrial hypermetabolism precedes impaired autophagy and synaptic disorganization in App knock-in Alzheimer mouse models. Mol Psychiatry 2023; 28:3966-3981. [PMID: 37907591 PMCID: PMC10730401 DOI: 10.1038/s41380-023-02289-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 09/05/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
Accumulation of amyloid β-peptide (Aβ) is a driver of Alzheimer's disease (AD). Amyloid precursor protein (App) knock-in mouse models recapitulate AD-associated Aβ pathology, allowing elucidation of downstream effects of Aβ accumulation and their temporal appearance upon disease progression. Here we have investigated the sequential onset of AD-like pathologies in AppNL-F and AppNL-G-F knock-in mice by time-course transcriptome analysis of hippocampus, a region severely affected in AD. Strikingly, energy metabolism emerged as one of the most significantly altered pathways already at an early stage of pathology. Functional experiments in isolated mitochondria from hippocampus of both AppNL-F and AppNL-G-F mice confirmed an upregulation of oxidative phosphorylation driven by the activity of mitochondrial complexes I, IV and V, associated with higher susceptibility to oxidative damage and Ca2+-overload. Upon increasing pathologies, the brain shifts to a state of hypometabolism with reduced abundancy of mitochondria in presynaptic terminals. These late-stage mice also displayed enlarged presynaptic areas associated with abnormal accumulation of synaptic vesicles and autophagosomes, the latter ultimately leading to local autophagy impairment in the synapses. In summary, we report that Aβ-induced pathways in App knock-in mouse models recapitulate key pathologies observed in AD brain, and our data herein adds a comprehensive understanding of the pathologies including dysregulated metabolism and synapses and their timewise appearance to find new therapeutic approaches for AD.
Collapse
Affiliation(s)
- Luana Naia
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Centre for Translational Microbiome Research and National Pandemic Center, Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xidan Li
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Jianping Liu
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Richeng Jiang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Otolaryngology Head and Neck Surgery, The First Hospital of Jilin University, Changchun, China
| | - Romain Giraud
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Nuno Santos Leal
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Catarina Moreira Pinho
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Erik Berger
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Victoria Lim Falk
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Giacomo Dentoni
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Maria Ankarcrona
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Moradi F, van den Berg M, Mirjebreili M, Kosten L, Verhoye M, Amiri M, Keliris GA. Early classification of Alzheimer's disease phenotype based on hippocampal electrophysiology in the TgF344-AD rat model. iScience 2023; 26:107454. [PMID: 37599835 PMCID: PMC10432721 DOI: 10.1016/j.isci.2023.107454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 04/27/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
The hippocampus plays a vital role in navigation, learning, and memory, and is affected in Alzheimer's disease (AD). This study investigated the classification of AD-transgenic rats versus wild-type littermates using electrophysiological activity recorded from the hippocampus at an early, presymptomatic stage of the disease (6 months old) in the TgF344-AD rat model. The recorded signals were filtered into low frequency (LFP) and high frequency (spiking activity) signals, and machine learning classifiers were employed to identify the rat genotype (TG vs. WT). By analyzing specific frequency bands in the low frequency signals and calculating distance metrics between spike trains in the high frequency signals, accurate classification was achieved. Gamma band power emerged as a valuable signal for classification, and combining information from both low and high frequency signals improved the accuracy further. These findings provide valuable insights into the early stage effects of AD on different regions of the hippocampus.
Collapse
Affiliation(s)
- Faraz Moradi
- Faculty of Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Monica van den Berg
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | | | - Lauren Kosten
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Mahmood Amiri
- Medical Technology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Georgios A. Keliris
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Institute of Computer Science, Foundation for Research & Technology - Hellas, Heraklion, Crete, Greece
| |
Collapse
|
15
|
Fabietti M, Mahmud M, Lotfi A, Leparulo A, Fontana R, Vassanelli S, Fasolato C. Early Detection of Alzheimer's Disease From Cortical and Hippocampal Local Field Potentials Using an Ensembled Machine Learning Model. IEEE Trans Neural Syst Rehabil Eng 2023; 31:2839-2848. [PMID: 37347628 DOI: 10.1109/tnsre.2023.3288835] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Early diagnosis of Alzheimer's disease (AD) is a very challenging problem and has been attempted through data-driven methods in recent years. However, considering the inherent complexity in decoding higher cognitive functions from spontaneous neuronal signals, these data-driven methods benefit from the incorporation of multimodal data. This work proposes an ensembled machine learning model with explainability (EXML) to detect subtle patterns in cortical and hippocampal local field potential signals (LFPs) that can be considered as a potential marker for AD in the early stage of the disease. The LFPs acquired from healthy and two types of AD animal models (n = 10 each) using linear multielectrode probes were endorsed by electrocardiogram and respiration signals for their veracity. Feature sets were generated from LFPs in temporal, spatial and spectral domains and were fed into selected machine-learning models for each domain. Using late fusion, the EXML model achieved an overall accuracy of 99.4%. This provided insights into the amyloid plaque deposition process as early as 3 months of the disease onset by identifying the subtle patterns in the network activities. Lastly, the individual and ensemble models were found to be robust when evaluated by randomly masking channels to mimic the presence of artefacts.
Collapse
|
16
|
Andrade-Talavera Y, Fisahn A, Rodríguez-Moreno A. Timing to be precise? An overview of spike timing-dependent plasticity, brain rhythmicity, and glial cells interplay within neuronal circuits. Mol Psychiatry 2023; 28:2177-2188. [PMID: 36991134 PMCID: PMC10611582 DOI: 10.1038/s41380-023-02027-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/31/2023]
Abstract
In the mammalian brain information processing and storage rely on the complex coding and decoding events performed by neuronal networks. These actions are based on the computational ability of neurons and their functional engagement in neuronal assemblies where precise timing of action potential firing is crucial. Neuronal circuits manage a myriad of spatially and temporally overlapping inputs to compute specific outputs that are proposed to underly memory traces formation, sensory perception, and cognitive behaviors. Spike-timing-dependent plasticity (STDP) and electrical brain rhythms are suggested to underlie such functions while the physiological evidence of assembly structures and mechanisms driving both processes continues to be scarce. Here, we review foundational and current evidence on timing precision and cooperative neuronal electrical activity driving STDP and brain rhythms, their interactions, and the emerging role of glial cells in such processes. We also provide an overview of their cognitive correlates and discuss current limitations and controversies, future perspectives on experimental approaches, and their application in humans.
Collapse
Affiliation(s)
- Yuniesky Andrade-Talavera
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, ES-41013, Seville, Spain.
| | - André Fisahn
- Department of Biosciences and Nutrition and Department of Women's and Children's Health, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, ES-41013, Seville, Spain.
| |
Collapse
|
17
|
Wang P, Wang X, Wang Q, Jiao Y, Wang X, Chen C, Chen H, Song T. Cognitive improvement via a modulated rhythmic pulsed magnetic field in D-galactose-induced accelerated aging mice. Brain Res 2023; 1810:148372. [PMID: 37094765 DOI: 10.1016/j.brainres.2023.148372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 04/26/2023]
Abstract
Rhythmic physical stimulations have emerged as effective noninvasive intervention strategies in the treatment of pathological cognitive deficits. Transcranial magnetic stimulation (TMS) can regulate neural firing and improve the learning and memory abilities of rodents or patients with cognitive deterioration. However, the effects of elaborate magnetic stimulation with low intensity during aging or other neurological disordering processes on cognitive decline remain unclear. In this study, we developed an elaborate modulated pulsed magnetic field (PMF) stimulation with a complex pattern in the theta repeated frequency and gamma carrier frequency and then determined the effects of this rhythmic PMF on the cognitive function of accelerated aging mice established by chronic subcutaneous injection of D-galactose (D-gal). The results of the Morris water maze (MWM) test showed that mice treated with modulated PMF displayed shorter swimming distance and latency time in the spatial exploration acquisition trial and exhibited a significant preference in the target presumptive platform area in the probe trial, all of which indicated the enhancement in spatial learning and memory abilities upon PMF stimulation of the accelerated aging mice. The novel object recognition (NOR) test results showed a similar tendency as the MWM results although without statistical significance. Further determination of histological structures demonstrated that the cognitive function-related hippocampal CA3 neurons degenerated upon D-gal injection, which could also be partially rescued by PMF application. In comparison with the high-intensity TMS approach, low-intensity magnetic stimulation could be much safer and allow deeper penetration without adverse effects such as seizure. In summary, modulated PMF, even with low intensity, could effectively improve rodent cognitive functions impaired by D-gal-induced accelerated aging, which might provide a new safe therapeutic strategy for cognitive deficits as well as other neurological disorders.
Collapse
Affiliation(s)
- Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xue Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qingmeng Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Yangkun Jiao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xuting Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
18
|
Kumar R, Arroyo-García LE, Manchanda S, Adam L, Pizzirusso G, Biverstål H, Nilsson P, Fisahn A, Johansson J, Abelein A. Molecular Mechanisms of Amyloid-β Self-Assembly Seeded by In Vivo-Derived Fibrils and Inhibitory Effects of the BRICHOS Chaperone. ACS Chem Neurosci 2023; 14. [PMID: 37023330 PMCID: PMC10119923 DOI: 10.1021/acschemneuro.3c00044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Self-replication of amyloid-β-peptide (Aβ) fibril formation is a hallmark in Alzheimer's disease (AD). Detailed insights have been obtained in Aβ self-assembly in vitro, yet whether similar mechanisms are relevant in vivo has remained elusive. Here, we investigated the ability of in vivo-derived Aβ fibrils from two different amyloid precursor protein knock-in AD mouse models to seed Aβ42 aggregation, where we quantified the microscopic rate constants. We found that the nucleation mechanism of in vivo-derived fibril-seeded Aβ42 aggregation can be described with the same kinetic model as that in vitro. Further, we identified the inhibitory mechanism of the anti-amyloid BRICHOS chaperone on seeded Aβ42 fibrillization, revealing a suppression of secondary nucleation and fibril elongation, which is strikingly similar as observed in vitro. These findings hence provide a molecular understanding of the Aβ42 nucleation process triggered by in vivo-derived Aβ42 propagons, providing a framework for the search for new AD therapeutics.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Luis Enrique Arroyo-García
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Department
of Women’s and Children’s Health, Karolinska Institutet, 171 64 Solna, Sweden
| | - Shaffi Manchanda
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Laurène Adam
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Giusy Pizzirusso
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Department
of Women’s and Children’s Health, Karolinska Institutet, 171 64 Solna, Sweden
| | - Henrik Biverstål
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Per Nilsson
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - André Fisahn
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Jan Johansson
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Axel Abelein
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
19
|
Meftah S, Gan J. Alzheimer's disease as a synaptopathy: Evidence for dysfunction of synapses during disease progression. Front Synaptic Neurosci 2023; 15:1129036. [PMID: 36970154 PMCID: PMC10033629 DOI: 10.3389/fnsyn.2023.1129036] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
The synapse has consistently been considered a vulnerable and critical target within Alzheimer's disease, and synapse loss is, to date, one of the main biological correlates of cognitive decline within Alzheimer's disease. This occurs prior to neuronal loss with ample evidence that synaptic dysfunction precedes this, in support of the idea that synaptic failure is a crucial stage within disease pathogenesis. The two main pathological hallmarks of Alzheimer's disease, abnormal aggregates of amyloid or tau proteins, have had demonstrable effects on synaptic physiology in animal and cellular models of Alzheimer's disease. There is also growing evidence that these two proteins may have a synergistic effect on neurophysiological dysfunction. Here, we review some of the main findings of synaptic alterations in Alzheimer's disease, and what we know from Alzheimer's disease animal and cellular models. First, we briefly summarize some of the human evidence to suggest that synapses are altered, including how this relates to network activity. Subsequently, animal and cellular models of Alzheimer's disease are considered, highlighting mouse models of amyloid and tau pathology and the role these proteins may play in synaptic dysfunction, either in isolation or examining how the two pathologies may interact in dysfunction. This specifically focuses on neurophysiological function and dysfunction observed within these animal models, typically measured using electrophysiology or calcium imaging. Following synaptic dysfunction and loss, it would be impossible to imagine that this would not alter oscillatory activity within the brain. Therefore, this review also discusses how this may underpin some of the aberrant oscillatory patterns seen in animal models of Alzheimer's disease and human patients. Finally, an overview of some key directions and considerations in the field of synaptic dysfunction in Alzheimer's disease is covered. This includes current therapeutics that are targeted specifically at synaptic dysfunction, but also methods that modulate activity to rescue aberrant oscillatory patterns. Other important future avenues of note in this field include the role of non-neuronal cell types such as astrocytes and microglia, and mechanisms of dysfunction independent of amyloid and tau in Alzheimer's disease. The synapse will certainly continue to be an important target within Alzheimer's disease for the foreseeable future.
Collapse
Affiliation(s)
- Soraya Meftah
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jian Gan
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Advances in Molecular Psychiatry - March 2023: mitochondrial function, stress, neuroinflammation - bipolar disorder, psychosis, and Alzheimer's disease. Mol Psychiatry 2023; 28:968-971. [PMID: 36899214 DOI: 10.1038/s41380-023-01968-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 03/12/2023]
|
21
|
Arroyo-García LE, Bachiller S, Ruiz R, Boza-Serrano A, Rodríguez-Moreno A, Deierborg T, Andrade-Talavera Y, Fisahn A. Targeting galectin-3 to counteract spike-phase uncoupling of fast-spiking interneurons to gamma oscillations in Alzheimer's disease. Transl Neurodegener 2023; 12:6. [PMID: 36740709 PMCID: PMC9901156 DOI: 10.1186/s40035-023-00338-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive multifaceted neurodegenerative disorder for which no disease-modifying treatment exists. Neuroinflammation is central to the pathology progression, with evidence suggesting that microglia-released galectin-3 (gal3) plays a pivotal role by amplifying neuroinflammation in AD. However, the possible involvement of gal3 in the disruption of neuronal network oscillations typical of AD remains unknown. METHODS Here, we investigated the functional implications of gal3 signaling on experimentally induced gamma oscillations ex vivo (20-80 Hz) by performing electrophysiological recordings in the hippocampal CA3 area of wild-type (WT) mice and of the 5×FAD mouse model of AD. In addition, the recorded slices from WT mice under acute gal3 application were analyzed with RT-qPCR to detect expression of some neuroinflammation-related genes, and amyloid-β (Aβ) plaque load was quantified by immunostaining in the CA3 area of 6-month-old 5×FAD mice with or without Gal3 knockout (KO). RESULTS Gal3 application decreased gamma oscillation power and rhythmicity in an activity-dependent manner, which was accompanied by impairment of cellular dynamics in fast-spiking interneurons (FSNs) and pyramidal cells. We found that the gal3-induced disruption was mediated by the gal3 carbohydrate-recognition domain and prevented by the gal3 inhibitor TD139, which also prevented Aβ42-induced degradation of gamma oscillations. Furthermore, the 5×FAD mice lacking gal3 (5×FAD-Gal3KO) exhibited WT-like gamma network dynamics and decreased Aβ plaque load. CONCLUSIONS We report for the first time that gal3 impairs neuronal network dynamics by spike-phase uncoupling of FSNs, inducing a network performance collapse. Moreover, our findings suggest gal3 inhibition as a potential therapeutic strategy to counteract the neuronal network instability typical of AD and other neurological disorders encompassing neuroinflammation and cognitive decline.
Collapse
Affiliation(s)
- Luis Enrique Arroyo-García
- grid.465198.7Neuronal Oscillations Laboratory, Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Sara Bachiller
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, BMC B11, 221 84 Lund, Sweden ,grid.9224.d0000 0001 2168 1229Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Rocío Ruiz
- grid.9224.d0000 0001 2168 1229Department of Biochemistry and Molecular Biology, University of Seville, Calle Profesor García González Nº2, 41012 Seville, Spain
| | - Antonio Boza-Serrano
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, BMC B11, 221 84 Lund, Sweden ,grid.9224.d0000 0001 2168 1229Department of Biochemistry and Molecular Biology, University of Seville, Calle Profesor García González Nº2, 41012 Seville, Spain
| | - Antonio Rodríguez-Moreno
- grid.15449.3d0000 0001 2200 2355Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cellular Biology, Universidad Pablo de Olavide, Carretera de Utrera Km-1, 41013 Seville, Spain
| | - Tomas Deierborg
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, BMC B11, 221 84 Lund, Sweden
| | - Yuniesky Andrade-Talavera
- Neuronal Oscillations Laboratory, Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164, Solna, Sweden. .,Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cellular Biology, Universidad Pablo de Olavide, Carretera de Utrera Km-1, 41013, Seville, Spain.
| | - André Fisahn
- Neuronal Oscillations Laboratory, Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164, Solna, Sweden. .,Department of Biosciences and Nutrition, Neo, Karolinska Institutet, 141 83, Huddinge, Sweden.
| |
Collapse
|
22
|
Manchanda S, Galan-Acosta L, Abelein A, Tambaro S, Chen G, Nilsson P, Johansson J. Intravenous treatment with a molecular chaperone designed against β-amyloid toxicity improves Alzheimer's disease pathology in mouse models. Mol Ther 2023; 31:487-502. [PMID: 35982621 PMCID: PMC9931549 DOI: 10.1016/j.ymthe.2022.08.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/12/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Attempts to treat Alzheimer's disease with immunotherapy against the β-amyloid (Aβ) peptide or with enzyme inhibitors to reduce Aβ production have not yet resulted in effective treatment, suggesting that alternative strategies may be useful. Here we explore the possibility of targeting the toxicity associated with Aβ aggregation by using the recombinant human (rh) Bri2 BRICHOS chaperone domain, mutated to act selectively against Aβ42 oligomer generation and neurotoxicity in vitro. We find that treatment of Aβ precursor protein (App) knockin mice with repeated intravenous injections of rh Bri2 BRICHOS R221E, from an age close to the start of development of Alzheimer's disease-like pathology, improves recognition and working memory, as assessed using novel object recognition and Y maze tests, and reduces Aβ plaque deposition and activation of astrocytes and microglia. When treatment was started about 4 months after Alzheimer's disease-like pathology was already established, memory improvement was not detected, but Aβ plaque deposition and gliosis were reduced, and substantially reduced astrocyte accumulation in the vicinity of Aβ plaques was observed. The degrees of treatment effects observed in the App knockin mouse models apparently correlate with the amounts of Bri2 BRICHOS detected in brain sections after the end of the treatment period.
Collapse
Affiliation(s)
- Shaffi Manchanda
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, 141 83 Huddinge, Sweden; Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Lorena Galan-Acosta
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, 141 83 Huddinge, Sweden; Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Axel Abelein
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, 141 83 Huddinge, Sweden; Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Gefei Chen
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, 141 83 Huddinge, Sweden; Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Jan Johansson
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, 141 83 Huddinge, Sweden; Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 64 Stockholm, Sweden.
| |
Collapse
|
23
|
Morrissey ZD, Gao J, Zhan L, Li W, Fortel I, Saido T, Saito T, Bakker A, Mackin S, Ajilore O, Lazarov O, Leow AD. Hippocampal functional connectivity across age in an App knock-in mouse model of Alzheimer's disease. Front Aging Neurosci 2023; 14:1085989. [PMID: 36711209 PMCID: PMC9878347 DOI: 10.3389/fnagi.2022.1085989] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive neurodegenerative disease. The early processes of AD, however, are not fully understood and likely begin years before symptoms manifest. Importantly, disruption of the default mode network, including the hippocampus, has been implicated in AD. METHODS To examine the role of functional network connectivity changes in the early stages of AD, we performed resting-state functional magnetic resonance imaging (rs-fMRI) using a mouse model harboring three familial AD mutations (App NL-G-F/NL-G-F knock-in, APPKI) in female mice in early, middle, and late age groups. The interhemispheric and intrahemispheric functional connectivity (FC) of the hippocampus was modeled across age. RESULTS We observed higher interhemispheric functional connectivity (FC) in the hippocampus across age. This was reduced, however, in APPKI mice in later age. Further, we observed loss of hemispheric asymmetry in FC in APPKI mice. DISCUSSION Together, this suggests that there are early changes in hippocampal FC prior to heavy onset of amyloid β plaques, and which may be clinically relevant as an early biomarker of AD.
Collapse
Affiliation(s)
- Zachery D. Morrissey
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL, United States
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
- Department of Anatomy & Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Jin Gao
- Department of Electrical and Computer Engineering, University of Illinois at Chicago, Chicago, IL, United States
- Preclinical Imaging Core, University of Illinois at Chicago, Chicago, IL, United States
| | - Liang Zhan
- Department of Electrical and Computer Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Weiguo Li
- Preclinical Imaging Core, University of Illinois at Chicago, Chicago, IL, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
- Department of Radiology, Northwestern University, Chicago, IL, United States
| | - Igor Fortel
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya, Japan
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Scott Mackin
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, United States
| | - Olusola Ajilore
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Orly Lazarov
- Department of Anatomy & Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Alex D. Leow
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
- Department of Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
24
|
Yao J, Chen SRW. R-carvedilol, a potential new therapy for Alzheimer's disease. Front Pharmacol 2022; 13:1062495. [PMID: 36532759 PMCID: PMC9756136 DOI: 10.3389/fphar.2022.1062495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
For decades, the amyloid cascade hypothesis has been the leading hypothesis in studying Alzheimer's disease (AD) pathology and drug development. However, a growing body of evidence indicates that simply removing amyloid plaques may not significantly affect AD progression. Alternatively, it has been proposed that AD progression is driven by increased neuronal excitability. Consistent with this alternative hypothesis, recent studies showed that pharmacologically limiting ryanodine receptor 2 (RyR2) open time with the R-carvedilol enantiomer prevented and reversed neuronal hyperactivity, memory impairment, and neuron loss in AD mouse models without affecting the accumulation of ß-amyloid (Aβ). These data indicate that R-carvedilol could be a potential new therapy for AD.
Collapse
Affiliation(s)
- Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Jinjing Yao, ; S. R. Wayne Chen,
| | - S. R. Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Jinjing Yao, ; S. R. Wayne Chen,
| |
Collapse
|
25
|
Ratas I, Pyragas K. Interplay of different synchronization modes and synaptic plasticity in a system of class I neurons. Sci Rep 2022; 12:19631. [PMID: 36385488 PMCID: PMC9668974 DOI: 10.1038/s41598-022-24001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
We analyze the effect of spike-timing-dependent plasticity (STDP) on a system of pulse-coupled class I neurons. Our research begins with a system of two mutually connected quadratic integrate-and-fire (QIF) neurons, which are canonical representatives of class I neurons. Along with various asymptotic modes previously observed in other neuronal models with plastic synapses, we found a stable synchronous mode characterized by unidirectional link from a slower neuron to a faster neuron. In this frequency-locked mode, the faster neuron emits multiple spikes per cycle of the slower neuron. We analytically obtain the Arnold tongues for this mode without STDP and with STDP. We also consider larger plastic networks of QIF neurons and show that the detected mode can manifest itself in such a way that slow neurons become pacemakers. As a result, slow and fast neurons can form large synchronous clusters that generate low-frequency oscillations. We demonstrate the generality of the results obtained with two connected QIF neurons using Wang-Buzsáki and Morris-Lecar biophysically plausible class I neuron models.
Collapse
Affiliation(s)
- Irmantas Ratas
- Center for Physical Sciences and Technology, 10257, Vilnius, Lithuania.
| | - Kestutis Pyragas
- Center for Physical Sciences and Technology, 10257, Vilnius, Lithuania
| |
Collapse
|
26
|
Chauhan K, Khaledi-Nasab A, Neiman AB, Tass PA. Dynamics of phase oscillator networks with synaptic weight and structural plasticity. Sci Rep 2022; 12:15003. [PMID: 36056151 PMCID: PMC9440105 DOI: 10.1038/s41598-022-19417-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/29/2022] [Indexed: 11/08/2022] Open
Abstract
We study the dynamics of Kuramoto oscillator networks with two distinct adaptation processes, one varying the coupling strengths and the other altering the network structure. Such systems model certain networks of oscillatory neurons where the neuronal dynamics, synaptic weights, and network structure interact with and shape each other. We model synaptic weight adaptation with spike-timing-dependent plasticity (STDP) that runs on a longer time scale than neuronal spiking. Structural changes that include addition and elimination of contacts occur at yet a longer time scale than the weight adaptations. First, we study the steady-state dynamics of Kuramoto networks that are bistable and can settle in synchronized or desynchronized states. To compare the impact of adding structural plasticity, we contrast the network with only STDP to one with a combination of STDP and structural plasticity. We show that the inclusion of structural plasticity optimizes the synchronized state of a network by allowing for synchronization with fewer links than a network with STDP alone. With non-identical units in the network, the addition of structural plasticity leads to the emergence of correlations between the oscillators' natural frequencies and node degrees. In the desynchronized regime, the structural plasticity decreases the number of contacts, leading to a sparse network. In this way, adding structural plasticity strengthens both synchronized and desynchronized states of a network. Second, we use desynchronizing coordinated reset stimulation and synchronizing periodic stimulation to induce desynchronized and synchronized states, respectively. Our findings indicate that a network with a combination of STDP and structural plasticity may require stronger and longer stimulation to switch between the states than a network with STDP only.
Collapse
Affiliation(s)
- Kanishk Chauhan
- Department of Physics and Astronomy, Ohio University, Athens, OH, 45701, USA.
- Neuroscience Program, Ohio University, Athens, OH, 45701, USA.
| | - Ali Khaledi-Nasab
- Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA
| | - Alexander B Neiman
- Department of Physics and Astronomy, Ohio University, Athens, OH, 45701, USA
- Neuroscience Program, Ohio University, Athens, OH, 45701, USA
| | - Peter A Tass
- Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
27
|
Synaptic Effects of Palmitoylethanolamide in Neurodegenerative Disorders. Biomolecules 2022; 12:biom12081161. [PMID: 36009055 PMCID: PMC9405819 DOI: 10.3390/biom12081161] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence strongly supports the key role of neuroinflammation in the pathophysiology of neurodegenerative diseases, such as Alzheimer’s disease, frontotemporal dementia, and amyotrophic lateral sclerosis. Neuroinflammation may alter synaptic transmission contributing to the progression of neurodegeneration, as largely documented in animal models and in patients’ studies. In the last few years, palmitoylethanolamide (PEA), an endogenous lipid mediator, and its new composite, which is a formulation constituted of PEA and the well-recognized antioxidant flavonoid luteolin (Lut) subjected to an ultra-micronization process (co-ultraPEALut), has been identified as a potential therapeutic agent in different disorders by exerting potential beneficial effects on neurodegeneration and neuroinflammation by modulating synaptic transmission. In this review, we will show the potential therapeutic effects of PEA in animal models and in patients affected by neurodegenerative disorders.
Collapse
|
28
|
Tok S, Maurin H, Delay C, Crauwels D, Manyakov NV, Van Der Elst W, Moechars D, Drinkenburg WHIM. Pathological and neurophysiological outcomes of seeding human-derived tau pathology in the APP-KI NL-G-F and NL-NL mouse models of Alzheimer's Disease. Acta Neuropathol Commun 2022; 10:92. [PMID: 35739575 PMCID: PMC9219251 DOI: 10.1186/s40478-022-01393-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
The two main histopathological hallmarks that characterize Alzheimer’s Disease are the presence of amyloid plaques and neurofibrillary tangles. One of the current approaches to studying the consequences of amyloid pathology relies on the usage of transgenic animal models that incorporate the mutant humanized form of the amyloid precursor protein (hAPP), with animal models progressively developing amyloid pathology as they age. However, these mice models generally overexpress the hAPP protein to facilitate the development of amyloid pathology, which has been suggested to elicit pathological and neuropathological changes unrelated to amyloid pathology. In this current study, we characterized APP knock-in (APP-KI) animals, that do not overexpress hAPP but still develop amyloid pathology to understand the influence of protein overexpression. We also induced tau pathology via human-derived tau seeding material to understand the neurophysiological effects of amyloid and tau pathology. We report that tau-seeded APP-KI animals progressively develop tau pathology, exacerbated by the presence of amyloid pathology. Interestingly, older amyloid-bearing, tau-seeded animals exhibited more amyloid pathology in the entorhinal area, isocortex and hippocampus, but not thalamus, which appeared to correlate with impairments in gamma oscillations before seeding. Tau-seeded animals also featured immediate deficits in power spectra values and phase-amplitude indices in the hippocampus after seeding, with gamma power spectra deficits persisting in younger animals. Both deficits in hippocampal phase-amplitude coupling and gamma power differentiate tau-seeded, amyloid-positive animals from buffer controls. Based on our results, impairments in gamma oscillations appear to be strongly associated with the presence and development of amyloid and tau pathology, and may also be an indicator of neuropathology, network dysfunction, and even potential disposition to the future development of amyloid pathology.
Collapse
Affiliation(s)
- S Tok
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - H Maurin
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - C Delay
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - D Crauwels
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - N V Manyakov
- Data Sciences, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - W Van Der Elst
- Quantitative Sciences Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - D Moechars
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - W H I M Drinkenburg
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium. .,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
29
|
Olah VJ, Goettemoeller AM, Rayaprolu S, Dammer EB, Seyfried NT, Rangaraju S, Dimidschstein J, Rowan MJM. Biophysical Kv3 channel alterations dampen excitability of cortical PV interneurons and contribute to network hyperexcitability in early Alzheimer's. eLife 2022; 11:75316. [PMID: 35727131 PMCID: PMC9278953 DOI: 10.7554/elife.75316] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
In Alzheimer’s disease (AD), a multitude of genetic risk factors and early biomarkers are known. Nevertheless, the causal factors responsible for initiating cognitive decline in AD remain controversial. Toxic plaques and tangles correlate with progressive neuropathology, yet disruptions in circuit activity emerge before their deposition in AD models and patients. Parvalbumin (PV) interneurons are potential candidates for dysregulating cortical excitability as they display altered action potential (AP) firing before neighboring excitatory neurons in prodromal AD. Here, we report a novel mechanism responsible for PV hypoexcitability in young adult familial AD mice. We found that biophysical modulation of Kv3 channels, but not changes in their mRNA or protein expression, were responsible for dampened excitability in young 5xFAD mice. These K+ conductances could efficiently regulate near-threshold AP firing, resulting in gamma-frequency-specific network hyperexcitability. Thus, biophysical ion channel alterations alone may reshape cortical network activity prior to changes in their expression levels. Our findings demonstrate an opportunity to design a novel class of targeted therapies to ameliorate cortical circuit hyperexcitability in early AD.
Collapse
Affiliation(s)
- Viktor J Olah
- Department of Cell Biology, Emory University, Atlanta, United States
| | | | - Sruti Rayaprolu
- Department of Neurology, Emory University, Atlanta, United States
| | - Eric B Dammer
- Department of Biochemistry, Emory University, Atlanta, United States
| | | | | | | | - Matthew J M Rowan
- Department of Cell Biology, Emory University, Atlanta, United States
| |
Collapse
|
30
|
Mackenzie-Gray Scott CA, Pelkey KA, Caccavano AP, Abebe D, Lai M, Black KN, Brown ND, Trevelyan AJ, McBain CJ. Resilient Hippocampal Gamma Rhythmogenesis and Parvalbumin-Expressing Interneuron Function Before and After Plaque Burden in 5xFAD Alzheimer's Disease Model. Front Synaptic Neurosci 2022; 14:857608. [PMID: 35645763 PMCID: PMC9131009 DOI: 10.3389/fnsyn.2022.857608] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/14/2022] [Indexed: 12/04/2022] Open
Abstract
Recent studies have implicated impaired Parvalbumin Fast-Spiking Interneuron (PVIN) function as a precipitating factor underlying abnormalities in network synchrony, oscillatory rhythms, and cognition associated with Alzheimer's disease (AD). However, a complete developmental investigation of potential gamma deficits, induced by commonly used carbachol or kainate in ex vivo slice preparations, within AD model mice is lacking. We examined gamma oscillations using field recordings in acute hippocampal slices from 5xFAD and control mice, through the period of developing pathology, starting at 3 months of age, when there is minimal plaque presence in the hippocampus, through to 12+ months of age, when plaque burden is high. In addition, we examined PVIN participation in gamma rhythms using targeted cell-attached recordings of genetically-reported PVINs, in both wild type and mutant mice. In parallel, a developmental immunohistochemical characterisation probing the PVIN-associated expression of PV and perineuronal nets (PNNs) was compared between control and 5xFAD mice. Remarkably, this comprehensive longitudinal evaluation failed to reveal any obvious correlations between PVIN deficits (electrical and molecular), circuit rhythmogenesis (gamma frequency and power), and Aβ deposits/plaque formation. By 6-12 months, 5xFAD animals have extensive plaque formation throughout the hippocampus. However, a deficit in gamma oscillatory power was only evident in the oldest 5xFAD animals (12+ months), and only when using kainate, and not carbachol, to induce the oscillations. We found no difference in PV firing or phase preference during kainate-induced oscillations in younger or older 5xFAD mice compared to control, and a reduction of PV and PNNs only in the oldest 5xFAD mice. The lack of a clear relationship between PVIN function, network rhythmicity, and plaque formation in our study highlights an unexpected resilience in PVIN function in the face of extensive plaque pathology associated with this model, calling into question the presumptive link between PVIN pathology and Alzheimer's progression.
Collapse
Affiliation(s)
- Connie A. Mackenzie-Gray Scott
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kenneth A. Pelkey
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Adam P. Caccavano
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel Abebe
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Mandy Lai
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Khayla N. Black
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Nicolette D. Brown
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Andrew J. Trevelyan
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chris J. McBain
- Section on Cellular and Synaptic Physiology, NICHD - Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
31
|
Theta and gamma oscillatory dynamics in mouse models of Alzheimer's disease: A path to prospective therapeutic intervention. Neurosci Biobehav Rev 2022; 136:104628. [PMID: 35331816 DOI: 10.1016/j.neubiorev.2022.104628] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/26/2022]
Abstract
Understanding the neural basis of cognitive deficits, a key feature of Alzheimer's disease (AD), is imperative for achieving the therapy of the disease. Rhythmic oscillatory activities in neural systems are a fundamental mechanism for diverse brain functions, including cognition. In several neurological conditions like AD, aberrant neural oscillations have been shown to play a central role. Furthermore, manipulation of brain oscillations in animals has confirmed their impact on cognition and disease. In this article, we review the evidence from mouse models that shows how synchronized oscillatory activity is intricately linked to AD machinery. We primarily focus on recent reports showing abnormal oscillatory activities at theta and gamma frequencies in AD condition and their influence on cellular disturbances and cognitive impairments. A thorough comprehension of the role that neuronal oscillations play in AD pathology should pave the way to therapeutic interventions that can curb the disease.
Collapse
|
32
|
Emre C, Arroyo-García LE, Do KV, Jun B, Ohshima M, Alcalde SG, Cothern ML, Maioli S, Nilsson P, Hjorth E, Fisahn A, Bazan NG, Schultzberg M. Intranasal delivery of pro-resolving lipid mediators rescues memory and gamma oscillation impairment in App NL-G-F/NL-G-F mice. Commun Biol 2022; 5:245. [PMID: 35314851 PMCID: PMC8938447 DOI: 10.1038/s42003-022-03169-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
Sustained microglial activation and increased pro-inflammatory signalling cause chronic inflammation and neuronal damage in Alzheimer’s disease (AD). Resolution of inflammation follows neutralization of pathogens and is a response to limit damage and promote healing, mediated by pro-resolving lipid mediators (LMs). Since resolution is impaired in AD brains, we decided to test if intranasal administration of pro-resolving LMs in the AppNL-G-F/NL-G-F mouse model for AD could resolve inflammation and ameliorate pathology in the brain. A mixture of the pro-resolving LMs resolvin (Rv) E1, RvD1, RvD2, maresin 1 (MaR1) and neuroprotectin D1 (NPD1) was administered to stimulate their respective receptors. We examined amyloid load, cognition, neuronal network oscillations, glial activation and inflammatory factors. The treatment ameliorated memory deficits accompanied by a restoration of gamma oscillation deficits, together with a dramatic decrease in microglial activation. These findings open potential avenues for therapeutic exploration of pro-resolving LMs in AD, using a non-invasive route. Intranasal administration of pro-resolving lipid mediators improves memory deficits and reduce microglial activation in a mouse model for Alzheimer’s disease, suggesting a future therapy.
Collapse
Affiliation(s)
- Ceren Emre
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.
| | - Luis E Arroyo-García
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Khanh V Do
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA.,Faculty of Medicine, PHENIKAA University, Hanoi, 12116, Vietnam.,PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi, 11313, Vietnam
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Makiko Ohshima
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Silvia Gómez Alcalde
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Megan L Cothern
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Erik Hjorth
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - André Fisahn
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Marianne Schultzberg
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Cope ZA, Murai T, Sukoff Rizzo SJ. Emerging Electroencephalographic Biomarkers to Improve Preclinical to Clinical Translation in Alzheimer's Disease. Front Aging Neurosci 2022; 14:805063. [PMID: 35250541 PMCID: PMC8891809 DOI: 10.3389/fnagi.2022.805063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
Abstract
Continually emerging data indicate that sub-clinical, non-convulsive epileptiform activity is not only prevalent in Alzheimer's disease (AD) but is detectable early in the course of the disease and predicts cognitive decline in both humans and animal models. Epileptiform activity and other electroencephalographic (EEG) measures may hold powerful, untapped potential to improve the translational validity of AD-related biomarkers in model animals ranging from mice, to rats, and non-human primates. In this review, we will focus on studies of epileptiform activity, EEG slowing, and theta-gamma coupling in preclinical models, with particular focus on its role in cognitive decline and relevance to AD. Here, each biomarker is described in the context of the contemporary literature and recent findings in AD relevant animal models are discussed.
Collapse
Affiliation(s)
| | | | - Stacey J. Sukoff Rizzo
- Aging Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
34
|
Klemz A, Wildner F, Tütüncü E, Gerevich Z. Regulation of Hippocampal Gamma Oscillations by Modulation of Intrinsic Neuronal Excitability. Front Neural Circuits 2022; 15:778022. [PMID: 35177966 PMCID: PMC8845518 DOI: 10.3389/fncir.2021.778022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Ion channels activated around the subthreshold membrane potential determine the likelihood of neuronal firing in response to synaptic inputs, a process described as intrinsic neuronal excitability. Long-term plasticity of chemical synaptic transmission is traditionally considered the main cellular mechanism of information storage in the brain; however, voltage- and calcium-activated channels modulating the inputs or outputs of neurons are also subjects of plastic changes and play a major role in learning and memory formation. Gamma oscillations are associated with numerous higher cognitive functions such as learning and memory, but our knowledge of their dependence on intrinsic plasticity is by far limited. Here we investigated the roles of potassium and calcium channels activated at near subthreshold membrane potentials in cholinergically induced persistent gamma oscillations measured in the CA3 area of rat hippocampal slices. Among potassium channels, which are responsible for the afterhyperpolarization in CA3 pyramidal cells, we found that blockers of SK (KCa2) and KV7.2/7.3 (KCNQ2/3), but not the BK (KCa1.1) and IK (KCa3.1) channels, increased the power of gamma oscillations. On the contrary, activators of these channels had an attenuating effect without affecting the frequency. Pharmacological blockade of the low voltage-activated T-type calcium channels (CaV3.1–3.3) reduced gamma power and increased the oscillation peak frequency. Enhancement of these channels also inhibited the peak power without altering the frequency of the oscillations. The presented data suggest that voltage- and calcium-activated ion channels involved in intrinsic excitability strongly regulate the power of hippocampal gamma oscillations. Targeting these channels could represent a valuable pharmacological strategy against cognitive impairment.
Collapse
|
35
|
Mu L, Cai J, Gu B, Yu L, Li C, Liu QS, Zhao L. Treadmill Exercise Prevents Decline in Spatial Learning and Memory in 3×Tg-AD Mice through Enhancement of Structural Synaptic Plasticity of the Hippocampus and Prefrontal Cortex. Cells 2022; 11:244. [PMID: 35053360 PMCID: PMC8774241 DOI: 10.3390/cells11020244] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 01/08/2022] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by deficits in learning and memory. A pathological feature of AD is the alterations in the number and size of synapses, axon length, dendritic complexity, and dendritic spine numbers in the hippocampus and prefrontal cortex. Treadmill exercise can enhance synaptic plasticity in mouse or rat models of stroke, ischemia, and dementia. The aim of this study was to examine the effects of treadmill exercise on learning and memory, and structural synaptic plasticity in 3×Tg-AD mice, a mouse model of AD. Here, we show that 12 weeks treadmill exercise beginning in three-month-old mice improves spatial working memory in six-month-old 3×Tg-AD mice, while non-exercise six-month-old 3×Tg-AD mice exhibited impaired spatial working memory. To investigate potential mechanisms for the treadmill exercise-induced improvement of spatial learning and memory, we examined structural synaptic plasticity in the hippocampus and prefrontal cortex of six-month-old 3×Tg-AD mice that had undergone 12 weeks of treadmill exercise. We found that treadmill exercise led to increases in synapse numbers, synaptic structural parameters, the expression of synaptophysin (Syn, a presynaptic marker), the axon length, dendritic complexity, and the number of dendritic spines in 3×Tg-AD mice and restored these parameters to similar levels of non-Tg control mice without treadmill exercise. In addition, treadmill exercise also improved these parameters in non-Tg control mice. Strengthening structural synaptic plasticity may represent a potential mechanism by which treadmill exercise prevents decline in spatial learning and memory and synapse loss in 3×Tg-AD mice.
Collapse
Affiliation(s)
- Lianwei Mu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Jiajia Cai
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Boya Gu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Laikang Yu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Cui Li
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
- School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou 450001, China
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| |
Collapse
|
36
|
Licinio J, Wong ML. Molecular Psychiatry special issue: advances in Alzheimer's disease. Mol Psychiatry 2021; 26:5467-5470. [PMID: 35027660 DOI: 10.1038/s41380-021-01434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Julio Licinio
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Ma-Li Wong
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| |
Collapse
|