1
|
Jiang M, Li Q, Chen J, Li R, Yao J, Hu Y, Zhang H, Cai L, Luo M, Sun Y, Zeng W. Microglial MS4A4A Protects against Epileptic Seizures in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417733. [PMID: 40349168 DOI: 10.1002/advs.202417733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/16/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is a predominant neurodegenerative disorder worldwide, with epileptic seizures being a common comorbidity that can exacerbate cognitive deterioration in affected individuals, thus highlighting the importance of early therapeutic intervention. It is determined that deletion of Ms4a4a, an AD-associated gene, exacerbates seizures in amyloid β (Aβ)-driven AD mouse model. MS4A4A is significantly upregulated in brain lesions in patients with epilepsy. Single-cell sequencing reveals that MS4A4A is highly expressed in microglia within these lesions, linked to enhanced phagocytic activity. Mechanistic investigation delineates that deletion of Ms4a4a impairs microglial phagocytosis, accompanied by diminished calcium influx and disruptions in mitochondrial metabolic fitness. The cytosolic fragment of Ms4a4a is anchored to the cytoskeletal components, supporting its critical role in mediating phagocytosis. Induction of Ms4a4a through central delivery of LNP-Il4 alleviates seizure conditions. Collectively, these findings identify Ms4a4a as a potential therapeutic target for managing seizures in AD treatment.
Collapse
Affiliation(s)
- Meng Jiang
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
| | - Qingqing Li
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
| | - Jianhui Chen
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
| | - Ruochong Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jun Yao
- ENO Bio mRNA Innovation Institute, Shenzhen Rhegen Biotechnology Co. Ltd, Shenzhen, 518000, China
| | - Yong Hu
- ENO Bio mRNA Innovation Institute, Shenzhen Rhegen Biotechnology Co. Ltd, Shenzhen, 518000, China
| | - Haizheng Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lixin Cai
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, 100034, China
| | - Maoguo Luo
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yu Sun
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, 100034, China
| | - Wenwen Zeng
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, 030001, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| |
Collapse
|
2
|
Orozco Valero A, Rodríguez-González V, Montobbio N, Casal MA, Tlaie A, Pelayo F, Morillas C, Poza J, Gómez C, Martínez-Cañada P. A Python toolbox for neural circuit parameter inference. NPJ Syst Biol Appl 2025; 11:45. [PMID: 40346107 PMCID: PMC12064716 DOI: 10.1038/s41540-025-00527-9] [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] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
Computational research tools have reached a level of maturity that enables efficient simulation of neural activity across diverse scales. Concurrently, experimental neuroscience is experiencing an unprecedented scale of data generation. Despite these advancements, our understanding of the precise mechanistic relationship between neural recordings and key aspects of neural activity remains insufficient, including which specific features of electrophysiological population dynamics (i.e., putative biomarkers) best reflect properties of the underlying microcircuit configuration. We present ncpi, an open-source Python toolbox that serves as an all-in-one solution, effectively integrating well-established methods for both forward and inverse modeling of extracellular signals based on single-neuron network model simulations. Our tool serves as a benchmarking resource for model-driven interpretation of electrophysiological data and the evaluation of candidate biomarkers that plausibly index changes in neural circuit parameters. Using mouse LFP data and human EEG recordings, we demonstrate the potential of ncpi to uncover imbalances in neural circuit parameters during brain development and in Alzheimer's Disease.
Collapse
Affiliation(s)
- Alejandro Orozco Valero
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain
| | - Víctor Rodríguez-González
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valladolid, Spain
| | - Noemi Montobbio
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Miguel A Casal
- Research Center for Information and Communication Technologies (CITIC), University of A Coruña, A Coruña, Spain
| | - Alejandro Tlaie
- Ernst Strüngmann Institute for Neuroscience, Frankfurt am Main, Germany
| | - Francisco Pelayo
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain
| | - Christian Morillas
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain
| | - Jesús Poza
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valladolid, Spain
- IMUVA, Instituto de Investigación en Matemáticas, University of Valladolid, Valladolid, Spain
| | - Carlos Gómez
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valladolid, Spain
| | - Pablo Martínez-Cañada
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain.
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain.
| |
Collapse
|
3
|
Papanikolaou A, Graykowski D, Lee BI, Yang M, Ellingford R, Zünkler J, Bond SA, Rowland JM, Rajani RM, Harris SS, Sharp DJ, Busche MA. Selectively vulnerable deep cortical layer 5/6 fast-spiking interneurons in Alzheimer's disease models in vivo. Neuron 2025:S0896-6273(25)00293-4. [PMID: 40345184 DOI: 10.1016/j.neuron.2025.04.010] [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: 09/26/2024] [Revised: 03/03/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Alzheimer's disease (AD) is initiated by amyloid-beta (Aβ) accumulation in the neocortex; however, the cortical layers and neuronal cell types first susceptible to Aβ remain unknown. Using in vivo two-photon Ca2+ imaging in the visual cortex of AD mouse models, we found that cortical layer 5 neurons displayed abnormally prolonged Ca2+ transients before substantial plaque formation. Neuropixels recordings revealed that these abnormal transients were associated with reduced spiking and impaired visual tuning of parvalbumin (PV)-positive fast-spiking interneurons (FSIs) in layers 5/6, whereas PV-FSIs in superficial layers remained unaffected. These dysfunctions occurred alongside a deep-layer-specific reduction in neuronal pentraxin 2 (NPTX2) within excitatory neurons, decreased GluA4 in PV-FSIs, and fewer excitatory synapses onto PV-FSIs. Notably, NPTX2 overexpression increased excitatory input onto layers 5/6 PV-FSIs and rectified their spiking activity. Thus, our findings reveal an early selective impairment of deep cortical layers 5/6 in AD models and identify deep-layer PV-FSIs as therapeutic targets.
Collapse
Affiliation(s)
| | - David Graykowski
- UK Dementia Research Institute at University College London, London, UK
| | - Byung Il Lee
- UK Dementia Research Institute at University College London, London, UK
| | - Mengke Yang
- UK Dementia Research Institute at University College London, London, UK
| | - Robert Ellingford
- UK Dementia Research Institute at University College London, London, UK
| | - Jana Zünkler
- UK Dementia Research Institute at University College London, London, UK
| | - Suraya A Bond
- UK Dementia Research Institute at University College London, London, UK
| | - James M Rowland
- UK Dementia Research Institute at University College London, London, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at University College London, London, UK; British Heart Foundation - UK Dementia Research Institute Centre for Vascular Dementia Research at The University of Edinburgh, Edinburgh, UK
| | - Samuel S Harris
- UK Dementia Research Institute at University College London, London, UK
| | - David J Sharp
- UK Dementia Research Institute Care Research & Technology Centre and Department of Brain Sciences, Imperial College London, London, UK
| | - Marc Aurel Busche
- UK Dementia Research Institute at University College London, London, UK; Department of Neurodegenerative Diseases, University Hospital of Geriatric Medicine FELIX PLATTER and University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Yuan J, Huang R, Nao J, Dong X. The role of semaphorin 3A in the pathogenesis and progression of Alzheimer's disease and other aging-related diseases: A comprehensive review. Pharmacol Res 2025; 215:107732. [PMID: 40222695 DOI: 10.1016/j.phrs.2025.107732] [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: 02/01/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Aging serves as a pivotal factor in the etiology of numerous diseases, such as Alzheimer's disease (AD), Parkinson's disease, diabetes, osteoarthritis, atherosclerosis and aging-related macular degeneration. Notably, these diseases often interact with AD through various pathways, facilitating the onset or progression of one another. Semaphorin 3 A (Sema3A), a protein that is essential for axonal guidance during neural development, has recently been identified as a novel regulator in the pathogenesis and progression of multiple aging-related diseases. This article provides a comprehensive review of the expression patterns and mechanisms of action of Sema3A in these diseases. Specifically, Sema3A influences the occurrence and development of aging-related diseases by participating in oxidative stress, inflammatory responses, apoptosis, and synaptic plasticity. Therefore, therapeutic strategies targeting Sema3A present promising avenues for delaying the progression of aging-related diseases and offer novel insights and strategies for their treatment.
Collapse
Affiliation(s)
- Jiayu Yuan
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| |
Collapse
|
5
|
Fischer L, Adams JN, Molloy EN, Vockert N, Tremblay-Mercier J, Remz J, Pichet Binette A, Villeneuve S, Maass A. Differential effects of aging, Alzheimer's pathology, and APOE4 on longitudinal functional connectivity and episodic memory in older adults. Alzheimers Res Ther 2025; 17:91. [PMID: 40281595 PMCID: PMC12023467 DOI: 10.1186/s13195-025-01742-6] [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: 12/30/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Both aging and Alzheimer's disease (AD) affect brain networks, with early disruptions occurring in regions involved in episodic memory. Few studies have, however, focused on distinguishing region-specific effects of AD-biomarker negative "normal" aging and early amyloid- and tau pathology on functional connectivity. Further, longitudinal studies combining imaging, biomarkers, and cognition are rare. METHODS We assessed resting-state functional connectivity (rsFC) strength and graph measures in the episodic memory network including the medial temporal lobe (MTL), posteromedial cortex (PMC), and medial prefrontal cortex alongside cognition over two years. For this preregistered study, we included 100 older adults who were amyloid- and tau-negative using CSF and PET measurements to investigate "normal" aging, and 70 older adults who had longitudinal CSF data available to investigate functional changes related to early AD pathology. All participants were cognitively unimpaired older adults from the PREVENT-AD cohort. We used region of interest (ROI)-to-ROI bivariate correlations, graph analysis, and multiple regression models. RESULTS In the amyloid- and tau-negative sample, rsFC strength within PMC, between parahippocampal cortex and inferomedial precuneus, and between posterior hippocampus and inferomedial precuneus decreased over time. Additionally, we observed a longitudinal decrease in global efficiency. Further, there was a steeper longitudinal decrease in rsFC and global efficiency with higher baseline age particularly of parahippocampal-gyrus regions. Further, lower rsFC strength within PMC was associated with poorer longitudinal episodic memory performance. In the sample with available CSF data, a steeper increase in rsFC between anterior hippocampus and superior precuneus was related to higher baseline AD pathology. Higher MTL-PMC rsFC strength was differentially associated with episodic memory trajectories depending on APOE4 genotype. CONCLUSIONS Our findings suggest differential effects of aging and AD pathology. Hypoconnectivity within PMC was related to aging and cognitive decline. MTL-PMC hyperconnectivity was related to early AD pathology and cognitive decline in APOE4 carriers. Future studies should investigate more diverse samples, nonetheless, our approach allowed us to identify longitudinal functional changes related to aging and early AD pathology, enhancing cross-sectional research. Hyperconnectivity has been proposed as a mechanism related to early AD pathology before, we now contribute specific functional connections to focus on in future research.
Collapse
Affiliation(s)
- Larissa Fischer
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
- Department of Neurobiology and Behavior, University of California, Irvine, USA.
| | - Jenna N Adams
- Department of Neurobiology and Behavior, University of California, Irvine, USA
| | - Eóin N Molloy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto Von Guericke University, Magdeburg, Germany
| | - Niklas Vockert
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Jennifer Tremblay-Mercier
- StoP-AD Centre, Douglas Mental Health Institute Research Centre, McGill University, Montréal, Canada
| | - Jordana Remz
- StoP-AD Centre, Douglas Mental Health Institute Research Centre, McGill University, Montréal, Canada
| | - Alexa Pichet Binette
- Department of Physiology and Pharmacology, Université de Montréal, Montréal, Canada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, Canada
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Sylvia Villeneuve
- StoP-AD Centre, Douglas Mental Health Institute Research Centre, McGill University, Montréal, Canada
- Department of Psychiatry, McGill University, Montréal, Canada
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
- Institute for Biology, Otto Von Guericke University, Magdeburg, Germany.
| |
Collapse
|
6
|
Harris SS, Ellingford R, Hartmann J, Dasgupta D, Kehring M, Rajani RM, Graykowski D, Quittot N, Sivasankaran D, Commins C, Fan Z, Bond SA, Wolf F, Dupret D, Dolan RJ, Konnerth A, Neef A, Hyman BT, Busche MA. Alzheimer's disease patient-derived high-molecular-weight tau impairs bursting in hippocampal neurons. Cell 2025:S0092-8674(25)00408-8. [PMID: 40300603 DOI: 10.1016/j.cell.2025.04.006] [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: 10/17/2024] [Revised: 02/21/2025] [Accepted: 04/03/2025] [Indexed: 05/01/2025]
Abstract
Tau accumulation is closely related to cognitive symptoms in Alzheimer's disease (AD). However, the cellular drivers of tau-dependent decline of memory-based cognition remain elusive. Here, we employed in vivo Neuropixels and patch-clamp recordings in mouse models and demonstrate that tau, independent of β-amyloid, selectively debilitates complex-spike burst firing of CA1 hippocampal neurons, a fundamental cellular mechanism underpinning learning and memory. Impaired bursting was associated with altered hippocampal network activities that are coupled to burst firing patterns (i.e., theta rhythms and high-frequency ripples) and was concurrent with reduced neuronal expression of CaV2.3 calcium channels, which are essential for burst firing in vivo. We subsequently identify soluble high molecular weight (HMW) tau, isolated from human AD brain, as the tau species responsible for suppression of burst firing. These data provide a cellular mechanism for tau-dependent cognitive decline in AD and implicate a rare species of intracellular HMW tau as a therapeutic target.
Collapse
Affiliation(s)
- Samuel S Harris
- UK Dementia Research Institute at University College London, London, UK.
| | - Robert Ellingford
- UK Dementia Research Institute at University College London, London, UK
| | - Jana Hartmann
- UK Dementia Research Institute at University College London, London, UK
| | - Debanjan Dasgupta
- UK Dementia Research Institute at University College London, London, UK
| | - Marten Kehring
- UK Dementia Research Institute at University College London, London, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at University College London, London, UK; British Heart Foundation - UK Dementia Research Institute Centre for Vascular Dementia Research at The University of Edinburgh, Edinburgh, UK
| | - David Graykowski
- UK Dementia Research Institute at University College London, London, UK
| | - Noé Quittot
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Dhanush Sivasankaran
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zhanyun Fan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Suraya A Bond
- UK Dementia Research Institute at University College London, London, UK
| | - Fred Wolf
- Göttingen Campus Institute for Dynamics of Biological Networks, Göttingen, Germany
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Raymond J Dolan
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, London, UK
| | - Arthur Konnerth
- Institute of Neuroscience, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Andreas Neef
- Göttingen Campus Institute for Dynamics of Biological Networks, Göttingen, Germany
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Marc Aurel Busche
- UK Dementia Research Institute at University College London, London, UK; Department of Neurodegenerative Diseases, University Hospital of Geriatric Medicine FELIX PLATTER and University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
7
|
Décarie-Labbé L, Mellah S, Dialahy IZ, Belleville S. Predicting cognitive change using functional, structural, and neuropsychological predictors. Brain Commun 2025; 7:fcaf155. [PMID: 40337465 PMCID: PMC12056721 DOI: 10.1093/braincomms/fcaf155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
To effectively address Alzheimer's disease, it is crucial to understand its earliest manifestations, underlying mechanisms and early markers of progression. Recent findings of very early brain activation anomalies highlight their potential for early disease characterization and predicting future cognitive decline. Our objective was to evaluate the value of brain activation-both individually and in combination with structural and neuropsychological measures-for predicting cognitive change. The study included 105 individuals from the Consortium for the Early Identification of Alzheimer's Disease-Quebec cohort who exhibited subjective cognitive decline or mild cognitive impairment. Cognitive decline was assessed by calculating the slope of Montreal Cognitive Assessment scores using regression models across successive assessments, and individuals were characterized as either decliners or stable based on clinically reliable change. We evaluated cognitive decline predictions using unimodal models for each class of predictors and multimodal models that combined these predictors. Functional activation emerged as a strong predictor of cognitive change (R²=52.5%), with 87.6% accuracy and 98.7% specificity, performing comparably to structural and neuropsychological measures. Although the unimodal functional model exhibited high specificity, indicating that functional abnormalities frequently predict future decline, it had low sensitivity (60%), meaning that the absence of abnormalities does not rule out future decline. Multimodal models provided greater explanatory power than unimodal models and greater sensitivity than the functional model. These findings highlight the potential role of early brain activation anomalies in the early detection of future cognitive changes, offering valuable insights for clinicians and researchers in assessing cognitive decline risk and refining clinical trial criteria.
Collapse
Affiliation(s)
- Laurie Décarie-Labbé
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
- Department of Psychology, Université de Montréal, Montreal, Quebec, Canada, H3C 3J7
| | - Samira Mellah
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
| | - Isaora Z Dialahy
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
| | - Sylvie Belleville
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
- Department of Psychology, Université de Montréal, Montreal, Quebec, Canada, H3C 3J7
| |
Collapse
|
8
|
Lu K, Baker J, Nicholas JM, Street RE, Keuss SE, Coath W, James SN, Keshavan A, Weston PSJ, Murray-Smith H, Cash DM, Malone IB, Wong A, Fox NC, Richards M, Crutch SJ, Schott JM. Associations between accelerated forgetting, amyloid deposition and brain atrophy in older adults. Brain 2025; 148:1302-1315. [PMID: 39423292 PMCID: PMC11969454 DOI: 10.1093/brain/awae316] [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: 01/26/2024] [Revised: 08/14/2024] [Accepted: 09/22/2024] [Indexed: 10/21/2024] Open
Abstract
Accelerated long-term forgetting (ALF) is the phenomenon whereby material is retained normally over short intervals (e.g. minutes) but forgotten abnormally rapidly over longer periods (days or weeks). ALF might be an early marker of cognitive decline, but little is known about its relationships with preclinical Alzheimer's disease pathology and how memory selectivity might influence which material is forgotten. We assessed ALF in 'Insight 46', a sub-study of the MRC National Survey of Health and Development (a population-based cohort born during the same week in 1946) (n = 429; 47% female; assessed at age ∼73 years). ALF assessment comprised visual and verbal memory tests: complex figure drawing and the face-name associative memory exam (FNAME). ALF scores were calculated as the percentage of material retained after 7 days, relative to 30 min. In 306 cognitively normal participants, we investigated effects on ALF of β-amyloid pathology (quantified using 18F-Florbetapir-PET, classified as positive/negative) and whole-brain and hippocampal atrophy rate (quantified from serial T1-MRI over ∼2.4 years preceding the ALF assessment), in addition to interactions between these pathologies. We categorized complex figure drawing items as 'outline' or 'detail', to test our hypothesis that forgetting the outline of the structure would be more sensitive to the effect of brain pathologies. We also investigated associations between ALF and subjective cognitive decline, measured with the MyCog questionnaire. Complex figure 'outline' items were better retained than 'detail' items (mean retention over 7 days = 94% versus 72%). Amyloid-positive participants showed greater forgetting of the complex figure outline compared with amyloid-negative participants (90% versus 95%; P < 0.01). There were interactions between amyloid pathology and cerebral atrophy, such that whole-brain and hippocampal atrophy predicted greater ALF on complex figure drawing among amyloid-positive participants only [e.g. 1.9 percentage-points lower retention per ml/year of whole-brain atrophy (95% confidence intervals 0.5, 3.7); P < 0.05]. Greater ALF on FNAME was associated with increased rate of hippocampal atrophy. ALF on complex figure drawing was also correlated with subjective cognitive decline [-0.45 percentage-points per MyCog point (-0.85, -0.05); P < 0.05]. These results provide evidence of associations between some measures of ALF and biomarkers of brain pathologies and subjective cognitive decline in cognitively normal older adults. On complex figure drawing, 'outline' items were better remembered than 'detail' items, illustrating the strategic role of memory selectivity, but 'outline' items were also relatively more vulnerable to ALF in individuals with amyloid pathology. Overall, our findings suggest that ALF might be a sensitive marker of cognitive changes in preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Kirsty Lu
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - John Baker
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Jennifer M Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Rebecca E Street
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Sarah E Keuss
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - William Coath
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Sarah-Naomi James
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Philip S J Weston
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - Heidi Murray-Smith
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - Ian B Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Sebastian J Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| |
Collapse
|
9
|
Peng J, Tang Q, Li Y, Liu L, Biswal BB, Wang P. Neuromorphic deviations associated with transcriptomic expression and specific cell type in Alzheimer's disease. Sci Rep 2025; 15:7460. [PMID: 40032887 PMCID: PMC11876660 DOI: 10.1038/s41598-025-90872-w] [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] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is known to be associated with cortical anatomical atrophy and neurodegeneration across various brain regions. However, the relationships between brain structural changes in AD and gene expression remain unclear. We perform the morphometric similarity network (MSN) analysis to reveal the consistent cortical structural differences in individuals with AD compared to controls, and investigate the associations between brain-wide gene expression and morphometric changes. Furthermore, we identify abnormally MSN-related genes linked to specific cell types as the major contributors to transcriptomic relationships. MSN-related structural changes are located in the lateral ventral prefrontal cortex, temporal pole and medial prefrontal lobe, which are highly associated with the AD's cognitive decline. Analysis of gene expression shows the spatial correlations between AD-related genes and MSN differences. Examination of cell type-specific signature genes indicates that changes in microglia and neuronal transcriptional profiles largely contribute to AD-specific MSN differences. The study map the disease-specific structural alterations in AD down to the cellular level, offering a novel perspective on the linking surface-level changes to molecular mechanisms.
Collapse
Affiliation(s)
- Jinzhong Peng
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Qin Tang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Yilu Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Lin Liu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Bharat Bhusan Biswal
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China.
- Department of Biomedical Engineering, New Jersey Institute of Technology, 607 Fenster Hall, University Height, Newark, NJ, 07102, USA.
| | - Pan Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China.
| |
Collapse
|
10
|
Wan L, Zhong P, Li P, Ren Y, Wang W, Yu M, Feng HY, Yan Z. CRISPR-based epigenetic editing of Gad1 improves synaptic inhibition and cognitive behavior in a Tauopathy mouse model. Neurobiol Dis 2025; 206:106826. [PMID: 39894446 DOI: 10.1016/j.nbd.2025.106826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025] Open
Abstract
GABAergic signaling in the brain plays a key role in regulating synaptic transmission, neuronal excitability, and cognitive processes. Large-scale sequencing has revealed the diminished expression of GABA-related genes in Alzheimer's disease (AD), however, it is largely unclear about the epigenetic mechanisms that dysregulate the transcription of these genes in AD. We confirmed that GABA synthesizing enzymes, GAD1 and GAD2, were significantly downregulated in prefrontal cortex (PFC) of AD human postmortem tissues. A tauopathy mouse model also had the significantly reduced expression of GABA-related genes, as well as the diminished GABAergic synaptic transmission in PFC pyramidal neurons. To elevate endogenous Gad1 levels, we used the CRISPR/Cas9-based epigenome editing technology to recruit histone acetyltransferase p300 to Gad1. Cells transfected with a fusion protein consisting of the nuclease-null dCas9 protein and the catalytic core of p300 (dCas9p300), as well as a guide RNA targeting Gad1 promoter (gRNAGad1), had significantly increased Gad1 mRNA expression and histone acetylation at Gad1 promoter. Furthermore, the tauopathy mouse model with PFC injection of dCas9p300 and gRNAGad1 lentiviruses had significantly elevated GABAergic synaptic currents and improved spatial memory. These results have provided an epigenetic editing-based gene-targeting strategy to restore synaptic inhibition and cognitive function in AD and related disorders.
Collapse
Affiliation(s)
- Lei Wan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Pei Li
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Yong Ren
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Wei Wang
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Mingjun Yu
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Henry Y Feng
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
11
|
Scheijbeler EP, de Haan W, Coomans EM, den Braber A, Tomassen J, ten Kate M, Konijnenberg E, Collij LE, van de Giessen E, Barkhof F, Visser PJ, Stam CJ, Gouw AA. Amyloid-β deposition predicts oscillatory slowing of magnetoencephalography signals and a reduction of functional connectivity over time in cognitively unimpaired adults. Brain Commun 2025; 7:fcaf018. [PMID: 40008329 PMCID: PMC11851009 DOI: 10.1093/braincomms/fcaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/11/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
With the ongoing developments in the field of anti-amyloid therapy for Alzheimer's disease, it is crucial to better understand the longitudinal associations between amyloid-β deposition and altered network activity in the living human brain. We included 110 cognitively unimpaired individuals (67.9 ± 5.7 years), who underwent [18F]flutemetamol (amyloid-β)-PET imaging and resting-state magnetoencephalography (MEG) recording at baseline and 4-year follow-up. We tested associations between baseline amyloid-β deposition and MEG measures (oscillatory power and functional connectivity). Next, we examined the relationship between baseline amyloid-β deposition and longitudinal MEG measures, as well as between baseline MEG measures and longitudinal amyloid-β deposition. Finally, we assessed associations between longitudinal changes in both amyloid-β deposition and MEG measures. Analyses were performed using linear mixed models corrected for age, sex and family. At baseline, amyloid-β deposition in orbitofrontal-posterior cingulate regions (i.e. early Alzheimer's disease regions) was associated with higher theta (4-8 Hz) power (β = 0.17, P < 0.01) in- and lower functional connectivity [inverted Joint Permutation Entropy (JPEinv) theta, β = -0.24, P < 0.001] of these regions, lower whole-brain beta (13-30 Hz) power (β = -0.13, P < 0.05) and lower whole-brain functional connectivity (JPEinv theta, β = -0.18, P < 0.001). Whole-brain amyloid-β deposition was associated with higher whole-brain theta power (β = 0.17, P < 0.05), lower whole-brain beta power (β = -0.13, P < 0.05) and lower whole-brain functional connectivity (JPEinv theta, β = -0.21, P < 0.001). Baseline amyloid-β deposition in early Alzheimer's disease regions also predicted future oscillatory slowing, reflected by increased theta power over time in early Alzheimer's disease regions and across the whole brain (β = 0.11, β = 0.08, P < 0.001), as well as decreased whole-brain beta power over time (β = -0.04, P < 0.05). Baseline amyloid-β deposition in early Alzheimer's disease regions also predicted a reduction in functional connectivity between these regions and the rest of the brain over time (JPEinv theta, β = -0.07, P < 0.05). Baseline whole-brain amyloid-β deposition was associated with increased whole-brain theta power over time (β = 0.08, P < 0.01). Baseline MEG measures were not associated with longitudinal amyloid-β deposition. Longitudinal changes in amyloid-β deposition in early Alzheimer's disease regions were associated with longitudinal changes in functional connectivity of early Alzheimer's disease regions (JPEinv theta, β = -0.19, P < 0.05) and the whole brain [corrected amplitude envelope correlations alpha (8-13 Hz), β = -0.22, P < 0.05]. Finally, longitudinal changes in whole-brain amyloid-β deposition were associated with longitudinal changes in whole-brain relative theta power (β = 0.21, P < 0.05). Disruptions of oscillatory power and functional connectivity appear to represent early functional consequences of emerging amyloid-β deposition in cognitively unimpaired individuals. These findings suggest a role for neurophysiology in monitoring disease progression and potential treatment effects in pre-clinical Alzheimer's disease.
Collapse
Affiliation(s)
- Elliz P Scheijbeler
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Neurophysiology & MEG Center, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - Willem de Haan
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Emma M Coomans
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Mara ten Kate
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
| | - Elles Konijnenberg
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 202 13 Malmö, Sweden
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, WC1N 3BG London, UK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Cornelis J Stam
- Department of Clinical Neurophysiology & MEG Center, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - Alida A Gouw
- Department of Clinical Neurophysiology & MEG Center, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
12
|
Santi A, Moore S, Fogelson KA, Wang A, Lawlor J, Amato J, Burke K, Lauer AM, Kuchibhotla KV. Revealing hidden knowledge in amnestic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632026. [PMID: 39829851 PMCID: PMC11741257 DOI: 10.1101/2025.01.09.632026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease (AD) is a form of dementia in which memory and cognitive decline is thought to arise from underlying neurodegeneration. These cognitive impairments, however, are transient when they first appear and can fluctuate across disease progression. Here, we investigate the neural mechanisms underlying fluctuations of performance in amnestic mice. We trained APP/PS1+ mice on an auditory go/no-go task that dissociated learning of task contingencies (knowledge) from its more variable expression under reinforcement (performance). APP/PS1+ exhibited significant performance deficits compared to control mice. Using large-scale two-photon imaging of 6,216 excitatory neurons in 8 mice, we found that auditory cortical networks were more suppressed, less selective to the sensory cues, and exhibited aberrant higher-order encoding of reward prediction compared to control mice. A small sub-population of neurons, however, displayed the opposite phenotype, reflecting a potential compensatory mechanism. Volumetric analysis demonstrated that deficits were concentrated near Aβ plaques. Strikingly, we found that these cortical deficits were reversed almost instantaneously on probe (non-reinforced) trials when APP/PS1+ performed as well as control mice, providing neural evidence for intact stimulus-action knowledge despite variable ongoing performance. A biologically-plausible reinforcement learning model recapitulated these results and showed that synaptic weights from sensory-to-decision neurons were preserved (i.e. intact stimulus-action knowledge) despite poor performance that was due to inadequate contextual scaling (i.e. impaired performance). Our results suggest that the amnestic phenotype is transient, contextual, and endogenously reversible, with the underlying neural circuits retaining the underlying stimulus-action associations. Thus, memory deficits commonly observed in amnestic mouse models, and potentially at early stages of dementia in humans, relate more to contextual drivers of performance rather than degeneration of the underlying memory traces.
Collapse
|
13
|
Anton PE, Maphis NM, Linsenbardt DN, Coleman LG. Excessive Alcohol Use as a Risk Factor for Alzheimer's Disease: Epidemiological and Preclinical Evidence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:211-242. [PMID: 40128481 DOI: 10.1007/978-3-031-81908-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Alcohol use has recently emerged as a modifiable risk factor for Alzheimer's disease (AD). However, the neurobiological mechanisms by which alcohol interacts with AD pathogenesis remain poorly understood. In this chapter, we review the epidemiological and preclinical support for the interaction between alcohol use and AD. We hypothesize that alcohol use increases the rate of accumulation of specific AD-relevant pathologies during the prodromal phase and exacerbates dementia onset and progression. We find that alcohol consumption rates are increasing in adolescence, middle age, and aging populations. In tandem, rates of AD are also on the rise, potentially as a result of this increased alcohol use throughout the lifespan. We then review the biological processes in common between alcohol use disorder and AD as a means to uncover potential mechanisms by which they interact; these include oxidative stress, neuroimmune function, metabolism, pathogenic tauopathy development and spread, and neuronal excitatory/inhibitory balance (EIB). Finally, we provide some forward-thinking suggestions we believe this field should consider. In particular, the inclusion of alcohol use assessments in longitudinal studies of AD and more preclinical studies on alcohol's impacts using better animal models of late-onset Alzheimer's disease (LOAD).
Collapse
Affiliation(s)
- Paige E Anton
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Nicole M Maphis
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - David N Linsenbardt
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Ziontz J, Harrison TM, Fonseca C, Giorgio J, Han F, Lee J, Jagust WJ, Alzheimer's Disease Neuroimaging Initiative. Connectivity, Pathology, and ApoE4 Interactions Predict Longitudinal Tau Spatial Progression and Memory. Hum Brain Mapp 2024; 45:e70083. [PMID: 39651679 PMCID: PMC11626484 DOI: 10.1002/hbm.70083] [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: 09/06/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/11/2024] Open
Abstract
Tau pathology spread into neocortex indicates a transition from healthy aging to Alzheimer's disease (AD). Connectivity between tau epicenters and later accumulating regions of cortex has been proposed as a mechanism of tau spread, but how this relationship changes with greater AD pathology burden or genotype is not understood. We investigated tau accumulation in two key regions, precuneus and inferior temporal cortex, using resting state functional connectivity (rsFC) and longitudinal PET imaging from a multicohort sample of cognitively unimpaired older adults. We examined how baseline tau PET, Aβ PET, and ApoE4 genotype status interact with rsFC between hippocampus and these downstream regions to predict rate of tau accumulation in neocortex. We found that the 3-way interaction between connectivity, baseline tau, and baseline Aβ or ApoE4 status was associated with neocortical tau accumulation in precuneus and inferior temporal cortex. In addition, baseline tau, Aβ, and ApoE4 status also moderated the association between connectivity and rate of memory decline. Together, these results suggest that the extent and distribution of future tau accumulation may be predicted by the interaction of baseline connectivity, AD pathology, and genetic risk.
Collapse
Affiliation(s)
- Jacob Ziontz
- Department of NeuroscienceUC BerkeleyBerkeleyCaliforniaUSA
| | | | | | - Joseph Giorgio
- Department of NeuroscienceUC BerkeleyBerkeleyCaliforniaUSA
- School of Psychological Sciences, College of Engineering, Science and the Environment, University of NewcastleNewcastleNew South WalesAustralia
| | - Feng Han
- Department of NeuroscienceUC BerkeleyBerkeleyCaliforniaUSA
| | - JiaQie Lee
- Department of NeuroscienceUC BerkeleyBerkeleyCaliforniaUSA
| | - William J. Jagust
- Department of NeuroscienceUC BerkeleyBerkeleyCaliforniaUSA
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | | |
Collapse
|
15
|
Heneka MT, Morgan D, Jessen F. Passive anti-amyloid β immunotherapy in Alzheimer's disease-opportunities and challenges. Lancet 2024; 404:2198-2208. [PMID: 39549715 DOI: 10.1016/s0140-6736(24)01883-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 11/18/2024]
Abstract
With the advent of the first disease-modifying, anti-amyloid β-directed passive immunotherapy for Alzheimer's disease, questions arise who, when, and how to treat. This paper describes shortly the pathogenic basis of and preclinical data, which have, more than two decades ago, initiated the development of this vaccination therapy. We discuss clinical trial results of aducanumab, lecanemab, and donanemab. We also review appropriate use recommendations of these novel treatments on patient selection and safety monitoring. Furthermore, estimations of numbers of patient who will qualify for treatment regarding inclusion and exclusion criteria and estimations on readiness of health-care systems for identifying the right patients and for providing the treatment are reported. In our view, we are experiencing a fundamental shift from syndrome-based Alzheimer's dementia care to early, biomarker-guided treatment of Alzheimer's disease. This shift requires substantial adjustments of infrastructure and resources, but also holds promise of eventually achieving substantial slowing of disease progression and delaying dementia.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg; Departmnet of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany; German Center for Neurodegenerative Diseases, Bonn, Cologne, Germany; Excellence cluster on cellular stress response in aging associated disease, University of Cologne, Cologne, Germany
| |
Collapse
|
16
|
van Nifterick AM, de Haan W, Stam CJ, Hillebrand A, Scheltens P, van Kesteren RE, Gouw AA. Functional network disruption in cognitively unimpaired autosomal dominant Alzheimer's disease: a magnetoencephalography study. Brain Commun 2024; 6:fcae423. [PMID: 39713236 PMCID: PMC11660908 DOI: 10.1093/braincomms/fcae423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
Understanding the nature and onset of neurophysiological changes, and the selective vulnerability of central hub regions in the functional network, may aid in managing the growing impact of Alzheimer's disease on society. However, the precise neurophysiological alterations occurring in the pre-clinical stage of human Alzheimer's disease remain controversial. This study aims to provide increased insights on quantitative neurophysiological alterations during a true early stage of Alzheimer's disease. Using high spatial resolution source-reconstructed magnetoencephalography, we investigated regional and whole-brain neurophysiological changes in a unique cohort of 11 cognitively unimpaired individuals with pathogenic mutations in the presenilin-1 or amyloid precursor protein gene and a 1:3 matched control group (n = 33) with a median age of 49 years. We examined several quantitative magnetoencephalography measures that have been shown robust in detecting differences in sporadic Alzheimer's disease patients and are sensitive to excitation-inhibition imbalance. This includes spectral power and functional connectivity in different frequency bands. We also investigated hub vulnerability using the hub disruption index. To understand how magnetoencephalography measures change as the disease progresses through its pre-clinical stage, correlations between magnetoencephalography outcomes and various clinical variables like age were analysed. A comparison of spectral power between mutation carriers and controls revealed oscillatory slowing, characterized by widespread higher theta (4-8 Hz) power, a lower posterior peak frequency and lower occipital alpha 2 (10-13 Hz) power. Functional connectivity analyses presented a lower whole-brain (amplitude-based) functional connectivity in the alpha (8-13 Hz) and beta (13-30 Hz) bands, predominantly located in parieto-temporal hub regions. Furthermore, we found a significant hub disruption index for (phase-based) functional connectivity in the theta band, attributed to both higher functional connectivity in 'non-hub' regions alongside a hub disruption. Neurophysiological changes did not correlate with indicators of pre-clinical disease progression in mutation carriers after multiple comparisons correction. Our findings provide evidence that oscillatory slowing and functional connectivity differences occur before cognitive impairment in individuals with autosomal dominant mutations leading to early onset Alzheimer's disease. The nature and direction of these alterations are comparable to those observed in the clinical stages of Alzheimer's disease, suggest an early excitation-inhibition imbalance, and fit with the activity-dependent functional degeneration hypothesis. These insights may prove useful for early diagnosis and intervention in the future.
Collapse
Affiliation(s)
- Anne M van Nifterick
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Clinical Neurophysiology and MEG Center, Neurology, Amsterdam UMC Location VUmc, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Willem de Haan
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Cornelis J Stam
- Clinical Neurophysiology and MEG Center, Neurology, Amsterdam UMC Location VUmc, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Arjan Hillebrand
- Clinical Neurophysiology and MEG Center, Neurology, Amsterdam UMC Location VUmc, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Systems and Network Neurosciences, 1081 HV Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Alida A Gouw
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Clinical Neurophysiology and MEG Center, Neurology, Amsterdam UMC Location VUmc, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Systems and Network Neurosciences, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
17
|
Lam AD, Thibault EG, Mayblyum DV, Hsieh S, Pellerin KR, Sternberg EJ, Viswanathan A, Buss S, Sarkis RA, Jacobs HIL, Johnson KA, Sperling RA. Association of Seizure Foci and Location of Tau and Amyloid Deposition and Brain Atrophy in Patients With Alzheimer Disease and Seizures. Neurology 2024; 103:e209920. [PMID: 39331846 PMCID: PMC11441794 DOI: 10.1212/wnl.0000000000209920] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/22/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Alzheimer disease (AD) is associated with a 2 to 3-fold increased risk of developing late-onset focal epilepsy, yet it remains unclear how development of focal epilepsy in AD is related to AD pathology. The objective of this study was to examine spatial relationships between the epileptogenic zone and tau deposition, amyloid deposition, and brain atrophy in individuals with AD who developed late-onset, otherwise unexplained focal epilepsy. We hypothesized that if network hyperexcitability is mechanistically linked to AD pathology, then there would be increased tau and amyloid deposition within the epileptogenic hemisphere. METHODS In this cross-sectional study, we performed tau and amyloid PET imaging, brain MRI, and overnight scalp EEG in individuals with early clinical stages of AD who developed late-onset, otherwise unexplained focal epilepsy (AD-Ep). Participants were referred from epilepsy and memory disorders clinics at our institutions. We determined epilepsy localization based on EEG findings and seizure semiology. We quantified tau deposition, amyloid deposition, and atrophy across brain regions and calculated asymmetry indices for these measures. We compared findings in AD-Ep with those in a control AD group without epilepsy (AD-NoEp). RESULTS The AD-Ep group included 8 individuals with a mean age of 69.5 ± 4.2 years at PET imaging. The AD-NoEp group included 14 individuals with a mean age of 71.7 ± 9.8 years at PET imaging. In AD-Ep, we found a highly asymmetric pattern of tau deposition, with significantly greater tau in the epileptogenic hemisphere. Amyloid deposition and cortical atrophy were also greater in the epileptogenic hemisphere, although the magnitudes of asymmetry were reduced compared with tau. Compared with AD-NoEp, the AD-Ep group had significantly greater tau asymmetry and trends toward greater asymmetry of amyloid and atrophy. AD-Ep also had significantly greater amyloid burden bilaterally and trends toward greater tau burden within the epileptogenic hemisphere, compared with AD-NoEp. DISCUSSION Our results reveal a spatial association between the epileptogenic focus and tau deposition, amyloid deposition, and neurodegeneration in early clinical stages of AD. Within the limitations of a cross-sectional study with small sample sizes, these findings contribute to our understanding of the clinicopathologic heterogeneity of AD, demonstrating an association between focal epilepsy and lateralized pathology in AD.
Collapse
Affiliation(s)
- Alice D Lam
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Emma G Thibault
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Danielle V Mayblyum
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Stephanie Hsieh
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Kyle R Pellerin
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Eliezer J Sternberg
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Anand Viswanathan
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Stephanie Buss
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Rani A Sarkis
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Heidi I L Jacobs
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Keith A Johnson
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| | - Reisa A Sperling
- From the Department of Neurology (A.D.L., S.H., K.R.P., A.V., K.A.J.), Massachusetts General Hospital, Boston; Harvard Medical School (A.D.L., A.V., S.B., R.A. Sarkis, H.L.J., K.A.J., R.A. Sperling), Boston; Department of Radiology (E.G.T., D.V.M., H.L.J., K.A.J.), Massachusetts General Hospital, Boston; Department of Neurology (E.J.S.), Milford Regional Medical Center; Department of Neurology (S.B.), Beth Israel Deaconess Medical Center, Boston; and Department of Neurology (R.A.Sarkis, R.A.Sperling), Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
18
|
Benitez MJ, Retana D, Ordoñez-Gutiérrez L, Colmena I, Goméz MJ, Álvarez R, Ciorraga M, Dopazo A, Wandosell F, Garrido JJ. Transcriptomic alterations in APP/PS1 mice astrocytes lead to early postnatal axon initial segment structural changes. Cell Mol Life Sci 2024; 81:444. [PMID: 39485512 PMCID: PMC11530419 DOI: 10.1007/s00018-024-05485-9] [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: 10/10/2023] [Revised: 09/20/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024]
Abstract
Alzheimer´s disease (AD) is characterized by neuronal function loss and degeneration. The integrity of the axon initial segment (AIS) is essential to maintain neuronal function and output. AIS alterations are detected in human post-mortem AD brains and mice models, as well as, neurodevelopmental and mental disorders. However, the mechanisms leading to AIS deregulation in AD and the extrinsic glial origin are elusive. We studied early postnatal differences in AIS cellular/molecular mechanisms in wild-type or APP/PS1 mice and combined neuron-astrocyte co-cultures. We observed AIS integrity alterations, reduced ankyrinG expression and shortening, in APP/PS1 mice from P21 and loss of AIS integrity at 21 DIV in wild-type and APP/PS1 neurons in the presence of APP/PS1 astrocytes. AnkyrinG decrease is due to mRNAs and protein reduction of retinoic acid synthesis enzymes Rdh1 and Aldh1b1, as well as ADNP (Activity-dependent neuroprotective protein) in APP/PS1 astrocytes. This effect was mimicked by wild-type astrocytes expressing ADNP shRNA. In the presence of APP/PS1 astrocytes, wild-type neurons AIS is recovered by inhibition of retinoic acid degradation, and Adnp-derived NAP peptide (NAPVSIPQ) addition or P2X7 receptor inhibition, both regulated by retinoic acid levels. Moreover, P2X7 inhibitor treatment for 2 months impaired AIS disruption in APP/PS1 mice. Our findings extend current knowledge on AIS regulation, providing data to support the role of astrocytes in early postnatal AIS modulation. In conclusion, AD onset may be related to very early glial cell alterations that induce AIS and neuronal function changes, opening new therapeutic approaches to detect and avoid neuronal function loss.
Collapse
Affiliation(s)
- María José Benitez
- Instituto Cajal, CSIC, Madrid, Spain
- Departamento de Química Física Aplicada, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Lara Ordoñez-Gutiérrez
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Inés Colmena
- Instituto Cajal, CSIC, Madrid, Spain
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain
| | | | - Rebeca Álvarez
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Wandosell
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan José Garrido
- Instituto Cajal, CSIC, Madrid, Spain.
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-ISCIII), Madrid, Spain.
| |
Collapse
|
19
|
Corriveau-Lecavalier N, Adams JN, Fischer L, Molloy EN, Maass A. Cerebral hyperactivation across the Alzheimer's disease pathological cascade. Brain Commun 2024; 6:fcae376. [PMID: 39513091 PMCID: PMC11542485 DOI: 10.1093/braincomms/fcae376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
Neuronal dysfunction in specific brain regions or across distributed brain networks is a known feature of Alzheimer's disease. An often reported finding in the early stage of the disease is the presence of increased functional MRI (fMRI) blood oxygenation level-dependent signal under task conditions relative to cognitively normal controls, a phenomenon known as 'hyperactivation'. However, research in the past decades yielded complex, sometimes conflicting results. The magnitude and topology of fMRI hyperactivation patterns have been found to vary across the preclinical and clinical spectrum of Alzheimer's disease, including concomitant 'hypoactivation' in some cases. These incongruences are likely due to a range of factors, including the disease stage at which the cohort is examined, the brain areas or networks studied and the fMRI paradigm utilized to evoke these functional abnormalities. Additionally, a perennial question pertains to the nature of hyperactivation in the context of Alzheimer's disease. Some propose it reflects compensatory mechanisms to sustain cognitive performance, while others suggest it is linked to the pathological disruption of a highly regulated homeostatic cycle that contributes to, or even drives, disease progression. Providing a coherent narrative for these empirical and conceptual discrepancies is paramount to develop disease models, understand the synergy between hyperactivation and the Alzheimer's disease pathological cascade and tailor effective interventions. We first provide a comprehensive overview of functional brain changes spanning the course from normal ageing to the clinical spectrum of Alzheimer's disease. We then highlight evidence supporting a close relationship between fMRI hyperactivation and in vivo markers of Alzheimer's pathology. We primarily focus on task-based fMRI studies in humans, but also consider studies using different functional imaging techniques and animal models. We then discuss the potential mechanisms underlying hyperactivation in the context of Alzheimer's disease and provide a testable framework bridging hyperactivation, ageing, cognition and the Alzheimer's disease pathological cascade. We conclude with a discussion of future challenges and opportunities to advance our understanding of the fundamental disease mechanisms of Alzheimer's disease, and the promising development of therapeutic interventions incorporating or aimed at hyperactivation and large-scale functional systems.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota 55902 USA
| | - Jenna N Adams
- Department of Neurobiology and Behavior, University of California, Irvine 92697, CA, USA
| | - Larissa Fischer
- German Center for Neurodegenerative Diseases, Magdeburg 39120, Germany
| | - Eóin N Molloy
- German Center for Neurodegenerative Diseases, Magdeburg 39120, Germany
- Division of Nuclear Medicine, Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases, Magdeburg 39120, Germany
- Institute for Biology, Otto-von-Guericke University Magdeburg, Magdeburg 39120, Germany
| |
Collapse
|
20
|
Maksour S, Finol-Urdaneta RK, Hulme AJ, Cabral-da-Silva MEC, Targa Dias Anastacio H, Balez R, Berg T, Turner C, Sanz Muñoz S, Engel M, Kalajdzic P, Lisowski L, Sidhu K, Sachdev PS, Dottori M, Ooi L. Alzheimer's disease induced neurons bearing PSEN1 mutations exhibit reduced excitability. Front Cell Neurosci 2024; 18:1406970. [PMID: 39444394 PMCID: PMC11497635 DOI: 10.3389/fncel.2024.1406970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative condition that affects memory and cognition, characterized by neuronal loss and currently lacking a cure. Mutations in PSEN1 (Presenilin 1) are among the most common causes of early-onset familial AD (fAD). While changes in neuronal excitability are believed to be early indicators of AD progression, the link between PSEN1 mutations and neuronal excitability remains to be fully elucidated. This study examined iPSC-derived neurons (iNs) from fAD patients with PSEN1 mutations S290C or A246E, alongside CRISPR-corrected isogenic cell lines, to investigate early changes in excitability. Electrophysiological profiling revealed reduced excitability in both PSEN1 mutant iNs compared to their isogenic controls. Neurons bearing S290C and A246E mutations exhibited divergent passive membrane properties compared to isogenic controls, suggesting distinct effects of PSEN1 mutations on neuronal excitability. Additionally, both PSEN1 backgrounds exhibited higher current density of voltage-gated potassium (Kv) channels relative to their isogenic iNs, while displaying comparable voltage-gated sodium (Nav) channel current density. This suggests that the Nav/Kv imbalance contributes to impaired neuronal firing in fAD iNs. Deciphering these early cellular and molecular changes in AD is crucial for understanding disease pathogenesis.
Collapse
Affiliation(s)
- Simon Maksour
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Rocio K. Finol-Urdaneta
- School of Medical and Indigenous Health Science and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Amy J. Hulme
- School of Medical and Indigenous Health Science and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | | | - Helena Targa Dias Anastacio
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Rachelle Balez
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Tracey Berg
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Calista Turner
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Sonia Sanz Muñoz
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Martin Engel
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Predrag Kalajdzic
- Translational Vectorology Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
- Australian Genome Therapeutics Centre, Children’s Medical Research Institute and Sydney Children’s Hospitals Network, Westmead, NSW, Australia
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine – National Research Institute, Warsaw, Poland
| | - Kuldip Sidhu
- Centre for Healthy Brain Ageing, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Mirella Dottori
- School of Medical and Indigenous Health Science and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Lezanne Ooi
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
21
|
Chen J, Peng G, Sun B. Alzheimer's disease and sleep disorders: A bidirectional relationship. Neuroscience 2024; 557:12-23. [PMID: 39137870 DOI: 10.1016/j.neuroscience.2024.08.008] [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: 06/16/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent dementia, pathologically featuring abnormal accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, while sleep, divided into rapid eye movement sleep (REM) and nonrapid eye movement sleep (NREM), plays a key role in consolidating social and spatial memory. Emerging evidence has revealed that sleep disorders such as circadian disturbances and disruption of neuronal rhythm activity are considered as both candidate risks and consequence of AD, suggesting a bidirectional relationship between sleep and AD. This review will firstly grasp basic knowledge of AD pathogenesis, then highlight macrostructural and microstructural alteration of sleep along with AD progression, explain the interaction between accumulation of Aβ and hyperphosphorylated tau, which are two critical neuropathological processes of AD, as well as neuroinflammation and sleep, and finally introduce several methods of sleep enhancement as strategies to reduce AD-associated neuropathology. Although theories about the bidirectional relationship and relevant therapeutic methods in mice have been well developed in recent years, the knowledge in human is still limited. More studies on how to effectively ameliorate AD pathology in patients by sleep enhancement and what specific roles of sleep play in AD are needed.
Collapse
Affiliation(s)
- Junhua Chen
- Chu Kochen Honors College of Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Binggui Sun
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
22
|
Michaud F, Francavilla R, Topolnik D, Iloun P, Tamboli S, Calon F, Topolnik L. Altered firing output of VIP interneurons and early dysfunctions in CA1 hippocampal circuits in the 3xTg mouse model of Alzheimer's disease. eLife 2024; 13:RP95412. [PMID: 39264364 PMCID: PMC11392531 DOI: 10.7554/elife.95412] [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] [Indexed: 09/13/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive memory decline, and alterations in hippocampal function are among the earliest pathological features observed in human and animal studies. GABAergic interneurons (INs) within the hippocampus coordinate network activity, among which type 3 interneuron-specific (I-S3) cells expressing vasoactive intestinal polypeptide and calretinin play a crucial role. These cells provide primarily disinhibition to principal excitatory cells (PCs) in the hippocampal CA1 region, regulating incoming inputs and memory formation. However, it remains unclear whether AD pathology induces changes in the activity of I-S3 cells, impacting the hippocampal network motifs. Here, using young adult 3xTg-AD mice, we found that while the density and morphology of I-S3 cells remain unaffected, there were significant changes in their firing output. Specifically, I-S3 cells displayed elongated action potentials and decreased firing rates, which was associated with a reduced inhibition of CA1 INs and their higher recruitment during spatial decision-making and object exploration tasks. Furthermore, the activation of CA1 PCs was also impacted, signifying early disruptions in CA1 network functionality. These findings suggest that altered firing patterns of I-S3 cells might initiate early-stage dysfunction in hippocampal CA1 circuits, potentially influencing the progression of AD pathology.
Collapse
Affiliation(s)
- Felix Michaud
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Ruggiero Francavilla
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Dimitry Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Parisa Iloun
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Suhel Tamboli
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Frederic Calon
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| |
Collapse
|
23
|
Saha O, Melo de Farias AR, Pelletier A, Siedlecki-Wullich D, Landeira BS, Gadaut J, Carrier A, Vreulx AC, Guyot K, Shen Y, Bonnefond A, Amouyel P, Tcw J, Kilinc D, Queiroz CM, Delahaye F, Lambert JC, Costa MR. The Alzheimer's disease risk gene BIN1 regulates activity-dependent gene expression in human-induced glutamatergic neurons. Mol Psychiatry 2024; 29:2634-2646. [PMID: 38514804 PMCID: PMC11420064 DOI: 10.1038/s41380-024-02502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Bridging Integrator 1 (BIN1) is the second most important Alzheimer's disease (AD) risk gene, but its physiological roles in neurons and its contribution to brain pathology remain largely elusive. In this work, we show that BIN1 plays a critical role in the regulation of calcium homeostasis, electrical activity, and gene expression of glutamatergic neurons. Using single-cell RNA-sequencing on cerebral organoids generated from isogenic BIN1 wild type (WT), heterozygous (HET) and homozygous knockout (KO) human-induced pluripotent stem cells (hiPSCs), we show that BIN1 is mainly expressed by oligodendrocytes and glutamatergic neurons, like in the human brain. Both BIN1 HET and KO cerebral organoids show specific transcriptional alterations, mainly associated with ion transport and synapses in glutamatergic neurons. We then demonstrate that BIN1 cell-autonomously regulates gene expression in glutamatergic neurons by using a novel protocol to generate pure culture of hiPSC-derived induced neurons (hiNs). Using this system, we also show that BIN1 plays a key role in the regulation of neuronal calcium transients and electrical activity via its interaction with the L-type voltage-gated calcium channel Cav1.2. BIN1 KO hiNs show reduced activity-dependent internalization and higher Cav1.2 expression compared to WT hiNs. Pharmacological blocking of this channel with clinically relevant doses of nifedipine, a calcium channel blocker, partly rescues electrical and gene expression alterations in BIN1 KO glutamatergic neurons. Further, we show that transcriptional alterations in BIN1 KO hiNs that affect biological processes related to calcium homeostasis are also present in glutamatergic neurons of the human brain at late stages of AD pathology. Together, these findings suggest that BIN1-dependent alterations in neuronal properties could contribute to AD pathophysiology and that treatment with low doses of clinically approved calcium blockers should be considered as an option to slow disease-onset and progression.
Collapse
Affiliation(s)
- Orthis Saha
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Ana Raquel Melo de Farias
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Alexandre Pelletier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Dolores Siedlecki-Wullich
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Bruna Soares Landeira
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Johanna Gadaut
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Arnaud Carrier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Anaïs-Camille Vreulx
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Karine Guyot
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Yun Shen
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Amelie Bonnefond
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Julia Tcw
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
- Bioinformatics Program, Faculty of Computing & Data Sciences, Boston University, Boston, MA, 02115, USA
| | - Devrim Kilinc
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Claudio Marcos Queiroz
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Fabien Delahaye
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Marcos R Costa
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France.
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil.
| |
Collapse
|
24
|
Ishiguro T, Kasuga K. Alzheimer's Disease-Related Cerebrospinal Fluid Biomarkers in Progressive Supranuclear Palsy. Brain Sci 2024; 14:859. [PMID: 39335355 PMCID: PMC11430815 DOI: 10.3390/brainsci14090859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Progressive Supranuclear Palsy (PSP) is the most common four-repeat tauopathy. PSP cases are typically characterized by vertical gaze palsy and postural instability; however, various phenotypes have been reported, making antemortem diagnosis based on clinical symptoms challenging. The development of biomarkers reflecting brain pathology and the ability to diagnose patients based on these biomarkers are essential for developing future intervention strategies, including disease-modifying therapies. However, despite many dedicated efforts, no highly specific fluid biomarker for PSP has yet been established. Conversely, several cerebrospinal fluid (CSF) biomarkers of Alzheimer's Disease (AD) have been established, and an AT(N) classification system has been proposed. Typically, among patients with AD, CSF amyloid β42 (Aβ42), but not Aβ40, is decreased, resulting in a reduction in the Aβ42/Aβ40 ratio, while tau phosphorylated at threonine 181 (p-tau181) and total tau (t-tau) are increased. Interestingly, the core CSF AD biomarkers show unique patterns in patients with PSP. Furthermore, reports have indicated that the CSF levels of both Aβ42 and Aβ40 are decreased independently of Aβ accumulation in PSP. Therefore, the Aβ42/Aβ40 ratio could potentially be used to differentiate PSP from AD. Additionally, studies have reported that CSF p-tau and t-tau are reduced in PSP, and that the neurofilament light chain is remarkably increased compared to healthy controls and patients with AD, even though PSP is a neurodegenerative disease associated with tau accumulation. These PSP-specific changes in AD-related core biomarkers may reflect the pathology of PSP and contribute to its diagnosis. As such, elucidating the mechanisms underlying the observed decreases in Aβ and tau levels could facilitate a better understanding of the pathogenesis of PSP.
Collapse
Affiliation(s)
- Takanobu Ishiguro
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| |
Collapse
|
25
|
Ma LH, Li S, Jiao XH, Li ZY, Zhou Y, Zhou CR, Zhou CH, Zheng H, Wu YQ. BLA-involved circuits in neuropsychiatric disorders. Ageing Res Rev 2024; 99:102363. [PMID: 38838785 DOI: 10.1016/j.arr.2024.102363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/04/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
The basolateral amygdala (BLA) is the subregion of the amygdala located in the medial of the temporal lobe, which is connected with a wide range of brain regions to achieve diverse functions. Recently, an increasing number of studies have focused on the participation of the BLA in many neuropsychiatric disorders from the neural circuit perspective, aided by the rapid development of viral tracing methods and increasingly specific neural modulation technologies. However, how to translate this circuit-level preclinical intervention into clinical treatment using noninvasive or minor invasive manipulations to benefit patients struggling with neuropsychiatric disorders is still an inevitable question to be considered. In this review, we summarized the role of BLA-involved circuits in neuropsychiatric disorders including Alzheimer's disease, perioperative neurocognitive disorders, schizophrenia, anxiety disorders, depressive disorders, posttraumatic stress disorders, autism spectrum disorders, and pain-associative affective states and cognitive dysfunctions. Additionally, we provide insights into future directions and challenges for clinical translation.
Collapse
Affiliation(s)
- Lin-Hui Ma
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xin-Hao Jiao
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Zi-Yi Li
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Yue Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Chen-Rui Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Cheng-Hua Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
26
|
Liu Z, Liu M, Xiong Y, Wang Y, Bu X. Crosstalk between bone and brain in Alzheimer's disease: Mechanisms, applications, and perspectives. Alzheimers Dement 2024; 20:5720-5739. [PMID: 38824621 PMCID: PMC11350061 DOI: 10.1002/alz.13864] [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: 02/05/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain-derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone-derived hormones, bone marrow-derived cells, bone-derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment. HIGHLIGHTS: The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis. Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms. Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.
Collapse
Affiliation(s)
- Zhuo‐Ting Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
| | - Ming‐Han Liu
- Department of OrthopaedicsXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Yan Xiong
- Department of OrthopaedicsDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| |
Collapse
|
27
|
Rajani RM, Ellingford R, Hellmuth M, Harris SS, Taso OS, Graykowski D, Lam FKW, Arber C, Fertan E, Danial JSH, Swire M, Lloyd M, Giovannucci TA, Bourdenx M, Klenerman D, Vassar R, Wray S, Sala Frigerio C, Busche MA. Selective suppression of oligodendrocyte-derived amyloid beta rescues neuronal dysfunction in Alzheimer's disease. PLoS Biol 2024; 22:e3002727. [PMID: 39042667 PMCID: PMC11265669 DOI: 10.1371/journal.pbio.3002727] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Reduction of amyloid beta (Aβ) has been shown to be effective in treating Alzheimer's disease (AD), but the underlying assumption that neurons are the main source of pathogenic Aβ is untested. Here, we challenge this prevailing belief by demonstrating that oligodendrocytes are an important source of Aβ in the human brain and play a key role in promoting abnormal neuronal hyperactivity in an AD knock-in mouse model. We show that selectively suppressing oligodendrocyte Aβ production improves AD brain pathology and restores neuronal function in the mouse model in vivo. Our findings suggest that targeting oligodendrocyte Aβ production could be a promising therapeutic strategy for treating AD.
Collapse
Affiliation(s)
- Rikesh M. Rajani
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Robert Ellingford
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Mariam Hellmuth
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Samuel S. Harris
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Orjona S. Taso
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - David Graykowski
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Francesca Kar Wey Lam
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Charles Arber
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Emre Fertan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute at University of Cambridge, Cambridge, United Kingdom
| | - John S. H. Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute at University of Cambridge, Cambridge, United Kingdom
- School of Physics and Astronomy, University of St Andrews, St. Andrews, United Kingdom
| | - Matthew Swire
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Marcus Lloyd
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Tatiana A. Giovannucci
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Mathieu Bourdenx
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute at University of Cambridge, Cambridge, United Kingdom
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Selina Wray
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Carlo Sala Frigerio
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Marc Aurel Busche
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| |
Collapse
|
28
|
Zhao J, Wei M, Guo M, Wang M, Niu H, Xu T, Zhou Y. GSK3: A potential target and pending issues for treatment of Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14818. [PMID: 38946682 PMCID: PMC11215492 DOI: 10.1111/cns.14818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Glycogen synthase kinase-3 (GSK3), consisting of GSK3α and GSK3β subtypes, is a complex protein kinase that regulates numerous substrates. Research has observed increased GSK3 expression in the brains of Alzheimer's disease (AD) patients and models. AD is a neurodegenerative disorder with diverse pathogenesis and notable cognitive impairments, characterized by Aβ aggregation and excessive tau phosphorylation. This article provides an overview of GSK3's structure and regulation, extensively analyzing its relationship with AD factors. GSK3 overactivation disrupts neural growth, development, and function. It directly promotes tau phosphorylation, regulates amyloid precursor protein (APP) cleavage, leading to Aβ formation, and directly or indirectly triggers neuroinflammation and oxidative damage. We also summarize preclinical research highlighting the inhibition of GSK3 activity as a primary therapeutic approach for AD. Finally, pending issues like the lack of highly specific and affinity-driven GSK3 inhibitors, are raised and expected to be addressed in future research. In conclusion, GSK3 represents a target in AD treatment, filled with hope, challenges, opportunities, and obstacles.
Collapse
Affiliation(s)
- Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Mengying Wei
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Future Health Laboratory, Innovation Center of Yangtze River DeltaZhejiang UniversityJiaxingChina
| | - Minsong Guo
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouChina
| | - Mengyao Wang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Hongxia Niu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Laboratory of Blood‐stasis‐toxin Syndrome of Zhejiang ProvinceHangzhouChina
| | - Tengfei Xu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Laboratory of Blood‐stasis‐toxin Syndrome of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
29
|
Stephens GS, Park J, Eagle A, You J, Silva-Pérez M, Fu CH, Choi S, Romain CPS, Sugimoto C, Buffington SA, Zheng Y, Costa-Mattioli M, Liu Y, Robison AJ, Chin J. Persistent ∆FosB expression limits recurrent seizure activity and provides neuroprotection in the dentate gyrus of APP mice. Prog Neurobiol 2024; 237:102612. [PMID: 38642602 PMCID: PMC11406539 DOI: 10.1016/j.pneurobio.2024.102612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 03/14/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024]
Abstract
Recurrent seizures lead to accumulation of the activity-dependent transcription factor ∆FosB in hippocampal dentate granule cells in both mouse models of epilepsy and mouse models of Alzheimer's disease (AD), which is also associated with increased incidence of seizures. In patients with AD and related mouse models, the degree of ∆FosB accumulation corresponds with increasing severity of cognitive deficits. We previously found that ∆FosB impairs spatial memory in mice by epigenetically regulating expression of target genes such as calbindin that are involved in synaptic plasticity. However, the suppression of calbindin in conditions of neuronal hyperexcitability has been demonstrated to provide neuroprotection to dentate granule cells, indicating that ∆FosB may act over long timescales to coordinate neuroprotective pathways. To test this hypothesis, we used viral-mediated expression of ∆JunD to interfere with ∆FosB signaling over the course of several months in transgenic mice expressing mutant human amyloid precursor protein (APP), which exhibit spontaneous seizures and develop AD-related neuropathology and cognitive deficits. Our results demonstrate that persistent ∆FosB activity acts through discrete modes of hippocampal target gene regulation to modulate neuronal excitability, limit recurrent seizure activity, and provide neuroprotection to hippocampal dentate granule cells in APP mice.
Collapse
Affiliation(s)
| | - Jin Park
- Department of Neuroscience, Baylor College of Medicine, USA
| | - Andrew Eagle
- Department of Physiology, Michigan State University, USA
| | - Jason You
- Department of Neuroscience, Baylor College of Medicine, USA
| | | | - Chia-Hsuan Fu
- Department of Neuroscience, Baylor College of Medicine, USA
| | - Sumin Choi
- Department of Neuroscience, Baylor College of Medicine, USA
| | | | - Chiho Sugimoto
- Department of Physiology, Michigan State University, USA
| | - Shelly A Buffington
- Center for Precision Environmental Health, Department of Neuroscience, Baylor College of Medicine, USA
| | - Yi Zheng
- Department of Neuroscience, Baylor College of Medicine, USA
| | | | - Yin Liu
- Department of Neurobiology and Anatomy, McGovern Medical School at UT Health, USA
| | - A J Robison
- Department of Physiology, Michigan State University, USA
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, USA.
| |
Collapse
|
30
|
Karimani F, Asgari Taei A, Abolghasemi-Dehaghani MR, Safari MS, Dargahi L. Impairment of entorhinal cortex network activity in Alzheimer's disease. Front Aging Neurosci 2024; 16:1402573. [PMID: 38882526 PMCID: PMC11176617 DOI: 10.3389/fnagi.2024.1402573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
The entorhinal cortex (EC) stands out as a critical brain region affected in the early phases of Alzheimer's disease (AD), with some of the disease's pathological processes originating from this area, making it one of the most crucial brain regions in AD. Recent research highlights disruptions in the brain's network activity, characterized by heightened excitability and irregular oscillations, may contribute to cognitive impairment. These disruptions are proposed not only as potential therapeutic targets but also as early biomarkers for AD. In this paper, we will begin with a review of the anatomy and function of EC, highlighting its selective vulnerability in AD. Subsequently, we will discuss the disruption of EC network activity, exploring changes in excitability and neuronal oscillations in this region during AD and hypothesize that, considering the advancements in neuromodulation techniques, addressing the disturbances in the network activity of the EC could offer fresh insights for both the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Farnaz Karimani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mir-Shahram Safari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Alexandersen CG, Goriely A, Bick C. Neuronal activity induces symmetry breaking in neurodegenerative disease spreading. J Math Biol 2024; 89:3. [PMID: 38740613 PMCID: PMC11614967 DOI: 10.1007/s00285-024-02103-x] [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: 09/29/2023] [Revised: 03/01/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Dynamical systems on networks typically involve several dynamical processes evolving at different timescales. For instance, in Alzheimer's disease, the spread of toxic protein throughout the brain not only disrupts neuronal activity but is also influenced by neuronal activity itself, establishing a feedback loop between the fast neuronal activity and the slow protein spreading. Motivated by the case of Alzheimer's disease, we study the multiple-timescale dynamics of a heterodimer spreading process on an adaptive network of Kuramoto oscillators. Using a minimal two-node model, we establish that heterogeneous oscillatory activity facilitates toxic outbreaks and induces symmetry breaking in the spreading patterns. We then extend the model formulation to larger networks and perform numerical simulations of the slow-fast dynamics on common network motifs and on the brain connectome. The simulations corroborate the findings from the minimal model, underscoring the significance of multiple-timescale dynamics in the modeling of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, UK.
| | - Christian Bick
- Mathematical Institute, University of Oxford, Oxford, UK
- Department of Mathematics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience - Systems and Network Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
32
|
L'esperance OJ, McGhee J, Davidson G, Niraula S, Smith AS, Sosunov A, Yan SS, Subramanian J. Functional connectivity favors aberrant visual network c-Fos expression accompanied by cortical synapse loss in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.05.522900. [PMID: 36712054 PMCID: PMC9881957 DOI: 10.1101/2023.01.05.522900] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
While Alzheimer's disease (AD) has been extensively studied with a focus on cognitive networks, sensory network dysfunction has received comparatively less attention despite compelling evidence of its significance in both Alzheimer's disease patients and mouse models. We recently found that neurons in the primary visual cortex of an AD mouse model expressing human amyloid protein precursor with the Swedish and Indiana mutations (hAPP mutations) exhibit aberrant c-Fos expression and altered synaptic structures at a pre-amyloid plaque stage. However, it is unclear whether aberrant c-Fos expression and synaptic pathology vary across the broader visual network and to what extent c-Fos abnormality in the cortex is inherited through functional connectivity. Using both sexes of 4-6-month AD model mice with hAPP mutations (J20[PDGF-APPSw, Ind]), we found that cortical regions of the visual network show aberrant c-Fos expression and impaired experience-dependent modulation while subcortical regions do not. Interestingly, the average network-wide functional connectivity strength of a brain region in wild type (WT) mice significantly predicts its aberrant c-Fos expression, which in turn correlates with impaired experience-dependent modulation in the AD model. Using in vivo two-photon and ex vivo imaging of presynaptic termini, we observed a subtle yet selective weakening of excitatory cortical synapses in the visual cortex. Intriguingly, the change in the size distribution of cortical boutons in the AD model is downscaled relative to those in WT mice, suggesting that synaptic weakening may reflect an adaptation to aberrant activity. Our observations suggest that cellular and synaptic abnormalities in the AD model represent a maladaptive transformation of the baseline physiological state seen in WT conditions rather than entirely novel and unrelated manifestations.
Collapse
|
33
|
Bonifazi G, Luchena C, Gaminde-Blasco A, Ortiz-Sanz C, Capetillo-Zarate E, Matute C, Alberdi E, De Pittà M. A nonlinear meccano for Alzheimer's emergence by amyloid β-mediated glutamatergic hyperactivity. Neurobiol Dis 2024; 194:106473. [PMID: 38493903 DOI: 10.1016/j.nbd.2024.106473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/10/2024] [Accepted: 03/10/2024] [Indexed: 03/19/2024] Open
Abstract
The pathophysiological process of Alzheimer's disease (AD) is believed to begin many years before the formal diagnosis of AD dementia. This protracted preclinical phase offers a crucial window for potential therapeutic interventions, yet its comprehensive characterization remains elusive. Accumulating evidence suggests that amyloid-β (Aβ) may mediate neuronal hyperactivity in circuit dysfunction in the early stages of AD. At the same time, neural activity can also facilitate Aβ accumulation through intricate feed-forward interactions, complicating elucidating the conditions governing Aβ-dependent hyperactivity and its diagnostic utility. In this study, we use biophysical modeling to shed light on such conditions. Our analysis reveals that the inherently nonlinear nature of the underlying molecular interactions can give rise to the emergence of various modes of hyperactivity. This diversity in the mechanisms of hyperactivity may ultimately account for a spectrum of AD manifestations.
Collapse
Affiliation(s)
- Giulio Bonifazi
- Basque Center for Applied Mathematics, Alameda Mazarredo 14, Bilbao 48009, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto M5T 0S8, ON, Canada
| | - Celia Luchena
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain
| | - Adhara Gaminde-Blasco
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain
| | - Carolina Ortiz-Sanz
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain
| | - Estibaliz Capetillo-Zarate
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain
| | - Elena Alberdi
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Achucarro Basque Center for Neuroscience, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain
| | - Maurizio De Pittà
- Basque Center for Applied Mathematics, Alameda Mazarredo 14, Bilbao 48009, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country, Barrio Sarriena, s/n, Leioa 48940, Bizkaia, Spain; Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto M5T 0S8, ON, Canada; Department of Physiology, University of Toronto, 1 King's College Circle, Toronto M5S 1A8, ON, Canada.
| |
Collapse
|
34
|
Ziontz J, Harrison TM, Chen X, Giorgio J, Adams JN, Wang Z, Jagust W, Alzheimer’s Disease Neuroimaging Initiative. Behaviorally meaningful functional networks mediate the effect of Alzheimer's pathology on cognition. Cereb Cortex 2024; 34:bhae134. [PMID: 38602736 PMCID: PMC11008686 DOI: 10.1093/cercor/bhae134] [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: 09/13/2023] [Revised: 01/25/2024] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
Tau pathology is associated with cognitive impairment in both aging and Alzheimer's disease, but the functional and structural bases of this relationship remain unclear. We hypothesized that the integrity of behaviorally meaningful functional networks would help explain the relationship between tau and cognitive performance. Using resting state fMRI, we identified unique networks related to episodic memory and executive function cognitive domains. The episodic memory network was particularly related to tau pathology measured with positron emission tomography in the entorhinal and temporal cortices. Further, episodic memory network strength mediated the relationship between tau pathology and cognitive performance above and beyond neurodegeneration. We replicated the association between these networks and tau pathology in a separate cohort of older adults, including both cognitively unimpaired and mildly impaired individuals. Together, these results suggest that behaviorally meaningful functional brain networks represent a functional mechanism linking tau pathology and cognition.
Collapse
Affiliation(s)
- Jacob Ziontz
- Helen Wills Neuroscience Institute, UC Berkeley, 250 Warren Hall, 2195 Hearst Ave, Berkeley, CA 94720, United States
| | - Theresa M Harrison
- Helen Wills Neuroscience Institute, UC Berkeley, 250 Warren Hall, 2195 Hearst Ave, Berkeley, CA 94720, United States
| | - Xi Chen
- Helen Wills Neuroscience Institute, UC Berkeley, 250 Warren Hall, 2195 Hearst Ave, Berkeley, CA 94720, United States
| | - Joseph Giorgio
- Helen Wills Neuroscience Institute, UC Berkeley, 250 Warren Hall, 2195 Hearst Ave, Berkeley, CA 94720, United States
- School of Psychological Sciences, College of Engineering, Science and the Environment, University of Newcastle, University Dr, Callaghan, Newcastle, NSW 2305, Australia
| | - Jenna N Adams
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, 1400 Biological Sciences III, University of California, Irvine, Irvine, CA 92697, United States
| | - Zehao Wang
- Helen Wills Neuroscience Institute, UC Berkeley, 250 Warren Hall, 2195 Hearst Ave, Berkeley, CA 94720, United States
| | - William Jagust
- Helen Wills Neuroscience Institute, UC Berkeley, 250 Warren Hall, 2195 Hearst Ave, Berkeley, CA 94720, United States
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, United States
| | | |
Collapse
|
35
|
Verma P, Ranasinghe K, Prasad J, Cai C, Xie X, Lerner H, Mizuiri D, Miller B, Rankin K, Vossel K, Cheung SW, Nagarajan SS, Raj A. Impaired long-range excitatory time scale predicts abnormal neural oscillations and cognitive deficits in Alzheimer's disease. Alzheimers Res Ther 2024; 16:62. [PMID: 38504361 PMCID: PMC10953266 DOI: 10.1186/s13195-024-01426-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/04/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia, progressively impairing cognitive abilities. While neuroimaging studies have revealed functional abnormalities in AD, how these relate to aberrant neuronal circuit mechanisms remains unclear. Using magnetoencephalography imaging we documented abnormal local neural synchrony patterns in patients with AD. To identify global abnormal biophysical mechanisms underlying the spatial and spectral electrophysiological patterns in AD, we estimated the parameters of a biophysical spectral graph model (SGM). METHODS SGM is an analytic neural mass model that describes how long-range fiber projections in the brain mediate the excitatory and inhibitory activity of local neuronal subpopulations. Unlike other coupled neuronal mass models, the SGM is linear, available in closed-form, and parameterized by a small set of biophysical interpretable global parameters. This facilitates their rapid and unambiguous inference which we performed here on a well-characterized clinical population of patients with AD (N = 88, age = 62.73 +/- 8.64 years) and a cohort of age-matched controls (N = 88, age = 65.07 +/- 9.92 years). RESULTS Patients with AD showed significantly elevated long-range excitatory neuronal time scales, local excitatory neuronal time scales and local inhibitory neural synaptic strength. The long-range excitatory time scale had a larger effect size, compared to local excitatory time scale and inhibitory synaptic strength and contributed highest for the accurate classification of patients with AD from controls. Furthermore, increased long-range time scale was associated with greater deficits in global cognition. CONCLUSIONS These results demonstrate that long-range excitatory time scale of neuronal activity, despite being a global measure, is a key determinant in the local spectral signatures and cognition in the human brain, and how it might be a parsimonious factor underlying altered neuronal activity in AD. Our findings provide new insights into mechanistic links between abnormal local spectral signatures and global connectivity measures in AD.
Collapse
Affiliation(s)
- Parul Verma
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Kamalini Ranasinghe
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | | | - Chang Cai
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Xihe Xie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Hannah Lerner
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Danielle Mizuiri
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Bruce Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Katherine Rankin
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Keith Vossel
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Steven W Cheung
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA, USA
- Surgical Services, Veterans Affairs, San Francisco, USA
| | - Srikantan S Nagarajan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
36
|
Kamondi A, Grigg-Damberger M, Löscher W, Tanila H, Horvath AA. Epilepsy and epileptiform activity in late-onset Alzheimer disease: clinical and pathophysiological advances, gaps and conundrums. Nat Rev Neurol 2024; 20:162-182. [PMID: 38356056 DOI: 10.1038/s41582-024-00932-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
A growing body of evidence has demonstrated a link between Alzheimer disease (AD) and epilepsy. Late-onset epilepsy and epileptiform activity can precede cognitive deterioration in AD by years, and its presence has been shown to predict a faster disease course. In animal models of AD, amyloid and tau pathology are linked to cortical network hyperexcitability that precedes the first signs of memory decline. Thus, detection of epileptiform activity in AD has substantial clinical importance as a potential novel modifiable risk factor for dementia. In this Review, we summarize the epidemiological evidence for the complex bidirectional relationship between AD and epilepsy, examine the effect of epileptiform activity and seizures on cognition in people with AD, and discuss the precision medicine treatment strategies based on the latest research in human and animal models. Finally, we outline some of the unresolved questions of the field that should be addressed by rigorous research, including whether particular clinicopathological subtypes of AD have a stronger association with epilepsy, and the sequence of events between epileptiform activity and amyloid and tau pathology.
Collapse
Affiliation(s)
- Anita Kamondi
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary.
- Department of Neurology, Semmelweis University, Budapest, Hungary.
| | | | - Wolfgang Löscher
- Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Andras Attila Horvath
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
37
|
Giorgio J, Adams JN, Maass A, Jagust WJ, Breakspear M. Amyloid induced hyperexcitability in default mode network drives medial temporal hyperactivity and early tau accumulation. Neuron 2024; 112:676-686.e4. [PMID: 38096815 PMCID: PMC10922797 DOI: 10.1016/j.neuron.2023.11.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/01/2023] [Accepted: 11/14/2023] [Indexed: 02/24/2024]
Abstract
In early Alzheimer's disease (AD) β-amyloid (Aβ) deposits throughout association cortex and tau appears in the entorhinal cortex (EC). Why these initially appear in disparate locations is not understood. Using task-based fMRI and multimodal PET imaging, we assess the impact of local AD pathology on network-to-network interactions. We show that AD pathologies flip interactions between the default mode network (DMN) and the medial temporal lobe (MTL) from inhibitory to excitatory. The DMN is hyperexcited with increasing levels of Aβ, which drives hyperexcitability within the MTL and this directed hyperexcitation of the MTL by the DMN predicts the rate of tau accumulation within the EC. Our results support a model whereby Aβ induces disruptions to local excitatory-inhibitory balance in the DMN, driving hyperexcitability in the MTL, leading to tau accumulation. We propose that Aβ-induced disruptions to excitatory-inhibitory balance is a candidate causal route between Aβ and remote EC-tau accumulation.
Collapse
Affiliation(s)
- Joseph Giorgio
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; School of Psychological Sciences, College of Engineering, Science, and the Environment, University of Newcastle, Newcastle, NSW 2305, Australia.
| | - Jenna N Adams
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael Breakspear
- School of Psychological Sciences, College of Engineering, Science, and the Environment, University of Newcastle, Newcastle, NSW 2305, Australia; Discipline of Psychiatry, College of Health, Medicine, and Wellbeing, The University of Newcastle, Newcastle, NSW 2305, Australia
| |
Collapse
|
38
|
Luppi JJ, Stam CJ, Scheltens P, de Haan W. Virtual neural network-guided optimization of non-invasive brain stimulation in Alzheimer's disease. PLoS Comput Biol 2024; 20:e1011164. [PMID: 38232116 PMCID: PMC10824453 DOI: 10.1371/journal.pcbi.1011164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/29/2024] [Accepted: 12/19/2023] [Indexed: 01/19/2024] Open
Abstract
Transcranial direct current stimulation (tDCS) is a non-invasive brain stimulation technique with potential for counteracting disrupted brain network activity in Alzheimer's disease (AD) to improve cognition. However, the results of tDCS studies in AD have been variable due to different methodological choices such as electrode placement. To address this, a virtual brain network model of AD was used to explore tDCS optimization. We compared a large, representative set of virtual tDCS intervention setups, to identify the theoretically optimized tDCS electrode positions for restoring functional network features disrupted in AD. We simulated 20 tDCS setups using a computational dynamic network model of 78 neural masses coupled according to human structural topology. AD network damage was simulated using an activity-dependent degeneration algorithm. Current flow modeling was used to estimate tDCS-targeted cortical regions for different electrode positions, and excitability of the pyramidal neurons of the corresponding neural masses was modulated to simulate tDCS. Outcome measures were relative power spectral density (alpha bands, 8-10 Hz and 10-13 Hz), total spectral power, posterior alpha peak frequency, and connectivity measures phase lag index (PLI) and amplitude envelope correlation (AEC). Virtual tDCS performance varied, with optimized strategies improving all outcome measures, while others caused further deterioration. The best performing setup involved right parietal anodal stimulation, with a contralateral supraorbital cathode. A clear correlation between the network role of stimulated regions and tDCS success was not observed. This modeling-informed approach can guide and perhaps accelerate tDCS therapy development and enhance our understanding of tDCS effects. Follow-up studies will compare the general predictions to personalized virtual models and validate them with tDCS-magnetoencephalography (MEG) in a clinical AD patient cohort.
Collapse
Affiliation(s)
- Janne J. Luppi
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Clinical Neurophysiology and MEG, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Cornelis J. Stam
- Department of Clinical Neurophysiology and MEG, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Willem de Haan
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Clinical Neurophysiology and MEG, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Yu H, Ding Y, Wei Y, Dyrba M, Wang D, Kang X, Xu W, Zhao K, Liu Y, for the Alzheimer's Disease Neuroimaging Initiative. Morphological connectivity differences in Alzheimer's disease correlate with gene transcription and cell-type. Hum Brain Mapp 2023; 44:6364-6374. [PMID: 37846762 PMCID: PMC10681645 DOI: 10.1002/hbm.26512] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/10/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most prevalent forms of dementia in older individuals. Convergent evidence suggests structural connectome abnormalities in specific brain regions are linked to AD progression. The biological basis underpinnings of these connectome changes, however, have remained elusive. We utilized an individual regional mean connectivity strength (RMCS) derived from a regional radiomics similarity network to capture altered morphological connectivity in 1654 participants (605 normal controls, 766 mild cognitive impairment [MCI], and 283 AD). Then, we also explored the biological basis behind these morphological changes through gene enrichment analysis and cell-specific analysis. We found that RMCS probes of the hippocampus and medial temporal lobe were significantly altered in AD and MCI, with these differences being spatially related to the expression of AD-risk genes. In addition, gene enrichment analysis revealed that the modulation of chemical synaptic transmission is the most relevant biological process associated with the altered RMCS in AD. Notably, neuronal cells were found to be the most pertinent cells in the altered RMCS. Our findings shed light on understanding the biological basis of structural connectome changes in AD, which may ultimately lead to more effective diagnostic and therapeutic strategies for this devastating disease.
Collapse
Affiliation(s)
- Huiying Yu
- School of Information Science and EngineeringShandong Normal UniversityJinanChina
| | - Yanhui Ding
- School of Information Science and EngineeringShandong Normal UniversityJinanChina
| | - Yongbin Wei
- School of Artificial IntelligenceBeijing University of Posts and TelecommunicationsBeijingChina
| | - Martin Dyrba
- German Center for Neurodegenerative Diseases (DZNE)RostockGermany
| | - Dong Wang
- School of Information Science and EngineeringShandong Normal UniversityJinanChina
| | - Xiaopeng Kang
- School of Artificial IntelligenceUniversity of Chinese Academy of SciencesBeijingChina
| | - Weizhi Xu
- School of Information Science and EngineeringShandong Normal UniversityJinanChina
| | - Kun Zhao
- School of Artificial IntelligenceBeijing University of Posts and TelecommunicationsBeijingChina
| | - Yong Liu
- School of Artificial IntelligenceBeijing University of Posts and TelecommunicationsBeijingChina
- School of Artificial IntelligenceUniversity of Chinese Academy of SciencesBeijingChina
| | | |
Collapse
|
40
|
Lu Z, Fu J, Wu G, Yang Z, Wu X, Wang D, You Z, Nie Z, Sheng Q. Neuroprotection and Mechanism of Gas-miR36-5p from Gastrodia elata in an Alzheimer's Disease Model by Regulating Glycogen Synthase Kinase-3β. Int J Mol Sci 2023; 24:17295. [PMID: 38139125 PMCID: PMC10744203 DOI: 10.3390/ijms242417295] [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: 11/02/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease (AD) is currently the most common neurodegenerative disease. Glycogen synthase kinase 3β (GSK-3β) is a pivotal factor in AD pathogenesis. Recent research has demonstrated that plant miRNAs exert cross-kingdom regulation on the target genes in animals. Gastrodia elata (G. elata) is a valuable traditional Chinese medicine that has significant pharmacological activity against diseases of the central nervous system (CNS). Our previous studies have indicated that G. elata-specific miRNA plays a cross-kingdom regulatory role for the NF-κB signaling pathway in mice. In this study, further bioinformatics analysis suggested that Gas-miR36-5p targets GSK-3β. Through western blot, RT-qPCR, and assessments of T-AOC, SOD, and MDA levels, Gas-miR36-5p demonstrated its neuroprotective effects in an AD cell model. Furthermore, Gas-miR36-5p was detected in the murine brain tissues. The results of the Morris water maze test and western blot analysis provided positive evidence for reversing the learning deficits and hyperphosphorylation of Tau in AD mice, elucidating significant neuroprotective effects in an AD model following G. elata RNA administration. Our research emphasizes Gas-miR36-5p as a novel G. elata-specific miRNA with neuroprotective properties in Alzheimer's disease by targeting GSK-3β. Consequently, our findings provide valuable insights into the cross-kingdom regulatory mechanisms underlying G. elata-specific miRNA, presenting a novel perspective for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qing Sheng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
41
|
Dastgheib ZA, Lithgow BJ, Moussavi ZK. Evaluating the Diagnostic Value of Electrovestibulography (EVestG) in Alzheimer's Patients with Mixed Pathology: A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2091. [PMID: 38138194 PMCID: PMC10744488 DOI: 10.3390/medicina59122091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/24/2023] [Accepted: 11/26/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: Diagnosis of dementia subtypes caused by different brain pathophysiologies, particularly Alzheimer's disease (AD) from AD mixed with levels of cerebrovascular disease (CVD) symptomology (AD-CVD), is challenging due to overlapping symptoms. In this pilot study, the potential of Electrovestibulography (EVestG) for identifying AD, AD-CVD, and healthy control populations was investigated. Materials and Methods: A novel hierarchical multiclass diagnostic algorithm based on the outcomes of its lower levels of binary classifications was developed using data of 16 patients with AD, 13 with AD-CVD, and 24 healthy age-matched controls, and then evaluated on a blind testing dataset made up of a new population of 12 patients diagnosed with AD, 9 with AD-CVD, and 8 healthy controls. Multivariate analysis was run to test the between population differences while controlling for sex and age covariates. Results: The accuracies of the multiclass diagnostic algorithm were found to be 85.7% and 79.6% for the training and blind testing datasets, respectively. While a statistically significant difference was found between the populations after accounting for sex and age, no significant effect was found for sex or age covariates. The best characteristic EVestG features were extracted from the upright sitting and supine up/down stimulus responses. Conclusions: Two EVestG movements (stimuli) and their most informative features that are best selective of the above-populations' separations were identified, and a hierarchy diagnostic algorithm was developed for three-way classification. Given that the two stimuli predominantly stimulate the otholithic organs, physiological and experimental evidence supportive of the results are presented. Disruptions of inhibition associated with GABAergic activity might be responsible for the changes in the EVestG features.
Collapse
Affiliation(s)
| | | | - Zahra K. Moussavi
- Diagnostic and Neurological Processing Research Laboratory, Biomedical Engineering Program, University of Manitoba, Riverview Health Centre, Winnipeg, MB R3L 2P4, Canada; (Z.A.D.); (B.J.L.)
| |
Collapse
|
42
|
Zegarra-Valdivia JA, Pignatelli J, Nuñez A, Torres Aleman I. The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer's Disease. Int J Mol Sci 2023; 24:16440. [PMID: 38003628 PMCID: PMC10671249 DOI: 10.3390/ijms242216440] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Despite decades of intense research, disease-modifying therapeutic approaches for Alzheimer's disease (AD) are still very much needed. Apart from the extensively analyzed tau and amyloid pathological cascades, two promising avenues of research that may eventually identify new druggable targets for AD are based on a better understanding of the mechanisms of resilience and vulnerability to this condition. We argue that insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. We postulate that preserved brain IGF-I activity contributes to resilience to AD pathology as this growth factor intervenes in all the major pathological cascades considered to be involved in AD, including metabolic impairment, altered proteostasis, and inflammation, to name the three that are considered to be the most important ones. Conversely, disturbed IGF-I activity is found in many AD risk factors, such as old age, type 2 diabetes, imbalanced diet, sedentary life, sociality, stroke, stress, and low education, whereas the Apolipoprotein (Apo) E4 genotype and traumatic brain injury may also be influenced by brain IGF-I activity. Accordingly, IGF-I activity should be taken into consideration when analyzing these processes, while its preservation will predictably help prevent the progress of AD pathology. Thus, we need to define IGF-I activity in all these conditions and develop a means to preserve it. However, defining brain IGF-I activity cannot be solely based on humoral or tissue levels of this neurotrophic factor, and new functionally based assessments need to be developed.
Collapse
Affiliation(s)
- Jonathan A. Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- School of Medicine, Universidad Señor de Sipán, Chiclayo 14000, Peru
| | - Jaime Pignatelli
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Cajal Institute (CSIC), 28002 Madrid, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain;
- Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
43
|
Chen L, Christenson Wick Z, Vetere LM, Vaughan N, Jurkowski A, Galas A, Diego KS, Philipsberg PA, Soler I, Feng Y, Cai DJ, Shuman T. Progressive Excitability Changes in the Medial Entorhinal Cortex in the 3xTg Mouse Model of Alzheimer's Disease Pathology. J Neurosci 2023; 43:7441-7454. [PMID: 37714705 PMCID: PMC10621765 DOI: 10.1523/jneurosci.1204-23.2023] [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: 06/29/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by memory loss and progressive cognitive impairments. In mouse models of AD pathology, studies have found neuronal and synaptic deficits in hippocampus, but less is known about changes in medial entorhinal cortex (MEC), which is the primary spatial input to the hippocampus and an early site of AD pathology. Here, we measured neuronal intrinsic excitability and synaptic activity in MEC layer II (MECII) stellate cells, MECII pyramidal cells, and MEC layer III (MECIII) excitatory neurons at 3 and 10 months of age in the 3xTg mouse model of AD pathology, using male and female mice. At 3 months of age, before the onset of memory impairments, we found early hyperexcitability in intrinsic properties of MECII stellate and pyramidal cells, but this was balanced by a relative reduction in synaptic excitation (E) compared with inhibition (I; E/I ratio), suggesting intact homeostatic mechanisms regulating MECII activity. Conversely, MECIII neurons had reduced intrinsic excitability at this early time point with no change in synaptic E/I ratio. By 10 months of age, after the onset of memory deficits, neuronal excitability of MECII pyramidal cells and MECIII excitatory neurons was largely normalized in 3xTg mice. However, MECII stellate cells remained hyperexcitable, and this was further exacerbated by an increased synaptic E/I ratio. This observed combination of increased intrinsic and synaptic hyperexcitability suggests a breakdown in homeostatic mechanisms specifically in MECII stellate cells at this postsymptomatic time point, which may contribute to the emergence of memory deficits in AD.SIGNIFICANCE STATEMENT AD causes cognitive deficits, but the specific neural circuits that are damaged to drive changes in memory remain unknown. Using a mouse model of AD pathology that expresses both amyloid and tau transgenes, we found that neurons in the MEC have altered excitability. Before the onset of memory impairments, neurons in layer 2 of MEC had increased intrinsic excitability, but this was balanced by reduced inputs onto the cell. However, after the onset of memory impairments, stellate cells in MEC became further hyperexcitable, with increased excitability exacerbated by increased synaptic inputs. Thus, it appears that MEC stellate cells are uniquely disrupted during the progression of memory deficits and may contribute to cognitive deficits in AD.
Collapse
Affiliation(s)
- Lingxuan Chen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California 92697
| | - Zoé Christenson Wick
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Lauren M Vetere
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Nick Vaughan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Albert Jurkowski
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Hunter College, City University of New York, New York, New York 10065
| | - Angelina Galas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- New York University, New York, New York 10012
| | - Keziah S Diego
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Paul A Philipsberg
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Ivan Soler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Yu Feng
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Denise J Cai
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Tristan Shuman
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
44
|
Quintela-López T, Lezmy J. Homeostatic plasticity of axonal excitable sites in Alzheimer's disease. Front Neurosci 2023; 17:1277251. [PMID: 37937068 PMCID: PMC10626477 DOI: 10.3389/fnins.2023.1277251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Affiliation(s)
| | - Jonathan Lezmy
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
45
|
Chen H, Cai J, Wang A, Su W, Ji C, Zhao L. Treadmill exercise prevents the hyperexcitability of pyramidal neurons in medial entorhinal cortex in the 3xTg-AD mouse model of Alzheimer's disease. Exp Gerontol 2023; 182:112309. [PMID: 37832802 DOI: 10.1016/j.exger.2023.112309] [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: 09/06/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Neuronal hyperactivity is a key abnormality in early stage Alzheimer's disease (AD). Medial entorhinal cortex (mEC) plays a vital role in memory function and is affected early in AD. Growing evidence indicates benefits of regular exercise on memory and cognitive function in humans with AD, although, the underlying mechanisms are not clear. Therefore, this study was designed to test the effects of 16 weeks treadmill exercise on spatial learning memory and the underlying cellular mechanisms in 6-month-old 3xTg-AD mice. Whole-cell patch clamp was used to examine neuronal intrinsic excitability, spontaneous excitatory postsynaptic currents (sEPSCs) and spontaneous inhibitory postsynaptic currents (sIPSCs) of mEC layer II/III pyramidal neurons in the following groups: wild type (WT + sham), 3xTg-AD (AD+sham), WT receiving exercise (WT + Ex), and AD receiving exercise (AD+Ex). We found that at a behavioral level, treadmill exercise decreased working memory errors in radial arm maze (RAM) test in 6-month-old AD mice. At a cellular level, we found that treadmill exercise prevented the abnormal increase in mEC pyramidal neuron input resistance and action potential firing in 6-month-old 3xTg-AD mice compared with WT + sham and AD+Ex mice; further, sEPSC amplitude and frequency were normal in AD+Ex but overactive in AD+sham; additionally, GABAergic inhibition was normal in AD+Ex mice but reduced in AD+sham. In conclusion, our results indicate that treadmill exercise improves spatial learning memory and prevents network hyperexcitability in mEC by reducing pyramidal neuronal intrinsic excitability and normalizing excitatory and inhibitory synaptic transmission in 3xTg-AD mice.
Collapse
Affiliation(s)
- Huimin Chen
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China.
| | - Jiajia Cai
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Aozhe Wang
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Wantang Su
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China.
| | - Chunyan Ji
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China.
| |
Collapse
|
46
|
Grieco SF, Holmes TC, Xu X. Probing neural circuit mechanisms in Alzheimer's disease using novel technologies. Mol Psychiatry 2023; 28:4407-4420. [PMID: 36959497 PMCID: PMC10827671 DOI: 10.1038/s41380-023-02018-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
The study of Alzheimer's Disease (AD) has traditionally focused on neuropathological mechanisms that has guided therapies that attenuate neuropathological features. A new direction is emerging in AD research that focuses on the progressive loss of cognitive function due to disrupted neural circuit mechanisms. Evidence from humans and animal models of AD show that dysregulated circuits initiate a cascade of pathological events that culminate in functional loss of learning, memory, and other aspects of cognition. Recent progress in single-cell, spatial, and circuit omics informs this circuit-focused approach by determining the identities, locations, and circuitry of the specific cells affected by AD. Recently developed neuroscience tools allow for precise access to cell type-specific circuitry so that their functional roles in AD-related cognitive deficits and disease progression can be tested. An integrated systems-level understanding of AD-associated neural circuit mechanisms requires new multimodal and multi-scale interrogations that longitudinally measure and/or manipulate the ensemble properties of specific molecularly-defined neuron populations first susceptible to AD. These newly developed technological and conceptual advances present new opportunities for studying and treating circuits vulnerable in AD and represent the beginning of a new era for circuit-based AD research.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
47
|
Vogel JW, Corriveau-Lecavalier N, Franzmeier N, Pereira JB, Brown JA, Maass A, Botha H, Seeley WW, Bassett DS, Jones DT, Ewers M. Connectome-based modelling of neurodegenerative diseases: towards precision medicine and mechanistic insight. Nat Rev Neurosci 2023; 24:620-639. [PMID: 37620599 DOI: 10.1038/s41583-023-00731-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
Neurodegenerative diseases are the most common cause of dementia. Although their underlying molecular pathologies have been identified, there is substantial heterogeneity in the patterns of progressive brain alterations across and within these diseases. Recent advances in neuroimaging methods have revealed that pathological proteins accumulate along specific macroscale brain networks, implicating the network architecture of the brain in the system-level pathophysiology of neurodegenerative diseases. However, the extent to which 'network-based neurodegeneration' applies across the wide range of neurodegenerative disorders remains unclear. Here, we discuss the state-of-the-art of neuroimaging-based connectomics for the mapping and prediction of neurodegenerative processes. We review findings supporting brain networks as passive conduits through which pathological proteins spread. As an alternative view, we also discuss complementary work suggesting that network alterations actively modulate the spreading of pathological proteins between connected brain regions. We conclude this Perspective by proposing an integrative framework in which connectome-based models can be advanced along three dimensions of innovation: incorporating parameters that modulate propagation behaviour on the basis of measurable biological features; building patient-tailored models that use individual-level information and allowing model parameters to interact dynamically over time. We discuss promises and pitfalls of these strategies for improving disease insights and moving towards precision medicine.
Collapse
Affiliation(s)
- Jacob W Vogel
- Department of Clinical Sciences, SciLifeLab, Lund University, Lund, Sweden.
| | - Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Acadamy, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Neuro Division, Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden
| | - Jesse A Brown
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Dani S Bassett
- Departments of Bioengineering, Electrical and Systems Engineering, Physics and Astronomy, Neurology and Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
48
|
Lam AD. Linking Late-Onset Epilepsy With Alzheimer Disease: Insights From Plasma Amyloid Measurements. Neurology 2023; 101:551-552. [PMID: 37541840 DOI: 10.1212/wnl.0000000000207683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 08/06/2023] Open
Affiliation(s)
- Alice D Lam
- From the Department of Neurology (A.D.L.), Massachusetts General Hospital; and Harvard Medical School (A.D.L.), Boston, MA.
| |
Collapse
|
49
|
Melo de Farias AR, Pelletier A, Iohan LCC, Saha O, Bonnefond A, Amouyel P, Delahaye F, Lambert JC, Costa MR. Amyloid-Beta Peptides Trigger Premature Functional and Gene Expression Alterations in Human-Induced Neurons. Biomedicines 2023; 11:2564. [PMID: 37761004 PMCID: PMC10526858 DOI: 10.3390/biomedicines11092564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia in the elderly, characterized by the presence of amyloid-beta (Aβ) plaques, neurofibrillary tangles, neuroinflammation, synapse loss and neurodegeneration in the brain. The amyloid cascade hypothesis postulates that deposition of Aβ peptides is the causative agent of AD pathology, but we still lack comprehensive understanding of the molecular mechanisms connecting Aβ peptides to neuronal dysfunctions in AD. In this work, we investigate the early effects of Aβ peptide accumulation on the functional properties and gene expression profiles of human-induced neurons (hiNs). We show that hiNs acutely exposed to low concentrations of both cell-secreted Aβ peptides or synthetic Aβ1-42 exhibit alterations in the frequency of calcium transients suggestive of increased neuronal excitability. Using single-cell RNA sequencing, we also show that cell-secreted Aβ up-regulates the expression of several synapse-related genes and down-regulates the expression of genes associated with metabolic stress mainly in glutamatergic neurons and, to a lesser degree, in GABAergic neurons and astrocytes. These neuronal alterations correlate with activation of the SEMA5, EPHA and NECTIN signaling pathways, which are important regulators of synaptic plasticity. Altogether, our findings indicate that slight elevations in Aβ concentrations are sufficient to elicit transcriptional changes in human neurons, which can contribute to early alterations in neural network activity.
Collapse
Affiliation(s)
- Ana Raquel Melo de Farias
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019 Lille, France; (A.R.M.d.F.); (O.S.); (P.A.); (J.-C.L.)
- Brain Institute, Federal University of Rio Grande do Norte, Campus Universitário Lagoa Nova, Av. Senador Salgado Filho, 3000, Natal 59078-970, Brazil
| | - Alexandre Pelletier
- Université de Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, CEDEX, 59045 Lille, France; (A.P.); (A.B.); (F.D.)
| | - Lukas Cruz Carvalho Iohan
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil;
| | - Orthis Saha
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019 Lille, France; (A.R.M.d.F.); (O.S.); (P.A.); (J.-C.L.)
| | - Amélie Bonnefond
- Université de Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, CEDEX, 59045 Lille, France; (A.P.); (A.B.); (F.D.)
| | - Philippe Amouyel
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019 Lille, France; (A.R.M.d.F.); (O.S.); (P.A.); (J.-C.L.)
| | - Fabien Delahaye
- Université de Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, CEDEX, 59045 Lille, France; (A.P.); (A.B.); (F.D.)
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019 Lille, France; (A.R.M.d.F.); (O.S.); (P.A.); (J.-C.L.)
| | - Marcos R. Costa
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019 Lille, France; (A.R.M.d.F.); (O.S.); (P.A.); (J.-C.L.)
- Brain Institute, Federal University of Rio Grande do Norte, Campus Universitário Lagoa Nova, Av. Senador Salgado Filho, 3000, Natal 59078-970, Brazil
| |
Collapse
|
50
|
Martínez‐Cañada P, Perez‐Valero E, Minguillon J, Pelayo F, López‐Gordo MA, Morillas C. Combining aperiodic 1/f slopes and brain simulation: An EEG/MEG proxy marker of excitation/inhibition imbalance in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12477. [PMID: 37662693 PMCID: PMC10474329 DOI: 10.1002/dad2.12477] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/27/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023]
Abstract
INTRODUCTION Accumulation and interaction of amyloid-beta (Aβ) and tau proteins during progression of Alzheimer's disease (AD) are shown to tilt neuronal circuits away from balanced excitation/inhibition (E/I). Current available techniques for noninvasive interrogation of E/I in the intact human brain, for example, magnetic resonance spectroscopy (MRS), are highly restrictive (i.e., limited spatial extent), have low temporal and spatial resolution and suffer from the limited ability to distinguish accurately between different neurotransmitters complicating its interpretation. As such, these methods alone offer an incomplete explanation of E/I. Recently, the aperiodic component of neural power spectrum, often referred to in the literature as the '1/f slope', has been described as a promising and scalable biomarker that can track disruptions in E/I potentially underlying a spectrum of clinical conditions, such as autism, schizophrenia, or epilepsy, as well as developmental E/I changes as seen in aging. METHODS Using 1/f slopes from resting-state spectral data and computational modeling, we developed a new method for inferring E/I alterations in AD. RESULTS We tested our method on recent freely and publicly available electroencephalography (EEG) and magnetoencephalography (MEG) datasets of patients with AD or prodromal disease and demonstrated the method's potential for uncovering regional patterns of abnormal excitatory and inhibitory parameters. DISCUSSION Our results provide a general framework for investigating circuit-level disorders in AD and developing therapeutic interventions that aim to restore the balance between excitation and inhibition.
Collapse
Affiliation(s)
- Pablo Martínez‐Cañada
- Department of Computer EngineeringAutomation and RoboticsUniversity of GranadaGranadaSpain
- Research Centre for Information and Communications Technologies (CITIC)University of GranadaGranadaSpain
| | - Eduardo Perez‐Valero
- Department of Computer EngineeringAutomation and RoboticsUniversity of GranadaGranadaSpain
- Research Centre for Information and Communications Technologies (CITIC)University of GranadaGranadaSpain
| | - Jesus Minguillon
- Research Centre for Information and Communications Technologies (CITIC)University of GranadaGranadaSpain
- Department of Signal TheoryTelematics and CommunicationsUniversity of GranadaGranadaSpain
| | - Francisco Pelayo
- Department of Computer EngineeringAutomation and RoboticsUniversity of GranadaGranadaSpain
- Research Centre for Information and Communications Technologies (CITIC)University of GranadaGranadaSpain
| | - Miguel A. López‐Gordo
- Research Centre for Information and Communications Technologies (CITIC)University of GranadaGranadaSpain
- Department of Signal TheoryTelematics and CommunicationsUniversity of GranadaGranadaSpain
| | - Christian Morillas
- Department of Computer EngineeringAutomation and RoboticsUniversity of GranadaGranadaSpain
- Research Centre for Information and Communications Technologies (CITIC)University of GranadaGranadaSpain
| |
Collapse
|