1
|
Madadi Asl M, Valizadeh A. Entrainment by transcranial alternating current stimulation: Insights from models of cortical oscillations and dynamical systems theory. Phys Life Rev 2025; 53:147-176. [PMID: 40106964 DOI: 10.1016/j.plrev.2025.03.008] [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: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Signature of neuronal oscillations can be found in nearly every brain function. However, abnormal oscillatory activity is linked with several brain disorders. Transcranial alternating current stimulation (tACS) is a non-invasive brain stimulation technique that can potentially modulate neuronal oscillations and influence behavior both in health and disease. Yet, a complete understanding of how interacting networks of neurons are affected by tACS remains elusive. Entrainment effects by which tACS synchronizes neuronal oscillations is one of the main hypothesized mechanisms, as evidenced in animals and humans. Computational models of cortical oscillations may shed light on the entrainment effects of tACS, but current modeling studies lack specific guidelines to inform experimental investigations. This study addresses the existing gap in understanding the mechanisms of tACS effects on rhythmogenesis within the brain by providing a comprehensive overview of both theoretical and experimental perspectives. We explore the intricate interactions between oscillators and periodic stimulation through the lens of dynamical systems theory. Subsequently, we present a synthesis of experimental findings that demonstrate the effects of tACS on both individual neurons and collective oscillatory patterns in animal models and humans. Our review extends to computational investigations that elucidate the interplay between tACS and neuronal dynamics across diverse cortical network models. To illustrate these concepts, we conclude with a simple oscillatory neuron model, showcasing how fundamental theories of oscillatory behavior derived from dynamical systems, such as phase response of neurons to external perturbation, can account for the entrainment effects observed with tACS. Studies reviewed here render the necessity of integrated experimental and computational approaches for effective neuromodulation by tACS in health and disease.
Collapse
Affiliation(s)
- Mojtaba Madadi Asl
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran; Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran, Iran.
| | - Alireza Valizadeh
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran, Iran; Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran; The Zapata-Briceño Institute of Neuroscience, Madrid, Spain
| |
Collapse
|
2
|
Srivastava I, Goikolea J, Ayberk Kaya T, Latorre-Leal M, Eroli F, Pereira Iglesias M, Álvarez-Jiménez L, Arroyo-García LE, Shimozawa M, Nilsson P, Fisahn A, Lindskog M, Maioli S, Loera-Valencia R. Reactive Astrocytes with Reduced Function of Glutamate Transporters in the AppNL-G-F Knock-in Mice. ACS Chem Neurosci 2025. [PMID: 40421586 DOI: 10.1021/acschemneuro.4c00714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Alzheimer's disease (AD) is associated with synaptic and memory dysfunction. One of the hallmarks of AD is reactive astrogliosis, with reactive astrocytes surrounding amyloid plaques in the brain. Astrocytes have also been shown to be actively involved in disease progression, nevertheless, mechanistic information about their role in synaptic transmission during AD pathology is lacking. Astrocytes maintain synaptic transmission by taking up extracellular glutamate during synaptic activity through astrocytic glutamate transporter GLT-1, but its function has been difficult to measure in real-time in AD pathology. Here, we used an App knock-in AD model (AppNL-G-F) carrying the Swedish, Arctic and Beyreuther mutations associated with AD and exhibiting AD-like Aβ plaque deposition and memory impairment. Using immunohistochemistry, patch-clamp of astrocytes, and Western blot from tissue and FACS isolated synaptosomes, we found that AppNL-G-F mice at 6-8 months of age have astrocytes with clearly altered morphology compared to wild-type (WT). Moreover, astrocyte glutamate clearance function in AppNL-G-F mice, measured as electrophysiological recordings of glutamate transporter currents, was severely impaired compared to WT animals. The reduction of glutamate uptake by astrocytes cannot be explained by GLT-1 protein levels, which were unchanged in synaptosomes and hippocampus of AppNL-G-F mice. Our data suggest that astrocytic glutamate transporters are affected by excess Aβ42 in the brain contributing to synaptic dysfunction in the hippocampus. This data contributes to the notion of restoring astrocyte synaptic function as a potential therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Ipsit Srivastava
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Julen Goikolea
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Tamer Ayberk Kaya
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - María Latorre-Leal
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Francesca Eroli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Marta Pereira Iglesias
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Laura Álvarez-Jiménez
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Luis Enrique Arroyo-García
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Makoto Shimozawa
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - André Fisahn
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Maria Lindskog
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Silvia Maioli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Raúl Loera-Valencia
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Chihuahua, Av. H. Colegio Militar 4700, Nombre de Dios, 31150 Chihuahua, Chih. Mexico
| |
Collapse
|
3
|
Nong Y, Kim JS, Jia L, Arancio O, Wang Q. The interaction between neurotransmitter receptor activity and amyloid-β pathology in Alzheimer's disease. J Alzheimers Dis 2025:13872877251342273. [PMID: 40388923 DOI: 10.1177/13872877251342273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
The accumulation of amyloid-β (Aβ) peptides is a hallmark of Alzheimer's disease (AD). Central to AD pathology is the production of Aβ peptides through the amyloidogenic processing of amyloid-β protein precursor (AβPP) by β-secretase (BACE-1) and γ-secretase. Recent studies have shifted focus from Aβ plaque deposits to the more toxic soluble Aβ oligomers. One significant way in which Aβ peptides impair neuronal information processing is by influencing neurotransmitter receptor function. These receptors, including adrenergic, acetylcholine, dopamine, 5-HT, glutamate, and gamma-aminobutyric acid (GABA) receptors, play a crucial role in regulating synaptic transmission, which underlies perceptual and cognitive functions. This review explores how Aβ interacts with these key neurotransmitter receptors and how these interactions contribute to neural dysfunction in AD. Moreover, we examine how agonists and antagonists of these receptors influence Aβ pathology, offering new perspectives on potential therapeutic strategies to curb AD progression effectively and improve patients' quality of life.
Collapse
Affiliation(s)
- Yuhan Nong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jung Soo Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Litian Jia
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ottavio Arancio
- Departments of Pathology & Cell Biology, and Medicine, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Neurosurgery, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Alcantara-Gonzalez D, Kennedy M, Criscuolo C, Botterill J, Scharfman HE. Increased excitability of dentate gyrus mossy cells occurs early in life in the Tg2576 model of Alzheimer's disease. Alzheimers Res Ther 2025; 17:105. [PMID: 40375112 PMCID: PMC12079945 DOI: 10.1186/s13195-025-01747-1] [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: 03/07/2024] [Accepted: 04/22/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Hyperexcitability in Alzheimer's disease (AD) is proposed to emerge early and contribute to disease progression. The dentate gyrus (DG) and its primary cell type, granule cells (GCs) are implicated in hyperexcitability in AD. Hence, we hypothesized that mossy cells (MCs), important regulators of GC excitability, contribute to early hyperexcitability in AD. Indeed, MCs and GCs are linked to hyperexcitability in epilepsy. METHODS Using the Tg2576 model of AD and WT mice (~ 1 month-old), we compared MCs and GCs electrophysiologically and morphologically, assessed the activity marker c-Fos, Aβ expression and a hippocampal- and MC-dependent memory task that is impaired at 3-4 months of age in Tg2576 mice. RESULTS Tg2576 MCs had increased spontaneous excitatory events (sEPSP/Cs) and decreased spontaneous inhibitory currents (sIPSCs), increasing the excitation/inhibition ratio. Additionally, Tg2576 MC intrinsic excitability was enhanced. Consistent with in vitro results, Tg2576 MCs showed enhanced c-Fos protein expression. Tg2576 MCs had increased intracellular Aβ expression, suggesting a reason for increased excitability. GCs showed increased excitatory and inhibitory input without changes in intrinsic properties, consistent with effects of increased MC activity. In support, increased GC activity was normalized by an antagonist of MC input to GCs. Also in support, Tg2576 MC axons showed sprouting to the area of GC dendrites. These effects occurred before an impairment in the memory task, suggesting they are extremely early alterations. CONCLUSIONS Alterations in Tg2576 MCs and GCs early in life suggest an early role for MCs in increased GC excitability. MCs may be a novel target to intervene in AD pathophysiology at early stages.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA.
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York City, NY, 10016, USA.
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York City, NY, 10016, USA
| | - Justin Botterill
- College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd. Bldg. 39, Orangeburg, NY, 10962, USA.
- Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York City, NY, 10016, USA.
- Neuroscience Institute, New York University Langone Health, New York City, NY, 10016, USA.
| |
Collapse
|
5
|
Sarti G, Traini C, Magni G, Attorre S, Tognozzi G, Calussi E, Giovannini MG, Vannucchi MG, Lana D. Chronic administration of prebiotics and probiotics prevent pathophysiological hallmarks of Alzheimer's disease in the cortex of APP/PS1 mice. Front Pharmacol 2025; 16:1596469. [PMID: 40444050 PMCID: PMC12119559 DOI: 10.3389/fphar.2025.1596469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/30/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Dysbiosis is a characteristic of patients with Alzheimer's disease (AD). The disbalance between Gram-negative and Gram-positive bacteria causes increased production of beta-amyloid (Aβ) in the gut, which can contribute to brain accumulation of Aβ. Recovering microbiota composition with symbiotic administration of prebiotics and probiotics may be a strategy to prevent or reduce AD symptomathology. The aim of this research was to study whether chronic administration of pre- and probiotics modifies the histopathological signs of neurodegeneration in the cortex of APP/PS1 mice, a transgenic mouse model of AD. We focused on neuritic plaques deposition, neuronal degeneration and glia activation. Methods Transgenic (TG) mice and Wild type (WT) littermates were fed daily with a diet supplemented with prebiotics (a multi-extract of fibers and plant complexes, containing inulin/fruit-oligosaccharides) and probiotics (a 50%-50% mixture of Lactobacillus rhamnosus and Lactobacillus paracasei). The treatment started at 2 months of age and lasted for 6 months. Controls were WT and TG mice fed with a standard diet. All groups were evaluated qualitatively and quantitatively by immunofluorescence, confocal microscopy and digital imaging. Cortical sections were immunostained for neuritic plaques, neurons, astrocytes, microglia, and inflammatory proteins. Qualitative and quantitative analyses were carried out by immunofluorescence, confocal microscopy and digital imaging with ImageJ software. Results Quantitative analyses in TG mice demonstrated intense Aβ load and accumulation of neurofilament heavy polypeptide (NHP) in neuritic plaques, neuronal degeneration, shrinkage of the cortex, increase of GFAP expression, and microglia and astrocytes activation. All these effects were mainly evident in cortical Layer 5. The symbiotic treatment with pre- and probiotics decreased Aβ deposition and neuritic plaques in the frontoparietal cortex. In addition, the treatment decreased the degeneration of neurons, the cortical shrinkage, increased GFAP expression, and modified microglia phenomic, decreasing significantly microglia activation. The abovementioned effects of the treatment were mostly evident in cortical Layer 5. Discussion These data confirm that prolonged dietary regimen enriched with pre- and probiotics counteracts many of the histopathological hallmarks of AD, and poses the bases for a simple, affordable treatment that may help prevent AD.
Collapse
Affiliation(s)
- Giorgia Sarti
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Chiara Traini
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Giada Magni
- Cnr-Institute of Applied Physics “Nello Carrara”, Sesto Fiorentino, Italy
| | - Selene Attorre
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Giorgio Tognozzi
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Edoardo Calussi
- Section of Pathological Anatomy, Careggi University Hospital, Florence, Italy
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Maria Giuliana Vannucchi
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
6
|
Cao W, Tan Z, Berackey BT, Nguyen JK, Brown SR, Du S, Lin B, Ye Q, Seiler M, Holmes TC, Xu X. An AAV capsid proposed as microglia-targeting directs genetic expression in forebrain excitatory neurons. CELL REPORTS METHODS 2025:101054. [PMID: 40403726 DOI: 10.1016/j.crmeth.2025.101054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/18/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025]
Abstract
A newly developed capsid AAV-MG1.2 was reported to mediate specific microglial transduction. However, we find that AAV-MG1.2 actually enables specific genetic access to excitatory neurons in forebrain regions including hippocampal formation and visual cortex but does not confer expression in microglia or astrocytes in vivo. Furthermore, we find that AAV-MG1.2 specifically labels the deep layer of the CA1 pyramidal layer in a titer-dependent manner. We show that AAV-MG1.2-Cre can be used to genetically target excitatory neurons for cell-type-specific neural circuit mapping studies. We also find that AAV-MG1.2 conserves specificity for excitatory neurons in rat hippocampus. Thus, the AAV-MG1.2 presents a useful viral-genetic tool for targeting excitatory neurons in the forebrain across different species.
Collapse
Affiliation(s)
- Wenhao Cao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Zhiqun Tan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Bereket T Berackey
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Jason K Nguyen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Sara R Brown
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Shiyang Du
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Bin Lin
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Department of Physical Medicine & Rehabilitation, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Qiao Ye
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Magdalene Seiler
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Department of Physical Medicine & Rehabilitation, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
7
|
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
|
8
|
Cretin B. Epileptic variant in the spectrum of Alzheimer's disease - practical implications. Seizure 2025; 128:133-139. [PMID: 39343706 DOI: 10.1016/j.seizure.2024.09.015] [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: 03/30/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Alzheimer's disease (AD) is known to be associated with an increased risk of epilepsy, which is not exclusively related to the late stage of the disease - when a major cognitive impairment is observed, previously known as the dementia stage - but also to its prodromal stage (mild cognitive impairment). Moreover, published case reports and cohorts have shown that epilepsy may occur even earlier, at the preclinical stage of AD: Epileptic seizures may therefore be the sole objective manifestation of the disease. Such a situation is called the epileptic variant of AD (evAD). EvAD is one of the etiologies of late-onset epilepsy, which means that it carries a risk of later progression to dementia and that it can only be diagnosed by assessing amyloid and tau biomarkers. However, evAD is a window of therapeutic opportunity that is probably optimal for preventing, through antiseizure medication treatment, the accelerated cognitive decline associated with AD-related brain hyperexcitability (manifested by seizures or interictal epileptiform activities).
Collapse
Affiliation(s)
- Benjamin Cretin
- Centre Mémoire, de Ressources et de Recherche de Strasbourg, France; Unité de Neuropsychologie, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Strasbourg, France; University of Strasbourg and CNRS, ICube laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS/Neurocrypto Strasbourg, France; Centre de Compétences des démences rares des Hôpitaux Universitaires de Strasbourg, France.
| |
Collapse
|
9
|
Martín-Belmonte A, Aguado C, Alfaro-Ruíz R, Luján R. G protein-gated inwardly rectifying K + (GIRK/K ir3) channels: Molecular, cellular, and subcellular diversity. Histol Histopathol 2025; 40:597-620. [PMID: 39434650 DOI: 10.14670/hh-18-822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels are mainly expressed in excitable cells such as neurons and atrial myocytes, where they can respond to a wide variety of neurotransmitters. Four GIRK subunits have been found in mammals (GIRK1-4) and act as downstream targets for various Gαi/o-linked G protein-coupled receptors (GPCRs). Activation of GIRK channels produces a postsynaptic efflux of potassium from the cell, responsible for hyperpolarization/inhibition of the neuron. A growing body of evidence suggests that dysregulation of GIRK signalling can lead to excessive or deficient neuronal excitability, which contributes to neurological diseases and disorders. Therefore, GIRK channels are proposed as new pharmacological targets. The function of GIRK channels in neurons is not only determined by their biophysical properties but also by their cellular and subcellular localization patterns and densities on the neuronal surface. GIRK channels can be located within several subcellular compartments, where they have many different functional implications. This subcellular localization changes dynamically along the neuronal surface in response to drug intake. Ongoing research is focusing on determining the proteins that form macromolecular complexes with GIRK channels and are responsible for fast and precise signalling under physiological conditions, and how their alteration is implicated in pathological conditions. In this review, the distinct regional, cellular, and subcellular distribution of GIRK channel subunits in the brain will be discussed in view of their possible functional and pathological implications.
Collapse
Affiliation(s)
- Alejandro Martín-Belmonte
- Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| | - Rocio Alfaro-Ruíz
- Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB-UCLM), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Albacete, Spain.
| |
Collapse
|
10
|
Jin J, Fu C, Xia J, Luo H, Wang X, Chen S, Mao H, Yuan K, Lu L, Xiong W, Zou G. Cannabidiol ameliorates cognitive decline in 5×FAD mouse model of Alzheimer's disease through potentiating the function of extrasynaptic glycine receptors. Mol Psychiatry 2025; 30:1817-1827. [PMID: 39396064 DOI: 10.1038/s41380-024-02789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Emerging evidence supports the therapeutic potential of cannabinoids in Alzheimer's disease (AD), but the underlying mechanism upon how cannabinoids impact brain cognition and AD pathology remains unclear. Here we show that chronic cannabidiol (CBD) administration significantly mitigates cognitive deficiency and hippocampal β-amyloid (Aβ) pathology in 5×FAD mouse model of AD. CBD achieves its curative effect mainly through potentiating the function of inhibitory extrasynaptic glycine receptor (GlyR) in hippocampal dentate gyrus (DG). Based on the in vitro and in vivo electrophysiological recording and calcium imaging, CBD mediated anti-AD effects via GlyR are mainly accomplished by decreasing neuronal hyperactivity of granule cells in the DG of AD mice. Furthermore, the AAV-mediated ablation of DG GlyRα1, or the GlyRα1S296A mutation that exclusively disrupts CBD binding, significantly intercepts the anti-AD effect of CBD. These findings suggest a GlyR dependent mechanism underlying the therapeutic potential of CBD in the treatment of AD.
Collapse
Grants
- 32225020, 91849206, 91942315, 92049304, 32121002, 81901157, 82241032 National Natural Science Foundation of China (National Science Foundation of China)
- 32225020, 91849206, 91942315, 92049304, 32121002 National Natural Science Foundation of China (National Science Foundation of China)
Collapse
Affiliation(s)
- Jin Jin
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Chonglei Fu
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China
| | - Jing Xia
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Heyi Luo
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xianglian Wang
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Si Chen
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Huanhuan Mao
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, 100191, Beijing, China
| | - Lin Lu
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, 100191, Beijing, China.
| | - Wei Xiong
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China.
| |
Collapse
|
11
|
Mukherjee AG, Mishra S, Gopalakrishnan AV, Kannampuzha S, Murali R, Wanjari UR, B S, Vellingiri B, Madhyastha H, Kanagavel D, Vijayan M. Unraveling the mystery of citrate transporters in Alzheimer's disease: An updated review. Ageing Res Rev 2025; 107:102726. [PMID: 40073978 DOI: 10.1016/j.arr.2025.102726] [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: 06/25/2024] [Revised: 12/26/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
A key molecule in cellular metabolism, citrate is essential for lipid biosynthesis, energy production, and epigenetic control. The etiology of Alzheimer's disease (AD), a progressive neurodegenerative illness marked by memory loss and cognitive decline, may be linked to dysregulated citrate transport, according to recent research. Citrate transporters, which help citrate flow both inside and outside of cells, are becoming more and more recognized as possible participants in the molecular processes underlying AD. Citrate synthase (CS), a key enzyme in the tricarboxylic acid (TCA) cycle, supports mitochondrial function and neurotransmitter synthesis, particularly acetylcholine (ACh), essential for cognition. Changes in CS activity affect citrate availability, influencing energy metabolism and neurotransmitter production. Choline, a precursor for ACh, is crucial for neuronal function. Lipid metabolism, oxidative stress reactions, and mitochondrial function can all be affected by aberrant citrate transport, and these changes are linked to dementia. Furthermore, the two main pathogenic characteristics of AD, tau hyperphosphorylation and amyloid-beta (Aβ) aggregation, may be impacted by disturbances in citrate homeostasis. The goal of this review is to clarify the complex function of citrate transporters in AD and provide insight into how they contribute to the development and course of the illness. We aim to provide an in-depth idea of which particular transporters are dysregulated in AD and clarify the functional implications of these dysregulated transporters in brain cells. To reduce neurodegenerative processes and restore metabolic equilibrium, we have also discussed the therapeutic potential of regulating citrate transport. Gaining insight into the relationship between citrate transporters and the pathogenesis of AD may help identify new indicators for early detection and creative targets for treatment. This study offers hope for more potent ways to fight this debilitating illness and is a crucial step in understanding the metabolic foundations of AD.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Shatakshi Mishra
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Stany B
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Deepankumar Kanagavel
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
12
|
Le Gac B, Tournissac M, Belzic E, Picaud S, Dusart I, Soula H, Li D, Charpak S, Cauli B. Elevated pyramidal cell firing orchestrates arteriolar vasoconstriction through COX-2-derived prostaglandin E2 signaling. eLife 2025; 13:RP102424. [PMID: 40266667 PMCID: PMC12017770 DOI: 10.7554/elife.102424] [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: 04/24/2025] Open
Abstract
Neurovascular coupling, linking neuronal activity to cerebral blood flow, is essential for brain function and underpins functional brain imaging. Whereas mechanisms involved in vasodilation are well-documented, those controlling vasoconstriction remain overlooked. This study unravels the mechanisms by which pyramidal cells elicit arteriole vasoconstriction. Using patch-clamp recording, vascular and Ca2+ imaging in mouse cortical slices, we show that strong optogenetic activation of layer II/III pyramidal cells induces vasoconstriction, correlating with firing frequency and somatic Ca2+ increase. Ex vivo and in vivo pharmacological investigations indicate that this vasoconstriction predominantly recruits prostaglandin E2 through the cyclooxygenase-2 pathway, and activation of EP1 and EP3 receptors. We also present evidence that specific interneurons releasing neuropeptide Y, and astrocytes, through 20-hydroxyeicosatetraenoic acid, contribute to this process. By revealing the mechanisms by which pyramidal cells lead to vasoconstriction, our findings shed light on the complex regulation of neurovascular coupling.
Collapse
Affiliation(s)
- Benjamin Le Gac
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Marine Tournissac
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012ParisFrance
| | - Esther Belzic
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Sandrine Picaud
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Isabelle Dusart
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Hédi Soula
- Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches, NutriOmics, Research UnitParisFrance
| | - Dongdong Li
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| | - Serge Charpak
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012ParisFrance
| | - Bruno Cauli
- Sorbonne Université, CNRS, Inserm, Neuro-SUParisFrance
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-SeineParisFrance
| |
Collapse
|
13
|
Jeans AF, Padamsey Z, Collins H, Foster W, Allison S, Dierksmeier S, Klein WL, van den Maagdenberg AMJM, Emptage NJ. Ca V2.1 mediates presynaptic dysfunction induced by amyloid β oligomers. Cell Rep 2025; 44:115451. [PMID: 40127100 DOI: 10.1016/j.celrep.2025.115451] [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: 07/09/2024] [Revised: 02/06/2025] [Accepted: 02/28/2025] [Indexed: 03/26/2025] Open
Abstract
Synaptic dysfunction is an early pathological phenotype of Alzheimer's disease (AD) that is initiated by oligomers of amyloid β peptide (Aβos). Treatments aimed at correcting synaptic dysfunction could be beneficial in preventing disease progression, but mechanisms underlying Aβo-induced synaptic defects remain incompletely understood. Here, we uncover an epithelial sodium channel (ENaC) - CaV2.3 - protein kinase C (PKC) - glycogen synthase kinase-3β (GSK-3β) signal transduction pathway that is engaged by Aβos to enhance presynaptic CaV2.1 voltage-gated Ca2+ channel activity, resulting in pathological potentiation of action-potential-evoked synaptic vesicle exocytosis. We present evidence that the pathway is active in human APP transgenic mice in vivo and in human AD brains, and we show that either pharmacological CaV2.1 inhibition or genetic CaV2.1 haploinsufficiency is sufficient to restore normal neurotransmitter release. These findings reveal a previously unrecognized mechanism driving synaptic dysfunction in AD and identify multiple potentially tractable therapeutic targets.
Collapse
Affiliation(s)
- Alexander F Jeans
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Zahid Padamsey
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Helen Collins
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - William Foster
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Sally Allison
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Steven Dierksmeier
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - William L Klein
- Department of Neurobiology and Physiology, Northwestern University, Evanston, IL 60208, USA
| | | | - Nigel J Emptage
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
14
|
Wang Y, Liu J, Yue S, Chen L, Singh A, Yu T, Calipari ES, Wang ZJ. Prefrontal cortex excitatory neurons show distinct response to heroin-associated cue and social stimulus after prolonged heroin abstinence in mice. Neuropsychopharmacology 2025:10.1038/s41386-025-02102-6. [PMID: 40223131 DOI: 10.1038/s41386-025-02102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Substance use disorder (SUD) has been linked with social impairments. The social cognitive dysfunctions can further increase the risk of the development of SUD or relapse. Therefore, understanding the neural mechanism of substance exposure-associated social impairments is beneficial for the development of novel prevention or treatment strategies for SUD. The prefrontal cortex (PFC) is a key brain region involved in both social cognition and drug addiction. Specifically, the prelimbic part of PFC (PrL) regulates social interaction and heroin-seeking behavior. Therefore, in this study, we explored how PFC excitatory neurons respond to social stimuli after prolonged abstinence from heroin self-administration (SA). Using fiber photometry calcium imaging, we monitored calcium-dependent fluorescent signals in PrL CaMKII-expressing neurons during drug seeking and social interaction tests following 14 days of abstinence from heroin SA. We found that GCaMP6f signals in PrL CaMKII-expressing neurons were increased when heroin-associated cues were presented during drug-seeking tests in both male and female mice after prolonged heroin abstinence, although the baseline neuronal activity in home cage is lower in the heroin group. Conversely, the calcium signals in PrL CaMKII-expressing neurons during social investigation were decreased after heroin abstinence in both sexes, along with reduced total social interaction time. In addition, drug-seeking behavior is partially negatively correlated with social investigation time. These findings provide direct evidence showing that opioid exposure impairs the PFC functional response to social stimuli, which may potentially increase the risk for opioid relapse.
Collapse
Affiliation(s)
- Yunwanbin Wang
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Junting Liu
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Shuwen Yue
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Lu Chen
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Archana Singh
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Tianshi Yu
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Zi-Jun Wang
- Department of Pharmacology & Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA.
- Cofrin Logan Center for Addiction Research and Treatment, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
15
|
Acosta Ingram D, Turkes E, Kim TY, Vo S, Sweeney N, Bonte MA, Rutherford R, Julian DL, Pan M, Marsh J, Argouarch AR, Wu M, Scharre DW, Bell EH, Honig LS, Vonsattel JP, Serrano GE, Beach TG, Karch CM, Kao AW, Hester ME, Han X, Fu H. GRAMD1B is a regulator of lipid homeostasis, autophagic flux and phosphorylated tau. Nat Commun 2025; 16:3312. [PMID: 40204713 PMCID: PMC11982250 DOI: 10.1038/s41467-025-58585-w] [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: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Lipid dyshomeostasis and tau pathology are present in frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD). However, the relationship between lipid dyshomeostasis and tau pathology remains unclear. We report that GRAM Domain Containing 1B (GRAMD1B), a nonvesicular cholesterol transporter, is increased in excitatory neurons of human neural organoids (HNOs) with the MAPT R406W mutation. Human FTLD, AD cases, and PS19 tau mice also have increased GRAMD1B expression. We show that overexpression of GRAMD1B increases levels of free cholesterol, lipid droplets, and impairs autophagy flux. Modulating GRAMD1B in iPSC-derived neurons also alters key autophagy-related components such as PI3K, phospho-AKT, and p62, as well as phosphorylated tau, and CDK5R1. Blocking GRAMD1B function decreases free cholesterol and lipid droplets. Knocking down GRAMD1B additionally reduces phosphorylated tau, and CDK5R1 expression. Our findings elucidate the role of GRAMD1B in the nervous system and highlight its relevance to FTLD and AD.
Collapse
Affiliation(s)
- Diana Acosta Ingram
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Emir Turkes
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Tae Yeon Kim
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Sheeny Vo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicholas Sweeney
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Marie-Amandine Bonte
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Ryan Rutherford
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Dominic L Julian
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jacob Marsh
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea R Argouarch
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Min Wu
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas W Scharre
- Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Erica H Bell
- Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Lawrence S Honig
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jean Paul Vonsattel
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Aimee W Kao
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Mark E Hester
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
16
|
Walls AB, Andersen JV, Waagepetersen HS, Bak LK. Fueling Brain Inhibition: Integrating GABAergic Neurotransmission and Energy Metabolism. Neurochem Res 2025; 50:136. [PMID: 40189668 DOI: 10.1007/s11064-025-04384-0] [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: 01/29/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025]
Abstract
Despite decades of research in brain energy metabolism and to what extent different cell types utilize distinct substrates for their energy metabolism, this topic remains a vibrant area of neuroscience research. In this review, we focus on the substrates utilized by the inhibitory GABAergic neurons, which has been less explored than glutamatergic neurons. First, we discuss how GABAergic neurons may utilize both glucose, lactate, or ketone bodies under different functional conditions, and provide some preliminary data suggesting that unlike glutamatergic neurons, GABAergic neurons work well when substrate supply is restricted to lactate. We end by discussing the role of GABAergic neuron energy metabolism in pathologies where failure of inhibitory function play a central role, namely epilepsy, hepatic encephalopathy, and Alzheimer's disease.
Collapse
Affiliation(s)
- Anne B Walls
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Capital Region Hospital Pharmacy, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Lasse K Bak
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
- Translational Research Center (TRACE), Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
17
|
Yang H, Niu L, Tian L, Hu Y, Cheng C, Li S, Le W. Circadian rhythm disturbances in Alzheimer's disease: insights from plaque-free and plaque-burdened stages in APP SWE/PS1 dE9 mice. Alzheimers Res Ther 2025; 17:76. [PMID: 40188157 PMCID: PMC11971749 DOI: 10.1186/s13195-025-01724-8] [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: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Disruptions in circadian rhythms are commonly observed in patients with Alzheimer's disease (AD) and could potentially accelerate the progression of the condition. However, the relationship between circadian rhythm disruptions and AD development, as well as the mechanisms involved, remain poorly understood. METHODS This study investigated the circadian behavior, rhythmic gene expression in multiple brain regions, and its correlation with sleep architecture of AD mice at two disease stages: plaque-free stage (2-month-old) and plaque-burdened stage (10-month-old) as compared to age-matched wild-type (WT) mice. RESULTS Two-month-old AD mice already displayed alteration in the activity patterns compared to WT mice, showing increased activity during the light phase and decreased activity during the dark phase, and the change in the activity pattern of 10-month-old AD mice was more significant. Further, electroencephalogram (EEG) examination showed increased wakefulness and reduced non-rapid eye movement (NREM) sleep in 2- and 10-month-old AD mice. In addition, we documented a significant change in circadian core clock genes in the suprachiasmatic nucleus (SCN), hippocampus, and cortex of 2- and 10-month-old AD mice. Correlation analyses demonstrated the close relationship between circadian clock gene expression level and specific sleep-wake parameters, especially within the SCN and hippocampus. CONCLUSIONS These findings revealed that circadian rhythm disturbances in AD mice preceded Aβ deposition. The circadian rhythm disturbances observed in the early AD might be attributed to the abnormal expression of core clock genes in the brain regions involved in circadian rhythm regulation.
Collapse
Affiliation(s)
- Huijia Yang
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Long Niu
- Department of Neurology, Heping Hospital affiliated to Changzhi Medical College, Changzhi, China
| | - Lulu Tian
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Yiying Hu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Cheng Cheng
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Weidong Le
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
18
|
Zhao X, Ma C, Sun Q, Huang X, Qu W, Chen Y, Liu Z, Bao A, Sun B, Yang Y, Li X. Mettl3 regulates the pathogenesis of Alzheimer's disease via fine-tuning Lingo2. Mol Psychiatry 2025:10.1038/s41380-025-02984-4. [PMID: 40169805 DOI: 10.1038/s41380-025-02984-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and diverse factors contribute to its pathogenesis. Previous studies have suggested the dysregulation of m6A modification involves in AD, but the underlying mechanism and targets remain largely unknown. In the present study, we have shown that the levels of Mettl3 and m6A modification are increased in specific brain regions of 5xFAD mice and post-mortem AD patients, respectively. Heterozygous deletion of neuronal Mettl3 (AD::Mettl3+/-) reduced Aβ plaques and inflammation, and improved learning and memory of AD mice, and vice versa for Mettl3 knock in (AD::Mettl3-KI). Mechanistically, we observed that the level of m6A modification of Lingo2 increased in 5xFAD mice and AD patients, which promoted the binding of Ythdf2 and enhanced the degradation of Lingo2 mRNA. The decreased level of Lingo2 promoted the interaction between APP and β-site amyloid precursor protein cleaving enzyme (Bace1), and subsequently enhanced Aβ production in AD mice, which can be inhibited by Mettl3 depletion. Both ectopic Lingo2 and the administration of Mettl3 inhibitor STM2457 significantly alleviated the neuropathology and behavioral deficits of AD mice. In summary, our study has revealed the important function of Mettl3 and m6A in the pathogenesis of AD and provided novel insight for the underlying mechanisms. Our study also suggests that m6A and Lingo2 could be potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Xingsen Zhao
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Chengyi Ma
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Qihang Sun
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xiaoli Huang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Wenzheng Qu
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Yusheng Chen
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Ziqin Liu
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Aimin Bao
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Binggui Sun
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| | - Ying Yang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Xuekun Li
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China.
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
19
|
Rozeboom A, Broekaart DWM, Anink JJ, Boonkamp L, Idema S, Teunissen CE, Aronica E, Gorter JA, van Vliet EA. Cellular expression of low-density lipoprotein receptor-related protein 1 and amyloid beta deposition in human and rat epileptogenic brain. Exp Neurol 2025; 386:115149. [PMID: 39842492 DOI: 10.1016/j.expneurol.2025.115149] [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: 11/01/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Decreased capillary expression of low-density lipoprotein receptor-related protein 1 (LRP1) has been linked to increased brain amyloid beta (Aβ) accumulation in Alzheimer's disease (AD). Aβ accumulation has also been observed in (a subset of) temporal lobe epilepsy (TLE) patients, suggesting a potential link between epilepsy and AD. This study examines cellular LRP1 expression in human and rat epileptogenic brain tissue to explore LRP1's role in epilepsy. LRP1 expression and localization were analyzed in hippocampal sections from patients with status epilepticus (SE, n = 12), TLE (n = 12), autopsy controls (n = 20), and AD (n = 10) using immunohistochemistry. Soluble Aβ levels and deposits were compared across TLE, AD, and control tissues. LRP1 expression was also studied in an electrical post-SE rat model of TLE. Decreased capillary LRP1 expression was found in both human and rat brain tissue (SE and TLE). Higher LRP1 expression was detected in CA1 neurons (only in human TLE) and glial cells (SE and TLE). Aβ deposits were observed in only one out of 12 TLE patients, and soluble Aβ levels were not significantly elevated. In contrast, AD patients showed decreased capillary LRP1 expression accompanied by Aβ plaques and increased soluble Aβ40/42 levels. The significant reduction in LRP1 expression in brain capillaries in both adult human and rat TLE was not clearly associated with notable Aβ accumulation implying that alternative amyloid clearance mechanisms beyond LRP1 in blood vessels might be at play. It also supports previous findings indicating that Aβ pathology may be less prominent in adult TLE than some studies suggest.
Collapse
Affiliation(s)
- Annemieke Rozeboom
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Diede W M Broekaart
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jasper J Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lynn Boonkamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Sander Idema
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Erwin A van Vliet
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
20
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Neurotrophic factor alpha 1 gene therapy in Alzheimer's disease: scope and advancements. Front Mol Neurosci 2025; 18:1518868. [PMID: 40235693 PMCID: PMC11996844 DOI: 10.3389/fnmol.2025.1518868] [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: 10/29/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, accounting for 60-80% of all cases globally. Hallmark pathologies of AD include the accumulation of amyloid β peptide and phosphorylated tau, leading to neuronal circuit dysfunction, defective axonal transport, and neurotransmitter system (NTS) abnormalities. Disruptions in acetylcholine, GABA, dopamine, serotonin, and glutamate levels, as well as the loss of cholinergic, GABAergic, and monoaminergic neurons, contribute to the progression of AD. Additionally, neurotrophic factors like brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) are significantly reduced in AD, impacting neuronal health and synaptic integrity. This review highlights the emerging role of neurotrophic factor alpha 1 (NF-α1), also known as carboxypeptidase E, in AD. NF-α1 shows neuroprotective and neurogenesis-promoting properties, offering potential for therapeutic interventions. The review compares NF-α1 gene therapy with other neurotrophin-based treatments, providing insights into its efficacy in AD management.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Severin D, Koh MT, Moreno C, Contreras D, Contreras A, Wesselborg C, Bridi M, Atufa J, Branch A, Worley P, Gallagher M, Kirkwood A. NPTX2 transfection improves synaptic E/I balance and performance in learning impaired aged rats. Prog Neurobiol 2025; 247:102746. [PMID: 40057261 DOI: 10.1016/j.pneurobio.2025.102746] [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: 12/08/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Excessive neural activity in the medial temporal lobe commonly associates with cognitive decline in elderly humans and also in rodents.An attractive model pathway to study synaptic mechanisms underlying age-dependent circuit hyperexcitability is the connection made by lateral entorhinal cortex cells onto the dentate gyrus (LEC→DG). Both structures are particularly affected by age and, importantly, in behaviorally characterized aged rats, learning impairment correlates with diminished feedforward inhibition of granule cells recruited by LEC inputs. In this rat model of aging, we evaluated how overexpression of Neuronal Pentraxin 2 (NPTX2) in the LEC, essential for stabilizing excitatory inputs onto fast-spiking inhibitory interneurons (FS-INs), enhances feedforward inhibition and improves spatial memory in impaired individuals. In addition, we found that FS-INs from unimpaired aged individuals have an increased excitatory drive compared to young individuals. These findings support the notion that NPTX2-mediated compensatory mechanisms to enhance the recruitment of FS-INs are crucial to maintaining proficient memory performance during aging.
Collapse
Affiliation(s)
- Daniel Severin
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Ming Teng Koh
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Cristian Moreno
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Darwin Contreras
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Altagracia Contreras
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Christian Wesselborg
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Michelle Bridi
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Jala Atufa
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Audrey Branch
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Paul Worley
- Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA.
| | - Alfredo Kirkwood
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA; Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA.
| |
Collapse
|
22
|
Wu M, Zhang R, Fu P, Mei Y. Disrupted astrocyte-neuron signaling reshapes brain activity in epilepsy and Alzheimer's disease. Neuroscience 2025; 570:132-151. [PMID: 39986432 DOI: 10.1016/j.neuroscience.2025.02.029] [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: 11/26/2024] [Revised: 01/20/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Astrocytes establish dynamic interactions with surrounding neurons and synchronize neuronal networks within a specific range. However, these reciprocal astrocyte-neuronal interactions are selectively disrupted in epilepsy and Alzheimer's disease (AD), which contributes to the initiation and progression of network hypersynchrony. Deciphering how disrupted astrocyte-neuronal signaling reshapes brain activity is crucial to prevent subclinical epileptiform activity in epilepsy and AD. In this review, we provide an overview of the diverse astrocyte-neuronal crosstalk in maintaining of network activity via homeostatic control of extracellular ions and transmitters, synapse formation and elimination. More importantly, since AD and epilepsy share the common symptoms of neuronal hyperexcitability and astrogliosis, we then explore the crosstalk between astrocytes and neurons in the context of epilepsy and AD and discuss how these disrupted interactions reshape brain activity in pathological conditions. Collectively, this review sheds light on how disrupted astrocyte-neuronal signaling reshapes brain activity in epilepsy and AD, and highlights that modifying astrocyte-neuronal signaling could be a therapeutic approach to prevent epileptiform activity in AD.
Collapse
Affiliation(s)
- Mengjie Wu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ruonan Zhang
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Peng Fu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
23
|
Simonyan K, Darbinyan L, Hambardzumyan L, Manukyan L, Chavushyan V. Teucrium Polium ameliorates amyloid β-induced brain network disorders in rats: electrophysiological and behavioral studies. BMC Complement Med Ther 2025; 25:116. [PMID: 40148951 PMCID: PMC11948851 DOI: 10.1186/s12906-024-04715-8] [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: 10/15/2023] [Accepted: 11/22/2024] [Indexed: 03/29/2025] Open
Abstract
Synaptic failure in specific cholinergic networks in rat brains has been implicated in amyloid β-induced neurodegeneration. Teucrium polium is a promising candidate for drug development against Alzheimer's disease (AD) and similar disorders. However, the protective effect of Teucrium polium against amyloid β-induced impairment of short-term synaptic plasticity is still poorly understood. In this study, we used in vivo extracellular single-unit recordings to investigate the preventive efficacy of Teucrium polium on Aβ(25-35)-induced aberrant neuronal activity in the hippocampus and basolateral amygdala of rats, in response to high-frequency stimulation of the cholinergic nucleus basalis magnocellularis (NBM). After 12 weeks of intracerebroventricular administration of Aβ(25-35), alterations such as decreased excitatory responses and increased inhibitory synaptic activity were observed in the NBM-hippocampus and NBM-basolateral amygdala cholinergic circuits. Treatment with Teucrium polium improved the balance of excitatory and inhibitory responses by modulating synaptic transmission strength and restoring short-term plasticity. Acute injection of a therapeutic dose of Teucrium temporarily inhibited spiking activity in single NBM neurons. Open field tests revealed that amyloid-injected rats displayed anxiety and reduced exploratory drive. Treatment with Teucrium polium improved these behaviors, reducing anxiety and increasing exploration. Teucrium polium mitigated amyloid β-induced alterations in cholinergic circuits by enhancing the adaptive capacity of short-term synaptic plasticity. These findings suggest that Teucrium polium could serve as a preventive strategy to delay the progression of cholinergic neurodegeneration.
Collapse
Affiliation(s)
- Karen Simonyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, Yerevan, 0028, Armenia
| | - Lilit Darbinyan
- Sensorimotor Integration Lab, Orbeli Institute of Physiology NAS RA, Yerevan, 0028, Armenia.
| | - Lilia Hambardzumyan
- Sensorimotor Integration Lab, Orbeli Institute of Physiology NAS RA, Yerevan, 0028, Armenia
| | - Larisa Manukyan
- Sensorimotor Integration Lab, Orbeli Institute of Physiology NAS RA, Yerevan, 0028, Armenia
| | - Vergine Chavushyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, Yerevan, 0028, Armenia
| |
Collapse
|
24
|
Bergamasco MI, Ozturk E, Casillas-Espinosa PM, Garnham AL, Abeysekera W, Wimmer VC, Rajasekhar P, Vanyai HK, Whitehead L, Blewitt ME, Rogers K, Vogel AP, Hannan AJ, Smyth GK, Jones NC, Thomas T, Voss AK. KAT6B overexpression in mice causes aggression, anxiety, and epilepsy. iScience 2025; 28:111953. [PMID: 40083716 PMCID: PMC11904597 DOI: 10.1016/j.isci.2025.111953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 10/20/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Loss of the gene encoding the histone acetyltransferase KAT6B (MYST4/MORF/QKF) causes developmental brain abnormalities as well as behavioral and cognitive defects in mice. In humans, heterozygous variants in the KAT6B gene cause two cognitive disorders, Say-Barber-Biesecker-Young-Simpson syndrome (SBBYSS; OMIM:603736) and genitopatellar syndrome (GTPTS; OMIM:606170). Although the effects of KAT6B homozygous and heterozygous mutations have been documented in humans and mice, KAT6B gain-of-function effects have not been reported. Here, we show that overexpression of the Kat6b gene in mice caused aggression, anxiety, and spontaneous epilepsy. Kat6b overexpression led to an increase in histone H3 lysine 9 acetylation and upregulation of genes driving nervous system development and neuronal differentiation. Kat6b overexpression additionally promoted neural stem cell proliferation and favored neuronal over astrocyte differentiation in vivo and in vitro. Our results suggest that, in addition to loss-of-function alleles, gain-of-function KAT6B alleles may be detrimental for brain development.
Collapse
Affiliation(s)
- Maria I. Bergamasco
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Ezgi Ozturk
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville VIC 3052, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Melbourne, VIC 3004, Australia
| | - Pablo M. Casillas-Espinosa
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville VIC 3052, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Melbourne, VIC 3004, Australia
| | - Alexandra L. Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Waruni Abeysekera
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Verena C. Wimmer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Pradeep Rajasekhar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Hannah K. Vanyai
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Marnie E. Blewitt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Kelly Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Adam P. Vogel
- Centre for Neurosciences of Speech, The University of Melbourne, Melbourne, VIC 3052, Australia
- Redenlab Inc, Melbourne, VIC 3000, Australia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Gordon K. Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Nigel C. Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville VIC 3052, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Melbourne, VIC 3004, Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Anne K. Voss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
25
|
Quiles LE, Quintay PKP, Anlacan VM, Espiritu A, Mendoza V. Prevalence and factors associated with seizures among patients with dementia: A retrospective clinic-based study. Epilepsia Open 2025. [PMID: 40100088 DOI: 10.1002/epi4.70013] [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: 12/05/2023] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE Chronic diseases associated with aging, such as dementia and seizures, are expected to rise significantly in the Philippines' growing elderly population. This study aims to determine the frequency, demographic characteristics, and clinical profile of dementia patients who developed new-onset seizures in an outpatient setting. METHODS This descriptive, retrospective, cumulative prevalence study included 245 patients diagnosed with dementia at a tertiary hospital in Manila from February 2010 to February 2020, according to DSM-5 criteria. Patients were stratified into those who developed seizures and those who did not. Data on demographics, type, dementia severity, comorbidities, and seizure characteristics were collected and analyzed using descriptive statistics, bivariate, and multivariate logistic regression analyses. RESULTS The study included 245 dementia patients, of whom 10 (4.1%) developed seizures, with a higher likelihood observed in those with severe dementia. Most patients were diagnosed with Alzheimer's disease, and seizures were mostly seen in individuals between the ages of 65 and 79. The majority of the seizures were classified as generalized (50%). Compared to mild cases, patients with moderate dementia are about 1.5 times more likely to experience seizures, whereas patients with severe dementia are about 10 times more likely to experience seizures compared to patients with mild dementia. The association is statistically significant for severe cases of dementia. SIGNIFICANCE This study revealed that 4.1% of Filipino patients diagnosed with dementia in an outpatient setting at a tertiary hospital developed new-onset seizures. Seizures were mostly reported in patients with severe Alzheimer's disease. Conventional understanding of seizures among patients with dementia is important to identify features and predictors to provide efficient management among these patients to possibly improve their quality of life. PLAIN LANGUAGE SUMMARY With the aging Filipino population, there is an expected rise in chronic diseases such as dementia and seizures. This study looked at dementia patients in an outpatient setting over 10 years and found that 4.1% developed seizures. Most patients had Alzheimer's disease, and seizures were more common in severe dementia cases.
Collapse
Affiliation(s)
- Liz Edenberg Quiles
- Department of Neurology, Institute of Neurosciences, The Medical City, Pasig City, Philippines
| | | | - Veeda Michelle Anlacan
- Department of Neurology, Institute of Neurosciences, The Medical City, Pasig City, Philippines
- Department of Neurosciences, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
- Center for Memory and Cognition, Department of Neurosciences, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Adrian Espiritu
- Department of Neurosciences, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Viel Mendoza
- Center for Memory and Cognition, Department of Neurosciences, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
26
|
Sánchez-Garrido Campos G, Zafra ÁM, Estévez-Rodríguez M, Cordones I, Ruffini G, Márquez-Ruiz J. Preclinical insights into gamma-tACS: foundations for clinical translation in neurodegenerative diseases. Front Neurosci 2025; 19:1549230. [PMID: 40143845 PMCID: PMC11936909 DOI: 10.3389/fnins.2025.1549230] [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: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Gamma transcranial alternating current stimulation (gamma-tACS) represents a novel neuromodulation technique with promising therapeutic applications across neurodegenerative diseases. This mini-review consolidates recent preclinical and clinical findings, examining the mechanisms by which gamma-tACS influences neural oscillations, enhances synaptic plasticity, and modulates neuroimmune responses. Preclinical studies have demonstrated the capacity of gamma-tACS to synchronize neuronal firing, support long-term neuroplasticity, and reduce markers of neuroinflammation, suggesting its potential to counteract neurodegenerative processes. Early clinical studies indicate that gamma-tACS may improve cognitive functions and network connectivity, underscoring its ability to restore disrupted oscillatory patterns central to cognitive performance. Given the intricate and multifactorial nature of gamma oscillations, the development of tailored, optimized tACS protocols informed by extensive animal research is crucial. Overall, gamma-tACS presents a promising avenue for advancing treatments that support cognitive resilience in a range of neurodegenerative conditions.
Collapse
Affiliation(s)
| | - Ángela M. Zafra
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Marta Estévez-Rodríguez
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Isabel Cordones
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Giulio Ruffini
- Brain Modeling Department, Neuroelectrics Barcelona, Barcelona, Spain
| | - Javier Márquez-Ruiz
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| |
Collapse
|
27
|
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
|
28
|
Tinston J, Hudson MR, Harutyunyan A, Chen Z, Jones NC. Forty-hertz sensory entrainment impedes kindling epileptogenesis and reduces amyloid pathology in an Alzheimer disease mouse model. Epilepsia 2025; 66:886-898. [PMID: 39737719 DOI: 10.1111/epi.18222] [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: 07/21/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025]
Abstract
OBJECTIVE The 5xFAD mouse model of Alzheimer disease (AD) recapitulates amyloid-beta (Aβ) deposition and pronounced seizure susceptibility observed in patients with AD. Forty-hertz audiovisual stimulation is a noninvasive technique that entrains gamma neural oscillations and can reduce Aβ pathology and modulate glial expression in AD models. We hypothesized that 40-Hz sensory stimulation would improve seizure susceptibility in 5xFAD mice and this would be associated with reduction of plaques and modulation of glial phenotypes. METHODS 5xFAD mice and wild-type (WT) littermates received 1 h/day 40-Hz audiovisual stimulation or sham (n = 7-11/group), beginning 2 weeks before and continuing throughout amygdala kindling epileptogenesis. Postmortem analyses included Aβ pathology and morphology of astrocytes and microglia. RESULTS 5xFAD mice exhibited enhanced susceptibility to seizures compared to WT, evidenced by fewer stimulations to reach kindling endpoint (incidence rate ratio [IRR] = 1.46, p < .0001) and a trend to higher seizure severity (odds ratio [OR] = .34, p = .059). Forty-hertz stimulation reduced the behavioral severity of the first seizure (OR = 4.04, p = .02) and delayed epileptogenesis, increasing the number of stimulations required to reach kindling endpoint (IRR = .82, p = .01) compared to sham, regardless of genotype. 5xFAD mice receiving sensory stimulation exhibited ~50% reduction in amyloid pathology compared to sham. Furthermore, markers of astrocytes and microglia were upregulated in both genotypes receiving 40-Hz stimulation. SIGNIFICANCE Forty-hertz sensory entrainment slows epileptogenesis in the mouse amygdala kindling model. Although this intervention improves Aβ pathology in 5xFAD mice, the observed antiepileptogenic effect may also relate to effects on glia, because mice without Aβ plaques (i.e., WT) also experienced antiepileptogenic effects of the intervention.
Collapse
Affiliation(s)
- Jennifer Tinston
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Matthew R Hudson
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anna Harutyunyan
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Zhibin Chen
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Ye W, Tao Y, Wang W, Yu Y, Li X. Periodontitis associated with brain function impairment in middle-aged and elderly individuals with normal cognition. J Periodontol 2025; 96:290-300. [PMID: 39565645 DOI: 10.1002/jper.24-0264] [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: 09/18/2024] [Accepted: 09/22/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND The present study aimed to investigate changes in intranetwork functional connectivity (FC) and internetwork FC in middle-aged and elderly individuals with normal cognition (NC) and varying degrees of periodontitis to determine the effects of periodontitis on brain function. METHODS Periodontal findings and resting-state functional magnetic resonance imaging data were acquired from 51 subjects with NC. Independent component analysis and correlation analysis were used for the statistical analysis of the data. RESULTS Differences in intranetwork FC were observed among groups in the anterior default-mode network (aDMN), dorsal attention network and dorsal sensorimotor network (dSMN). Compared with the nonperiodontitis (NP) group or the mild-periodontitis group, the analysis of internetwork FC showed increased FC between the auditory network and the ventral attention network (VAN), between the aDMN and the salience network (SN), and between the SN and the VAN and decreased FC between the posterior default-mode network and the right frontoparietal network in the moderate-to-severe periodontitis group. Additionally, internetwork FC between the dSMN and the VAN was also increased in the moderate-to-severe periodontitis group compared to the NP group. The altered intra- and internetwork FC were significantly correlated with the periodontal clinical index. CONCLUSION Our results confirmed that periodontitis was associated with both intra- and internetwork FC changes even in NC. The present study indicates that periodontitis might be a potential risk factor for brain damage and provides a theoretical clue and a new treatment target for the early prevention of Alzheimer disease. PLAIN LANGUAGE SUMMARY Recent research has proposed that periodontitis is a potential risk factor for Alzheimer disease (AD). However, the relationship between periodontitis and the brain function of middle-aged and elderly individuals with normal cognition (NC) remains unclear. Analyzing the effect of periodontitis on brain function in the NC stage can provide clues to AD development and help achieve early prevention of dementia. The present study aimed to investigate changes in brain functional connectivity (FC) in NC with different severity of periodontitis to determine the effects of periodontitis on brain function. Both changed intranetwork FC and internetwork FC were found in the moderate-to-severe periodontitis group, and periodontitis was associated with brain network function impairment in NC. The present study indicates that periodontitis might be a potential risk factor for brain damage even in NC stage, and provides a theoretical clue and a new treatment target for the early prevention of AD.
Collapse
Affiliation(s)
- Wei Ye
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yufei Tao
- Department of Periodontics, Hefei Stomatological Clinic College, Anhui Medical University & Stomatological Hospital, Hefei, China
| | - Wenrui Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoshu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Zhang X, Zhang Y, Zhang T, Wang J, Liu C, Shang Q, Wei X, Zhu H, Shen H, Sun B. HCN2 deficiency correlates with memory deficits and hyperexcitability of dCA1 pyramidal neurons in Alzheimer's disease. Alzheimers Res Ther 2025; 17:55. [PMID: 40016780 PMCID: PMC11866685 DOI: 10.1186/s13195-025-01704-y] [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: 09/18/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Abnormal excitability of hippocampal neurons may lead to dysfunction of neural circuits and then causes cognitive impairments in Alzheimer's disease (AD). However, the underlying mechanisms remain to be fully elucidated. METHODS Electrophysiology was performed to examine the intrinsic excitability of CA1 neurons and the activity of the hyperpolarization-activated cyclic nucleotide-gated ion channels (HCNs) of CA1 neurons in wild type (WT) and hAPP-J20 mice. The activity of CA1 pyramidal neurons (PNs) was modulated with chemogenetics. The activity of HCNs was regulated with nonselective facilitator (cAMP) or inhibitor (ZD7288) of HCNs. Immunohistochemical staining or western blotting were performed to examine the expression of HCN1 and HCN2 in the hippocampus of WT and hAPP-J20 mice, or AD patients and non-AD controls. AAVs were injected to specifically modulate the expression of HCN2 in dorsal CA1 (dCA1) PNs. Cognitive performance of mice was assessed with behavioral tests. RESULTS dCA1 PNs were more excitable in hAPP-J20 mice, but the excitability of PNs in the ventral CA1 (vCA1) or PV neurons was comparable between WT and hAPP-J20 mice. The activity of the HCNs was reduced in dCA1 PNs of hAPP-J20 mice, and pharmacologically increasing the activity of HCNs attenuated the hyperexcitability of dCA1 PNs in hAPP-J20 mice, suggesting that the reduced activity of HCNs is associated with the hyperexcitability of dCA1 PNs in hAPP-J20 mice. The expression of HCN2 but not HCN1 was reduced in the hippocampus of hAPP-J20 mice, and the expression of HCN2 was also reduced in the hippocampus of AD patients, suggesting that dysregulation of HCN2 is associated with the reduced activity of HCNs in AD. Overexpressing HCN2 rescued the activity of HCNs, attenuated the hyperexcitability of dCA1 PNs and improved memory of hAPP-J20 mice, and knocking down HCN2 impaired the function of HCNs, increased the excitability of dCA1 PNs and led to memory deficits in WT mice. CONCLUSIONS Our data suggest that dysregulation of HCNs, particularly HCN2, contributes to the abnormal excitability of CA1 PNs in AD mice and probably in AD patients as well, and thus provide new insights into the mechanisms underlying the aberrant activity or excitability of hippocampal neurons in AD.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Yiping Zhang
- 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; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Ting Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Jing Wang
- 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; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Chang Liu
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, Zhejiang Province, 315211, China
| | - Xiaojie Wei
- 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; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Huaqiang Zhu
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang Province, 315100, China
| | - Haowei Shen
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, 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; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|
31
|
Dybowski FP, Scott DS, Tamminga CA. Pharmacological reduction of reverse-translated hippocampal hyperactivity in mouse: relevance for psychosis. Neuropsychopharmacology 2025:10.1038/s41386-025-02077-4. [PMID: 40016366 DOI: 10.1038/s41386-025-02077-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Hippocampal hyperactivity (HH) is a potential biomarker in schizophrenia psychosis, which also appears in several other brain disorders, compromising specificity. We hypothesized that the reversal of HH in an established, reverse-translational animal preparation, coupled with a behavioral marker of psychosis may be a predictor of antipsychotic efficacy of a medication. We used a chemogenetic reverse-translational mouse preparation relevant to schizophrenia psychosis which shows HH and aberrant psychosis-relevant behaviors, specifically disrupted social recognition memory (SRM). Mice with and without HH were treated with three drugs; two known antipsychotics and one HH-reducing anticonvulsant, to assess their effects on both HH and SRM performance. All animals received one of the four treatments: vehicle (N = 15-24), haloperidol (N = 8-15), xanomeline (N = 8-13) or levetiracetam (N = 6-15) and were subsequently tested for baseline c-Fos protein expression within the hippocampal subfields (CA3 and CA1) as a measure of neuronal activity, or tested with the SRM task as a measure of social memory. All three drugs acutely reduced baseline HH compared to vehicle treatment. Subacute administration of haloperidol or xanomeline, the two drugs known to have antipsychotic activity, but not levetiracetam, normalized the SRM behavior to control levels. These results suggest that the reversal of HH alone cannot be a predictor of antipsychotic efficacy of an experimental drug and HH as a biomarker could benefit from a more sensitive readout approach.
Collapse
Affiliation(s)
- Filip P Dybowski
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
- O'Donnell Brain Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Daniel S Scott
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- O'Donnell Brain Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carol A Tamminga
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
- O'Donnell Brain Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
32
|
Bu SH, Hu XZ, Su Z, Li LT. Advances in the study of apathy related to cerebral small vessel disease. Front Neurol 2025; 16:1513574. [PMID: 40013000 PMCID: PMC11860071 DOI: 10.3389/fneur.2025.1513574] [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: 10/18/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Cerebral small vessel disease (CSVD) is a complex clinical-imaging pathological syndrome caused by small vessel lesions in the brain, which is characterized by aging-related, insidious onset and slow progression. Apathy is a key component of the common neuropsychiatric symptoms among CSVD patients, severely affecting their daily lives and social functioning. Moreover, there are fewer studies on CSVD-related apathy, and greater attention should be paid to this condition in clinical practice. This article describes the latest research advances in the concept, epidemiological features, pathogenesis, assessment and diagnosis, imaging and biomarkers, and treatment of CSVD-related apathy, aiming to serve as a reference for the clinical diagnosis and prevention of CSVD-related apathy.
Collapse
Affiliation(s)
- Shuo-han Bu
- Department of Neurology, Hebei North College, Zhangjiakou, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Xin-zhu Hu
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Zhe Su
- Department of Neurology, Hebei North College, Zhangjiakou, China
| | - Li-tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
33
|
Futácsi A, Rusznák K, Szarka G, Völgyi B, Wiborg O, Czéh B. Quantification and correlation of amyloid-β plaque load, glial activation, GABAergic interneuron numbers, and cognitive decline in the young TgF344-AD rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1542229. [PMID: 40013092 PMCID: PMC11860898 DOI: 10.3389/fnagi.2025.1542229] [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: 12/09/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025] Open
Abstract
Background Animal models of Alzheimer's disease (AD) are essential tools for investigating disease pathophysiology and conducting preclinical drug testing. In this study, we examined neuronal and glial alterations in the hippocampus and medial prefrontal cortex (mPFC) of young TgF344-AD rats and correlated these changes with cognitive decline and amyloid-β plaque load. Methods We compared TgF344-AD and non-transgenic littermate rats aged 7-8 months of age. We systematically quantified β-amyloid plaques, astrocytes, microglia, four different subtypes of GABAergic interneurons (calretinin-, cholecystokinin-, parvalbumin-, and somatostatin-positive neurons), and newly generated neurons in the hippocampus. Spatial learning and memory were assessed using the Barnes maze test. Results Young TgF344-AD rats had a large number of amyloid plaques in both the hippocampus and mPFC, together with a pronounced increase in microglial cell numbers. Astrocytic activation was significant in the mPFC. Cholecystokinin-positive cell numbers were decreased in the hippocampus of transgenic rats, but calretinin-, parvalbumin-, and somatostatin-positive cell numbers were not altered. Adult neurogenesis was not affected by genotype. TgF344-AD rats had spatial learning and memory impairments, but this cognitive deficit did not correlate with amyloid plaque number or cellular changes in the brain. In the hippocampus, amyloid plaque numbers were negatively correlated with cholecystokinin-positive neuron and microglial cell numbers. In the mPFC, amyloid plaque number was negatively correlated with the number of astrocytes. Conclusion Pronounced neuropathological changes were found in the hippocampus and mPFC of young TgF344-AD rats, including the loss of hippocampal cholecystokinin-positive interneurons. Some of these neuropathological changes were negatively correlated with amyloid-β plaque load, but not with cognitive impairment.
Collapse
Affiliation(s)
- Anett Futácsi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Kitti Rusznák
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
34
|
Qu T. The effects of amyloidosis and aging on glutamatergic and GABAergic synapses, and interneurons in the barrel cortex and non-neocortical brain regions. Front Neuroanat 2025; 19:1526962. [PMID: 40012738 PMCID: PMC11863279 DOI: 10.3389/fnana.2025.1526962] [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: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Previous studies on changes in the distribution of GABAergic interneurons and excitation/inhibition (E/I) balance in Alzheimer's disease (AD) and aging were mainly conducted in the neocortex and hippocampus. However, the limbic system is the primary and crucial location for AD progression. Therefore, in this study, we utilized AD and aging mouse models to investigate the E/I balance and the distribution of parvalbumin (PV)- and somatostatin (SST)-expressing cells in S1BF (barrel field of primary somatosensory cortex, barrel cortex), CA1 hippocampal area and brain regions beyond the neocortex and hippocampus, including retrosplenial cortex (RSC, which is composed of RSG and RSA), piriform cortex (Pir), amygdala (BMA), and hypothalamus (DM). We discovered that amyloidosis may disrupt the alignment of excitatory pre- and postsynaptic quantities. Amyloidosis reduces the quantity of synapses and SST cells, but does not impact the counts of PV cells. By contrast, aging is linked to a decline in synapses, I/E ratios, SST and PV cells. Amyloidosis affects the S1BF and BMA, while aging may harm all studied regions, including the S1BF, RSC, hippocampus, Pir, BMA, and DM. Aging mostly affects synapses and I/E ratios in Pir, BMA, and DM, and PV and SST interneurons in the hippocampus.
Collapse
Affiliation(s)
- Tao Qu
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
35
|
Pierson SR, Fiock KL, Wang R, Balasubramanian N, Reinhardt J, Khan KM, James TD, Hunter ML, Cooper BJ, Williamsen HR, Betters R, Deniz K, Lee G, Aldridge G, Hefti MM, Marcinkiewcz CA. Tau pathology in the dorsal raphe may be a prodromal indicator of Alzheimer's disease. Mol Psychiatry 2025; 30:532-546. [PMID: 39143322 PMCID: PMC12010729 DOI: 10.1038/s41380-024-02664-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 06/22/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
Protein aggregation in brainstem nuclei is thought to occur in the early stages of Alzheimer's disease (AD), but its specific role in driving prodromal symptoms and disease progression is largely unknown. The dorsal raphe nucleus (DRN) contains a large population of serotonin (5-hydroxytryptamine; 5-HT) neurons that regulate mood, reward-related behavior, and sleep, which are all disrupted in AD. We report here that tau pathology is present in the DRN of individuals 25-80 years old without a known history of dementia, and its prevalence was comparable to the locus coeruleus (LC). By comparison, fewer cases were positive for other pathological proteins including α-synuclein, β-amyloid, and TDP-43. To evaluate how early tau pathology impacts behavior, we overexpressed human P301L-tau in the DRN of mice and observed depressive-like behaviors and hyperactivity without deficits in spatial memory. Tau pathology was predominantly found in neurons relative to glia and colocalized with a significant proportion of Tph2-expressing neurons in the DRN. 5-HT neurons were also hyperexcitable in P301L-tauDRN mice, and there was an increase in the amplitude of excitatory post-synaptic currents (EPSCs). Moreover, astrocytic density was elevated in the DRN and accompanied by an increase in IL-1α and Frk expression, which suggests increased inflammatory signaling. Additionally, tau pathology was detected in axonal processes in the thalamus, hypothalamus, amygdala, and caudate putamen. A significant proportion of this tau pathology colocalized with the serotonin reuptake transporter (SERT), suggesting that tau may spread in an anterograde manner to regions outside the DRN. Together these results indicate that tau pathology accumulates in the DRN in a subset of individuals over 50 years and may lead to behavioral dysregulation, 5-HT neuronal dysfunction, and activation of local astrocytes which may be prodromal indicators of AD.
Collapse
Affiliation(s)
- Samantha R Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kimberly L Fiock
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Jessica Reinhardt
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kanza M Khan
- Psychological Sciences Department, Daemen University, Amherst, NY, 14226, USA
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Mikayla L Hunter
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin J Cooper
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Ryan Betters
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kaancan Deniz
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
| | - Gloria Lee
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Georgina Aldridge
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
36
|
Barker-Haliski M. Seizing the opportunity to therapeutically address neuronal hyperexcitability in Alzheimer's disease. J Alzheimers Dis 2025; 103:662-665. [PMID: 39784685 DOI: 10.1177/13872877241305740] [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] [Indexed: 01/12/2025]
Abstract
Seizures in people with Alzheimer's disease are increasingly recognized to worsen disease burden and accelerate functional decline. Harnessing established antiseizure medicine discovery strategies in rodents with Alzheimer's disease associated risk genes represents a novel way to uncover disease modifying treatments that may benefit these Alzheimer's disease patients. This commentary discusses the recent evaluation by Dejakaisaya and colleagues to assess the antiseizure and disease-modifying potential of the repurposed cephalosporin antibiotic, ceftriaxone, in the Tg2576 mouse model. The use of established epilepsy models in Alzheimer's disease research carries the potential to advance novel disease-modifying treatments.
Collapse
|
37
|
Calvin-Dunn KN, Mcneela A, Leisgang Osse A, Bhasin G, Ridenour M, Kinney JW, Hyman JM. Electrophysiological insights into Alzheimer's disease: A review of human and animal studies. Neurosci Biobehav Rev 2025; 169:105987. [PMID: 39732222 DOI: 10.1016/j.neubiorev.2024.105987] [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: 01/22/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
This review highlights the crucial role of neuroelectrophysiology in illuminating the mechanisms underlying Alzheimer's disease (AD) pathogenesis and progression, emphasizing its potential to inform the development of effective treatments. Electrophysiological techniques provide unparalleled precision in exploring the intricate networks affected by AD, offering insights into the synaptic dysfunction, network alterations, and oscillatory abnormalities that characterize the disease. We discuss a range of electrophysiological methods, from non-invasive clinical techniques like electroencephalography and magnetoencephalography to invasive recordings in animal models. By drawing on findings from these studies, we demonstrate how electrophysiological research has deepened our understanding of AD-related network disruptions, paving the way for targeted therapeutic interventions. Moreover, we underscore the potential of electrophysiological modalities to play a pivotal role in evaluating treatment efficacy. Integrating electrophysiological data with clinical neuroimaging and longitudinal studies holds promise for a more comprehensive understanding of AD, enabling early detection and the development of personalized treatment strategies. This expanded research landscape offers new avenues for unraveling the complexities of AD and advancing therapeutic approaches.
Collapse
Affiliation(s)
- Kirsten N Calvin-Dunn
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, United States.
| | - Adam Mcneela
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States
| | - A Leisgang Osse
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - G Bhasin
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| | - M Ridenour
- Department of Psychology, University of Nevada, Las Vegas, United States
| | - J W Kinney
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - J M Hyman
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| |
Collapse
|
38
|
Sabec MH, Savage QR, Wood JL, Maskos U. Targeting high-affinity nicotinic receptors protects against the functional consequences of β-amyloid in mouse hippocampus. Mol Psychiatry 2025; 30:556-566. [PMID: 39164528 DOI: 10.1038/s41380-024-02666-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/22/2024]
Abstract
The accumulation of β-amyloid oligomers is a hallmark of Alzheimer's disease, inducing neural and network dysfunction in the early stages of pathology. The hippocampus is affected early in the pathogenesis of AD, however the impact of soluble β-amyloid on the dentate gyrus (DG) subregion of the hippocampus and its interaction with nicotinic acetylcholine receptors (nAChRs) within this region are not known. Using a localized model of over-expression, we show that β-amyloid induces early-onset neuronal hyperactivity and hippocampal-dependent memory deficits in mice. Further, we find the DG region to be under potent and sub-type specific nicotinic control in both healthy and pathophysiological conditions, with targeted receptor inhibition leading to a mnemonic rescue against localized amyloidosis. We show that while neurogenesis and synaptic functions are not severely affected in our model, reducing β2-containing nAChR function is associated with the promotion of young adult-born neurons within the pathological network, suggesting a possible protective mechanism. Our data thus reveal the DG network level changes which occur in the early-stages of β-amyloid accumulation and highlight the downstream consequences of targeted nicotinic neuromodulation.
Collapse
Affiliation(s)
- Marie H Sabec
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems, 75015, Paris, France.
- Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| | - Quentin R Savage
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, 76798, USA
| | - John L Wood
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, 76798, USA
| | - Uwe Maskos
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems, 75015, Paris, France.
| |
Collapse
|
39
|
Kim TA, Cruz G, Syty MD, Wang F, Wang X, Duan A, Halterman M, Xiong Q, Palop JJ, Ge S. Neural circuit mechanisms underlying aberrantly prolonged functional hyperemia in young Alzheimer's disease mice. Mol Psychiatry 2025; 30:367-378. [PMID: 39043843 PMCID: PMC11750623 DOI: 10.1038/s41380-024-02680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
Neurovascular defects are one of the most common alterations in Alzheimer's disease (AD) pathogenesis, but whether these deficits develop before the onset of amyloid beta (Aβ) accumulation remains to be determined. Using in vivo optical imaging in freely moving mice, we explored activity-induced hippocampal microvascular blood flow dynamics in AppSAA knock-in and J20 mouse models of AD at early stages of disease progression. We found that prior to the onset of Aβ accumulation, there was a pathologically elevated blood flow response to context exploration, termed functional hyperemia. After the onset of Aβ accumulation, this context exploration-induced hyperemia declined rapidly relative to that in control mice. Using in vivo electrophysiology recordings to explore the neural circuit mechanism underlying this blood flow alteration, we found that hippocampal interneurons before the onset of Aβ accumulation were hyperactive during context exploration. Chemogenetic tests suggest that hyperactive activation of inhibitory neurons accounted for the elevated functional hyperemia. The suppression of nitric oxide (NO) produced from hippocampal interneurons in young AD mice decreased the accumulation of Aβ. Together, these findings reveal that neurovascular coupling is aberrantly elevated before Aβ deposition, and this hyperactive functional hyperemia declines rapidly upon Aβ accumulation.
Collapse
Affiliation(s)
- Thomas A Kim
- Medical Scientist Training Program (MSTP), Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794, USA
| | - George Cruz
- Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Michelle D Syty
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Faye Wang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Xinxing Wang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Alexandra Duan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Marc Halterman
- Department of Neurology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Jorge J Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
40
|
Juvenal G, Higa GSV, Bonfim Marques L, Tessari Zampieri T, Costa Viana FJ, Britto LR, Tang Y, Illes P, di Virgilio F, Ulrich H, de Pasquale R. Regulation of GABAergic neurotransmission by purinergic receptors in brain physiology and disease. Purinergic Signal 2025; 21:149-177. [PMID: 39046648 PMCID: PMC11958915 DOI: 10.1007/s11302-024-10034-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: 02/28/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Purinergic receptors regulate the processing of neural information in the hippocampus and cerebral cortex, structures related to cognitive functions. These receptors are activated when astrocytic and neuronal populations release adenosine triphosphate (ATP) in an autocrine and paracrine manner, following sustained patterns of neuronal activity. The modulation by these receptors of GABAergic transmission has only recently been studied. Through their ramifications, astrocytes and GABAergic interneurons reach large groups of excitatory pyramidal neurons. Their inhibitory effect establishes different synchronization patterns that determine gamma frequency rhythms, which characterize neural activities related to cognitive processes. During early life, GABAergic-mediated synchronization of excitatory signals directs the experience-driven maturation of cognitive development, and dysfunctions concerning this process have been associated with neurological and neuropsychiatric diseases. Purinergic receptors timely modulate GABAergic control over ongoing neural activity and deeply affect neural processing in the hippocampal and neocortical circuitry. Stimulation of A2 receptors increases GABA release from presynaptic terminals, leading to a considerable reduction in neuronal firing of pyramidal neurons. A1 receptors inhibit GABAergic activity but only act in the early postnatal period when GABA produces excitatory signals. P2X and P2Y receptors expressed in pyramidal neurons reduce the inhibitory tone by blocking GABAA receptors. Finally, P2Y receptor activation elicits depolarization of GABAergic neurons and increases GABA release, thus favoring the emergence of gamma oscillations. The present review provides an overall picture of purinergic influence on GABAergic transmission and its consequences on neural processing, extending the discussion to receptor subtypes and their involvement in the onset of brain disorders, including epilepsy and Alzheimer's disease.
Collapse
Affiliation(s)
- Guilherme Juvenal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Guilherme Shigueto Vilar Higa
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lucas Bonfim Marques
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Thais Tessari Zampieri
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Felipe José Costa Viana
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luiz R Britto
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Yong Tang
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany
| | | | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Roberto de Pasquale
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
41
|
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
|
42
|
Bonzanni M, Braga A, Saito T, Saido TC, Tesco G, Haydon PG. Adenosine deficiency facilitates CA1 synaptic hyperexcitability in the presymptomatic phase of a knockin mouse model of Alzheimer disease. iScience 2025; 28:111616. [PMID: 39850358 PMCID: PMC11754081 DOI: 10.1016/j.isci.2024.111616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 01/25/2025] Open
Abstract
The disease's trajectory of Alzheimer disease (AD) is associated with and negatively correlated to hippocampal hyperexcitability. Here, we show that during the asymptomatic stage in a knockin (KI) mouse model of Alzheimer disease (APPNL-G-F/NL-G-F; APPKI), hippocampal hyperactivity occurs at the synaptic compartment, propagates to the soma, and is manifesting at low frequencies of stimulation. We show that this aberrant excitability is associated with a deficient adenosine tone, an inhibitory neuromodulator, driven by reduced levels of CD39/73 enzymes, responsible for the extracellular ATP-to-adenosine conversion. Both pharmacologic (adenosine kinase inhibitor) and non-pharmacologic (ketogenic diet) restorations of the adenosine tone successfully normalize hippocampal neuronal activity. Our results demonstrated that neuronal hyperexcitability during the asymptomatic stage of a KI model of Alzheimer disease originated at the synaptic compartment and is associated with adenosine deficient tone. These results extend our comprehension of the hippocampal vulnerability associated with the asymptomatic stage of Alzheimer disease.
Collapse
Affiliation(s)
- Mattia Bonzanni
- Department of Neuroscience, Tufts University, Boston, MA 02111, USA
| | - Alice Braga
- Department of Neuroscience, Tufts University, Boston, MA 02111, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Giuseppina Tesco
- Department of Neuroscience, Tufts University, Boston, MA 02111, USA
| | - Philip G. Haydon
- Department of Neuroscience, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
43
|
Vetere LM, Galas AM, Vaughan N, Feng Y, Wick ZC, Philipsberg PA, Liobimova O, Fernandez-Ruiz A, Cai DJ, Shuman T. Medial entorhinal-hippocampal desynchronization parallels the emergence of memory impairment in a mouse model of Alzheimer's disease pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633171. [PMID: 39868201 PMCID: PMC11761809 DOI: 10.1101/2025.01.15.633171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive impairments in episodic and spatial memory, as well as circuit and network-level dysfunction. While functional impairments in medial entorhinal cortex (MEC) and hippocampus (HPC) have been observed in patients and rodent models of AD, it remains unclear how communication between these regions breaks down in disease, and what specific physiological changes are associated with the onset of memory impairment. We used silicon probes to simultaneously record neural activity in MEC and hippocampus before or after the onset of spatial memory impairment in the 3xTg mouse model of AD pathology. We found that reduced hippocampal theta power, reduced MEC-CA1 theta coherence, and altered phase locking of MEC and hippocampal neurons all coincided with the emergence of spatial memory impairment in 3xTg mice. Together, these findings indicate that disrupted temporal coordination of neural activity in the MEC-hippocampal system parallels the emergence of memory impairment in a model of AD pathology.
Collapse
Affiliation(s)
| | | | - Nick Vaughan
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yu Feng
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | | | - Denise J Cai
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | |
Collapse
|
44
|
Li G, Hsu LM, Wu Y, Bozoki AC, Shih YYI, Yap PT. Revealing excitation-inhibition imbalance in Alzheimer's disease using multiscale neural model inversion of resting-state functional MRI. COMMUNICATIONS MEDICINE 2025; 5:17. [PMID: 39814858 PMCID: PMC11735810 DOI: 10.1038/s43856-025-00736-7] [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: 09/21/2023] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a serious neurodegenerative disorder without a clear understanding of pathophysiology. Recent experimental data have suggested neuronal excitation-inhibition (E-I) imbalance as an essential element of AD pathology, but E-I imbalance has not been systematically mapped out for either local or large-scale neuronal circuits in AD, precluding precise targeting of E-I imbalance in AD treatment. METHOD In this work, we apply a Multiscale Neural Model Inversion (MNMI) framework to the resting-state functional MRI data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) to identify brain regions with disrupted E-I balance in a large network during AD progression. RESULTS We observe that both intra-regional and inter-regional E-I balance is progressively disrupted from cognitively normal individuals, to mild cognitive impairment (MCI) and to AD. Also, we find that local inhibitory connections are more significantly impaired than excitatory ones and the strengths of most connections are reduced in MCI and AD, leading to gradual decoupling of neural populations. Moreover, we reveal a core AD network comprised mainly of limbic and cingulate regions. These brain regions exhibit consistent E-I alterations across MCI and AD, and thus may represent important AD biomarkers and therapeutic targets. Lastly, the E-I balance of multiple brain regions in the core AD network is found to be significantly correlated with the cognitive test score. CONCLUSIONS Our study constitutes an important attempt to delineate E-I imbalance in large-scale neuronal circuits during AD progression, which may facilitate the development of new treatment paradigms to restore physiological E-I balance in AD.
Collapse
Affiliation(s)
- Guoshi Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ye Wu
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrea C Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pew-Thian Yap
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
45
|
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
|
46
|
Schneeweis A, Hillyer D, Lama T, Kim D, Palka C, Djemil S, Abdel-Ghani M, Mandella K, Zhu W, Alvarez N, Stefansson L, Yasuda R, Ma J, Pak DTS. Mass spectrometry identifies tau C-terminal phosphorylation cluster during neuronal hyperexcitation. J Neurochem 2025; 169:e16221. [PMID: 39308063 DOI: 10.1111/jnc.16221] [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: 04/03/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 12/20/2024]
Abstract
Tau is a microtubule-associated protein implicated in Alzheimer's disease (AD) and other neurodegenerative disorders termed tauopathies. Pathological, aggregated forms of tau form neurofibrillary tangles (NFTs), impairing its ability to stabilize microtubules and promoting neurotoxicity. Indeed, NFTs correlate with neuronal loss and cognitive impairment. Hyperphosphorylation of tau is seen in all tauopathies and mirrors disease progression, suggesting an essential role in pathogenesis. However, hyperphosphorylation remains a generic and ill-defined term, obscuring the functional importance of specific sites in different physiological or pathological settings. Here, we focused on global mapping of tau phosphorylation specifically during conditions of neuronal hyperexcitation. Hyperexcitation is a property of AD and other tauopathies linked to human cognitive deficits and increased risk of developing seizures and epilepsy. Moreover, hyperexcitation promotes extracellular secretion and trans-synaptic propagation of tau. Using unbiased mass spectrometry, we identified a novel phosphorylation signature in the C-terminal domain of tau detectable only during neuronal hyperactivity in primary cultured rat hippocampal neurons. These sites influenced tau localization to dendrites as well as the size of excitatory postsynaptic sites. These results demonstrate novel physiological tau functions at synapses and the utility of comprehensive analysis of tau phosphorylation during specific signaling contexts.
Collapse
Affiliation(s)
- Amanda Schneeweis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Dawson Hillyer
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Tsering Lama
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Daeun Kim
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Charles Palka
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Mai Abdel-Ghani
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Kelly Mandella
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - William Zhu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Nicole Alvarez
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Lara Stefansson
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Robert Yasuda
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Junfeng Ma
- Mass Spectrometry and Analytical Pharmacology Shared Resource, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
47
|
Sokolova D, Ghansah SA, Puletti F, Georgiades T, De Schepper S, Zheng Y, Crowley G, Wu L, Rueda-Carrasco J, Koutsiouroumpa A, Muckett P, Freeman OJ, Khakh BS, Hong S. Astrocyte-derived MFG-E8 facilitates microglial synapse elimination in Alzheimer's disease mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.31.606944. [PMID: 39257734 PMCID: PMC11383703 DOI: 10.1101/2024.08.31.606944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Region-specific synapse loss is an early pathological hallmark in Alzheimer's disease (AD). Emerging data in mice and humans highlight microglia, the brain-resident macrophages, as cellular mediators of synapse loss; however, the upstream modulators of microglia-synapse engulfment remain elusive. Here, we report a distinct subset of astrocytes, which are glial cells essential for maintaining synapse homeostasis, appearing in a region-specific manner with age and amyloidosis at onset of synapse loss. These astrocytes are distinguished by their peri-synaptic processes which are 'bulbous' in morphology, contain accumulated p62-immunoreactive bodies, and have reduced territorial domains, resulting in a decrease of astrocyte-synapse coverage. Using integrated in vitro and in vivo approaches, we show that astrocytes upregulate and secrete phagocytic modulator, milk fat globule-EGF factor 8 (MFG-E8), which is sufficient and necessary for promoting microglia-synapse engulfment in their local milieu. Finally, we show that knocking down Mfge8 specifically from astrocytes using a viral CRISPR-saCas9 system prevents microglia-synapse engulfment and ameliorates synapse loss in two independent amyloidosis mouse models of AD. Altogether, our findings highlight astrocyte-microglia crosstalk in determining synapse fate in amyloid models and nominate astrocytic MFGE8 as a potential target to ameliorate synapse loss during the earliest stages of AD.
Collapse
Affiliation(s)
- Dimitra Sokolova
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
- Neuroscience BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Shari Addington Ghansah
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Francesca Puletti
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Tatiana Georgiades
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Sebastiaan De Schepper
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Yongjing Zheng
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Gerard Crowley
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Ling Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Javier Rueda-Carrasco
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Angeliki Koutsiouroumpa
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Philip Muckett
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Oliver J. Freeman
- Neuroscience BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Baljit S. Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| |
Collapse
|
48
|
Cai Y, Wang T. Regulation of presynaptic homeostatic plasticity by glial signalling in Alzheimer's disease. J Physiol 2024. [PMID: 39705214 DOI: 10.1113/jp286751] [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: 05/19/2024] [Accepted: 12/04/2024] [Indexed: 12/22/2024] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia among the elderly, affects numerous individuals worldwide. Despite advances in understanding the molecular underpinnings of AD pathology, effective treatments to prevent or cure the disease remain elusive. AD is characterized not only by pathological hallmarks such as amyloid plaques and neurofibrillary tangles but also by impairments in synaptic physiology, circuit activity and cognitive function. Synaptic homeostatic plasticity plays a vital role in maintaining the stability of synaptic and neural functions amid genetic and environmental disturbances. A key component of this regulation is presynaptic homeostatic potentiation, where increased presynaptic neurotransmitter release compensates for reduced postsynaptic glutamate receptor functionality, thereby stabilizing neuronal excitability. The role of presynaptic homeostatic plasticity in synapse stabilization in AD, however, remains unclear. Moreover, recent advances in transcriptomics have illuminated the complex roles of glial cells in regulating synaptic function in ageing brains and in the progression of neurodegenerative diseases. Yet, the impact of AD-related abnormalities in glial signalling on synaptic homeostatic plasticity has not been fully delineated. This review discusses recent findings on how glial dysregulation in AD affects presynaptic homeostatic plasticity. There is increasing evidence that disrupted glial signalling, particularly through aberrant histone acetylation and transcriptomic changes in glia, compromises this plasticity in AD. Notably, the sphingosine signalling pathway has been identified as being protective in stabilizing synaptic physiology through epigenetic and homeostatic mechanisms, presenting potential therapeutic targets for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Yimei Cai
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Tingting Wang
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., USA
| |
Collapse
|
49
|
Sollazzo R, Li Puma DD, Aceto G, Paciello F, Colussi C, Vita MG, Giuffrè GM, Pastore F, Casamassa A, Rosati J, Novelli A, Maietta S, Tiziano FD, Marra C, Ripoli C, Grassi C. Structural and functional alterations of neurons derived from sporadic Alzheimer's disease hiPSCs are associated with downregulation of the LIMK1-cofilin axis. Alzheimers Res Ther 2024; 16:267. [PMID: 39702316 DOI: 10.1186/s13195-024-01632-3] [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: 04/11/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by the accumulation of pathological proteins and synaptic dysfunction. This study aims to investigate the molecular and functional differences between human induced pluripotent stem cells (hiPSCs) derived from patients with sporadic AD (sAD) and age-matched controls (healthy subjects, HS), focusing on their neuronal differentiation and synaptic properties in order to better understand the cellular and molecular mechanisms underlying AD pathology. METHODS Skin fibroblasts from sAD patients (n = 5) and HS subjects (n = 5) were reprogrammed into hiPSCs using non-integrating Sendai virus vectors. Through karyotyping, we assessed pluripotency markers (OCT4, SOX2, TRA-1-60) and genomic integrity. Neuronal differentiation was evaluated by immunostaining for MAP2 and NEUN. Electrophysiological properties were measured using whole-cell patch-clamp, while protein expression of Aβ, phosphorylated tau, Synapsin-1, Synaptophysin, PSD95, and GluA1 was quantified by western blot. We then focused on PAK1-LIMK1-Cofilin signaling, which plays a key role in regulating synaptic structure and function, both of which are disrupted in neurodegenerative diseases such as AD. RESULTS sAD and HS hiPSCs displayed similar stemness features and genomic stability. However, they differed in neuronal differentiation and function. sAD-derived neurons (sAD-hNs) displayed increased levels of AD-related proteins, including Aβ and phosphorylated tau. Electrophysiological analyses revealed that while both sAD- and HS-hNs generated action potentials, sAD-hNs exhibited decreased spontaneous synaptic activity. Significant reductions in the expression of synaptic proteins such as Synapsin-1, Synaptophysin, PSD95, and GluA1 were found in sAD-hNs, which are also characterized by reduced neurite length, indicating impaired differentiation. Notably, sAD-hNs demonstrated a marked reduction in LIMK1 phosphorylation, which could be the underlying cause for the changes in cytoskeletal dynamics that we found, leading to the morphological and functional modifications observed in sAD-hNs. To further investigate the involvement of the LIMK1 pathway in the morphological and functional changes observed in sAD neurons, we conducted perturbation experiments using the specific LIMK1 inhibitor, BMS-5. Neurons obtained from healthy subjects treated with the inhibitor showed similar morphological changes to those observed in sAD neurons, confirming that LIMK1 activity is crucial for maintaining normal neuronal structure. Furthermore, administration of the inhibitor to sAD neurons did not exacerbate the morphological alterations, suggesting that LIMK1 activity is already compromised in these cells. CONCLUSION Our findings demonstrate that although sAD- and HS-hiPSCs are similar in their stemness and genomic stability, sAD-hNs exhibit distinct functional and structural anomalies mirroring AD pathology. These anomalies include synaptic dysfunction, altered cytoskeletal organization, and accumulation of AD-related proteins. Our study underscores the usefulness of hiPSCs in modeling AD and provides insights into the disease's molecular underpinnings, thus highlighting potential therapeutic targets.
Collapse
Affiliation(s)
- Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Claudia Colussi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Engineering, Istituto Di Analisi Dei Sistemi Ed Informatica "Antonio Ruberti", National Research Council, 00185, Rome, Italy
| | | | | | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Alessia Casamassa
- Cellular Reprogramming Unit, Fondazione IRCCS Casa, Sollievo Della Sofferenza, 71013 - San Giovanni, Rotondo, Italy
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa, Sollievo Della Sofferenza, 71013 - San Giovanni, Rotondo, Italy
- Saint Camillus International, University of Health Sciences, 00131, Rome, Italy
| | - Agnese Novelli
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sabrina Maietta
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Francesco Danilo Tiziano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Camillo Marra
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
50
|
Power SK, Venkatesan S, Qu S, McLaurin J, Lambe EK. Enhanced prefrontal nicotinic signaling as evidence of active compensation in Alzheimer's disease models. Transl Neurodegener 2024; 13:58. [PMID: 39623428 PMCID: PMC11613856 DOI: 10.1186/s40035-024-00452-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 08/22/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Cognitive reserve allows for resilience to neuropathology, potentially through active compensation. Here, we examine ex vivo electrophysiological evidence for active compensation in Alzheimer's disease (AD) focusing on the cholinergic innervation of layer 6 in prefrontal cortex. Cholinergic pathways are vulnerable to neuropathology in AD and its preclinical models, and their modulation of deep layer prefrontal cortex is essential for attention and executive function. METHODS We functionally interrogated cholinergic modulation of prefrontal layer 6 pyramidal neurons in two preclinical models: a compound transgenic AD mouse model that permits optogenetically-triggered release of endogenous acetylcholine and a transgenic AD rat model that closely recapitulates the human trajectory of AD. We then tested the impact of therapeutic interventions to further amplify the compensated responses and preserve the typical kinetic profile of cholinergic signaling. RESULTS In two AD models, we found potentially compensatory upregulation of functional cholinergic responses above non-transgenic controls after onset of pathology. To identify the locus of this enhanced cholinergic signal, we dissected key pre- and post-synaptic components with pharmacological strategies. We identified a significant and selective increase in post-synaptic nicotinic receptor signalling on prefrontal cortical neurons. To probe the additional impact of therapeutic intervention on the adapted circuit, we tested cholinergic and nicotinic-selective pro-cognitive treatments. Inhibition of acetylcholinesterase further enhanced endogenous cholinergic responses but greatly distorted their kinetics. Positive allosteric modulation of nicotinic receptors, by contrast, enhanced endogenous cholinergic responses and retained their rapid kinetics. CONCLUSIONS We demonstrate that functional nicotinic upregulation occurs within the prefrontal cortex in two AD models. Promisingly, this nicotinic signal can be further enhanced while preserving its rapid kinetic signature. Taken together, our work suggests that compensatory mechanisms are active within the prefrontal cortex that can be harnessed by nicotinic receptor positive allosteric modulation, highlighting a new direction for cognitive treatment in AD neuropathology.
Collapse
Affiliation(s)
- Saige K Power
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sridevi Venkatesan
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sarah Qu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - JoAnne McLaurin
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Evelyn K Lambe
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Obstetrics and Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5G 1E2, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada.
| |
Collapse
|