1
|
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
|
2
|
Perdok A, Van Acker ZP, Vrancx C, Sannerud R, Vorsters I, Verrengia A, Callaerts-Végh Z, Creemers E, Gutiérrez Fernández S, D'hauw B, Serneels L, Wierda K, Chávez-Gutiérrez L, Annaert W. Altered expression of Presenilin2 impacts endolysosomal homeostasis and synapse function in Alzheimer's disease-relevant brain circuits. Nat Commun 2024; 15:10412. [PMID: 39613768 DOI: 10.1038/s41467-024-54777-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
Rare mutations in the gene encoding presenilin2 (PSEN2) are known to cause familial Alzheimer's disease (FAD). Here, we explored how altered PSEN2 expression impacts on the amyloidosis, endolysosomal abnormalities, and synaptic dysfunction observed in female APP knock-in mice. We demonstrate that PSEN2 knockout (KO) as well as the FAD-associated N141IKI mutant accelerate AD-related pathologies in female mice. Both models showed significant deficits in working memory that linked to elevated PSEN2 expression in the hippocampal CA3 region. The mossy fiber circuit of APPxPSEN2KO and APPxFADPSEN2 mice had smaller pre-synaptic compartments, distinct changes in synaptic vesicle populations and significantly impaired long term potentiation compared to APPKI mice. At the cellular level, altered PSEN2 expression resulted in endolysosomal defects and lowered surface expression of synaptic proteins. As PSEN2/γ-secretase is restricted to late endosomes/lysosomes, we propose PSEN2 impacts endolysosomal homeostasis, affecting synaptic signaling in AD-relevant vulnerable brain circuits; which could explain how mutant PSEN2 accelerates AD pathogenesis.
Collapse
Affiliation(s)
- Anika Perdok
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Zoë P Van Acker
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Ragna Sannerud
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Inge Vorsters
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Assunta Verrengia
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
| | - Zsuzsanna Callaerts-Végh
- mINT Animal Behavior Facility, Faculty of Psychology, KU Leuven, Tiensestraat 102, Leuven, Belgium
| | - Eline Creemers
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Sara Gutiérrez Fernández
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
- Laboratory of Proteolytic Mechanisms mediating Neurodegeneration, Leuven, Belgium
| | - Britt D'hauw
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Lutgarde Serneels
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
- Mouse Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Keimpe Wierda
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Lucía Chávez-Gutiérrez
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium
- Laboratory of Proteolytic Mechanisms mediating Neurodegeneration, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium.
- Department of Neurosciences, KU Leuven, Herestraat 49box 602, Leuven, Belgium.
| |
Collapse
|
3
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [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: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- 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
| | - Yanjun Liu
- 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
| | - Chuhui Yin
- 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
| | - Yan Zeng
- 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
|
4
|
Vimbi V, Shaffi N, Mahmud M. Interpreting artificial intelligence models: a systematic review on the application of LIME and SHAP in Alzheimer's disease detection. Brain Inform 2024; 11:10. [PMID: 38578524 PMCID: PMC10997568 DOI: 10.1186/s40708-024-00222-1] [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: 09/02/2023] [Accepted: 03/04/2024] [Indexed: 04/06/2024] Open
Abstract
Explainable artificial intelligence (XAI) has gained much interest in recent years for its ability to explain the complex decision-making process of machine learning (ML) and deep learning (DL) models. The Local Interpretable Model-agnostic Explanations (LIME) and Shaply Additive exPlanation (SHAP) frameworks have grown as popular interpretive tools for ML and DL models. This article provides a systematic review of the application of LIME and SHAP in interpreting the detection of Alzheimer's disease (AD). Adhering to PRISMA and Kitchenham's guidelines, we identified 23 relevant articles and investigated these frameworks' prospective capabilities, benefits, and challenges in depth. The results emphasise XAI's crucial role in strengthening the trustworthiness of AI-based AD predictions. This review aims to provide fundamental capabilities of LIME and SHAP XAI frameworks in enhancing fidelity within clinical decision support systems for AD prognosis.
Collapse
Affiliation(s)
- Viswan Vimbi
- College of Computing and Information Sciences, University of Technology and Applied Sciences, OM 311, Sohar, Sultanate of Oman
| | - Noushath Shaffi
- College of Computing and Information Sciences, University of Technology and Applied Sciences, OM 311, Sohar, Sultanate of Oman
| | - Mufti Mahmud
- Department of Computer Science, Nottingham Trent University, Nottingham, NG11 8NS, UK.
- Medical Technologies Innovation Facility, Nottingham Trent University, Nottingham, NG11 8NS, UK.
- Computing and Informatics Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK.
| |
Collapse
|
5
|
García-Carlos CA, Basurto-Islas G, Perry G, Mondragón-Rodríguez S. Meta-Analysis in Transgenic Alzheimer's Disease Mouse Models Reveals Opposite Brain Network Effects of Amyloid-β and Phosphorylated Tau Proteins. J Alzheimers Dis 2024; 99:595-607. [PMID: 38669540 DOI: 10.3233/jad-231365] [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: 04/28/2024]
Abstract
Background Cognitive deficits observed in Alzheimer's disease (AD) patients have been correlated with altered hippocampal activity. Although the mechanism remains under extensive study, neurofibrillary tangles and amyloid plaques have been proposed as responsible for brain activity alterations. Aiming to unveil the mechanism, researchers have developed several transgenic models of AD. Nevertheless, the variability in hippocampal oscillatory alterations found in different genetic backgrounds and ages remains unclear. Objective To assess the oscillatory alterations in relation to animal developmental age and protein inclusion, amyloid-β (Aβ) load, and abnormally phosphorylated tau (pTau), we reviewed and analyzed the published data on peak power, frequency, and quantification of theta-gamma cross-frequency coupling (modulation index values). Methods To ensure that the search was as current as possible, a systematic review was conducted to locate and abstract all studies published from January 2000 to February 2023 that involved in vivo hippocampal local field potential recording in transgenic mouse models of AD. Results The presence of Aβ was associated with electrophysiological alterations that are mainly reflected in power increases, frequency decreases, and lower modulation index values. Concomitantly, pTau accumulation was associated with electrophysiological alterations that are mainly reflected in power decreases, frequency decreases, and no significant alterations in modulation index values. Conclusions In this study, we showed that electrophysiological parameters are altered from prodromal stages to the late stages of pathology. Thus, we found that Aβ deposition is associated with brain network hyperexcitability, whereas pTau deposition mainly leads to brain network hypoexcitability in transgenic models.
Collapse
Affiliation(s)
- Carlos Antonio García-Carlos
- UNAM Division of Neurosciences, Institute of Cellular Physiology, National Autonomous University of México, México City, México
| | | | - George Perry
- UTSA Neuroscience Institute and Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Siddhartha Mondragón-Rodríguez
- UAQ Centre for Applied Biomedical Research - CIBA, School of Medicine, Autonomous University of Querétaro, Querétaro, México
- CONAHCYT National Council for Science and Technology, México City, México
| |
Collapse
|
6
|
Victorino DB, Faber J, Pinheiro DJLL, Scorza FA, Almeida ACG, Costa ACS, Scorza CA. Toward the Identification of Neurophysiological Biomarkers for Alzheimer's Disease in Down Syndrome: A Potential Role for Cross-Frequency Phase-Amplitude Coupling Analysis. Aging Dis 2023; 14:428-449. [PMID: 37008053 PMCID: PMC10017148 DOI: 10.14336/ad.2022.0906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
Cross-frequency coupling (CFC) mechanisms play a central role in brain activity. Pathophysiological mechanisms leading to many brain disorders, such as Alzheimer's disease (AD), may produce unique patterns of brain activity detectable by electroencephalography (EEG). Identifying biomarkers for AD diagnosis is also an ambition among research teams working in Down syndrome (DS), given the increased susceptibility of people with DS to develop early-onset AD (DS-AD). Here, we review accumulating evidence that altered theta-gamma phase-amplitude coupling (PAC) may be one of the earliest EEG signatures of AD, and therefore may serve as an adjuvant tool for detecting cognitive decline in DS-AD. We suggest that this field of research could potentially provide clues to the biophysical mechanisms underlying cognitive dysfunction in DS-AD and generate opportunities for identifying EEG-based biomarkers with diagnostic and prognostic utility in DS-AD.
Collapse
Affiliation(s)
- Daniella B Victorino
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Federal University of São Paulo / Paulista Medical School, São Paulo, SP, Brazil.
| | - Jean Faber
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Federal University of São Paulo / Paulista Medical School, São Paulo, SP, Brazil.
| | - Daniel J. L. L Pinheiro
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Federal University of São Paulo / Paulista Medical School, São Paulo, SP, Brazil.
| | - Fulvio A Scorza
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Federal University of São Paulo / Paulista Medical School, São Paulo, SP, Brazil.
| | - Antônio C. G Almeida
- Department of Biosystems Engineering, Federal University of São João Del Rei, Minas Gerais, MG, Brazil.
| | - Alberto C. S Costa
- Division of Psychiatry, Case Western Reserve University, Cleveland, OH, United States.
- Department of Macromolecular Science and Engineering, Case Western Reserve University, Cleveland, OH, United States.
| | - Carla A Scorza
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Federal University of São Paulo / Paulista Medical School, São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Lia A, Sansevero G, Chiavegato A, Sbrissa M, Pendin D, Mariotti L, Pozzan T, Berardi N, Carmignoto G, Fasolato C, Zonta M. Rescue of astrocyte activity by the calcium sensor STIM1 restores long-term synaptic plasticity in female mice modelling Alzheimer's disease. Nat Commun 2023; 14:1590. [PMID: 36949142 PMCID: PMC10033875 DOI: 10.1038/s41467-023-37240-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 03/09/2023] [Indexed: 03/24/2023] Open
Abstract
Calcium dynamics in astrocytes represent a fundamental signal that through gliotransmitter release regulates synaptic plasticity and behaviour. Here we present a longitudinal study in the PS2APP mouse model of Alzheimer's disease (AD) linking astrocyte Ca2+ hypoactivity to memory loss. At the onset of plaque deposition, somatosensory cortical astrocytes of AD female mice exhibit a drastic reduction of Ca2+ signaling, closely associated with decreased endoplasmic reticulum Ca2+ concentration and reduced expression of the Ca2+ sensor STIM1. In parallel, astrocyte-dependent long-term synaptic plasticity declines in the somatosensory circuitry, anticipating specific tactile memory loss. Notably, we show that both astrocyte Ca2+ signaling and long-term synaptic plasticity are fully recovered by selective STIM1 overexpression in astrocytes. Our data unveil astrocyte Ca2+ hypoactivity in neocortical astrocytes as a functional hallmark of early AD stages and indicate astrocytic STIM1 as a target to rescue memory deficits.
Collapse
Affiliation(s)
- Annamaria Lia
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Gabriele Sansevero
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Angela Chiavegato
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Miriana Sbrissa
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Diana Pendin
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Letizia Mariotti
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, Padua, Italy
| | - Nicoletta Berardi
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Giorgio Carmignoto
- Neuroscience Institute, National Research Council (CNR), Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| | - Micaela Zonta
- Neuroscience Institute, National Research Council (CNR), Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
8
|
Fabietti M, Mahmud M, Lotfi A, Kaiser MS. ABOT: an open-source online benchmarking tool for machine learning-based artefact detection and removal methods from neuronal signals. Brain Inform 2022; 9:19. [PMID: 36048345 PMCID: PMC9437165 DOI: 10.1186/s40708-022-00167-3] [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: 04/25/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Brain signals are recorded using different techniques to aid an accurate understanding of brain function and to treat its disorders. Untargeted internal and external sources contaminate the acquired signals during the recording process. Often termed as artefacts, these contaminations cause serious hindrances in decoding the recorded signals; hence, they must be removed to facilitate unbiased decision-making for a given investigation. Due to the complex and elusive manifestation of artefacts in neuronal signals, computational techniques serve as powerful tools for their detection and removal. Machine learning (ML) based methods have been successfully applied in this task. Due to ML's popularity, many articles are published every year, making it challenging to find, compare and select the most appropriate method for a given experiment. To this end, this paper presents ABOT (Artefact removal Benchmarking Online Tool) as an online benchmarking tool which allows users to compare existing ML-driven artefact detection and removal methods from the literature. The characteristics and related information about the existing methods have been compiled as a knowledgebase (KB) and presented through a user-friendly interface with interactive plots and tables for users to search it using several criteria. Key characteristics extracted from over 120 articles from the literature have been used in the KB to help compare the specific ML models. To comply with the FAIR (Findable, Accessible, Interoperable and Reusable) principle, the source code and documentation of the toolbox have been made available via an open-access repository.
Collapse
Affiliation(s)
- Marcos Fabietti
- Department of Computer Science, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
| | - Mufti Mahmud
- Department of Computer Science, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK.
- Medical Technologies Innovation Facility, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK.
- Computing and Informatics Research Centre, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK.
| | - Ahmad Lotfi
- Department of Computer Science, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
| | - M Shamim Kaiser
- Institute of Information Technology, Jahangirnagar University, Dhaka, 1342, Savar, Bangladesh
| |
Collapse
|
9
|
Preininger MK, Kaufer D. Blood-Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging. Int J Mol Sci 2022; 23:6217. [PMID: 35682895 PMCID: PMC9180977 DOI: 10.3390/ijms23116217] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 01/27/2023] Open
Abstract
As the most abundant cell types in the brain, astrocytes form a tissue-wide signaling network that is responsible for maintaining brain homeostasis and regulating various brain activities. Here, we review some of the essential functions that astrocytes perform in supporting neurons, modulating the immune response, and regulating and maintaining the blood-brain barrier (BBB). Given their importance in brain health, it follows that astrocyte dysfunction has detrimental effects. Indeed, dysfunctional astrocytes are implicated in age-related neuropathology and participate in the onset and progression of neurodegenerative diseases. Here, we review two mechanisms by which astrocytes mediate neuropathology in the aging brain. First, age-associated blood-brain barrier dysfunction (BBBD) causes the hyperactivation of TGFβ signaling in astrocytes, which elicits a pro-inflammatory and epileptogenic phenotype. Over time, BBBD-associated astrocyte dysfunction results in hippocampal and cortical neural hyperexcitability and cognitive deficits. Second, senescent astrocytes accumulate in the brain with age and exhibit a decreased functional capacity and the secretion of senescent-associated secretory phenotype (SASP) factors, which contribute to neuroinflammation and neurotoxicity. Both BBBD and senescence progressively increase during aging and are associated with increased risk of neurodegenerative disease, but the relationship between the two has not yet been established. Thus, we discuss the potential relationship between BBBD, TGFβ hyperactivation, and senescence with respect to astrocytes in the context of aging and disease and identify future areas of investigation in the field.
Collapse
Affiliation(s)
- Marcela K. Preininger
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA;
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA;
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
10
|
Leparulo A, Bisio M, Redolfi N, Pozzan T, Vassanelli S, Fasolato C. Accelerated Aging Characterizes the Early Stage of Alzheimer's Disease. Cells 2022; 11:238. [PMID: 35053352 PMCID: PMC8774248 DOI: 10.3390/cells11020238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/12/2021] [Accepted: 01/08/2022] [Indexed: 02/01/2023] Open
Abstract
For Alzheimer's disease (AD), aging is the main risk factor, but whether cognitive impairments due to aging resemble early AD deficits is not yet defined. When working with mouse models of AD, the situation is just as complicated, because only a few studies track the progression of the disease at different ages, and most ignore how the aging process affects control mice. In this work, we addressed this problem by comparing the aging process of PS2APP (AD) and wild-type (WT) mice at the level of spontaneous brain electrical activity under anesthesia. Using local field potential recordings, obtained with a linear probe that traverses the posterior parietal cortex and the entire hippocampus, we analyzed how multiple electrical parameters are modified by aging in AD and WT mice. With this approach, we highlighted AD specific features that appear in young AD mice prior to plaque deposition or that are delayed at 12 and 16 months of age. Furthermore, we identified aging characteristics present in WT mice but also occurring prematurely in young AD mice. In short, we found that reduction in the relative power of slow oscillations (SO) and Low/High power imbalance are linked to an AD phenotype at its onset. The loss of SO connectivity and cortico-hippocampal coupling between SO and higher frequencies as well as the increase in UP-state and burst durations are found in young AD and old WT mice. We show evidence that the aging process is accelerated by the mutant PS2 itself and discuss such changes in relation to amyloidosis and gliosis.
Collapse
Affiliation(s)
- Alessandro Leparulo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.B.); (N.R.); (T.P.)
| | - Marta Bisio
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.B.); (N.R.); (T.P.)
| | - Nelly Redolfi
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.B.); (N.R.); (T.P.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.B.); (N.R.); (T.P.)
- Neuroscience Institute-Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2B, 35129 Padua, Italy
| | - Stefano Vassanelli
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.B.); (N.R.); (T.P.)
- Padua Neuroscience Center (PNC), University of Padua, Via G. Orus 2B, 35129 Padua, Italy
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.B.); (N.R.); (T.P.)
| |
Collapse
|
11
|
Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev 2021; 68:101335. [PMID: 33812051 PMCID: PMC8168445 DOI: 10.1016/j.arr.2021.101335] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/21/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023]
Abstract
Astrocyte reactivity is a hallmark of neuroinflammation that arises with Alzheimer’s disease (AD) and nearly every other neurodegenerative condition. While astrocytes certainly contribute to classic inflammatory processes (e.g. cytokine release, waste clearance, and tissue repair), newly emerging technologies for measuring and targeting cell specific activities in the brain have uncovered essential roles for astrocytes in synapse function, brain metabolism, neurovascular coupling, and sleep/wake patterns. In this review, we use a holistic approach to incorporate, and expand upon, classic neuroinflammatory concepts to consider how astrocyte dysfunction/reactivity modulates multiple pathological and clinical hallmarks of AD. Our ever-evolving understanding of astrocyte signaling in neurodegeneration is not only revealing new drug targets and treatments for dementia but is suggesting we reimagine AD pathophysiological mechanisms.
Collapse
Affiliation(s)
- Brittani R Price
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Lance A Johnson
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Physiology, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA.
| |
Collapse
|
12
|
Familial Alzheimer's disease presenilin-2 mutants affect Ca 2+ homeostasis and brain network excitability. Aging Clin Exp Res 2021; 33:1705-1708. [PMID: 31606858 DOI: 10.1007/s40520-019-01341-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/27/2019] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease (AD) is the most frequent cause of dementia in the elderly. Few cases are familial (FAD), due to autosomal dominant mutations in presenilin-1 (PS1), presenilin-2 (PS2) or amyloid precursor protein (APP). The three proteins are involved in the generation of amyloid-beta (Aβ) peptides, providing genetic support to the hypothesis of Aβ pathogenicity. However, clinical trials focused on the Aβ pathway failed in their attempt to modify disease progression, suggesting the existence of additional pathogenic mechanisms. Ca2+ dysregulation is a feature of cerebral aging, with an increased frequency and anticipated age of onset in several forms of neurodegeneration, including AD. Interestingly, FAD-linked PS1 and PS2 mutants alter multiple key cellular pathways, including Ca2+ signaling. By generating novel tools for measuring Ca2+ in living cells, and combining different approaches, we showed that FAD-linked PS2 mutants significantly alter cell Ca2+ signaling and brain network activity, as summarized below.
Collapse
|
13
|
Neuronal Network Excitability in Alzheimer's Disease: The Puzzle of Similar versus Divergent Roles of Amyloid β and Tau. eNeuro 2021; 8:ENEURO.0418-20.2020. [PMID: 33741601 PMCID: PMC8174042 DOI: 10.1523/eneuro.0418-20.2020] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most frequent neurodegenerative disorder that commonly causes dementia in the elderly. Recent evidence indicates that network abnormalities, including hypersynchrony, altered oscillatory rhythmic activity, interneuron dysfunction, and synaptic depression, may be key mediators of cognitive decline in AD. In this review, we discuss characteristics of neuronal network excitability in AD, and the role of Aβ and tau in the induction of network hyperexcitability. Many patients harboring genetic mutations that lead to increased Aβ production suffer from seizures and epilepsy before the development of plaques. Similarly, pathologic accumulation of hyperphosphorylated tau has been associated with hyperexcitability in the hippocampus. We present common and divergent roles of tau and Aβ on neuronal hyperexcitability in AD, and hypotheses that could serve as a template for future experiments.
Collapse
|
14
|
Rossi A, Galla L, Gomiero C, Zentilin L, Giacca M, Giorgio V, Calì T, Pozzan T, Greotti E, Pizzo P. Calcium Signaling and Mitochondrial Function in Presenilin 2 Knock-Out Mice: Looking for Any Loss-of-Function Phenotype Related to Alzheimer's Disease. Cells 2021; 10:204. [PMID: 33494218 PMCID: PMC7909802 DOI: 10.3390/cells10020204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder in which learning, memory and cognitive functions decline progressively. Familial forms of AD (FAD) are caused by mutations in amyloid precursor protein (APP), presenilin 1 (PSEN1) and presenilin 2 (PSEN2) genes. Presenilin 1 (PS1) and its homologue, presenilin 2 (PS2), represent, alternatively, the catalytic core of the γ-secretase complex that, by cleaving APP, produces neurotoxic amyloid beta (Aβ) peptides responsible for one of the histopathological hallmarks in AD brains, the amyloid plaques. Recently, PSEN1 FAD mutations have been associated with a loss-of-function phenotype. To investigate whether this finding can also be extended to PSEN2 FAD mutations, we studied two processes known to be modulated by PS2 and altered by FAD mutations: Ca2+ signaling and mitochondrial function. By exploiting neurons derived from a PSEN2 knock-out (PS2-/-) mouse model, we found that, upon IP3-generating stimulation, cytosolic Ca2+ handling is not altered, compared to wild-type cells, while mitochondrial Ca2+ uptake is strongly compromised. Accordingly, PS2-/- neurons show a marked reduction in endoplasmic reticulum-mitochondria apposition and a slight alteration in mitochondrial respiration, whereas mitochondrial membrane potential, and organelle morphology and number appear unchanged. Thus, although some alterations in mitochondrial function appear to be shared between PS2-/- and FAD-PS2-expressing neurons, the mechanisms leading to these defects are quite distinct between the two models. Taken together, our data appear to be difficult to reconcile with the proposal that FAD-PS2 mutants are loss-of-function, whereas the concept that PS2 plays a key role in sustaining mitochondrial function is here confirmed.
Collapse
Affiliation(s)
- Alice Rossi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
| | - Luisa Galla
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Chiara Gomiero
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (L.Z.); (M.G.)
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (L.Z.); (M.G.)
| | - Valentina Giorgio
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Department of Biomedical and Neuromotor Science, University of Bologna, 40112 Bologna, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| |
Collapse
|
15
|
Alcantara-Gonzalez D, Chartampila E, Criscuolo C, Scharfman HE. Early changes in synaptic and intrinsic properties of dentate gyrus granule cells in a mouse model of Alzheimer's disease neuropathology and atypical effects of the cholinergic antagonist atropine. Neurobiol Dis 2021; 152:105274. [PMID: 33484828 DOI: 10.1016/j.nbd.2021.105274] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
It has been reported that hyperexcitability occurs in a subset of patients with Alzheimer's disease (AD) and hyperexcitability could contribute to the disease. Several studies have suggested that the hippocampal dentate gyrus (DG) may be an important area where hyperexcitability occurs. Therefore, we tested the hypothesis that the principal DG cell type, granule cells (GCs), would exhibit changes at the single-cell level which would be consistent with hyperexcitability and might help explain it. We used the Tg2576 mouse, where it has been shown that hyperexcitability is robust at 2-3 months of age. GCs from 2 to 3-month-old Tg2576 mice were compared to age-matched wild type (WT) mice. Effects of muscarinic cholinergic antagonism were tested because previously we found that Tg2576 mice exhibited hyperexcitability in vivo that was reduced by the muscarinic cholinergic antagonist atropine, counter to the dogma that in AD one needs to boost cholinergic function. The results showed that GCs from Tg2576 mice exhibited increased frequency of spontaneous excitatory postsynaptic potentials/currents (sEPSP/Cs) and reduced frequency of spontaneous inhibitory synaptic events (sIPSCs) relative to WT, increasing the excitation:inhibition (E:I) ratio. There was an inward NMDA receptor-dependent current that we defined here as a novel synaptic current (nsC) in Tg2576 mice because it was very weak in WT mice. Intrinsic properties were distinct in Tg2576 GCs relative to WT. In summary, GCs of the Tg2576 mouse exhibit early electrophysiological alterations that are consistent with increased synaptic excitation, reduced inhibition, and muscarinic cholinergic dysregulation. The data support previous suggestions that the DG contributes to hyperexcitability and there is cholinergic dysfunction early in life in AD mouse models.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Elissavet Chartampila
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Chiara Criscuolo
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Helen E Scharfman
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY 10016, USA; Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
16
|
Noor MBT, Zenia NZ, Kaiser MS, Mamun SA, Mahmud M. Application of deep learning in detecting neurological disorders from magnetic resonance images: a survey on the detection of Alzheimer's disease, Parkinson's disease and schizophrenia. Brain Inform 2020; 7:11. [PMID: 33034769 PMCID: PMC7547060 DOI: 10.1186/s40708-020-00112-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroimaging, in particular magnetic resonance imaging (MRI), has been playing an important role in understanding brain functionalities and its disorders during the last couple of decades. These cutting-edge MRI scans, supported by high-performance computational tools and novel ML techniques, have opened up possibilities to unprecedentedly identify neurological disorders. However, similarities in disease phenotypes make it very difficult to detect such disorders accurately from the acquired neuroimaging data. This article critically examines and compares performances of the existing deep learning (DL)-based methods to detect neurological disorders-focusing on Alzheimer's disease, Parkinson's disease and schizophrenia-from MRI data acquired using different modalities including functional and structural MRI. The comparative performance analysis of various DL architectures across different disorders and imaging modalities suggests that the Convolutional Neural Network outperforms other methods in detecting neurological disorders. Towards the end, a number of current research challenges are indicated and some possible future research directions are provided.
Collapse
Affiliation(s)
- Manan Binth Taj Noor
- Institute of Information Technology, Jahangirnagar University, Savar, 1342, Dhaka, Bangladesh
| | - Nusrat Zerin Zenia
- Institute of Information Technology, Jahangirnagar University, Savar, 1342, Dhaka, Bangladesh
| | - M Shamim Kaiser
- Institute of Information Technology, Jahangirnagar University, Savar, 1342, Dhaka, Bangladesh.
| | - Shamim Al Mamun
- Institute of Information Technology, Jahangirnagar University, Savar, 1342, Dhaka, Bangladesh
| | - Mufti Mahmud
- Department of Computing & Technology, Nottingham Trent University, NG11 8NS, Nottingham, UK.
| |
Collapse
|
17
|
Pizzo P, Basso E, Filadi R, Greotti E, Leparulo A, Pendin D, Redolfi N, Rossini M, Vajente N, Pozzan T, Fasolato C. Presenilin-2 and Calcium Handling: Molecules, Organelles, Cells and Brain Networks. Cells 2020; 9:E2166. [PMID: 32992716 PMCID: PMC7601421 DOI: 10.3390/cells9102166] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Presenilin-2 (PS2) is one of the three proteins that are dominantly mutated in familial Alzheimer's disease (FAD). It forms the catalytic core of the γ-secretase complex-a function shared with its homolog presenilin-1 (PS1)-the enzyme ultimately responsible of amyloid-β (Aβ) formation. Besides its enzymatic activity, PS2 is a multifunctional protein, being specifically involved, independently of γ-secretase activity, in the modulation of several cellular processes, such as Ca2+ signalling, mitochondrial function, inter-organelle communication, and autophagy. As for the former, evidence has accumulated that supports the involvement of PS2 at different levels, ranging from organelle Ca2+ handling to Ca2+ entry through plasma membrane channels. Thus FAD-linked PS2 mutations impact on multiple aspects of cell and tissue physiology, including bioenergetics and brain network excitability. In this contribution, we summarize the main findings on PS2, primarily as a modulator of Ca2+ homeostasis, with particular emphasis on the role of its mutations in the pathogenesis of FAD. Identification of cell pathways and molecules that are specifically targeted by PS2 mutants, as well as of common targets shared with PS1 mutants, will be fundamental to disentangle the complexity of memory loss and brain degeneration that occurs in Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Paola Pizzo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Emy Basso
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Alessandro Leparulo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| | - Diana Pendin
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Nelly Redolfi
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| | - Michela Rossini
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| | - Nicola Vajente
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2B, 35131 Padua, Italy
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| |
Collapse
|
18
|
Prevention of age-associated neuronal hyperexcitability with improved learning and attention upon knockout or antagonism of LPAR2. Cell Mol Life Sci 2020; 78:1029-1050. [PMID: 32468095 PMCID: PMC7897625 DOI: 10.1007/s00018-020-03553-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/16/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022]
Abstract
Recent studies suggest that synaptic lysophosphatidic acids (LPAs) augment glutamate-dependent cortical excitability and sensory information processing in mice and humans via presynaptic LPAR2 activation. Here, we studied the consequences of LPAR2 deletion or antagonism on various aspects of cognition using a set of behavioral and electrophysiological analyses. Hippocampal neuronal network activity was decreased in middle-aged LPAR2−/− mice, whereas hippocampal long-term potentiation (LTP) was increased suggesting cognitive advantages of LPAR2−/− mice. In line with the lower excitability, RNAseq studies revealed reduced transcription of neuronal activity markers in the dentate gyrus of the hippocampus in naïve LPAR2−/− mice, including ARC, FOS, FOSB, NR4A, NPAS4 and EGR2. LPAR2−/− mice behaved similarly to wild-type controls in maze tests of spatial or social learning and memory but showed faster and accurate responses in a 5-choice serial reaction touchscreen task requiring high attention and fast spatial discrimination. In IntelliCage learning experiments, LPAR2−/− were less active during daytime but normally active at night, and showed higher accuracy and attention to LED cues during active times. Overall, they maintained equal or superior licking success with fewer trials. Pharmacological block of the LPAR2 receptor recapitulated the LPAR2−/− phenotype, which was characterized by economic corner usage, stronger daytime resting behavior and higher proportions of correct trials. We conclude that LPAR2 stabilizes neuronal network excitability upon aging and allows for more efficient use of resting periods, better memory consolidation and better performance in tasks requiring high selective attention. Therapeutic LPAR2 antagonism may alleviate aging-associated cognitive dysfunctions.
Collapse
|
19
|
5-Caffeoylquinic Acid Ameliorates Cognitive Decline and Reduces Aβ Deposition by Modulating Aβ Clearance Pathways in APP/PS2 Transgenic Mice. Nutrients 2020; 12:nu12020494. [PMID: 32075202 PMCID: PMC7071270 DOI: 10.3390/nu12020494] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 12/13/2022] Open
Abstract
The accumulation of amyloid β (Aβ) in the brain is a major pathological feature of Alzheimer's disease (AD). In our previous study, we demonstrated that coffee polyphenols (CPP) prevent cognitive dysfunction and Aβ deposition in the brain of an APP/PS2 transgenic mouse AD model. The underlying mechanisms, however, remain to be elucidated. Here, we investigated the effects of the chronic administration of 5-caffeoylquinic acid (5-CQA), the most abundant component of CPP, on cognitive dysfunction in APP/PS2 mice to identify the role of CPP in Aβ elimination. Relative to the untreated controls, the mice fed a 5-CQA-supplemented diet showed significant improvements in their cognitive function assessed by Y-maze and novel object recognition tests. Histochemical analysis revealed that 5-CQA substantially reduced Aβ plaque formation and neuronal loss in the hippocampi. Moreover, 5-CQA upregulated the gene encoding low-density lipoprotein receptor-related protein 1, an Aβ efflux receptor, and normalized the perivascular localization of aquaporin 4, which facilitates Aβ clearance along the paravascular pathway. These results suggest that 5-CQA reduces Aβ deposition in the brain by modulating the Aβ clearance pathways and ameliorating cognitive decline and neuronal loss in APP/PS2 mice. Thus, 5-CQA may be effective in preventing cognitive dysfunction in AD.
Collapse
|
20
|
Galla L, Redolfi N, Pozzan T, Pizzo P, Greotti E. Intracellular Calcium Dysregulation by the Alzheimer's Disease-Linked Protein Presenilin 2. Int J Mol Sci 2020; 21:E770. [PMID: 31991578 PMCID: PMC7037278 DOI: 10.3390/ijms21030770] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Even though most AD cases are sporadic, a small percentage is familial due to autosomal dominant mutations in amyloid precursor protein (APP), presenilin-1 (PSEN1), and presenilin-2 (PSEN2) genes. AD mutations contribute to the generation of toxic amyloid β (Aβ) peptides and the formation of cerebral plaques, leading to the formulation of the amyloid cascade hypothesis for AD pathogenesis. Many drugs have been developed to inhibit this pathway but all these approaches currently failed, raising the need to find additional pathogenic mechanisms. Alterations in cellular calcium (Ca2+) signaling have also been reported as causative of neurodegeneration. Interestingly, Aβ peptides, mutated presenilin-1 (PS1), and presenilin-2 (PS2) variously lead to modifications in Ca2+ homeostasis. In this contribution, we focus on PS2, summarizing how AD-linked PS2 mutants alter multiple Ca2+ pathways and the functional consequences of this Ca2+ dysregulation in AD pathogenesis.
Collapse
Affiliation(s)
- Luisa Galla
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Nelly Redolfi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35131 Padua, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| |
Collapse
|
21
|
Peña-Ortega F. Brain Arrhythmias Induced by Amyloid Beta and Inflammation: Involvement in Alzheimer’s Disease and Other Inflammation-related Pathologies. Curr Alzheimer Res 2020; 16:1108-1131. [DOI: 10.2174/1567205017666191213162233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/13/2022]
Abstract
A variety of neurological diseases, including Alzheimer’s disease (AD), involve amyloid beta (Aβ) accumulation and/or neuroinflammation, which can alter synaptic and neural circuit functions. Consequently, these pathological conditions induce changes in neural network rhythmic activity (brain arrhythmias), which affects many brain functions. Neural network rhythms are involved in information processing, storage and retrieval, which are essential for memory consolidation, executive functioning and sensory processing. Therefore, brain arrhythmias could have catastrophic effects on circuit function, underlying the symptoms of various neurological diseases. Moreover, brain arrhythmias can serve as biomarkers for a variety of brain diseases. The aim of this review is to provide evidence linking Aβ and inflammation to neural network dysfunction, focusing on alterations in brain rhythms and their impact on cognition and sensory processing. I reviewed the most common brain arrhythmias characterized in AD, in AD transgenic models and those induced by Aβ. In addition, I reviewed the modulations of brain rhythms in neuroinflammatory diseases and those induced by immunogens, interleukins and microglia. This review reveals that Aβ and inflammation produce a complex set of effects on neural network function, which are related to the induction of brain arrhythmias and hyperexcitability, both closely related to behavioral alterations. Understanding these brain arrhythmias can help to develop therapeutic strategies to halt or prevent these neural network alterations and treat not only the arrhythmias but also the symptoms of AD and other inflammation-related pathologies.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiologia del Desarrollo y Neurofisiologia, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Queretaro, Qro., 76230, Mexico
| |
Collapse
|
22
|
Leparulo A, Mahmud M, Scremin E, Pozzan T, Vassanelli S, Fasolato C. Dampened Slow Oscillation Connectivity Anticipates Amyloid Deposition in the PS2APP Mouse Model of Alzheimer's Disease. Cells 2019; 9:cells9010054. [PMID: 31878336 PMCID: PMC7016892 DOI: 10.3390/cells9010054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/21/2022] Open
Abstract
To fight Alzheimer's disease (AD), we should know when, where, and how brain network dysfunctions initiate. In AD mouse models, relevant information can be derived from brain electrical activity. With a multi-site linear probe, we recorded local field potentials simultaneously at the posterior-parietal cortex and hippocampus of wild-type and double transgenic AD mice, under anesthesia. We focused on PS2APP (B6.152H) mice carrying both presenilin-2 (PS2) and amyloid precursor protein (APP) mutations, at three and six months of age, before and after plaque deposition respectively. To highlight defects linked to either the PS2 or APP mutation, we included in the analysis age-matched PS2.30H and APP-Swedish mice, carrying each of the mutations individually. Our study also included PSEN2-/- mice. At three months, only predeposition B6.152H mice show a reduction in the functional connectivity of slow oscillations (SO) and in the power ratio between SO and delta waves. At six months, plaque-seeding B6.152H mice undergo a worsening of the low/high frequency power imbalance and show a massive loss of cortico-hippocampal phase-amplitude coupling (PAC) between SO and higher frequencies, a feature shared with amyloid-free PS2.30H mice. We conclude that the PS2 mutation is sufficient to impair SO PAC and accelerate network dysfunctions in amyloid-accumulating mice.
Collapse
Affiliation(s)
- Alessandro Leparulo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
| | - Mufti Mahmud
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
| | - Elena Scremin
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
- Neuroscience Institute-Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2B, 35129 Padua, Italy
| | - Stefano Vassanelli
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
- Padua Neuroscience Center (PNC), University of Padua, Via G. Orus 2B, 35129 Padua, Italy
- Correspondence: (S.V.); (C.F.); Tel.: +39-049-8275337 (S.V.); +39-049-8276065 (C.F.); Fax: +39-049-8276049 (S.V.); +39-049-8276049 (C.F.)
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (A.L.); (M.M.); (E.S.); (T.P.)
- Correspondence: (S.V.); (C.F.); Tel.: +39-049-8275337 (S.V.); +39-049-8276065 (C.F.); Fax: +39-049-8276049 (S.V.); +39-049-8276049 (C.F.)
| |
Collapse
|
23
|
Beckman M, Knox K, Koneval Z, Smith C, Jayadev S, Barker-Haliski M. Loss of presenilin 2 age-dependently alters susceptibility to acute seizures and kindling acquisition. Neurobiol Dis 2019; 136:104719. [PMID: 31862541 DOI: 10.1016/j.nbd.2019.104719] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/17/2019] [Accepted: 12/16/2019] [Indexed: 01/15/2023] Open
Abstract
Patients with Alzheimer's disease (AD) experience seizures at higher rates than the general population of that age, suggesting an underexplored role of hyperexcitability in AD. Genetic variants in presenilin (PSEN) 1 and 2 genes lead to autosomal dominant early-onset AD (ADAD); patients with PSEN gene variants also report seizures. Pharmacological control of seizures in AD may be disease-modifying. Preclinical efficacy of FDA-approved antiseizure drugs (ASDs) is well defined in young adult rodents; however, the efficacy of ASDs in aged rodents with chronic seizures is less clear. The mechanism by which ADAD genes lead to AD remains unclear, and even less studied is the pathogenesis of epilepsy in AD. PSEN variants generally all result in a biochemical loss of function (De Strooper, 2007). We herein determined whether well-established models of acute and chronic seizure could be used to explore the relationship between AD genes and seizures through investigating whether loss of normal PSEN2 function age-dependently influenced susceptibility to seizures and/or corneal kindling acquisition. PSEN2 knockout (KO) and age-matched wild-type (WT) mice were screened from 2- to 10-months-old to establish age-dependent focal seizure threshold. Additionally, PSEN2 KO and WT mice aged 2- and 8-months-old underwent corneal kindling such that mice were aged 3- and 9-months old at the beginning of ASD efficacy testing. We then defined the dose-dependent efficacy of mechanistically distinct ASDs on kindled seizures of young versus aged mice to better understand the applicability of corneal kindling to real-world use for geriatric patients. PSEN2 KO mice demonstrated early-life reductions in seizure threshold. However, kindling acquisition was delayed in 2-month-old PSEN2 KO versus WT mice. Young male WT mice took 24.3 ± 1.3 (S.E.M.) stimulations to achieve kindling criterion, whereas age-matched PSEN2 KO male mice took 41.2 ± 1.1 stimulations (p < .0001). The rate of kindling acquisition of 8-month-old mice was no longer different from WT. This study demonstrates that loss of normal PSEN2 function is associated with age-dependent changes in the in vivo susceptibility to acute seizures and kindling. Loss of normal PSEN2 function may be an underexplored molecular contributor to seizures. The use of validated models of chronic seizures in aged rodents may uncover age-related changes in susceptibility to epileptogenesis and/or ASD efficacy in mice with AD-associated genotypes, which may benefit the management of seizures in AD.
Collapse
Affiliation(s)
- Megan Beckman
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America
| | - Kevin Knox
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America
| | - Zachery Koneval
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America
| | - Carole Smith
- Department of Neurology, School of Medicine, University of Washington, United States of America
| | - Suman Jayadev
- Department of Neurology, School of Medicine, University of Washington, United States of America
| | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, United States of America.
| |
Collapse
|
24
|
Senatorov VV, Friedman AR, Milikovsky DZ, Ofer J, Saar-Ashkenazy R, Charbash A, Jahan N, Chin G, Mihaly E, Lin JM, Ramsay HJ, Moghbel A, Preininger MK, Eddings CR, Harrison HV, Patel R, Shen Y, Ghanim H, Sheng H, Veksler R, Sudmant PH, Becker A, Hart B, Rogawski MA, Dillin A, Friedman A, Kaufer D. Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction. Sci Transl Med 2019; 11:eaaw8283. [PMID: 31801886 DOI: 10.1126/scitranslmed.aaw8283] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/15/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022]
Abstract
Aging involves a decline in neural function that contributes to cognitive impairment and disease. However, the mechanisms underlying the transition from a young-and-healthy to aged-and-dysfunctional brain are not well understood. Here, we report breakdown of the vascular blood-brain barrier (BBB) in aging humans and rodents, which begins as early as middle age and progresses to the end of the life span. Gain-of-function and loss-of-function manipulations show that this BBB dysfunction triggers hyperactivation of transforming growth factor-β (TGFβ) signaling in astrocytes, which is necessary and sufficient to cause neural dysfunction and age-related pathology in rodents. Specifically, infusion of the serum protein albumin into the young rodent brain (mimicking BBB leakiness) induced astrocytic TGFβ signaling and an aged brain phenotype including aberrant electrocorticographic activity, vulnerability to seizures, and cognitive impairment. Furthermore, conditional genetic knockdown of astrocytic TGFβ receptors or pharmacological inhibition of TGFβ signaling reversed these symptomatic outcomes in aged mice. Last, we found that this same signaling pathway is activated in aging human subjects with BBB dysfunction. Our study identifies dysfunction in the neurovascular unit as one of the earliest triggers of neurological aging and demonstrates that the aging brain may retain considerable latent capacity, which can be revitalized by therapeutic inhibition of TGFβ signaling.
Collapse
Affiliation(s)
- Vladimir V Senatorov
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aaron R Friedman
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Rotem Saar-Ashkenazy
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Adiel Charbash
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Naznin Jahan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gregory Chin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Eszter Mihaly
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jessica M Lin
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Harrison J Ramsay
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ariana Moghbel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marcela K Preininger
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chelsy R Eddings
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen V Harrison
- School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rishi Patel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yizhuo Shen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hana Ghanim
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Huanjie Sheng
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Albert Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Barry Hart
- Innovation Pathways, Palo Alto, CA 94301, USA
| | - Michael A Rogawski
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Andrew Dillin
- Glenn Center for Aging Research, Howard Hughes Medical Institute, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA.
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
25
|
Ishida K, Yamamoto M, Misawa K, Nishimura H, Misawa K, Ota N, Shimotoyodome A. Coffee polyphenols prevent cognitive dysfunction and suppress amyloid β plaques in APP/PS2 transgenic mouse. Neurosci Res 2019; 154:35-44. [PMID: 31121203 DOI: 10.1016/j.neures.2019.05.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/26/2019] [Accepted: 05/09/2019] [Indexed: 12/31/2022]
Abstract
Epidemiological studies have found that habitual coffee consumption may reduce the risk of Alzheimer's disease. Coffee contains numerous phenolic compounds (coffee polyphenols) such as chlorogenic acids. However, evidence demonstrating the contribution of chlorogenic acids to the prevention of cognitive dysfunction induced by Alzheimer's disease is limited. The present study investigated the effect of chlorogenic acids on the prevention of cognitive dysfunction in APP/PS2 transgenic mouse model of Alzheimer's disease. Five-week-old APP/PS2 mice were administered a diet supplemented with coffee polyphenols daily for 5 months. The memory and cognitive function of mice was determined using the novel object recognition test, Morris water maze test, and the step-through passive avoidance test. Immunohistochemical analysis revealed that chronic treatment with coffee polyphenols prevented cognitive dysfunction and significantly reduced the amount of amyloid β (Aβ) plaques in the hippocampus. Furthermore, we determined that 5-caffeoylquinic acid, one of the primary coffee polyphenols, did not inhibit Aβ fibrillation; however, degraded Aβ fibrils. In conclusion, our results demonstrate that coffee polyphenols prevent cognitive deficits and reduce Aβ plaque deposition via disaggregation of Aβ in the APP/PS2 mouse.
Collapse
Affiliation(s)
- Keiko Ishida
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Masaki Yamamoto
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Kensuke Misawa
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Hitomi Nishimura
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Koichi Misawa
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Noriyasu Ota
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| | - Akira Shimotoyodome
- Health Care Food Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida, Tokyo, 131-8501, Japan.
| |
Collapse
|
26
|
Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in Aβ-Bearing Mice. J Neurosci 2017; 37:6132-6148. [PMID: 28559377 DOI: 10.1523/jneurosci.0877-17.2017] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 12/25/2022] Open
Abstract
Hyperexcitable neuronal networks are mechanistically linked to the pathologic and clinical features of Alzheimer's disease (AD). Astrocytes are a primary defense against hyperexcitability, but their functional phenotype during AD is poorly understood. Here, we found that activated astrocytes in the 5xFAD mouse model were strongly associated with proteolysis of the protein phosphatase calcineurin (CN) and the elevated expression of the CN-dependent transcription factor nuclear factor of activated T cells 4 (NFAT4). Intrahippocampal injections of adeno-associated virus vectors containing the astrocyte-specific promoter Gfa2 and the NFAT inhibitory peptide VIVIT reduced signs of glutamate-mediated hyperexcitability in 5xFAD mice, measured in vivo with microelectrode arrays and ex vivo brain slices, using whole-cell voltage clamp. VIVIT treatment in 5xFAD mice led to increased expression of the astrocytic glutamate transporter GLT-1 and to attenuated changes in dendrite morphology, synaptic strength, and NMDAR-dependent responses. The results reveal astrocytic CN/NFAT4 as a key pathologic mechanism for driving glutamate dysregulation and neuronal hyperactivity during AD.SIGNIFICANCE STATEMENT Neuronal hyperexcitability and excitotoxicity are increasingly recognized as important mechanisms for neurodegeneration and dementia associated with Alzheimer's disease (AD). Astrocytes are profoundly activated during AD and may lose their capacity to regulate excitotoxic glutamate levels. Here, we show that a highly active calcineurin (CN) phosphatase fragment and its substrate transcription factor, nuclear factor of activated T cells (NFAT4), appear in astrocytes in direct proportion to the extent of astrocyte activation. The blockade of astrocytic CN/NFAT signaling in a common mouse model of AD, using adeno-associated virus vectors normalized glutamate signaling dynamics, increased astrocytic glutamate transporter levels and alleviated multiple signs of neuronal hyperexcitability. The results suggest that astrocyte activation drives hyperexcitability during AD through a mechanism involving aberrant CN/NFAT signaling and impaired glutamate transport.
Collapse
|