1
|
Zeng J, Zhang R, Xu H, Zhang C, Lu L. Integrative single-cell RNA sequencing and mendelian randomization analysis reveal the potential role of synaptic vesicle cycling-related genes in Alzheimer's disease. J Prev Alzheimers Dis 2025; 12:100097. [PMID: 40021385 DOI: 10.1016/j.tjpad.2025.100097] [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: 12/25/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) involves alterations in synaptic vesicle cycling (SVC), which significantly affect neuronal communication and function. Therefore, a thorough investigation into the potential roles of SVC-related genes (SVCRGs) in AD can enhance the identification of critical biomarkers that may influence disease progression and treatment responses. METHODS The datasets used in this study were sourced exclusively from public databases. By integrating differential expression analysis with Mendelian randomization (MR), we identified SVCRGs as biomarkers for AD. Functional characterization of these biomarkers was performed, followed by integration into a nomogram. Further investigation of immune infiltration in AD patients and healthy individuals was carried out. Ultimately, the potential cellular mechanisms of AD were explored through single-cell RNA sequencing (scRNA-seq) analysis. RESULTS ATP6V1D, ATP6V1G2, CLTB, and NSF were identified as biomarkers, exhibiting a positive correlation with each other and a downregulated expression in AD. These markers were pinpointed as protective factors for AD [odds ratio (OR) < 1, P < 0.05], with potential to reduce the risk of the disease. Integrated into a nomogram, they demonstrated satisfactory diagnostic performance and clinical utility, surpassing the use of single gene. They were collectively enriched in pathways related to "interferon gamma response", "inflammatory response", and "TNFα signaling via NFκB". Additionally, an increase in infiltration of 17 immune cell types in AD was noted, particularly cells associated with neuroinflammation such as activated CD8 T cells and various dendritic cells (DCs), suggesting an inflammatory milieu in AD while also displaying a negative correlation with the biomarkers. The cell types were further annotated, revealing specific expressions of biomarkers and uncovering the heterogeneity of excitatory neurons. A significant reduction in the overall number of excitatory neurons under AD conditions was observed, alongside consistent expression of biomarkers during the developmental stages of excitatory neurons. CONCLUSION By using MR, we firstly identified four SVCRGs as protective factors for AD, functioning through pathways associated with mitochondrial dysfunction, chronic inflammation, immune dysregulation, and neuronal damage. These genes had the potential to modulate immune cell infiltration activated in AD patients and exhibited cell-type-specific expression profiles within AD-related cellular contexts. Their findings provide novel insights and valuable references for future research on AD pathogenesis and therapeutic strategies.
Collapse
Affiliation(s)
- Junfeng Zeng
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Ruihua Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Huihua Xu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Li Lu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Laboratory of Cellular Physiology of Chinese Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
2
|
Naganishi S, Hagihara H, Miyakawa T. Gene Expression Signatures of Immaturity, Decreased pH, and Neural Hyperexcitation in the Hippocampus of Alzheimer's Disease Model Mice. Neuropsychopharmacol Rep 2025; 45:e70001. [PMID: 39907034 PMCID: PMC11795175 DOI: 10.1002/npr2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
AIMS Alzheimer's disease (AD) is a leading cause of dementia, with increasing prevalence. Mutations in genes like MAPT, PSEN1, and PSEN2 are risk factors, leading to the development of several AD model mice. Recent hypotheses suggest AD brain pathology involves abnormal neurodevelopment, decreased pH, and neural hyperexcitation. However, it remains unclear to what extent these pathologies are reflected in the gene expression changes of AD models. This study aims to compare gene expression patterns in the brains of multiple AD model mice with those related to these three factors, evaluating the extent of overlap. METHODS We conducted a comprehensive search of public databases, collecting 20 gene expression datasets from the hippocampus of AD model mice. These datasets were compared with gene sets related to hippocampal maturation, brain pH, and neural hyperexcitation to statistically assess overlap. Pathway enrichment analysis explored the biological relevance of these gene expression changes. RESULTS The extent of overlap with maturity-, pH-, and hyperexcitation-associated genes varied across AD models, showing significant correlations between lower maturity, lower pH, and increased neural hyperexcitation. In MAPT mutant and APP+PSEN1 homozygous transgenic mice, these signatures became more pronounced with age. Pathway meta-analysis revealed that genes associated with maturity, pH, and hyperexcitation in AD models are involved in synaptic and channel functions, as well as inflammatory responses, consistent with previous studies. CONCLUSION These findings suggest that pathophysiological changes related to maturity, pH, and neural hyperexcitation play varying roles across individual AD model mice. Our recent study found a negative correlation between disease progression and actual pH levels in human AD patients. Considering the results presented in this study, maturity and neural hyperexcitation, which are correlated with pH, may also be linked to disease progression. Thus, gene expression changes in these factors could be useful markers for assessing the pathology in AD models.
Collapse
Affiliation(s)
- Sayaka Naganishi
- Department of Systems Medical ScienceFujita Health University Graduate School of MedicineToyoakeAichiJapan
- Division of Systems Medical Science, Center for Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Hideo Hagihara
- Division of Systems Medical Science, Center for Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical ScienceFujita Health UniversityToyoakeAichiJapan
| |
Collapse
|
3
|
Devulder A, Vanderlinden G, Van Langenhoven L, Testelmans D, Van Den Bossche M, De Winter FL, Vandenbulcke M, Vandenberghe R, Theys T, Van Laere K, Van Paesschen W. Epileptic activity on foramen ovale electrodes is associated with sleep and tau pathology in Alzheimer's disease. Brain 2025; 148:506-520. [PMID: 38990981 PMCID: PMC11788210 DOI: 10.1093/brain/awae231] [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: 02/14/2024] [Revised: 05/30/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Both sleep alterations and epileptiform activity are associated with the accumulation of amyloid-β and tau pathology and are currently investigated for potential therapeutic interventions in Alzheimer's disease. However, a bidirectional intertwining relationship between sleep and neuronal hyperexcitability might modulate the effects of Alzheimer's disease pathology on the corresponding associations. To investigate this, we performed multiple day simultaneous foramen ovale (FO) plus scalp EEG and polysomnography recordings and acquired 18F-MK6240 tau PET-MR in three patients in the prodromal stage of Alzheimer's disease and in two patients with mild and moderate dementia due to Alzheimer's disease, respectively. As an eligibility criterion for the present study, subjects either had a history of a recent seizure (n = 2) or subclinical epileptiform activity (SEA) on a previous scalp EEG taken in a research context (n = 3). The 18F-MK6240 standard uptake value ratio (SUVR) and asymmetry index (AI) were calculated in a priori-defined volumes of interest. Linear mixed-effects models were used to study associations between interictal epileptiform discharges (IEDs), polysomnography parameters and 18F-MK6240 SUVR. Epileptiform activity was bilateral but asymmetrically present on FO electrodes in all patients and ≥95% of IEDs were not visible on scalp EEG. In one patient, two focal seizures were detected on FO electrodes, both without visual scalp EEG correlate. We observed lateralized periodic discharges, brief potentially ictal rhythmic discharges and lateralized rhythmic delta activity on FO electrodes in four patients. Unlike scalp EEG, intracranial electrodes showed a lateralization of epileptiform activity. Although the amount of IEDs on intracranial electrodes was not associated to the 18F-MK6240 SUVR binding in different volumes of interest, there was a congruent asymmetry of the 18F-MK6240 binding towards the most epileptic hemisphere for the mesial (P = 0.007) and lateral temporal cortex (P = 0.006). IEDs on intracranial electrodes were most abundant during slow wave sleep (SWS) (92/h) and non-REM sleep 2 (N2, 81/h), followed by non-REM sleep 1 (N1, 33/h) and least frequent during wakefulness (17/h) and REM sleep (9/h). The extent of IEDs during sleep was not reflected in the relative time in each sleep stage spent [REM% (P = 0.415), N1% (P = 0.668), N2% (P = 0.442), SWS% (P = 0.988)], and not associated with the arousal index (P = 0.317), apnoea-hypopnoea index (P = 0.846) or oxygen desaturation index (P = 0.746). Together, our observations suggest a multi-directional interaction between sleep, epileptiform activity and tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Astrid Devulder
- Laboratory for Epilepsy Research, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven 3000, Belgium
| | - Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
| | - Leen Van Langenhoven
- Leuven Biostatistics and Statistical Bioinformatics Centre (L-BioStat), KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
| | - Dries Testelmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Pulmonary Diseases, University Hospitals Leuven, Leuven 3000, Belgium
| | - Maarten Van Den Bossche
- Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Geriatric Psychiatry, KUL University Psychiatric Center (UPC) KU Leuven, Leuven 3000, Belgium
| | - François-Laurent De Winter
- Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Geriatric Psychiatry, KUL University Psychiatric Center (UPC) KU Leuven, Leuven 3000, Belgium
| | - Mathieu Vandenbulcke
- Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Geriatric Psychiatry, KUL University Psychiatric Center (UPC) KU Leuven, Leuven 3000, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, Leuven 3000, Belgium
- Laboratory for Cognitive Neurology, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
| | - Tom Theys
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Neurosurgery, University Hospitals Leuven, Leuven 3000, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Division of Nuclear Medicine, University Hospitals Leuven, Leuven 3000, Belgium
| | - Wim Van Paesschen
- Laboratory for Epilepsy Research, KU Leuven Biomedical Sciences Group, Leuven 3000, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven 3000, Belgium
| |
Collapse
|
4
|
Roemer-Cassiano SN, Wagner F, Evangelista L, Rauchmann BS, Dehsarvi A, Steward A, Dewenter A, Biel D, Zhu Z, Pescoller J, Gross M, Perneczky R, Malpetti M, Ewers M, Schöll M, Dichgans M, Höglinger GU, Brendel M, Jäkel S, Franzmeier N. Amyloid-associated hyperconnectivity drives tau spread across connected brain regions in Alzheimer's disease. Sci Transl Med 2025; 17:eadp2564. [PMID: 39841807 DOI: 10.1126/scitranslmed.adp2564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/08/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
In Alzheimer's disease (AD), amyloid-β (Aβ) triggers the aggregation and spreading of tau pathology, which drives neurodegeneration and cognitive decline. However, the pathophysiological link between Aβ and tau remains unclear, which hinders therapeutic efforts to attenuate Aβ-related tau accumulation. Aβ has been found to trigger neuronal hyperactivity and hyperconnectivity, and preclinical research has shown that tau spreads across connected neurons in an activity-dependent manner. Here, we hypothesized that neuronal hyperactivity and hypersynchronicity, resulting in functional connectivity increases, constitute a crucial mechanism by which Aβ facilitates the spreading of tau pathology. By combining Aβ positron emission tomography (PET), resting-state functional magnetic resonance imaging, and longitudinal tau-PET in 69 cognitively normal amyloid-negative controls and 140 amyloid-positive patients covering the AD spectrum, we confirmed that Aβ induces hyperconnectivity of temporal lobe tau epicenters to posterior brain regions that are vulnerable to tau accumulation in AD. This was replicated in an independent sample of 55 controls and 345 individuals with preclinical AD and low cortical tau-PET uptake, suggesting that the emergence of Aβ-related hyperconnectivity precedes neocortical tau spreading . Last, using longitudinal tau-PET and mediation analysis, we confirmed that these Aβ-related connectivity increases in tau epicenters to typical tau-vulnerable brain regions in AD mediated the effect of Aβ on faster tau accumulation, unveiling increased connectivity as a potential causal link between the two AD hallmark pathologies. Together, these findings suggest that Aβ promotes tau spreading by eliciting neuronal hyperconnectivity and that targeting Aβ-related neuronal hyperconnectivity may attenuate tau spreading in AD.
Collapse
Affiliation(s)
- Sebastian N Roemer-Cassiano
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Max Planck School of Cognition, 04103 Leipzig, Germany
| | - Fabian Wagner
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Lisa Evangelista
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Neuroradiology, University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Zeyu Zhu
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Julia Pescoller
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Mattes Gross
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Aging Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, W6 8RP London, UK
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, S10 2HQ Sheffield, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, CB2 0PY Cambridge, UK
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Mölndal and Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Sarah Jäkel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Mölndal and Gothenburg, Sweden
| |
Collapse
|
5
|
Liu P, Xu J, Chen Y, Xu Q, Zhang W, Hu B, Li A, Zhu Q. Electrophysiological Signatures in Global Cerebral Ischemia: Neuroprotection Via Chemogenetic Inhibition of CA1 Pyramidal Neurons in Rats. J Am Heart Assoc 2024; 13:e036146. [PMID: 39673154 PMCID: PMC11935537 DOI: 10.1161/jaha.124.036146] [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: 09/09/2024] [Accepted: 11/19/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Although there has been limited research into the perturbation of electrophysiological activity in the brain after ischemia, the activity signatures during ischemia and reperfusion remain to be fully elucidated. We aim to comprehensively describe these electrophysiological signatures and interrogate their correlation with ischemic damage during global cerebral ischemia and reperfusion. METHODS AND RESULTS We used the 4-vessel occlusion method of inducing global cerebral ischemia in rats. We used in vivo electrophysiological techniques to simultaneously record single units, scalp electroencephalogram, and local field potentials in awake animals. Neuronal damage and astrocyte reactivation were examined by immunofluorescence, immunoblotting, and quantitative real-time reverse-transcription polymerase chain reaction under chemogenetic inhibition of glutamatergic neurons. Electroencephalogram/local field potentials power and phase-amplitude coupling of the theta and low-gamma bands were reduced during ischemia and the acute phase of reperfusion. The firing rate of single units was enhanced by ischemia-reperfusion, and the phase relationship between the local field potentials theta band and neuronal firing was altered. Precise inhibition of hippocampus CA1 pyramidal neuron hyperactivity by chemogenetics rescued the firing dysfunction, ischemic neuronal damage, and A1 astrocyte activation. CONCLUSIONS Our results provide a comprehensive description of the characteristics of electrophysiological activity that accompany ischemia-reperfusion and highlight the significance of this activity in ischemic damage.
Collapse
Affiliation(s)
- Penglai Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Jiang Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Yilan Chen
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Qi Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Wei Zhang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Bin Hu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| | - Qiuju Zhu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouChina
| |
Collapse
|
6
|
Rodriguez GA, Rothenberg EF, Shetler CO, Aoun A, Posani L, Vajram SV, Tedesco T, Fusi S, Hussaini SA. Impaired spatial coding and neuronal hyperactivity in the medial entorhinal cortex of aged App NL-G-F mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.624990. [PMID: 39651258 PMCID: PMC11623597 DOI: 10.1101/2024.11.26.624990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The progressive accumulation of amyloid beta (Aβ) pathology in the brain has been associated with aberrant neuronal network activity and poor cognitive performance in preclinical mouse models of Alzheimer's disease (AD). Presently, our understanding of the mechanisms driving pathology-associated neuronal dysfunction and impaired information processing in the brain remains incomplete. Here, we assessed the impact of advanced Aβ pathology on spatial information processing in the medial entorhinal cortex (MEC) of 18-month App NL-G-F/NL- G-F knock-in (APP KI) mice as they explored contextually novel and familiar open field arenas in a two-day, four-session recording paradigm. We tracked single unit firing activity across all sessions and found that spatial information scores were decreased in MEC neurons from APP KI mice versus those in age-matched C57BL/6J controls. MEC single unit spatial representations were also impacted in APP KI mice. Border cell firing preferences were unstable across sessions and spatial periodicity in putative grid cells was disrupted. In contrast, MEC border cells and grid cells in Control mice were intact and stable across sessions. We then quantified the stability of MEC spatial maps across sessions by utilizing a metric based on the Earth Mover's Distance (EMD). We found evidence for increased instability in spatially-tuned APP KI MEC neurons versus Controls when mice were re-exposed to familiar environments and exposed to a novel environment. Additionally, spatial decoding analysis of MEC single units revealed deficits in position and speed coding in APP KI mice in all session comparisons. Finally, MEC single unit analysis revealed a mild hyperactive phenotype in APP KI mice that appeared to be driven by narrow-spiking units (putative interneurons). These findings tie Aβ-associated dysregulation in neuronal firing to disruptions in spatial information processing that may underlie certain cognitive deficits associated with AD.
Collapse
|
7
|
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
|
8
|
Luppi AI, Sanz Perl Y, Vohryzek J, Mediano PAM, Rosas FE, Milisav F, Suarez LE, Gini S, Gutierrez-Barragan D, Gozzi A, Misic B, Deco G, Kringelbach ML. Competitive interactions shape brain dynamics and computation across species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619194. [PMID: 39484469 PMCID: PMC11526968 DOI: 10.1101/2024.10.19.619194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Adaptive cognition relies on cooperation across anatomically distributed brain circuits. However, specialised neural systems are also in constant competition for limited processing resources. How does the brain's network architecture enable it to balance these cooperative and competitive tendencies? Here we use computational whole-brain modelling to examine the dynamical and computational relevance of cooperative and competitive interactions in the mammalian connectome. Across human, macaque, and mouse we show that the architecture of the models that most faithfully reproduce brain activity, consistently combines modular cooperative interactions with diffuse, long-range competitive interactions. The model with competitive interactions consistently outperforms the cooperative-only model, with excellent fit to both spatial and dynamical properties of the living brain, which were not explicitly optimised but rather emerge spontaneously. Competitive interactions in the effective connectivity produce greater levels of synergistic information and local-global hierarchy, and lead to superior computational capacity when used for neuromorphic computing. Altogether, this work provides a mechanistic link between network architecture, dynamical properties, and computation in the mammalian brain.
Collapse
Affiliation(s)
- Andrea I. Luppi
- University of Oxford, Oxford, UK
- St John’s College, Cambridge, UK
- Montreal Neurological Institute, Montreal, Canada
| | | | | | | | | | | | | | - Silvia Gini
- Italian Institute of Technology, Rovereto, Italy
- Centre for Mind/Brain Sciences, University of Trento, Italy
| | | | | | | | | | | |
Collapse
|
9
|
Raghuraman R, Aoun A, Herman M, Shetler CO, Nahmani E, Hussaini SA. Lateral Entorhinal Cortex Dysfunction in Alzheimer's Disease Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589589. [PMID: 38659892 PMCID: PMC11042344 DOI: 10.1101/2024.04.15.589589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
In Alzheimer's disease (AD), the formation of amyloid beta and neurofibrillary tangles (NFTs) leads to neuronal loss in entorhinal cortex (EC), a crucial brain region involved in memory and navigation. These pathological changes are concurrent with the onset of memory-related issues in AD patients with symptoms of forgetfulness such as misplacing items, disorientation in familiar environments etc. The lateral EC (LEC) is associated with non-spatial memory processing including object recognition. Since in LEC, neurons fire in response to objects (object cells) and at locations previously occupied by objects (trace cells), pathology in this region could lead to dysfunction in object location coding. In this paper we show that a transgenic mouse model, EC-App/Tau, which expresses both APP and tau primarily in the EC region, have deficits in LEC-specific memory tasks. Using in vivo single-unit electrophysiology recordings we show that the LEC neurons are hyperactive with low information content and high sparsity compared to the controls indicating poor firing fidelity. We finally show that object cells and trace cells fire less precisely in the EC-App/Tau mice compared to controls indicating poor encoding of objects. Overall, we show that AD pathology causes erratic firing of LEC neurons and object coding defects leading to LEC-specific memory impairment.
Collapse
|
10
|
Lanni I, Chiacchierini G, Papagno C, Santangelo V, Campolongo P. Treating Alzheimer's disease with brain stimulation: From preclinical models to non-invasive stimulation in humans. Neurosci Biobehav Rev 2024; 165:105831. [PMID: 39074672 DOI: 10.1016/j.neubiorev.2024.105831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) is a severe and progressive neurodegenerative condition that exerts detrimental effects on brain function. As of now, there is no effective treatment for AD patients. This review explores two distinct avenues of research. The first revolves around the use of animal studies and preclinical models to gain insights into AD's underlying mechanisms and potential treatment strategies. Specifically, it delves into the effectiveness of interventions such as Optogenetics and Chemogenetics, shedding light on their implications for understanding pathophysiological mechanisms and potential therapeutic applications. The second avenue focuses on non-invasive brain stimulation (NiBS) techniques in the context of AD. Evidence suggests that NiBS can successfully modulate cognitive functions associated with various neurological and neuropsychiatric disorders, including AD, as demonstrated by promising findings. Here, we critically assessed recent findings in AD research belonging to these lines of research and discuss their potential impact on the clinical horizon of AD treatment. These multifaceted approaches offer hope for advancing our comprehension of AD pathology and developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Ilenia Lanni
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Chiacchierini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Costanza Papagno
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Valerio Santangelo
- Functional Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
11
|
Goettemoeller AM, Banks E, Kumar P, Olah VJ, McCann KE, South K, Ramelow CC, Eaton A, Duong DM, Seyfried NT, Weinshenker D, Rangaraju S, Rowan MJM. Entorhinal cortex vulnerability to human APP expression promotes hyperexcitability and tau pathology. Nat Commun 2024; 15:7918. [PMID: 39256379 PMCID: PMC11387477 DOI: 10.1038/s41467-024-52297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
Preventative treatment for Alzheimer's Disease (AD) is dire, yet mechanisms underlying early regional vulnerability remain unknown. In AD, one of the earliest pathophysiological correlates to cognitive decline is hyperexcitability, which is observed first in the entorhinal cortex. Why hyperexcitability preferentially emerges in specific regions in AD is unclear. Using regional, cell-type-specific proteomics and electrophysiology in wild-type mice, we uncovered a unique susceptibility of the entorhinal cortex to human amyloid precursor protein (hAPP). Entorhinal hyperexcitability resulted from selective vulnerability of parvalbumin (PV) interneurons, with respect to surrounding excitatory neurons. This effect was partially replicated with an APP chimera containing a humanized amyloid-beta sequence. EC hyperexcitability could be ameliorated by co-expression of human Tau with hAPP at the expense of increased pathological tau species, or by enhancing PV interneuron excitability in vivo. This study suggests early interventions targeting inhibitory neurons may protect vulnerable regions from the effects of APP/amyloid and tau pathology.
Collapse
Affiliation(s)
- Annie M Goettemoeller
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
- GDBBS Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Emmie Banks
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
- GDBBS Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Prateek Kumar
- Department of Neurology, Yale University, New Haven, CT, USA
| | - Viktor J Olah
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Katharine E McCann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly South
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
- GDBBS Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Christina C Ramelow
- GDBBS Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna Eaton
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Duc M Duong
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Matthew J M Rowan
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
12
|
Neřoldová M, Stuchlík A. Chemogenetic Tools and their Use in Studies of Neuropsychiatric Disorders. Physiol Res 2024; 73:S449-S470. [PMID: 38957949 PMCID: PMC11412350 DOI: 10.33549/physiolres.935401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Chemogenetics is a newly developed set of tools that allow for selective manipulation of cell activity. They consist of a receptor mutated irresponsive to endogenous ligands and a synthetic ligand that does not interact with the wild-type receptors. Many different types of these receptors and their respective ligands for inhibiting or excitating neuronal subpopulations were designed in the past few decades. It has been mainly the G-protein coupled receptors (GPCRs) selectively responding to clozapine-N-oxide (CNO), namely Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), that have been employed in research. Chemogenetics offers great possibilities since the activity of the receptors is reversible, inducible on demand by the ligand, and non-invasive. Also, specific groups or types of neurons can be selectively manipulated thanks to the delivery by viral vectors. The effect of the chemogenetic receptors on neurons lasts longer, and even chronic activation can be achieved. That can be useful for behavioral testing. The great advantage of chemogenetic tools is especially apparent in research on brain diseases since they can manipulate whole neuronal circuits and connections between different brain areas. Many psychiatric or other brain diseases revolve around the dysfunction of specific brain networks. Therefore, chemogenetics presents a powerful tool for investigating the underlying mechanisms causing the disease and revealing the link between the circuit dysfunction and the behavioral or cognitive symptoms observed in patients. It could also contribute to the development of more effective treatments.
Collapse
Affiliation(s)
- M Neřoldová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic. E-mail:
| | | |
Collapse
|
13
|
Goettemoeller AM, Banks E, Kumar P, Olah VJ, McCann KE, South K, Ramelow CC, Eaton A, Duong DM, Seyfried NT, Weinshenker D, Rangaraju S, Rowan MJ. Entorhinal cortex vulnerability to human APP expression promotes hyperexcitability and tau pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565629. [PMID: 39005389 PMCID: PMC11244896 DOI: 10.1101/2023.11.06.565629] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Preventative treatment for Alzheimer's Disease is of dire importance, and yet, cellular mechanisms underlying early regional vulnerability in Alzheimer's Disease remain unknown. In human patients with Alzheimer's Disease, one of the earliest observed pathophysiological correlates to cognitive decline is hyperexcitability. In mouse models, early hyperexcitability has been shown in the entorhinal cortex, the first cortical region impacted by Alzheimer's Disease. The origin of hyperexcitability in early-stage disease and why it preferentially emerges in specific regions is unclear. Using cortical-region and cell-type-specific proteomics coupled with ex vivo and in vivo electrophysiology, we uncovered differential susceptibility to human-specific amyloid precursor protein (hAPP) in a model of sporadic Alzheimer's. Unexpectedly, our findings reveal that early entorhinal hyperexcitability may result from intrinsic vulnerability of parvalbumin (PV) interneurons, rather than the suspected layer II excitatory neurons. This vulnerability of entorhinal PV interneurons is specific to hAPP, as it could not be recapitulated with increased murine APP expression. However, partial replication of the findings could be seen after introduction of a murine APP chimera containing a humanized amyloid-beta sequence. Surprisingly, neurons in the Somatosensory Cortex showed no such vulnerability to adult-onset hAPP expression. hAPP-induced hyperexcitability in entorhinal cortex could be ameliorated by enhancing PV interneuron excitability in vivo. Co-expression of human Tau with hAPP decreased circuit hyperexcitability, but at the expense of increased pathological tau species. This study suggests early disease interventions targeting non-excitatory cell types may protect regions with early vulnerability to pathological symptoms of Alzheimer's Disease and downstream cognitive decline.
Collapse
|
14
|
Ottoy J, Kang MS, Tan JXM, Boone L, Vos de Wael R, Park BY, Bezgin G, Lussier FZ, Pascoal TA, Rahmouni N, Stevenson J, Fernandez Arias J, Therriault J, Hong SJ, Stefanovic B, McLaurin J, Soucy JP, Gauthier S, Bernhardt BC, Black SE, Rosa-Neto P, Goubran M. Tau follows principal axes of functional and structural brain organization in Alzheimer's disease. Nat Commun 2024; 15:5031. [PMID: 38866759 PMCID: PMC11169286 DOI: 10.1038/s41467-024-49300-2] [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: 09/22/2023] [Accepted: 05/24/2024] [Indexed: 06/14/2024] Open
Abstract
Alzheimer's disease (AD) is a brain network disorder where pathological proteins accumulate through networks and drive cognitive decline. Yet, the role of network connectivity in facilitating this accumulation remains unclear. Using in-vivo multimodal imaging, we show that the distribution of tau and reactive microglia in humans follows spatial patterns of connectivity variation, the so-called gradients of brain organization. Notably, less distinct connectivity patterns ("gradient contraction") are associated with cognitive decline in regions with greater tau, suggesting an interaction between reduced network differentiation and tau on cognition. Furthermore, by modeling tau in subject-specific gradient space, we demonstrate that tau accumulation in the frontoparietal and temporo-occipital cortices is associated with greater baseline tau within their functionally and structurally connected hubs, respectively. Our work unveils a role for both functional and structural brain organization in pathology accumulation in AD, and supports subject-specific gradient space as a promising tool to map disease progression.
Collapse
Affiliation(s)
- Julie Ottoy
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Min Su Kang
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | | | - Lyndon Boone
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Reinder Vos de Wael
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Bo-Yong Park
- Department of Data Science, Inha University, Incheon, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Gleb Bezgin
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Neuroinformatics for Personalized Medicine lab, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nesrine Rahmouni
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Jaime Fernandez Arias
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Seok-Jun Hong
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bojana Stefanovic
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Serge Gauthier
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Boris C Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Sandra E Black
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Medicine (Division of Neurology), University of Toronto, Toronto, ON, Canada
| | - Pedro Rosa-Neto
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Maged Goubran
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Physical Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Karimani F, Asgari Taei A, Abolghasemi-Dehaghani MR, Safari MS, Dargahi L. Impairment of entorhinal cortex network activity in Alzheimer's disease. Front Aging Neurosci 2024; 16:1402573. [PMID: 38882526 PMCID: PMC11176617 DOI: 10.3389/fnagi.2024.1402573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
The entorhinal cortex (EC) stands out as a critical brain region affected in the early phases of Alzheimer's disease (AD), with some of the disease's pathological processes originating from this area, making it one of the most crucial brain regions in AD. Recent research highlights disruptions in the brain's network activity, characterized by heightened excitability and irregular oscillations, may contribute to cognitive impairment. These disruptions are proposed not only as potential therapeutic targets but also as early biomarkers for AD. In this paper, we will begin with a review of the anatomy and function of EC, highlighting its selective vulnerability in AD. Subsequently, we will discuss the disruption of EC network activity, exploring changes in excitability and neuronal oscillations in this region during AD and hypothesize that, considering the advancements in neuromodulation techniques, addressing the disturbances in the network activity of the EC could offer fresh insights for both the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Farnaz Karimani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mir-Shahram Safari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Sanchez-Rodriguez LM, Bezgin G, Carbonell F, Therriault J, Fernandez-Arias J, Servaes S, Rahmouni N, Tissot C, Stevenson J, Karikari TK, Ashton NJ, Benedet AL, Zetterberg H, Blennow K, Triana-Baltzer G, Kolb HC, Rosa-Neto P, Iturria-Medina Y. Personalized whole-brain neural mass models reveal combined Aβ and tau hyperexcitable influences in Alzheimer's disease. Commun Biol 2024; 7:528. [PMID: 38704445 PMCID: PMC11069569 DOI: 10.1038/s42003-024-06217-2] [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: 08/24/2023] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
Neuronal dysfunction and cognitive deterioration in Alzheimer's disease (AD) are likely caused by multiple pathophysiological factors. However, mechanistic evidence in humans remains scarce, requiring improved non-invasive techniques and integrative models. We introduce personalized AD computational models built on whole-brain Wilson-Cowan oscillators and incorporating resting-state functional MRI, amyloid-β (Aβ) and tau-PET from 132 individuals in the AD spectrum to evaluate the direct impact of toxic protein deposition on neuronal activity. This subject-specific approach uncovers key patho-mechanistic interactions, including synergistic Aβ and tau effects on cognitive impairment and neuronal excitability increases with disease progression. The data-derived neuronal excitability values strongly predict clinically relevant AD plasma biomarker concentrations (p-tau217, p-tau231, p-tau181, GFAP) and grey matter atrophy obtained through voxel-based morphometry. Furthermore, reconstructed EEG proxy quantities show the hallmark AD electrophysiological alterations (theta band activity enhancement and alpha reductions) which occur with Aβ-positivity and after limbic tau involvement. Microglial activation influences on neuronal activity are less definitive, potentially due to neuroimaging limitations in mapping neuroprotective vs detrimental activation phenotypes. Mechanistic brain activity models can further clarify intricate neurodegenerative processes and accelerate preventive/treatment interventions.
Collapse
Affiliation(s)
- Lazaro M Sanchez-Rodriguez
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, QC, Canada
| | - Gleb Bezgin
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | | | - Joseph Therriault
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Jaime Fernandez-Arias
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Stijn Servaes
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Cécile Tissot
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
- Lawrence Berkeley National Laboratory, Berkeley, USA
| | - Jenna Stevenson
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McGill University Research Centre for Studies in Aging, Douglas Research Centre, Montreal, QC, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada.
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, QC, Canada.
| |
Collapse
|
17
|
Kampmann M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat Rev Neurosci 2024; 25:351-371. [PMID: 38575768 DOI: 10.1038/s41583-024-00806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The selective vulnerability of specific neuronal subtypes is a hallmark of neurodegenerative diseases. In this Review, I summarize our current understanding of the brain regions and cell types that are selectively vulnerable in different neurodegenerative diseases and describe the proposed underlying cell-autonomous and non-cell-autonomous mechanisms. I highlight how recent methodological innovations - including single-cell transcriptomics, CRISPR-based screens and human cell-based models of disease - are enabling new breakthroughs in our understanding of selective vulnerability. An understanding of the molecular mechanisms that determine selective vulnerability and resilience would shed light on the key processes that drive neurodegeneration and point to potential therapeutic strategies to protect vulnerable cell populations.
Collapse
Affiliation(s)
- Martin Kampmann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Prabhu P, Morise H, Kudo K, Beagle A, Mizuiri D, Syed F, Kotegar KA, Findlay A, Miller BL, Kramer JH, Rankin KP, Garcia PA, Kirsch HE, Vossel K, Nagarajan SS, Ranasinghe KG. Abnormal gamma phase-amplitude coupling in the parahippocampal cortex is associated with network hyperexcitability in Alzheimer's disease. Brain Commun 2024; 6:fcae121. [PMID: 38665964 PMCID: PMC11043655 DOI: 10.1093/braincomms/fcae121] [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: 11/22/2023] [Revised: 03/08/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
While animal models of Alzheimer's disease (AD) have shown altered gamma oscillations (∼40 Hz) in local neural circuits, the low signal-to-noise ratio of gamma in the resting human brain precludes its quantification via conventional spectral estimates. Phase-amplitude coupling (PAC) indicating the dynamic integration between the gamma amplitude and the phase of low-frequency (4-12 Hz) oscillations is a useful alternative to capture local gamma activity. In addition, PAC is also an index of neuronal excitability as the phase of low-frequency oscillations that modulate gamma amplitude, effectively regulates the excitability of local neuronal firing. In this study, we sought to examine the local neuronal activity and excitability using gamma PAC, within brain regions vulnerable to early AD pathophysiology-entorhinal cortex and parahippocampus, in a clinical population of patients with AD and age-matched controls. Our clinical cohorts consisted of a well-characterized cohort of AD patients (n = 50; age, 60 ± 8 years) with positive AD biomarkers, and age-matched, cognitively unimpaired controls (n = 35; age, 63 ± 5.8 years). We identified the presence or the absence of epileptiform activity in AD patients (AD patients with epileptiform activity, AD-EPI+, n = 20; AD patients without epileptiform activity, AD-EPI-, n = 30) using long-term electroencephalography (LTM-EEG) and 1-hour long magnetoencephalography (MEG) with simultaneous EEG. Using the source reconstructed MEG data, we computed gamma PAC as the coupling between amplitude of the gamma frequency (30-40 Hz) with phase of the theta (4-8 Hz) and alpha (8-12 Hz) frequency oscillations, within entorhinal and parahippocampal cortices. We found that patients with AD have reduced gamma PAC in the left parahippocampal cortex, compared to age-matched controls. Furthermore, AD-EPI+ patients showed greater reductions in gamma PAC than AD-EPI- in bilateral parahippocampal cortices. In contrast, entorhinal cortices did not show gamma PAC abnormalities in patients with AD. Our findings demonstrate the spatial patterns of altered gamma oscillations indicating possible region-specific manifestations of network hyperexcitability within medial temporal lobe regions vulnerable to AD pathophysiology. Greater deficits in AD-EPI+ suggests that reduced gamma PAC is a sensitive index of network hyperexcitability in AD patients. Collectively, the current results emphasize the importance of investigating the role of neural circuit hyperexcitability in early AD pathophysiology and explore its potential as a modifiable contributor to AD pathobiology.
Collapse
Affiliation(s)
- Pooja Prabhu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Data science and Computer Applications, Manipal Institute of Technology, Manipal 576104, India
| | - Hirofumi Morise
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
- Medical Imaging Business Center, Ricoh Company Ltd., Kanazawa 920-0177, Japan
| | - Kiwamu Kudo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
- Medical Imaging Business Center, Ricoh Company Ltd., Kanazawa 920-0177, Japan
| | - Alexander Beagle
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Danielle Mizuiri
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Faatimah Syed
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Karunakar A Kotegar
- Department of Data science and Computer Applications, Manipal Institute of Technology, Manipal 576104, India
| | - Anne Findlay
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Katherine P Rankin
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Paul A Garcia
- Epilepsy Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Heidi E Kirsch
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
- Epilepsy Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Keith Vossel
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Srikantan S Nagarajan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kamalini G Ranasinghe
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
19
|
Zambon A, Rico LC, Herman M, Gundacker A, Telalovic A, Hartenberger LM, Kuehn R, Romanov RA, Hussaini SA, Harkany T, Pollak DD. Gestational immune activation disrupts hypothalamic neurocircuits of maternal care behavior. Mol Psychiatry 2024; 29:859-873. [PMID: 35581295 PMCID: PMC9112243 DOI: 10.1038/s41380-022-01602-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/27/2022]
Abstract
Immune activation is one of the most common complications during pregnancy, predominantly evoked by viral infections. Nevertheless, how immune activation affects mother-offspring relationships postpartum remains unknown. Here, by using the polyinosinic-polycytidylic acid (Poly I:C) model of gestational infection we show that viral-like immune activation at mid-gestation persistently changes hypothalamic neurocircuit parameters in mouse dams and, consequently, is adverse to parenting behavior. Poly I:C-exposed dams favor non-pup-directed exploratory behavior at the expense of pup retrieval. These behavioral deficits are underlain by dendrite pruning and lesser immediate early gene activation in Galanin (Gal)+ neurons with dam-specific transcriptional signatures that reside in the medial preoptic area (mPOA). Reduced activation of an exclusively inhibitory contingent of these distal-projecting Gal+ neurons allows for increased feed-forward inhibition onto putative dopaminergic neurons in the ventral tegmental area (VTA) in Poly I:C-exposed dams. Notably, destabilized VTA output specifically accompanies post-pup retrieval epochs. We suggest that gestational immunogenic insults bias both threat processing and reward perception, manifesting as disfavored infant caregiving.
Collapse
Affiliation(s)
- Alice Zambon
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Mathieu Herman
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amina Telalovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lisa-Marie Hartenberger
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rebekka Kuehn
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - S Abid Hussaini
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
20
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong DD, Seyfried NT, Wood LB, Rowan MJM, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies unique molecular signatures and vulnerabilities to early Alzheimer's pathology. Nat Commun 2024; 15:2823. [PMID: 38561349 PMCID: PMC10985119 DOI: 10.1038/s41467-024-47028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Dysfunction in fast-spiking parvalbumin interneurons (PV-INs) may represent an early pathophysiological perturbation in Alzheimer's Disease (AD). Defining early proteomic alterations in PV-INs can provide key biological and translationally-relevant insights. We used cell-type-specific in-vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state PV-IN proteomes. PV-IN proteomic signatures include high metabolic and translational activity, with over-representation of AD-risk and cognitive resilience-related proteins. In bulk proteomes, PV-IN proteins were associated with cognitive decline in humans, and with progressive neuropathology in humans and the 5xFAD mouse model of Aβ pathology. PV-IN CIBOP in early stages of Aβ pathology revealed signatures of increased mitochondria and metabolism, synaptic and cytoskeletal disruption and decreased mTOR signaling, not apparent in whole-brain proteomes. Furthermore, we demonstrated pre-synaptic defects in PV-to-excitatory neurotransmission, validating our proteomic findings. Overall, in this study we present native-state proteomes of PV-INs, revealing molecular insights into their unique roles in cognitive resiliency and AD pathogenesis.
Collapse
Affiliation(s)
- Prateek Kumar
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Annie M Goettemoeller
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Claudia Espinosa-Garcia
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Brendan R Tobin
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Ali Tfaily
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Ruth S Nelson
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Aditya Natu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric B Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Juliet V Santiago
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Sneha Malepati
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lihong Cheng
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Duc D Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Levi B Wood
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
- School of Chemical and Biological Engineering, GeoInsrgia titute of Technology, Atlanta, GA, 30322, USA
| | - Matthew J M Rowan
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
21
|
Slutsky I. Linking activity dyshomeostasis and sleep disturbances in Alzheimer disease. Nat Rev Neurosci 2024; 25:272-284. [PMID: 38374463 DOI: 10.1038/s41583-024-00797-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 02/21/2024]
Abstract
The presymptomatic phase of Alzheimer disease (AD) starts with the deposition of amyloid-β in the cortex and begins a decade or more before the emergence of cognitive decline. The trajectory towards dementia and neurodegeneration is shaped by the pathological load and the resilience of neural circuits to the effects of this pathology. In this Perspective, I focus on recent advances that have uncovered the vulnerability of neural circuits at early stages of AD to hyperexcitability, particularly when the brain is in a low-arousal states (such as sleep and anaesthesia). Notably, this hyperexcitability manifests before overt symptoms such as sleep and memory deficits. Using the principles of control theory, I analyse the bidirectional relationship between homeostasis of neuronal activity and sleep and propose that impaired activity homeostasis during sleep leads to hyperexcitability and subsequent sleep disturbances, whereas sleep disturbances mitigate hyperexcitability via negative feedback. Understanding the interplay among activity homeostasis, neuronal excitability and sleep is crucial for elucidating the mechanisms of vulnerability to and resilience against AD pathology and for identifying new therapeutic avenues.
Collapse
Affiliation(s)
- Inna Slutsky
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
22
|
Cabrera-Álvarez J, Stefanovski L, Martin L, Susi G, Maestú F, Ritter P. A Multiscale Closed-Loop Neurotoxicity Model of Alzheimer's Disease Progression Explains Functional Connectivity Alterations. eNeuro 2024; 11:ENEURO.0345-23.2023. [PMID: 38565295 PMCID: PMC11026343 DOI: 10.1523/eneuro.0345-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/05/2023] [Accepted: 12/22/2023] [Indexed: 04/04/2024] Open
Abstract
The accumulation of amyloid-β (Aβ) and hyperphosphorylated-tau (hp-tau) are two classical histopathological biomarkers in Alzheimer's disease (AD). However, their detailed interactions with the electrophysiological changes at the meso- and macroscale are not yet fully understood. We developed a mechanistic multiscale model of AD progression, linking proteinopathy to its effects on neural activity and vice-versa. We integrated a heterodimer model of prion-like protein propagation and a brain network model of Jansen-Rit neural masses derived from human neuroimaging data whose parameters varied due to neurotoxicity. Results showed that changes in inhibition guided the electrophysiological alterations found in AD, and these changes were mainly attributed to Aβ effects. Additionally, we found a causal disconnection between cellular hyperactivity and interregional hypersynchrony contrary to previous beliefs. Finally, we demonstrated that early Aβ and hp-tau depositions' location determine the spatiotemporal profile of the proteinopathy. The presented model combines the molecular effects of both Aβ and hp-tau together with a mechanistic protein propagation model and network effects within a closed-loop model. This holds the potential to enlighten the interplay between AD mechanisms on various scales, aiming to develop and test novel hypotheses on the contribution of different AD-related variables to the disease evolution.
Collapse
Affiliation(s)
- Jesús Cabrera-Álvarez
- Department of Experimental Psychology, Complutense University of Madrid, Pozuelo de Alarcón 28223, Spain
- Centre for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid 28040, Spain
| | - Leon Stefanovski
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Neurology with Experimental Neurology, Brain Simulation Section, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Leon Martin
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Neurology with Experimental Neurology, Brain Simulation Section, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Gianluca Susi
- Centre for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid 28040, Spain
- Department of Structure of Matter, Thermal Physics and Electronics, Complutense University of Madrid, Madrid 28040, Spain
| | - Fernando Maestú
- Department of Experimental Psychology, Complutense University of Madrid, Pozuelo de Alarcón 28223, Spain
- Centre for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid 28040, Spain
| | - Petra Ritter
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Neurology with Experimental Neurology, Brain Simulation Section, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin 10115, Germany
| |
Collapse
|
23
|
Rademacher K, Nakamura K. Role of dopamine neuron activity in Parkinson's disease pathophysiology. Exp Neurol 2024; 373:114645. [PMID: 38092187 DOI: 10.1016/j.expneurol.2023.114645] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023]
Abstract
Neural activity is finely tuned to produce normal behaviors, and disruptions in activity likely occur early in the course of many neurodegenerative diseases. However, how neural activity is altered, and how these changes influence neurodegeneration is poorly understood. Here, we focus on evidence that the activity of dopamine neurons is altered in Parkinson's disease (PD), either as a compensatory response to degeneration or as a result of circuit dynamics or pathologic proteins, based on available human data and studies in animal models of PD. We then discuss how this abnormal activity may augment other neurotoxic phenomena in PD, including mitochondrial deficits, protein aggregation and spread, dopamine toxicity, and excitotoxicity. A more complete picture of how activity is altered and the resulting effects on dopaminergic neuron health and function may inform future therapeutic interventions to target and protect dopamine neurons from degeneration.
Collapse
Affiliation(s)
- Katerina Rademacher
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, California, 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.; Graduate Program in Neuroscience, University of California San Francisco, San Francisco, California, 94158, USA
| | - Ken Nakamura
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, California, 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.; Graduate Program in Neuroscience, University of California San Francisco, San Francisco, California, 94158, USA; Graduate Program in Biomedical Sciences, University of California San Francisco, San Francisco, California, 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, California, 94158, USA.
| |
Collapse
|
24
|
Tian C, Reyes I, Masurkar AV. Impact of dendritic spine loss on excitability of hippocampal CA1 pyramidal neurons: a computational study of early Alzheimer disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576500. [PMID: 38328155 PMCID: PMC10849489 DOI: 10.1101/2024.01.20.576500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Synaptic spine loss is an early pathophysiologic hallmark of Alzheimer disease (AD) that precedes overt loss of dendritic architecture and frank neurodegeneration. While spine loss signifies a decreased engagement of postsynaptic neurons by presynaptic targets, the degree to which loss of spines and their passive components impacts the excitability of postsynaptic neurons and responses to surviving synaptic inputs is unclear. Using passive multicompartmental models of CA1 pyramidal neurons (PNs), implicated in early AD, we find that spine loss alone drives a boosting of remaining inputs to their proximal and distal dendrites, targeted by CA3 and entorhinal cortex (EC), respectively. This boosting effect is higher in distal versus proximal dendrites and can be mediated by spine loss restricted to the distal compartment, enough to impact synaptic input integration and somatodendritic backpropagation. This has particular relevance to very early stages of AD in which pathophysiology extends from EC to CA1.
Collapse
|
25
|
Aoun A, Shetler O, Raghuraman R, Rodriguez GA, Hussaini SA. Beyond Correlation: Optimal Transport Metrics For Characterizing Representational Stability and Remapping in Neurons Encoding Spatial Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548592. [PMID: 37503011 PMCID: PMC10369988 DOI: 10.1101/2023.07.11.548592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Spatial representations in the entorhinal cortex (EC) and hippocampus (HPC) are fundamental to cognitive functions like navigation and memory. These representations, embodied in spatial field maps, dynamically remap in response to environmental changes. However, current methods, such as Pearson's correlation coefficient, struggle to capture the complexity of these remapping events, especially when fields do not overlap, or transformations are non-linear. This limitation hinders our understanding and quantification of remapping, a key aspect of spatial memory function. To address this, we propose a family of metrics based on the Earth Mover's Distance (EMD) as a versatile framework for characterizing remapping. Applied to both normalized and unnormalized distributions, the EMD provides a granular, noise-resistant, and rate-robust description of remapping. This approach enables the identification of specific cell types and the characterization of remapping in various scenarios, including disease models. Furthermore, the EMD's properties can be manipulated to identify spatially tuned cell types and to explore remapping as it relates to alternate information forms such as spatiotemporal coding. By employing approximations of the EMD, we present a feasible, lightweight approach that complements traditional methods. Our findings underscore the potential of the EMD as a powerful tool for enhancing our understanding of remapping in the brain and its implications for spatial navigation, memory studies and beyond.
Collapse
Affiliation(s)
- Andrew Aoun
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
- Co-first author
| | - Oliver Shetler
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
- Co-first author
| | - Radha Raghuraman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Gustavo A. Rodriguez
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - S. Abid Hussaini
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
26
|
Goettemoeller AM, Banks E, McCann KE, Kumar P, South K, Olah VJ, Ramelow CC, Duong DM, Seyfried NT, Rangaraju S, Weinshenker D, Rowan MJM. Entorhinal cortex vulnerability to human APP expression promotes hyperexcitability and tau pathology. RESEARCH SQUARE 2023:rs.3.rs-3370607. [PMID: 37987015 PMCID: PMC10659529 DOI: 10.21203/rs.3.rs-3370607/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Preventative treatment for Alzheimer's Disease is of dire importance, and yet, cellular mechanisms underlying early regional vulnerability in Alzheimer's Disease remain unknown. In human patients with Alzheimer's Disease, one of the earliest observed pathophysiological correlates to cognitive decline is hyperexcitability1. In mouse models, early hyperexcitability has been shown in the entorhinal cortex, the first cortical region impacted by Alzheimer's Disease2-4. The origin of hyperexcitability in early-stage disease and why it preferentially emerges in specific regions is unclear. Using cortical-region and cell-type- specific proteomics and patch-clamp electrophysiology, we uncovered differential susceptibility to human-specific amyloid precursor protein (hAPP) in a model of sporadic Alzheimer's. Unexpectedly, our findings reveal that early entorhinal hyperexcitability may result from intrinsic vulnerability of parvalbumin interneurons, rather than the suspected layer II excitatory neurons. This vulnerability of entorhinal PV interneurons is specific to hAPP, as it could not be recapitulated with increased murine APP expression. Furthermore, the Somatosensory Cortex showed no such vulnerability to adult-onset hAPP expression, likely resulting from PV-interneuron variability between the two regions based on physiological and proteomic evaluations. Interestingly, entorhinal hAPP-induced hyperexcitability was quelled by co-expression of human Tau at the expense of increased pathological tau species. This study suggests early disease interventions targeting non-excitatory cell types may protect regions with early vulnerability to pathological symptoms of Alzheimer's Disease and downstream cognitive decline.
Collapse
Affiliation(s)
- Annie M Goettemoeller
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322
- GDBBS Graduate Program, Laney Graduate School, Emory University
| | - Emmie Banks
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322
- GDBBS Graduate Program, Laney Graduate School, Emory University
| | | | - Prateek Kumar
- Department of Neurology, Emory University School of Medicine
| | - Kelly South
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322
- GDBBS Graduate Program, Laney Graduate School, Emory University
| | - Viktor J Olah
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322
| | - Christina C Ramelow
- Department of Neurology, Emory University School of Medicine
- GDBBS Graduate Program, Laney Graduate School, Emory University
| | - Duc M Duong
- Department of Neurology, Emory University School of Medicine
- Department of Biochemistry, Emory University
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine
- Department of Biochemistry, Emory University
- Center for Neurodegenerative Disease, Emory University School of Medicine
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine
- GDBBS Graduate Program, Laney Graduate School, Emory University
| | | | - Matthew JM Rowan
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322
- Center for Neurodegenerative Disease, Emory University School of Medicine
| |
Collapse
|
27
|
Grieco SF, Holmes TC, Xu X. Probing neural circuit mechanisms in Alzheimer's disease using novel technologies. Mol Psychiatry 2023; 28:4407-4420. [PMID: 36959497 PMCID: PMC10827671 DOI: 10.1038/s41380-023-02018-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
The study of Alzheimer's Disease (AD) has traditionally focused on neuropathological mechanisms that has guided therapies that attenuate neuropathological features. A new direction is emerging in AD research that focuses on the progressive loss of cognitive function due to disrupted neural circuit mechanisms. Evidence from humans and animal models of AD show that dysregulated circuits initiate a cascade of pathological events that culminate in functional loss of learning, memory, and other aspects of cognition. Recent progress in single-cell, spatial, and circuit omics informs this circuit-focused approach by determining the identities, locations, and circuitry of the specific cells affected by AD. Recently developed neuroscience tools allow for precise access to cell type-specific circuitry so that their functional roles in AD-related cognitive deficits and disease progression can be tested. An integrated systems-level understanding of AD-associated neural circuit mechanisms requires new multimodal and multi-scale interrogations that longitudinally measure and/or manipulate the ensemble properties of specific molecularly-defined neuron populations first susceptible to AD. These newly developed technological and conceptual advances present new opportunities for studying and treating circuits vulnerable in AD and represent the beginning of a new era for circuit-based AD research.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
28
|
Vogel JW, Corriveau-Lecavalier N, Franzmeier N, Pereira JB, Brown JA, Maass A, Botha H, Seeley WW, Bassett DS, Jones DT, Ewers M. Connectome-based modelling of neurodegenerative diseases: towards precision medicine and mechanistic insight. Nat Rev Neurosci 2023; 24:620-639. [PMID: 37620599 DOI: 10.1038/s41583-023-00731-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
Neurodegenerative diseases are the most common cause of dementia. Although their underlying molecular pathologies have been identified, there is substantial heterogeneity in the patterns of progressive brain alterations across and within these diseases. Recent advances in neuroimaging methods have revealed that pathological proteins accumulate along specific macroscale brain networks, implicating the network architecture of the brain in the system-level pathophysiology of neurodegenerative diseases. However, the extent to which 'network-based neurodegeneration' applies across the wide range of neurodegenerative disorders remains unclear. Here, we discuss the state-of-the-art of neuroimaging-based connectomics for the mapping and prediction of neurodegenerative processes. We review findings supporting brain networks as passive conduits through which pathological proteins spread. As an alternative view, we also discuss complementary work suggesting that network alterations actively modulate the spreading of pathological proteins between connected brain regions. We conclude this Perspective by proposing an integrative framework in which connectome-based models can be advanced along three dimensions of innovation: incorporating parameters that modulate propagation behaviour on the basis of measurable biological features; building patient-tailored models that use individual-level information and allowing model parameters to interact dynamically over time. We discuss promises and pitfalls of these strategies for improving disease insights and moving towards precision medicine.
Collapse
Affiliation(s)
- Jacob W Vogel
- Department of Clinical Sciences, SciLifeLab, Lund University, Lund, Sweden.
| | - Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Acadamy, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Neuro Division, Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden
| | - Jesse A Brown
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Dani S Bassett
- Departments of Bioengineering, Electrical and Systems Engineering, Physics and Astronomy, Neurology and Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
29
|
Fortel I, Zhan L, Ajilore O, Wu Y, Mackin S, Leow A. Disrupted excitation-inhibition balance in cognitively normal individuals at risk of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554061. [PMID: 37662359 PMCID: PMC10473582 DOI: 10.1101/2023.08.21.554061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background Sex differences impact Alzheimer's disease (AD) neuropathology, but cell-to-network level dysfunctions in the prodromal phase are unclear. Alterations in hippocampal excitation-inhibition balance (EIB) have recently been linked to early AD pathology. Objective Examine how AD risk factors (age, APOE-ɛ4, amyloid-β) relate to hippocampal EIB in cognitively normal males and females using connectome-level measures. Methods Individuals from the OASIS-3 cohort (age 42-95) were studied (N = 437), with a subset aged 65+ undergoing neuropsychological testing (N = 231). Results In absence of AD risk factors (APOE-ɛ4/Aβ+), whole-brain EIB decreases with age more significantly in males than females (p = 0.021, β = -0.007). Regression modeling including APOE-ɛ4 allele carriers (Aβ-) yielded a significant positive AGE-by-APOE interaction in the right hippocampus for females only (p = 0.013, β = 0.014), persisting with inclusion of Aβ+ individuals (p = 0.012, β = 0.014). Partial correlation analyses of neuropsychological testing showed significant associations with EIB in females: positive correlations between right hippocampal EIB with categorical fluency and whole-brain EIB with the trail-making test (p < 0.05). Conclusion Sex differences in EIB emerge during normal aging and progresses differently with AD risk. Results suggest APOE-ɛ4 disrupts hippocampal balance more than amyloid in females. Increased excitation correlates positively with neuropsychological performance in the female group, suggesting a duality in terms of potential beneficial effects prior to cognitive impairment. This underscores the translational relevance of APOE-ɛ4 related hyperexcitation in females, potentially informing therapeutic targets or early interventions to mitigate AD progression in this vulnerable population.
Collapse
Affiliation(s)
- Igor Fortel
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL
| | - Liang Zhan
- Department of Electrical and Computer Engineering, University of Pittsburgh, Pittsburgh, PA
| | - Olusola Ajilore
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL
| | - Yichao Wu
- Department of Math, Statistics and Computer Science, University of Illinois at Chicago, Chicago, IL
| | - Scott Mackin
- Department of Psychiatry, University of California - San Francisco, San Francisco, CA
| | - Alex Leow
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
30
|
Yu SP, Jiang MQ, Shim SS, Pourkhodadad S, Wei L. Extrasynaptic NMDA receptors in acute and chronic excitotoxicity: implications for preventive treatments of ischemic stroke and late-onset Alzheimer's disease. Mol Neurodegener 2023; 18:43. [PMID: 37400870 DOI: 10.1186/s13024-023-00636-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/01/2023] [Indexed: 07/05/2023] Open
Abstract
Stroke and late-onset Alzheimer's disease (AD) are risk factors for each other; the comorbidity of these brain disorders in aging individuals represents a significant challenge in basic research and clinical practice. The similarities and differences between stroke and AD in terms of pathogenesis and pathophysiology, however, have rarely been comparably reviewed. Here, we discuss the research background and recent progresses that are important and informative for the comorbidity of stroke and late-onset AD and related dementia (ADRD). Glutamatergic NMDA receptor (NMDAR) activity and NMDAR-mediated Ca2+ influx are essential for neuronal function and cell survival. An ischemic insult, however, can cause rapid increases in glutamate concentration and excessive activation of NMDARs, leading to swift Ca2+ overload in neuronal cells and acute excitotoxicity within hours and days. On the other hand, mild upregulation of NMDAR activity, commonly seen in AD animal models and patients, is not immediately cytotoxic. Sustained NMDAR hyperactivity and Ca2+ dysregulation lasting from months to years, nevertheless, can be pathogenic for slowly evolving events, i.e. degenerative excitotoxicity, in the development of AD/ADRD. Specifically, Ca2+ influx mediated by extrasynaptic NMDARs (eNMDARs) and a downstream pathway mediated by transient receptor potential cation channel subfamily M member (TRPM) are primarily responsible for excitotoxicity. On the other hand, the NMDAR subunit GluN3A plays a "gatekeeper" role in NMDAR activity and a neuroprotective role against both acute and chronic excitotoxicity. Thus, ischemic stroke and AD share an NMDAR- and Ca2+-mediated pathogenic mechanism that provides a common receptor target for preventive and possibly disease-modifying therapies. Memantine (MEM) preferentially blocks eNMDARs and was approved by the Federal Drug Administration (FDA) for symptomatic treatment of moderate-to-severe AD with variable efficacy. According to the pathogenic role of eNMDARs, it is conceivable that MEM and other eNMDAR antagonists should be administered much earlier, preferably during the presymptomatic phases of AD/ADRD. This anti-AD treatment could simultaneously serve as a preconditioning strategy against stroke that attacks ≥ 50% of AD patients. Future research on the regulation of NMDARs, enduring control of eNMDARs, Ca2+ homeostasis, and downstream events will provide a promising opportunity to understand and treat the comorbidity of AD/ADRD and stroke.
Collapse
Affiliation(s)
- Shan P Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA.
| | - Michael Q Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Seong S Shim
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Soheila Pourkhodadad
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, 30033, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
31
|
Tombini M, Boscarino M, Di Lazzaro V. Tackling seizures in patients with Alzheimer's disease. Expert Rev Neurother 2023; 23:1131-1145. [PMID: 37946507 DOI: 10.1080/14737175.2023.2278487] [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: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION In past years, a possible bidirectional link between epilepsy and Alzheimer's disease (AD) has been proposed: if AD patients are more likely to develop epilepsy, people with late-onset epilepsy evidence an increased risk of dementia. Furthermore, current research suggested that subclinical epileptiform discharges may be more frequent in patients with AD and network hyperexcitability may hasten cognitive impairment. AREAS COVERED In this narrative review, the authors discuss the recent evidence linking AD and epilepsy as well as seizures semeiology and epileptiform activity observed in patients with AD. Finally, anti-seizure medications (ASMs) and therapeutic trials to tackle seizures and network hyperexcitability in this clinical scenario have been summarized. EXPERT OPINION There is growing experimental evidence demonstrating a strong connection between seizures, neuronal hyperexcitability, and AD. Epilepsy in AD has shown a good response to ASMs both at the late and prodromal stages. The new generation ASMs with fewer cognitive adverse effects seem to be a preferable option. Data on the possible effects of network hyperexcitability and ASMs on AD progression are still inconclusive. Further clinical trials are mandatory to identify clear guidelines about treatment of subclinical epileptiform discharges in patients with AD without seizures.
Collapse
Affiliation(s)
- Mario Tombini
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Marilisa Boscarino
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation Department, Milan, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
32
|
Brown J, Camporesi E, Lantero-Rodriguez J, Olsson M, Wang A, Medem B, Zetterberg H, Blennow K, Karikari TK, Wall M, Hill E. Tau in cerebrospinal fluid induces neuronal hyperexcitability and alters hippocampal theta oscillations. Acta Neuropathol Commun 2023; 11:67. [PMID: 37095572 PMCID: PMC10127378 DOI: 10.1186/s40478-023-01562-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Alzheimer's disease (AD) and other tauopathies are characterized by the aggregation of tau into soluble and insoluble forms (including tangles and neuropil threads). In humans, a fraction of both phosphorylated and non-phosphorylated N-terminal to mid-domain tau species, are secreted into cerebrospinal fluid (CSF). Some of these CSF tau species can be measured as diagnostic and prognostic biomarkers, starting from early stages of disease. While in animal models of AD pathology, soluble tau aggregates have been shown to disrupt neuronal function, it is unclear whether the tau species present in CSF will modulate neural activity. Here, we have developed and applied a novel approach to examine the electrophysiological effects of CSF from patients with a tau-positive biomarker profile. The method involves incubation of acutely-isolated wild-type mouse hippocampal brain slices with small volumes of diluted human CSF, followed by a suite of electrophysiological recording methods to evaluate their effects on neuronal function, from single cells through to the network level. Comparison of the toxicity profiles of the same CSF samples, with and without immuno-depletion for tau, has enabled a pioneering demonstration that CSF-tau potently modulates neuronal function. We demonstrate that CSF-tau mediates an increase in neuronal excitability in single cells. We then observed, at the network level, increased input-output responses and enhanced paired-pulse facilitation as well as an increase in long-term potentiation. Finally, we show that CSF-tau modifies the generation and maintenance of hippocampal theta oscillations, which have important roles in learning and memory and are known to be altered in AD patients. Together, we describe a novel method for screening human CSF-tau to understand functional effects on neuron and network activity, which could have far-reaching benefits in understanding tau pathology, thus allowing for the development of better targeted treatments for tauopathies in the future.
Collapse
Affiliation(s)
- Jessica Brown
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Maria Olsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Alice Wang
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Blanca Medem
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, WI, 53792, USA
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mark Wall
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Emily Hill
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
33
|
Morrone CD, Raghuraman R, Hussaini SA, Yu WH. Proteostasis failure exacerbates neuronal circuit dysfunction and sleep impairments in Alzheimer's disease. Mol Neurodegener 2023; 18:27. [PMID: 37085942 PMCID: PMC10119020 DOI: 10.1186/s13024-023-00617-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
Failed proteostasis is a well-documented feature of Alzheimer's disease, particularly, reduced protein degradation and clearance. However, the contribution of failed proteostasis to neuronal circuit dysfunction is an emerging concept in neurodegenerative research and will prove critical in understanding cognitive decline. Our objective is to convey Alzheimer's disease progression with the growing evidence for a bidirectional relationship of sleep disruption and proteostasis failure. Proteostasis dysfunction and tauopathy in Alzheimer's disease disrupts neurons that regulate the sleep-wake cycle, which presents behavior as impaired slow wave and rapid eye movement sleep patterns. Subsequent sleep loss further impairs protein clearance. Sleep loss is a defined feature seen early in many neurodegenerative disorders and contributes to memory impairments in Alzheimer's disease. Canonical pathological hallmarks, β-amyloid, and tau, directly disrupt sleep, and neurodegeneration of locus coeruleus, hippocampal and hypothalamic neurons from tau proteinopathy causes disruption of the neuronal circuitry of sleep. Acting in a positive-feedback-loop, sleep loss and circadian rhythm disruption then increase spread of β-amyloid and tau, through impairments of proteasome, autophagy, unfolded protein response and glymphatic clearance. This phenomenon extends beyond β-amyloid and tau, with interactions of sleep impairment with the homeostasis of TDP-43, α-synuclein, FUS, and huntingtin proteins, implicating sleep loss as an important consideration in an array of neurodegenerative diseases and in cases of mixed neuropathology. Critically, the dynamics of this interaction in the neurodegenerative environment are not fully elucidated and are deserving of further discussion and research. Finally, we propose sleep-enhancing therapeutics as potential interventions for promoting healthy proteostasis, including β-amyloid and tau clearance, mechanistically linking these processes. With further clinical and preclinical research, we propose this dynamic interaction as a diagnostic and therapeutic framework, informing precise single- and combinatorial-treatments for Alzheimer's disease and other brain disorders.
Collapse
Affiliation(s)
- Christopher Daniel Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
| | - Radha Raghuraman
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
| | - S Abid Hussaini
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
| | - Wai Haung Yu
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Geriatric Mental Health Research Services, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
34
|
Igarashi KM. Entorhinal cortex dysfunction in Alzheimer's disease. Trends Neurosci 2023; 46:124-136. [PMID: 36513524 PMCID: PMC9877178 DOI: 10.1016/j.tins.2022.11.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/31/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022]
Abstract
The entorhinal cortex (EC) is the brain region that often exhibits the earliest histological alterations in Alzheimer's disease (AD), including the formation of neurofibrillary tangles and cell death. Recently, brain imaging studies from preclinical AD patients and electrophysiological recordings from AD animal models have shown that impaired neuronal activity in the EC precedes neurodegeneration. This implies that memory impairments and spatial navigation deficits at the initial stage of AD are likely caused by activity dysfunction rather than by cell death. This review focuses on recent findings on EC dysfunction in AD, and discusses the potential pathways for mitigating AD progression by protecting the EC.
Collapse
Affiliation(s)
- Kei M Igarashi
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
35
|
Zamorano M, Alexander JF, Catania D, Dharmaraj S, Kavelaars A, Heijnen CJ. Nasal administration of mesenchymal stem cells prevents accelerated age-related tauopathy after chemotherapy in mice. Immun Ageing 2023; 20:5. [PMID: 36698170 PMCID: PMC9874182 DOI: 10.1186/s12979-023-00328-w] [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: 09/10/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND There is increasing concern that cancer and cancer treatment accelerate aging and the associated cognitive decline. We showed recently that treatment of 9-month-old male mice with cisplatin causes cognitive deficits that are associated with formation of tau deposits in the hippocampus. Here we explored the capacity of mesenchymal stem cells (MSC) given via the nose to prevent age-related brain tau deposits. Moreover, we more closely examined the cellular distribution of this hallmark of accelerated brain aging in response to treatment of 9-month-old female and male mice with cisplatin. RESULTS We show that cisplatin induces tau deposits in the entorhinal cortex and hippocampus in both sexes. The tau deposits colocalize with syndecan-2. Astrocytes surrounding tau deposits have increased glial fibrillary acidic protein glial fibrillary acidic protein (GFAP) expression. Most of the cisplatin-induced tau deposits were located in microtubule associated protein-2 (MAP-2)+ neurons that were surrounded by aquaporin 4+ (AQP4)+ neuron-facing membrane domains of astrocytes. In addition, some tau deposits were detected in the perinuclear region of GFAP+ astrocytes and in CD31+ endothelial cells. There were no morphological signs of activation of ionized calcium binding adaptor molecule-1+ (Iba-1)+ microglia and no increases in brain cytokine production. Nasal administration of MSC at 48 and 96 hours after cisplatin prevented formation of tau deposits and normalized syndecan-2 and GFAP expression. Behaviorally, cisplatin-induced tau cluster formation was associated with reduced executive functioning and working/spatial memory and nasal administration of MSC at 48 and 96 hours after cisplatin prevented these cognitive deficits. Notably, delayed MSC administration (1 month after cisplatin) also prevented tau cluster formation and cognitive deficits, in both sexes. CONCLUSION In summary, nasal administration of MSC to older mice at 2 days or 1 month after completion of cisplatin treatment prevents the accelerated development of tau deposits in entorhinal cortex and hippocampus and the associated cognitive deficits. Since MSC are already in clinical use for many other clinical indications, developing nasal MSC administration for treatment of accelerated brain aging and cognitive deficits in cancer survivors should be feasible and would greatly improve their quality of life.
Collapse
Affiliation(s)
- Miriam Zamorano
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA ,grid.267308.80000 0000 9206 2401Department of Pediatric Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Jenolyn F. Alexander
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA ,grid.410718.b0000 0001 0262 7331Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr, 55 Essen, Germany
| | - Desiree Catania
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Shruti Dharmaraj
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Annemieke Kavelaars
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Cobi J. Heijnen
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
36
|
Li S, Huang Y, Yu L, Ji X, Wu J. Impact of the Cannabinoid System in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:715-726. [PMID: 35105293 PMCID: PMC10207907 DOI: 10.2174/1570159x20666220201091006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
Cannabinoids are compounds isolated from cannabis and are also widely present in both nervous and immune systems of animals. In recent years, with in-depth research on cannabinoids, their clinical medicinal value has been evaluated, and many exciting achievements have been continuously accumulating, especially in the field of neurodegenerative disease. Alzheimer's disease is the most common type of neurodegenerative disease that causes dementia and has become a global health problem that seriously impacts human health today. In this review, we discuss the therapeutic potential of cannabinoids for the treatment of Alzheimer's disease. How cannabinoids act on different endocannabinoid receptor subtypes to regulate Alzheimer's disease and the roles of the endocannabinoid system in Alzheimer's disease are outlined, and the underlying mechanisms are discussed. Finally, we summarize the most relevant opportunities of cannabinoid pharmacology related to Alzheimer's disease and discuss the potential usefulness of cannabinoids in the clinical treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Shuangtao Li
- Shantou University Medical College, Brain Function and Disease Laboratory, Shantou, #22 Road Xinling, Guangdong 515041, China
| | - Yuanbing Huang
- Department of Neurology, Yunfu People’s Hospital, Yunfu, Guangdong 527300, China
| | - Lijun Yu
- Shantou University Medical College, Brain Function and Disease Laboratory, Shantou, #22 Road Xinling, Guangdong 515041, China
| | - Xiaoyu Ji
- Department of Neurology, Yunfu People’s Hospital, Yunfu, Guangdong 527300, China
| | - Jie Wu
- Shantou University Medical College, Brain Function and Disease Laboratory, Shantou, #22 Road Xinling, Guangdong 515041, China
| |
Collapse
|
37
|
Fortel I, Zhan L, Ajilore O, Wu Y, Mackin S, Leow A. Disrupted Excitation-Inhibition Balance in Cognitively Normal Individuals at Risk of Alzheimer's Disease. J Alzheimers Dis 2023; 95:1449-1467. [PMID: 37718795 PMCID: PMC11260287 DOI: 10.3233/jad-230035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
BACKGROUND Sex differences impact Alzheimer's disease (AD) neuropathology, but cell-to-network level dysfunctions in the prodromal phase are unclear. Alterations in hippocampal excitation-inhibition balance (EIB) have recently been linked to early AD pathology. OBJECTIVE Examine how AD risk factors (age, APOEɛ4, amyloid-β) relate to hippocampal EIB in cognitively normal males and females using connectome-level measures. METHODS Individuals from the OASIS-3 cohort (age 42-95) were studied (N = 437), with a subset aged 65+ undergoing neuropsychological testing (N = 231). RESULTS In absence of AD risk factors (APOEɛ4/Aβ+), whole-brain EIB decreases with age more significantly in males than females (p = 0.021, β= -0.007). Regression modeling including APOEɛ4 allele carriers (Aβ-) yielded a significant positive AGE-by-APOE interaction in the right hippocampus for females only (p = 0.013, β= 0.014), persisting with inclusion of Aβ+ individuals (p = 0.012, β= 0.014). Partial correlation analyses of neuropsychological testing showed significant associations with EIB in females: positive correlations between right hippocampal EIB with categorical fluency and whole-brain EIB with the Trail Making Test (p < 0.05). CONCLUSIONS Sex differences in EIB emerge during normal aging and progresses differently with AD risk. Results suggest APOEɛ4 disrupts hippocampal balance more than amyloid in females. Increased excitation correlates positively with neuropsychological performance in the female group, suggesting a duality in terms of potential beneficial effects prior to cognitive impairment. This underscores the translational relevance of APOEɛ4 related hyperexcitation in females, potentially informing therapeutic targets or early interventions to mitigate AD progression in this vulnerable population.
Collapse
Affiliation(s)
- Igor Fortel
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Liang Zhan
- Department of Electrical and Computer Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Olusola Ajilore
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Yichao Wu
- Department of Math, Statistics and Computer Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Scott Mackin
- Department of Psychiatry, University of California – San Francisco, San Francisco, CA, USA
| | - Alex Leow
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
38
|
Ravichandran S, Suhasini R, Madheswaran Deepa S, Selvaraj DB, Vergil Andrews JF, Thiagarajan V, Kandasamy M. Intertwining Neuropathogenic Impacts of Aberrant Circadian Rhythm and Impaired Neuroregenerative Plasticity in Huntington’s Disease: Neurotherapeutic Significance of Chemogenetics. JOURNAL OF MOLECULAR PATHOLOGY 2022; 3:355-371. [DOI: 10.3390/jmp3040030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024] Open
Abstract
Huntington’s disease (HD) is a progressive neurodegenerative disorder characterized by abnormal progressive involuntary movements, cognitive deficits, sleep disturbances, and psychiatric symptoms. The onset and progression of the clinical symptoms have been linked to impaired adult neurogenesis in the brains of subjects with HD, due to the reduced neurogenic potential of neural stem cells (NSCs). Among various pathogenic determinants, an altered clock pathway appears to induce the dysregulation of neurogenesis in neurodegenerative disorders. Notably, gamma-aminobutyric acid (GABA)-ergic neurons that express the vasoactive intestinal peptide (VIP) in the brain play a key role in the regulation of circadian rhythm and neuroplasticity. While an abnormal clock gene pathway has been associated with the inactivation of GABAergic VIP neurons, recent studies suggest the activation of this neuronal population in the brain positively contributes to neuroplasticity. Thus, the activation of GABAergic VIP neurons in the brain might help rectify the irregular circadian rhythm in HD. Chemogenetics refers to the incorporation of genetically engineered receptors or ion channels into a specific cell population followed by its activation using desired chemical ligands. The recent advancement of chemogenetic-based approaches represents a potential scientific tool to rectify the aberrant circadian clock pathways. Considering the facts, the defects in the circadian rhythm can be rectified by the activation of VIP-expressing GABAergic neurons using chemogenetics approaches. Thus, the chemogenetic-based rectification of an abnormal circadian rhythm may facilitate the neurogenic potentials of NSCs to restore the neuroregenerative plasticity in HD. Eventually, the increased neurogenesis in the brain can be expected to mitigate neuronal loss and functional deficits.
Collapse
Affiliation(s)
- Sowbarnika Ravichandran
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Ramalingam Suhasini
- Photonics and Biophotonics Lab, School of Chemistry, Bharathidasan University, Tiruchirappalli 620024, India
| | - Sudhiksha Madheswaran Deepa
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Divya Bharathi Selvaraj
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Jemi Feiona Vergil Andrews
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Viruthachalam Thiagarajan
- Photonics and Biophotonics Lab, School of Chemistry, Bharathidasan University, Tiruchirappalli 620024, India
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi 110002, India
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi 110002, India
| |
Collapse
|
39
|
Zhao R, Grunke SD, Wood CA, Perez GA, Comstock M, Li MH, Singh AK, Park KW, Jankowsky JL. Activity disruption causes degeneration of entorhinal neurons in a mouse model of Alzheimer's circuit dysfunction. eLife 2022; 11:e83813. [PMID: 36468693 PMCID: PMC9873254 DOI: 10.7554/elife.83813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are characterized by selective vulnerability of distinct cell populations; however, the cause for this specificity remains elusive. Here, we show that entorhinal cortex layer 2 (EC2) neurons are unusually vulnerable to prolonged neuronal inactivity compared with neighboring regions of the temporal lobe, and that reelin + stellate cells connecting EC with the hippocampus are preferentially susceptible within the EC2 population. We demonstrate that neuronal death after silencing can be elicited through multiple independent means of activity inhibition, and that preventing synaptic release, either alone or in combination with electrical shunting, is sufficient to elicit silencing-induced degeneration. Finally, we discovered that degeneration following synaptic silencing is governed by competition between active and inactive cells, which is a circuit refinement process traditionally thought to end early in postnatal life. Our data suggests that the developmental window for wholesale circuit plasticity may extend into adulthood for specific brain regions. We speculate that this sustained potential for remodeling by entorhinal neurons may support lifelong memory but renders them vulnerable to prolonged activity changes in disease.
Collapse
Affiliation(s)
- Rong Zhao
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Stacy D Grunke
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Caleb A Wood
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Gabriella A Perez
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Melissa Comstock
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Ming-Hua Li
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Anand K Singh
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Kyung-Won Park
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Joanna L Jankowsky
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
- Departments of Neurology, Neurosurgery, and Molecular and Cellular Biology, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
40
|
van Nifterick AM, Gouw AA, van Kesteren RE, Scheltens P, Stam CJ, de Haan W. A multiscale brain network model links Alzheimer’s disease-mediated neuronal hyperactivity to large-scale oscillatory slowing. Alzheimers Res Ther 2022; 14:101. [PMID: 35879779 PMCID: PMC9310500 DOI: 10.1186/s13195-022-01041-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 07/02/2022] [Indexed: 01/30/2023]
Abstract
Background Neuronal hyperexcitability and inhibitory interneuron dysfunction are frequently observed in preclinical animal models of Alzheimer’s disease (AD). This study investigates whether these microscale abnormalities explain characteristic large-scale magnetoencephalography (MEG) activity in human early-stage AD patients. Methods To simulate spontaneous electrophysiological activity, we used a whole-brain computational network model comprised of 78 neural masses coupled according to human structural brain topology. We modified relevant model parameters to simulate six literature-based cellular scenarios of AD and compare them to one healthy and six contrast (non-AD-like) scenarios. The parameters include excitability, postsynaptic potentials, and coupling strength of excitatory and inhibitory neuronal populations. Whole-brain spike density and spectral power analyses of the simulated data reveal mechanisms of neuronal hyperactivity that lead to oscillatory changes similar to those observed in MEG data of 18 human prodromal AD patients compared to 18 age-matched subjects with subjective cognitive decline. Results All but one of the AD-like scenarios showed higher spike density levels, and all but one of these scenarios had a lower peak frequency, higher spectral power in slower (theta, 4–8Hz) frequencies, and greater total power. Non-AD-like scenarios showed opposite patterns mainly, including reduced spike density and faster oscillatory activity. Human AD patients showed oscillatory slowing (i.e., higher relative power in the theta band mainly), a trend for lower peak frequency and higher total power compared to controls. Combining model and human data, the findings indicate that neuronal hyperactivity can lead to oscillatory slowing, likely due to hyperexcitation (by hyperexcitability of pyramidal neurons or greater long-range excitatory coupling) and/or disinhibition (by reduced excitability of inhibitory interneurons or weaker local inhibitory coupling strength) in early AD. Conclusions Using a computational brain network model, we link findings from different scales and models and support the hypothesis of early-stage neuronal hyperactivity underlying E/I imbalance and whole-brain network dysfunction in prodromal AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01041-4.
Collapse
|
41
|
Yang F, Chen L, Yu Y, Xu T, Chen L, Yang W, Wu Q, Han Y. Alzheimer's disease and epilepsy: An increasingly recognized comorbidity. Front Aging Neurosci 2022; 14:940515. [PMID: 36438002 PMCID: PMC9685172 DOI: 10.3389/fnagi.2022.940515] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/24/2022] [Indexed: 08/27/2023] Open
Abstract
Both Alzheimer's disease (AD) and epilepsy are common chronic diseases in older people. Seizures and epileptiform discharges are very prevalent in AD and can occur since any stage of AD. Increasing evidence indicates that AD and epilepsy may be comorbid. Several factors may be related to the underlying mechanism of the comorbidity. Identifying seizures in patients with AD is a challenge because seizures are often clinically non-motor and may overlap with some AD symptoms. Not only seizures but also epileptiform discharges may exacerbate the cognitive decline in AD patients, highlighting the importance of early recognition and treatment. This review provides a comprehensive overview of seizures in AD from multiple aspects to provide more insight.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yanbing Han
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
42
|
Delbono O, Wang Z, Messi ML. Brainstem noradrenergic neurons: Identifying a hub at the intersection of cognition, motility, and skeletal muscle regulation. Acta Physiol (Oxf) 2022; 236:e13887. [PMID: 36073023 PMCID: PMC9588743 DOI: 10.1111/apha.13887] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 01/29/2023]
Abstract
Brainstem noradrenergic neuron clusters form a node integrating efferents projecting to distinct areas such as those regulating cognition and skeletal muscle structure and function, and receive dissimilar afferents through established circuits to coordinate organismal responses to internal and environmental challenges. Genetic lineage tracing shows the remarkable heterogeneity of brainstem noradrenergic neurons, which may explain their varied functions. They project to the locus coeruleus, the primary source of noradrenaline in the brain, which supports learning and cognition. They also project to pre-ganglionic neurons, which lie within the spinal cord and form synapses onto post-ganglionic neurons. The synapse between descending brainstem noradrenergic neurons and pre-ganglionic spinal neurons, and these in turn with post-ganglionic noradrenergic neurons located at the paravertebral sympathetic ganglia, support an anatomical hierarchy that regulates skeletal muscle innervation, neuromuscular transmission, and muscle trophism. Whether any noradrenergic neuron subpopulation is more susceptible to damaged protein deposit and death with ageing and neurodegeneration is a relevant question that answer will help us to detect neurodegeneration at an early stage, establish prognosis, and anticipate disease progression. Loss of muscle mass and strength with ageing, termed sarcopenia, may predict impaired cognition with ageing and neurodegeneration and establish an early time to start interventions aimed at reducing central noradrenergic neurons hyperactivity. Complex multidisciplinary approaches, including genetic tracing, specific circuit labelling, optogenetics and chemogenetics, electrophysiology, and single-cell transcriptomics and proteomics, are required to test this hypothesis pre-clinical.
Collapse
Affiliation(s)
- Osvaldo Delbono
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Zhong‐Min Wang
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - María Laura Messi
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
43
|
Chemogenetics as a neuromodulatory approach to treating neuropsychiatric diseases and disorders. Mol Ther 2022; 30:990-1005. [PMID: 34861415 PMCID: PMC8899595 DOI: 10.1016/j.ymthe.2021.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/12/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Chemogenetics enables precise, non-invasive, and reversible modulation of neural activity via the activation of engineered receptors that are pharmacologically selective to endogenous or exogenous ligands. With recent advances in therapeutic gene delivery, chemogenetics is poised to support novel interventions against neuropsychiatric diseases and disorders. To evaluate its translational potential, we performed a scoping review of applications of chemogenetics that led to the reversal of molecular and behavioral deficits in studies relevant to neuropsychiatric diseases and disorders. In this review, we present these findings and discuss the potential and challenges for using chemogenetics as a precision medicine-based neuromodulation strategy.
Collapse
|
44
|
Moore BD, Levites Y, Xu G, Hampton H, Adamo MF, Croft CL, Futch HS, Moran C, Fromholt S, Janus C, Prokop S, Dickson D, Lewis J, Giasson BI, Golde TE, Borchelt DR. Soluble brain homogenates from diverse human and mouse sources preferentially seed diffuse Aβ plaque pathology when injected into newborn mouse hosts. FREE NEUROPATHOLOGY 2022; 3:9. [PMID: 35494163 PMCID: PMC9053163 DOI: 10.17879/freeneuropathology-2022-3766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/28/2022] [Indexed: 12/21/2022]
Abstract
Background Seeding of pathology related to Alzheimer's disease (AD) and Lewy body disease (LBD) by tissue homogenates or purified protein aggregates in various model systems has revealed prion-like properties of these disorders. Typically, these homogenates are injected into adult mice stereotaxically. Injection of brain lysates into newborn mice represents an alternative approach of delivering seeds that could direct the evolution of amyloid-β (Aβ) pathology co-mixed with either tau or α-synuclein (αSyn) pathology in susceptible mouse models. Methods Homogenates of human pre-frontal cortex were injected into the lateral ventricles of newborn (P0) mice expressing a mutant humanized amyloid precursor protein (APP), human P301L tau, human wild type αSyn, or combinations thereof. The homogenates were prepared from AD and AD/LBD cases displaying variable degrees of Aβ pathology and co-existing tau and αSyn deposits. Behavioral assessments of APP transgenic mice injected with AD brain lysates were conducted. For comparison, homogenates of aged APP transgenic mice that preferentially exhibit diffuse or cored deposits were similarly injected into the brains of newborn APP mice. Results We observed that lysates from the brains with AD (Aβ+, tau+), AD/LBD (Aβ+, tau+, αSyn+), or Pathological Aging (Aβ+, tau-, αSyn-) efficiently seeded diffuse Aβ deposits. Moderate seeding of cerebral amyloid angiopathy (CAA) was also observed. No animal of any genotype developed discernable tau or αSyn pathology. Performance in fear-conditioning cognitive tasks was not significantly altered in APP transgenic animals injected with AD brain lysates compared to nontransgenic controls. Homogenates prepared from aged APP transgenic mice with diffuse Aβ deposits induced similar deposits in APP host mice; whereas homogenates from APP mice with cored deposits induced similar cored deposits, albeit at a lower level. Conclusions These findings are consistent with the idea that diffuse Aβ pathology, which is a common feature of human AD, AD/LBD, and PA brains, may arise from a distinct strain of misfolded Aβ that is highly transmissible to newborn transgenic APP mice. Seeding of tau or αSyn comorbidities was inefficient in the models we used, indicating that additional methodological refinement will be needed to efficiently seed AD or AD/LBD mixed pathologies by injecting newborn mice.
Collapse
Affiliation(s)
- Brenda D. Moore
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Yona Levites
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Guilian Xu
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Hailey Hampton
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Munir F. Adamo
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Cara L. Croft
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Hunter S. Futch
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Corey Moran
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Susan Fromholt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Christopher Janus
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
- Department of Pathology, University of Florida, Gainesville, FLUnited States
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FLUnited States
| | - Dennis Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FLUnited States
| | - Jada Lewis
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Benoit I. Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| | - Todd E. Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
- Department of Neurology, College of Medicine, University of Florida, Gainesville FLUnited States
| | - David R. Borchelt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FLUnited States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FLUnited States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FLUnited States
| |
Collapse
|
45
|
Moulin TC, Rayêe D, Schiöth HB. Dendritic spine density changes and homeostatic synaptic scaling: a meta-analysis of animal studies. Neural Regen Res 2022; 17:20-24. [PMID: 34100421 PMCID: PMC8451564 DOI: 10.4103/1673-5374.314283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Mechanisms of homeostatic plasticity promote compensatory changes of cellular excitability in response to chronic changes in the network activity. This type of plasticity is essential for the maintenance of brain circuits and is involved in the regulation of neural regeneration and the progress of neurodegenerative disorders. One of the most studied homeostatic processes is synaptic scaling, where global synaptic adjustments take place to restore the neuronal firing rate to a physiological range by the modulation of synaptic receptors, neurotransmitters, and morphology. However, despite the comprehensive literature on the electrophysiological properties of homeostatic scaling, less is known about the structural adjustments that occur in the synapses and dendritic tree. In this study, we performed a meta-analysis of articles investigating the effects of chronic network excitation (synaptic downscaling) or inhibition (synaptic upscaling) on the dendritic spine density of neurons. Our results indicate that spine density is consistently reduced after protocols that induce synaptic scaling, independent of the intervention type. Then, we discuss the implication of our findings to the current knowledge on the morphological changes induced by homeostatic plasticity.
Collapse
Affiliation(s)
- Thiago C Moulin
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden; Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle Rayêe
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, NY, USA
| | - Helgi B Schiöth
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden; Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
46
|
Borcuk C, Héraud C, Herbeaux K, Diringer M, Panzer É, Scuto J, Hashimoto S, Saido TC, Saito T, Goutagny R, Battaglia D, Mathis C. Early memory deficits and extensive brain network disorganization in the AppNL-F/MAPT double knock-in mouse model of familial Alzheimer's disease. AGING BRAIN 2022; 2:100042. [PMID: 36908877 PMCID: PMC9997176 DOI: 10.1016/j.nbas.2022.100042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022] Open
Abstract
A critical challenge in current research on Alzheimer's disease (AD) is to clarify the relationship between network dysfunction and the emergence of subtle memory deficits in itspreclinical stage. The AppNL-F/MAPT double knock-in (dKI) model with humanized β-amyloid peptide (Aβ) and tau was used to investigate both memory and network dysfunctions at an early stage. Young male dKI mice (2 to 6 months) were tested in three tasks taxing different aspects of recognition memory affected in preclinical AD. An early deficit first appeared in the object-place association task at the age of 4 months, when increased levels of β-CTF and Aβ were detected in both the hippocampus and the medial temporal cortex, and tau pathology was found only in the medial temporal cortex. Object-place task-dependent c-Fos activation was then analyzed in 22 subregions across the medial prefrontal cortex, claustrum, retrosplenial cortex, and medial temporal lobe. Increased c-Fos activation was detected in the entorhinal cortex and the claustrum of dKI mice. During recall, network efficiency was reduced across cingulate regions with a major disruption of information flow through the retrosplenial cortex. Our findings suggest that early perirhinal-entorhinal pathology is associated with abnormal activity which may spread to downstream regions such as the claustrum, the medial prefrontal cortex and ultimately the key retrosplenial hub which relays information from frontal to temporal lobes. The similarity between our findings and those reported in preclinical stages of AD suggests that the AppNL-F/MAPT dKI model has a high potential for providing key insights into preclinical AD.
Collapse
Key Words
- AD, Alzheimer’s disease
- ADAD, autosomal dominant Alzheimer’s disease
- Associative memory
- CLA, claustrum
- Claustrum
- DMN, default mode network
- EI, exploration index
- FC, functional connectivity
- Functional connectivity
- MI, Memory index
- MTC, medial temporal cortex
- MTL, medial temporal lobe
- Medial temporal cortex
- NOR, novel object recognition
- OL, Object location
- OP, object-place
- PS, Pattern Separation
- Preclinical Alzheimer disease
- Retrosplenial cortex
- aMCI, amnestic mild cognitive impairment
- amyloid beta, Aβ
- dKI, AppNL-F/MAPT double knock-in
- ptau Thr 181, Thr181phosphorylated tau protein
Collapse
Affiliation(s)
- Christopher Borcuk
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Céline Héraud
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Karine Herbeaux
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Margot Diringer
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Élodie Panzer
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Jil Scuto
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Romain Goutagny
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Demian Battaglia
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France.,University of Strasbourg Institute for Advanced Studies (USIAS), F-67000 Strasbourg, France.,Université d'Aix-Marseille, Inserm, Institut de Neurosciences des Systèmes (INS) UMR_S 1106, F-13005 Marseille, France
| | - Chantal Mathis
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| |
Collapse
|
47
|
Kawai M, Imaizumi K, Ishikawa M, Shibata S, Shinozaki M, Shibata T, Hashimoto S, Kitagawa T, Ago K, Kajikawa K, Shibata R, Kamata Y, Ushiba J, Koga K, Furue H, Matsumoto M, Nakamura M, Nagoshi N, Okano H. Long-term selective stimulation of transplanted neural stem/progenitor cells for spinal cord injury improves locomotor function. Cell Rep 2021; 37:110019. [PMID: 34818559 DOI: 10.1016/j.celrep.2021.110019] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
In cell transplantation therapy for spinal cord injury (SCI), grafted human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) mainly differentiate into neurons, forming synapses in a process similar to neurodevelopment. In the developing nervous system, the activity of immature neurons has an important role in constructing and maintaining new synapses. Thus, we investigate how enhancing the activity of transplanted hiPSC-NS/PCs affects both the transplanted cells themselves and the host tissue. We find that chemogenetic stimulation of hiPSC-derived neural cells enhances cell activity and neuron-to-neuron interactions in vitro. In a rodent model of SCI, consecutive and selective chemogenetic stimulation of transplanted hiPSC-NS/PCs also enhances the expression of synapse-related genes and proteins in surrounding host tissues and prevents atrophy of the injured spinal cord, thereby improving locomotor function. These findings provide a strategy for enhancing activity within the graft to improve the efficacy of cell transplantation therapy for SCI.
Collapse
Affiliation(s)
- Momotaro Kawai
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kent Imaizumi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takahiro Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shogo Hashimoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takahiro Kitagawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kentaro Ago
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Keita Kajikawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Reo Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yasuhiro Kamata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Junichi Ushiba
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama 223-8522, Japan
| | - Keisuke Koga
- Department of Neurophysiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| |
Collapse
|
48
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
49
|
Tok S, Ahnaou A, Drinkenburg W. Functional Neurophysiological Biomarkers of Early-Stage Alzheimer's Disease: A Perspective of Network Hyperexcitability in Disease Progression. J Alzheimers Dis 2021; 88:809-836. [PMID: 34420957 PMCID: PMC9484128 DOI: 10.3233/jad-210397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Network hyperexcitability (NH) has recently been suggested as a potential neurophysiological indicator of Alzheimer’s disease (AD), as new, more accurate biomarkers of AD are sought. NH has generated interest as a potential indicator of certain stages in the disease trajectory and even as a disease mechanism by which network dysfunction could be modulated. NH has been demonstrated in several animal models of AD pathology and multiple lines of evidence point to the existence of NH in patients with AD, strongly supporting the physiological and clinical relevance of this readout. Several hypotheses have been put forward to explain the prevalence of NH in animal models through neurophysiological, biochemical, and imaging techniques. However, some of these hypotheses have been built on animal models with limitations and caveats that may have derived NH through other mechanisms or mechanisms without translational validity to sporadic AD patients, potentially leading to an erroneous conclusion of the underlying cause of NH occurring in patients with AD. In this review, we discuss the substantiation for NH in animal models of AD pathology and in human patients, as well as some of the hypotheses considering recently developed animal models that challenge existing hypotheses and mechanisms of NH. In addition, we provide a preclinical perspective on how the development of animal models incorporating AD-specific NH could provide physiologically relevant translational experimental data that may potentially aid the discovery and development of novel therapies for AD.
Collapse
Affiliation(s)
- Sean Tok
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, The Netherlands
| | - Abdallah Ahnaou
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Wilhelmus Drinkenburg
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, The Netherlands
| |
Collapse
|
50
|
Ying Y, Wang JZ. Illuminating Neural Circuits in Alzheimer's Disease. Neurosci Bull 2021; 37:1203-1217. [PMID: 34089505 PMCID: PMC8353043 DOI: 10.1007/s12264-021-00716-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and there is currently no cure. Neural circuit dysfunction is the fundamental mechanism underlying the learning and memory deficits in patients with AD. Therefore, it is important to understand the structural features and mechanisms underlying the deregulated circuits during AD progression, by which new tools for intervention can be developed. Here, we briefly summarize the most recently established cutting-edge experimental approaches and key techniques that enable neural circuit tracing and manipulation of their activity. We also discuss the advantages and limitations of these approaches. Finally, we review the applications of these techniques in the discovery of circuit mechanisms underlying β-amyloid and tau pathologies during AD progression, and as well as the strategies for targeted AD treatments.
Collapse
Affiliation(s)
- Yang Ying
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|