1
|
Li YB, Fu Q, Guo M, Du Y, Chen Y, Cheng Y. MicroRNAs: pioneering regulators in Alzheimer's disease pathogenesis, diagnosis, and therapy. Transl Psychiatry 2024; 14:367. [PMID: 39256358 PMCID: PMC11387755 DOI: 10.1038/s41398-024-03075-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
This article delves into Alzheimer's disease (AD), a prevalent neurodegenerative condition primarily affecting the elderly. It is characterized by progressive memory and cognitive impairments, severely disrupting daily life. Recent research highlights the potential involvement of microRNAs in the pathogenesis of AD. MicroRNAs (MiRNAs), short non-coding RNAs comprising 20-24 nucleotides, significantly influence gene regulation by hindering translation or promoting degradation of target genes. This review explores the role of specific miRNAs in AD progression, focusing on their impact on β-amyloid (Aβ) peptide accumulation, intracellular aggregation of hyperphosphorylated tau proteins, mitochondrial dysfunction, neuroinflammation, oxidative stress, and the expression of the APOE4 gene. Our insights contribute to understanding AD's pathology, offering new avenues for identifying diagnostic markers and developing novel therapeutic targets.
Collapse
Affiliation(s)
- Yao-Bo Li
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Qiang Fu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Mei Guo
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yang Du
- Institute of National Security, Minzu University of China, Beijing, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China.
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China.
| |
Collapse
|
2
|
Kramer M, Hoang TH, Yang H, Shchyglo O, Böge J, Neubacher U, Colitti-Klausnitzer J, Manahan-Vaughan D. Intracerebral inoculation of healthy non-transgenic rats with a single aliquot of oligomeric amyloid-β (1-42) profoundly and progressively alters brain function throughout life. Front Aging Neurosci 2024; 16:1397901. [PMID: 39156737 PMCID: PMC11327071 DOI: 10.3389/fnagi.2024.1397901] [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: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024] Open
Abstract
One of the puzzling aspects of sporadic Alzheimer's disease (AD) is how it commences. Changes in one key brain peptide, amyloid-beta (Aβ), accompany disease progression, but whether this comprises a trigger or a consequence of AD is still a topic of debate. It is clear however that the cerebral presence of oligomeric Aβ (1-42) is a key factor in early AD-pathogenesis. Furthermore, treatment of rodent brains with oligomeric Aβ (1-42) either in vitro or in vivo, acutely impairs hippocampal synaptic plasticity, creating a link between Aβ-pathology and learning impairments. Here, we show that a once-off inoculation of the brains of healthy adult rats with oligomeric Aβ (1-42) exerts debilitating effects on the long-term viability of the hippocampus, one of the primary targets of AD. Changes are progressive: months after treatment, synaptic plasticity, neuronal firing and spatial learning are impaired and expression of plasticity-related proteins are changed, in the absence of amyloid plaques. Early changes relate to activation of microglia, whereas later changes are associated with a reconstruction of astroglial morphology. These data suggest that a disruption of Aβ homeostasis may suffice to trigger an irreversible cascade, underlying progressive loss of hippocampal function, that parallels the early stages of AD.
Collapse
|
3
|
Tabuena DR, Jang SS, Grone B, Yip O, Aery Jones EA, Blumenfeld J, Liang Z, Koutsodendris N, Rao A, Ding L, Zhang AR, Hao Y, Xu Q, Yoon SY, Leon SD, Huang Y, Zilberter M. Neuronal APOE4-induced Early Hippocampal Network Hyperexcitability in Alzheimer's Disease Pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.28.555153. [PMID: 37693533 PMCID: PMC10491126 DOI: 10.1101/2023.08.28.555153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The full impact of apolipoprotein E4 (APOE4), the strongest genetic risk factor for Alzheimer's disease (AD), on neuronal and network function remains unclear. We found hippocampal region-specific network hyperexcitability in young APOE4 knock-in (E4-KI) mice which predicted cognitive deficits at old age. Network hyperexcitability in young E4-KI mice was mediated by hippocampal region-specific subpopulations of smaller and hyperexcitable neurons that were eliminated by selective removal of neuronal APOE4. Aged E4-KI mice exhibited hyperexcitable granule cells, a progressive inhibitory deficit, and E/I imbalance in the dentate gyrus, exacerbating hippocampal hyperexcitability. Single-nucleus RNA-sequencing revealed neuronal cell type-specific and age-dependent transcriptomic changes, including Nell2 overexpression in E4-KI mice. Reducing Nell2 expression in specific neuronal types of E4-KI mice with CRISPRi rescued their abnormal excitability phenotypes, implicating Nell2 overexpression as a cause of APOE4-induced hyperexcitability. These findings highlight the early transcriptomic and electrophysiological alterations underlying APOE4-induced hippocampal network dysfunction and its contribution to AD pathogenesis with aging.
Collapse
|
4
|
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: 1] [Impact Index Per Article: 0.5] [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
|
5
|
Piccialli I, Sisalli MJ, de Rosa V, Boscia F, Tedeschi V, Secondo A, Pannaccione A. Increased K V2.1 Channel Clustering Underlies the Reduction of Delayed Rectifier K + Currents in Hippocampal Neurons of the Tg2576 Alzheimer's Disease Mouse. Cells 2022; 11:cells11182820. [PMID: 36139395 PMCID: PMC9497218 DOI: 10.3390/cells11182820] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the progressive deterioration of cognitive functions. Cortical and hippocampal hyperexcitability intervenes in the pathological derangement of brain activity leading to cognitive decline. As key regulators of neuronal excitability, the voltage-gated K+ channels (KV) might play a crucial role in the AD pathophysiology. Among them, the KV2.1 channel, the main α subunit mediating the delayed rectifier K+ currents (IDR) and controlling the intrinsic excitability of pyramidal neurons, has been poorly examined in AD. In the present study, we investigated the KV2.1 protein expression and activity in hippocampal neurons from the Tg2576 mouse, a widely used transgenic model of AD. To this aim we performed whole-cell patch-clamp recordings, Western blotting, and immunofluorescence analyses. Our Western blotting results reveal that KV2.1 was overexpressed in the hippocampus of 3-month-old Tg2576 mice and in primary hippocampal neurons from Tg2576 mouse embryos compared with the WT counterparts. Electrophysiological experiments unveiled that the whole IDR were reduced in the Tg2576 primary neurons compared with the WT neurons, and that this reduction was due to the loss of the KV2.1 current component. Moreover, we found that the reduction of the KV2.1-mediated currents was due to increased channel clustering, and that glutamate, a stimulus inducing KV2.1 declustering, was able to restore the IDR to levels comparable to those of the WT neurons. These findings add new information about the dysregulation of ionic homeostasis in the Tg2576 AD mouse model and identify KV2.1 as a possible player in the AD-related alterations of neuronal excitability.
Collapse
|
6
|
Amyloid Beta Alters Prefrontal-dependent Functions Along with its Excitability and Synaptic Plasticity in Male Rats. Neuroscience 2022; 498:260-279. [PMID: 35839923 DOI: 10.1016/j.neuroscience.2022.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/20/2022] [Accepted: 07/07/2022] [Indexed: 12/17/2022]
Abstract
Prefrontal cortex (PFC)-related functions, such as working memory (WM) and cognitive flexibility (CF), are among the first to be altered at early stages of Alzheimer's disease (AD). Likewise, transgenic AD models carrying different AD-related mutations, mostly linked to the overproduction of amyloid beta (Aβ) and other peptides, show premature behavioral and functional symptoms associated with PFC alterations. However, little is known about the effects of intracerebral or intra-PFC Aβ infusion on WM and CF, as well as on pyramidal cell excitability and plasticity. Thus, here we evaluated the effects of a single Aβ injection, directly into the PFC, or its intracerebroventricular (icv) application, on PFC-dependent behaviors and on the intrinsic and synaptic properties of layer V pyramidal neurons in PFC slices. We found that a single icv Aβ infusion reduced learning and performance of a delayed non-matching-to-sample WM task and prevented reversal learning in a matching-to-sample version of the task, several weeks after its infusion. The inhibition of WM performance was reproduced more potently by a single PFC Aβ infusion and was associated with Aβ accumulation. This behavioral disruption was related to increased layer V pyramidal cell firing, larger sag membrane potential, increased fast after-hyperpolarization and a failure to sustain synaptic long-term potentiation, even leading to long-term depression, at both the hippocampal-PFC pathway and intracortical synapses. These findings show that Aβ can affect PFC excitability and synaptic plasticity balance, damaging PFC-dependent functions, which could constitute the foundations of the early alterations in executive functions in AD patients.
Collapse
|
7
|
Transcranial Magnetic Stimulation Indices of Cortical Excitability Enhance the Prediction of Response to Pharmacotherapy in Late-Life Depression. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:265-275. [PMID: 34311121 PMCID: PMC8783923 DOI: 10.1016/j.bpsc.2021.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Older adults with late-life depression (LLD) often experience incomplete or lack of response to first-line pharmacotherapy. The treatment of LLD could be improved using objective biological measures to predict response. Transcranial magnetic stimulation (TMS) can be used to measure cortical excitability, inhibition, and plasticity, which have been implicated in LLD pathophysiology and associated with brain stimulation treatment outcomes in younger adults with depression. TMS measures have not yet been investigated as predictors of treatment outcomes in LLD or pharmacotherapy outcomes in adults of any age with depression. METHODS We assessed whether pretreatment single-pulse and paired-pulse TMS measures, combined with clinical and demographic measures, predict venlafaxine treatment response in 76 outpatients with LLD. We compared the predictive performance of machine learning models including or excluding TMS predictors. RESULTS Two single-pulse TMS measures predicted venlafaxine response: cortical excitability (neuronal membrane excitability) and the variability of cortical excitability (dynamic fluctuations in excitability levels). In cross-validation, models using a combination of these TMS predictors, clinical markers of treatment resistance, and age classified patients with 73% ± 11% balanced accuracy (average correct classification rate of responders and nonresponders; permutation testing, p < .005); these models significantly outperformed (corrected t test, p = .025) models using clinical and demographic predictors alone (60% ± 10% balanced accuracy). CONCLUSIONS These preliminary findings suggest that single-pulse TMS measures of cortical excitability may be useful predictors of response to pharmacotherapy in LLD. Future studies are needed to confirm these findings and determine whether combining TMS predictors with other biomarkers further improves the accuracy of predicting LLD treatment outcome.
Collapse
|
8
|
Fernandez‐Perez EJ, Muñoz B, Bascuñan DA, Peters C, Riffo‐Lepe NO, Espinoza MP, Morgan PJ, Filippi C, Bourboulou R, Sengupta U, Kayed R, Epsztein J, Aguayo LG. Synaptic dysregulation and hyperexcitability induced by intracellular amyloid beta oligomers. Aging Cell 2021; 20:e13455. [PMID: 34409748 PMCID: PMC8441418 DOI: 10.1111/acel.13455] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Intracellular amyloid beta oligomer (iAβo) accumulation and neuronal hyperexcitability are two crucial events at early stages of Alzheimer's disease (AD). However, to date, no mechanism linking iAβo with an increase in neuronal excitability has been reported. Here, the effects of human AD brain-derived (h-iAβo) and synthetic (iAβo) peptides on synaptic currents and action potential firing were investigated in hippocampal neurons. Starting from 500 pM, iAβo rapidly increased the frequency of synaptic currents and higher concentrations potentiated the AMPA receptor-mediated current. Both effects were PKC-dependent. Parallel recordings of synaptic currents and nitric oxide (NO)-associated fluorescence showed that the increased frequency, related to pre-synaptic release, was dependent on a NO-mediated retrograde signaling. Moreover, increased synchronization in NO production was also observed in neurons neighboring those dialyzed with iAβo, indicating that iAβo can increase network excitability at a distance. Current-clamp recordings suggested that iAβo increased neuronal excitability via AMPA-driven synaptic activity without altering membrane intrinsic properties. These results strongly indicate that iAβo causes functional spreading of hyperexcitability through a synaptic-driven mechanism and offers an important neuropathological significance to intracellular species in the initial stages of AD, which include brain hyperexcitability and seizures.
Collapse
Affiliation(s)
| | - Braulio Muñoz
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Denisse A. Bascuñan
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Christian Peters
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Nicolas O. Riffo‐Lepe
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Maria P. Espinoza
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Peter J. Morgan
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Caroline Filippi
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Romain Bourboulou
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Jérôme Epsztein
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Luis G. Aguayo
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| |
Collapse
|
9
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
10
|
Van Egroo M, Chylinski D, Narbutas J, Besson G, Muto V, Schmidt C, Marzoli D, Cardone P, Vandeleene N, Grignard M, Luxen A, Salmon E, Lambert C, Bastin C, Collette F, Phillips C, Maquet P, Bahri MA, Balteau E, Vandewalle G. Early brainstem [18F]THK5351 uptake is linked to cortical hyperexcitability in healthy aging. JCI Insight 2021; 6:142514. [PMID: 33290274 PMCID: PMC7934880 DOI: 10.1172/jci.insight.142514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Neuronal hyperexcitability characterizes the early stages of Alzheimer’s disease (AD). In animals, early misfolded tau and amyloid-β (Aβ) protein accumulation — both central to AD neuropathology — promote cortical excitability and neuronal network dysfunction. In healthy humans, misfolded tau and Aβ aggregates are first detected, respectively, in the brainstem and frontomedial and temporobasal cortices, decades prior to the onset of AD cognitive symptoms. Whether cortical excitability is related to early brainstem tau — and its associated neuroinflammation — and cortical Aβ aggregations remains unknown. METHODS We probed frontal cortex excitability, using transcranial magnetic stimulation combined with electroencephalography, in a sample of 64 healthy late-middle–aged individuals (50–69 years; 45 women and 19 men). We assessed whole-brain [18F]THK5351 PET uptake as a proxy measure of tau/neuroinflammation, and we assessed whole-brain Aβ burden with [18F]Flutemetamol or [18F]Florbetapir radiotracers. RESULTS We found that higher [18F]THK5351 uptake in a brainstem monoaminergic compartment was associated with increased cortical excitability (r = 0.29, P = 0.02). By contrast, [18F]THK5351 PET signal in the hippocampal formation, although strongly correlated with brainstem signal in whole-brain voxel-based quantification analyses (P value corrected for family-wise error [PFWE-corrected] < 0.001), was not significantly associated with cortical excitability (r = 0.14, P = 0.25). Importantly, no significant association was found between early Aβ cortical deposits and cortical excitability (r = –0.20, P = 0.11). CONCLUSION These findings reveal potential brain substrates for increased cortical excitability in preclinical AD and may constitute functional in vivo correlates of early brainstem tau accumulation and neuroinflammation in humans. TRIAL REGISTRATION EudraCT 2016-001436-35. FUNDING F.R.S.-FNRS Belgium, Wallonie-Bruxelles International, ULiège, Fondation Simone et Pierre Clerdent, European Regional Development Fund.
Collapse
Affiliation(s)
| | | | - Justinas Narbutas
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Psychology and Cognitive Neuroscience Research Unit, University of Liège (ULiège), Liège, Belgium
| | | | - Vincenzo Muto
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Psychology and Cognitive Neuroscience Research Unit, University of Liège (ULiège), Liège, Belgium
| | - Christina Schmidt
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Psychology and Cognitive Neuroscience Research Unit, University of Liège (ULiège), Liège, Belgium
| | | | | | | | | | - André Luxen
- GIGA-Cyclotron Research Centre-In Vivo Imaging and
| | - Eric Salmon
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Psychology and Cognitive Neuroscience Research Unit, University of Liège (ULiège), Liège, Belgium.,Department of Neurology, University Hospital of Liège, Liège, Belgium
| | - Christian Lambert
- Wellcome Centre for Human Neuroimaging, University College London Institute of Neurology, London, United Kingdom
| | - Christine Bastin
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Psychology and Cognitive Neuroscience Research Unit, University of Liège (ULiège), Liège, Belgium
| | - Fabienne Collette
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Psychology and Cognitive Neuroscience Research Unit, University of Liège (ULiège), Liège, Belgium
| | - Christophe Phillips
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,GIGA-In Silico Medicine, ULiège, Liège, Belgium
| | - Pierre Maquet
- GIGA-Cyclotron Research Centre-In Vivo Imaging and.,Department of Neurology, University Hospital of Liège, Liège, Belgium
| | | | | | | |
Collapse
|
11
|
Lam J, Lee J, Liu CY, Lozano AM, Lee DJ. Deep Brain Stimulation for Alzheimer's Disease: Tackling Circuit Dysfunction. Neuromodulation 2020; 24:171-186. [PMID: 33377280 DOI: 10.1111/ner.13305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/07/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Treatments for Alzheimer's disease are urgently needed given its enormous human and economic costs and disappointing results of clinical trials targeting the primary amyloid and tau pathology. On the other hand, deep brain stimulation (DBS) has demonstrated success in other neurological and psychiatric disorders leading to great interest in DBS as a treatment for Alzheimer's disease. MATERIALS AND METHODS We review the literature on 1) circuit dysfunction in Alzheimer's disease and 2) DBS for Alzheimer's disease. Human and animal studies are reviewed individually. RESULTS There is accumulating evidence of neural circuit dysfunction at the structural, functional, electrophysiological, and neurotransmitter level. Recent evidence from humans and animals indicate that DBS has the potential to restore circuit dysfunction in Alzheimer's disease, similarly to other movement and psychiatric disorders, and may even slow or reverse the underlying disease pathophysiology. CONCLUSIONS DBS is an intriguing potential treatment for Alzheimer's disease, targeting circuit dysfunction as a novel therapeutic target. However, further exploration of the basic disease pathology and underlying mechanisms of DBS is necessary to better understand how circuit dysfunction can be restored. Additionally, robust clinical data in the form of ongoing phase III clinical trials are needed to validate the efficacy of DBS as a viable treatment.
Collapse
Affiliation(s)
- Jordan Lam
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| | - Justin Lee
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| | - Charles Y Liu
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| | - Andres M Lozano
- Division of Neurological Surgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Darrin J Lee
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| |
Collapse
|
12
|
Kwon HS, Koh SH. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener 2020; 9:42. [PMID: 33239064 PMCID: PMC7689983 DOI: 10.1186/s40035-020-00221-2] [Citation(s) in RCA: 1072] [Impact Index Per Article: 268.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
Neuroinflammation is associated with neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Microglia and astrocytes are key regulators of inflammatory responses in the central nervous system. The activation of microglia and astrocytes is heterogeneous and traditionally categorized as neurotoxic (M1-phenotype microglia and A1-phenotype astrocytes) or neuroprotective (M2-phenotype microglia and A2-phenotype astrocytes). However, this dichotomized classification may not reflect the various phenotypes of microglia and astrocytes. The relationship between these activated glial cells is also very complicated, and the phenotypic distribution can change, based on the progression of neurodegenerative diseases. A better understanding of the roles of microglia and astrocytes in neurodegenerative diseases is essential for developing effective therapies. In this review, we discuss the roles of inflammatory response in neurodegenerative diseases, focusing on the contributions of microglia and astrocytes and their relationship. In addition, we discuss biomarkers to measure neuroinflammation and studies on therapeutic drugs that can modulate neuroinflammation.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, Seoul, Republic of Korea.
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Klee JL, Kiliaan AJ, Lipponen A, Battaglia FP. Reduced firing rates of pyramidal cells in the frontal cortex of APP/PS1 can be restored by acute treatment with levetiracetam. Neurobiol Aging 2020; 96:79-86. [PMID: 32950781 DOI: 10.1016/j.neurobiolaging.2020.08.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 08/03/2020] [Accepted: 08/19/2020] [Indexed: 12/27/2022]
Abstract
In recent years, aberrant neural oscillations in various cortical areas have emerged as a common physiological hallmark across mouse models of amyloid pathology and patients with Alzheimer's disease. However, much less is known about the underlying effect of amyloid pathology on single cell activity. Here, we used high-density silicon probe recordings from frontal cortex area of 9-month-old APP/PS1 mice to show that local field potential power in the theta and beta band is increased in transgenic animals, whereas single-cell firing rates, specifically of putative pyramidal cells, are significantly reduced. At the same time, these sparsely firing pyramidal cells phase-lock their spiking activity more strongly to the ongoing theta and beta rhythms. Furthermore, we demonstrated that the antiepileptic drug, levetiracetam, counteracts these effects by increasing pyramidal cell firing rates in APP/PS1 mice and uncoupling pyramidal cells and interneurons. Overall, our results highlight reduced firing rates of cortical pyramidal cells as a pathophysiological phenotype in APP/PS1 mice and indicate a potentially beneficial effect of acute levetiracetam treatment.
Collapse
Affiliation(s)
- Jan L Klee
- Department of Neuroinformatics, Radboud University, Nijmegen, the Netherlands
| | - Amanda J Kiliaan
- Department of Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arto Lipponen
- Department of Psychology, University of Jyväskylä, Finland.
| | | |
Collapse
|
14
|
Li S, Hayden EY, Garcia VJ, Fuchs DT, Sheyn J, Daley DA, Rentsendorj A, Torbati T, Black KL, Rutishauser U, Teplow DB, Koronyo Y, Koronyo-Hamaoui M. Activated Bone Marrow-Derived Macrophages Eradicate Alzheimer's-Related Aβ 42 Oligomers and Protect Synapses. Front Immunol 2020; 11:49. [PMID: 32082319 PMCID: PMC7005081 DOI: 10.3389/fimmu.2020.00049] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Impaired synaptic integrity and function due to accumulation of amyloid β-protein (Aβ42) oligomers is thought to be a major contributor to cognitive decline in Alzheimer's disease (AD). However, the exact role of Aβ42 oligomers in synaptotoxicity and the ability of peripheral innate immune cells to rescue synapses remain poorly understood due to the metastable nature of oligomers. Here, we utilized photo-induced cross-linking to stabilize pure oligomers and study their effects vs. fibrils on synapses and protection by Aβ-phagocytic macrophages. We found that cortical neurons were more susceptible to Aβ42 oligomers than fibrils, triggering additional neuritic arborization retraction, functional alterations (hyperactivity and spike waveform), and loss of VGluT1- and PSD95-excitatory synapses. Co-culturing neurons with bone marrow-derived macrophages protected synapses against Aβ42 fibrils; moreover, immune activation with glatiramer acetate (GA) conferred further protection against oligomers. Mechanisms involved increased Aβ42 removal by macrophages, amplified by GA stimulation: fibrils were largely cleared through intracellular CD36/EEA1+-early endosomal proteolysis, while oligomers were primarily removed via extracellular/MMP-9 enzymatic degradation. In vivo studies in GA-immunized or CD115+-monocyte-grafted APPSWE/PS1ΔE9-transgenic mice followed by pre- and postsynaptic analyses of entorhinal cortex and hippocampal substructures corroborated our in vitro findings of macrophage-mediated synaptic preservation. Together, our data demonstrate that activated macrophages effectively clear Aβ42 oligomers and rescue VGluT1/PSD95 synapses, providing rationale for harnessing macrophages to treat AD.
Collapse
Affiliation(s)
- Songlin Li
- Institute of Neuroscience and Chemistry, Wenzhou University, Wenzhou, China
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Eric Y. Hayden
- Department of Neurology, David Geffen School of Medicine at UCLA, Mary S. Easton Center for Alzheimer's Disease Research at UCLA, Brain Research Institute, Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Veronica J. Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Julia Sheyn
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - David A. Daley
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Altan Rentsendorj
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Tania Torbati
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Ueli Rutishauser
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - David B. Teplow
- Department of Neurology, David Geffen School of Medicine at UCLA, Mary S. Easton Center for Alzheimer's Disease Research at UCLA, Brain Research Institute, Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yosef Koronyo
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine-Dunitz Neurosurgical Institute, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
15
|
Electrophysiological Characterization of Networks and Single Cells in the Hippocampal Region of a Transgenic Rat Model of Alzheimer's Disease. eNeuro 2019; 6:eN-NWR-0448-17. [PMID: 30809590 PMCID: PMC6390198 DOI: 10.1523/eneuro.0448-17.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 11/14/2018] [Accepted: 01/21/2019] [Indexed: 01/02/2023] Open
Abstract
The hippocampus and entorhinal cortex (EC) are areas affected early and severely in Alzheimer’s disease (AD), and this is associated with deficits in episodic memory. Amyloid-β (Aβ), the main protein found in amyloid plaques, can affect neuronal physiology and excitability, and several AD mouse models with memory impairments display aberrant network activity, including hyperexcitability and seizures. In this study, we investigated single cell physiology in EC and network activity in EC and dentate gyrus (DG) in the McGill-R-Thy1-APP transgenic rat model, using whole-cell patch clamp recordings and voltage-sensitive dye imaging (VSDI) in acute slices. In slices from transgenic animals up to 4 months of age, the majority of the principal neurons in Layer II of EC, fan cells and stellate cells, expressed intracellular Aβ (iAβ). Whereas the electrophysiological properties of fan cells were unaltered, stellate cells were more excitable in transgenic than in control rats. Stimulation in the DG resulted in comparable patterns in both groups at three and nine months, but at 12 months, the elicited responses in the transgenic group showed a significant preference for the enclosed blade, without any change in overall excitability. Only transient changes in the local network activity were seen in the medial EC (MEC). Although the observed changes in the McGill rat model are subtle, they are specific, pointing to a differential and selective involvement of specific parts of the hippocampal circuitry in Aβ pathology.
Collapse
|
16
|
Caravaglios G, Castro G, Muscoso EG, Crivelli D, Balconi M. Beta Responses in Healthy Elderly and in Patients With Amnestic Mild Cognitive Impairment During a Task of Temporal Orientation of Attention. Clin EEG Neurosci 2018; 49:258-271. [PMID: 27807013 DOI: 10.1177/1550059416676144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent studies demonstrated that beta oscillations are elicited during cognitive processes. To investigate their potential as electrophysiological markers of amnestic mild cognitive impairment (aMCI), we recorded beta EEG activity during resting and during an omitted tone task in patients and healthy elderly. Thirty participants were enrolled (15 patients, 15 healthy controls). In particular, we investigated event-related spectral perturbation and intertrial coherence indices. Analyses showed that ( a) healthy elderly presented greater beta power at rest than patients with aMCI patients; ( b) during the task, healthy elderly were more accurate than aMCI patients and presented greater beta power than aMCI patients; ( c) both groups showed qualitatively similar spectral perturbation responses during the task, but different spatiotemporal response patterns; and ( d) aMCI patients presented greater beta phase locking than healthy elderly during the task. Results indicate that beta activity in healthy elderly differs from that of patients with aMCI. Furthermore, the analysis of task-related EEG activity extends evidences obtained during resting and suggests that during the prodromal phase of Alzheimer's disease there is a reduced efficiency in information exchange by large-scale neural networks. The study for the first time shows the potential of task-related beta responses as early markers of aMCI impairments.
Collapse
Affiliation(s)
- Giuseppe Caravaglios
- 1 Department of Neurology, Center for AD Diagnosis and Care, Cannizzaro Hospital, Catania, Italy
| | - Giuseppe Castro
- 2 Local Health Department of Catania, Semi-residential Center for Dementia of Acireale, Acireale (CT), Italy
| | - Emma Gabriella Muscoso
- 1 Department of Neurology, Center for AD Diagnosis and Care, Cannizzaro Hospital, Catania, Italy
| | - Davide Crivelli
- 3 Research Unit in Affective and Social Neuroscience, Catholic University of the Sacred Heart, Milan, Italy.,4 Department of Psychology, Catholic University of the Sacred Heart, Milan, Italy
| | - Michela Balconi
- 3 Research Unit in Affective and Social Neuroscience, Catholic University of the Sacred Heart, Milan, Italy.,4 Department of Psychology, Catholic University of the Sacred Heart, Milan, Italy
| |
Collapse
|
17
|
Amyloid causes intermittent network disruptions in cognitively intact older subjects. Brain Imaging Behav 2018; 13:699-716. [DOI: 10.1007/s11682-018-9869-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
18
|
Mueller SG, Weiner MW. Amyloid Associated Intermittent Network Disruptions in Cognitively Intact Older Subjects: Structural Connectivity Matters. Front Aging Neurosci 2017; 9:418. [PMID: 29311904 PMCID: PMC5742224 DOI: 10.3389/fnagi.2017.00418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/06/2017] [Indexed: 01/20/2023] Open
Abstract
Observations in animal models suggest that amyloid can cause network hypersynchrony in the early preclinical phase of Alzheimer's disease (AD). The aim of this study was (a) to obtain evidence of paroxysmal hypersynchrony in cognitively intact subjects (CN) with increased brain amyloid load from task-free fMRI exams using a dynamic analysis approach, (b) to investigate if and how hypersynchrony interferes with memory performance, and (c) to describe its relationship with gray and white matter connectivity. Florbetapir-F18 PET and task-free 3T functional and structural MRI were acquired in 47 CN (age = 70.6 ± 6.6), 17 were amyloid pos (florbetapir SUVR >1.11). A parcellation scheme encompassing 382 regions of interest was used to extract regional gray matter volumes, FA-weighted fiber tracts and regional BOLD signals. Graph analysis was used to characterize the gray matter atrophy profile and the white matter connectivity of each subject. The fMRI data was processed using a combination of sliding windows, graph and hierarchical cluster analysis. Each activity cluster was characterized by identifying strength dispersion (difference between pos and neg strength) their maximal and minimal pos and neg strength rois and by investigating their distribution and association with memory performance and gray and white matter connectivity using spearman rank correlations (FDR p < 0.05). The cluster analysis identified eight different activity clusters. Cluster 8 was characterized by the largest strength dispersion indicating hypersynchrony. Its duration/subject was positively correlated with amyloid load (r = 0.42, p = 0.03) and negatively with memory performance (CVLT delayed recall r = -0.39 p = 0.04). The assessment of the regional strength distribution indicated a functional disconnection between mesial temporal structures and the rest of the brain. White matter connectivity was increased in left lateral and mesial temporal lobe and was positively correlated with strength dispersion in the cross-modality analysis suggesting that it enables widespread hypersynchrony. In contrast, precuneus, gray matter connectivity was decreased in the right fusiform gyrus and negatively correlated with high degrees of strength dispersion suggesting that progressing gray matter atrophy could prevent the generation of paroxysmal hypersynchrony in later stages of the disease.
Collapse
Affiliation(s)
- Susanne G Mueller
- Center for Imaging of Neurodegenerative Diseases, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Michael W Weiner
- Center for Imaging of Neurodegenerative Diseases, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
19
|
Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in Aβ-Bearing Mice. J Neurosci 2017; 37:6132-6148. [PMID: 28559377 DOI: 10.1523/jneurosci.0877-17.2017] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 12/25/2022] Open
Abstract
Hyperexcitable neuronal networks are mechanistically linked to the pathologic and clinical features of Alzheimer's disease (AD). Astrocytes are a primary defense against hyperexcitability, but their functional phenotype during AD is poorly understood. Here, we found that activated astrocytes in the 5xFAD mouse model were strongly associated with proteolysis of the protein phosphatase calcineurin (CN) and the elevated expression of the CN-dependent transcription factor nuclear factor of activated T cells 4 (NFAT4). Intrahippocampal injections of adeno-associated virus vectors containing the astrocyte-specific promoter Gfa2 and the NFAT inhibitory peptide VIVIT reduced signs of glutamate-mediated hyperexcitability in 5xFAD mice, measured in vivo with microelectrode arrays and ex vivo brain slices, using whole-cell voltage clamp. VIVIT treatment in 5xFAD mice led to increased expression of the astrocytic glutamate transporter GLT-1 and to attenuated changes in dendrite morphology, synaptic strength, and NMDAR-dependent responses. The results reveal astrocytic CN/NFAT4 as a key pathologic mechanism for driving glutamate dysregulation and neuronal hyperactivity during AD.SIGNIFICANCE STATEMENT Neuronal hyperexcitability and excitotoxicity are increasingly recognized as important mechanisms for neurodegeneration and dementia associated with Alzheimer's disease (AD). Astrocytes are profoundly activated during AD and may lose their capacity to regulate excitotoxic glutamate levels. Here, we show that a highly active calcineurin (CN) phosphatase fragment and its substrate transcription factor, nuclear factor of activated T cells (NFAT4), appear in astrocytes in direct proportion to the extent of astrocyte activation. The blockade of astrocytic CN/NFAT signaling in a common mouse model of AD, using adeno-associated virus vectors normalized glutamate signaling dynamics, increased astrocytic glutamate transporter levels and alleviated multiple signs of neuronal hyperexcitability. The results suggest that astrocyte activation drives hyperexcitability during AD through a mechanism involving aberrant CN/NFAT signaling and impaired glutamate transport.
Collapse
|
20
|
Aso E, Andrés-Benito P, Ferrer I. Delineating the Efficacy of a Cannabis-Based Medicine at Advanced Stages of Dementia in a Murine Model. J Alzheimers Dis 2016; 54:903-912. [DOI: 10.3233/jad-160533] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Ester Aso
- Institut de Neuropatologia, Servei d’Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, L’Hospitalet de Llobregat, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Spain
| | - Pol Andrés-Benito
- Institut de Neuropatologia, Servei d’Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, L’Hospitalet de Llobregat, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Spain
| | - Isidro Ferrer
- Institut de Neuropatologia, Servei d’Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, L’Hospitalet de Llobregat, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Spain
| |
Collapse
|
21
|
Maatuf Y, Stern EA, Slovin H. Abnormal Population Responses in the Somatosensory Cortex of Alzheimer's Disease Model Mice. Sci Rep 2016; 6:24560. [PMID: 27079783 PMCID: PMC4832196 DOI: 10.1038/srep24560] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 03/31/2016] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. One of the neuropathological hallmarks of AD is the accumulation of amyloid-β plaques. Overexpression of human amyloid precursor protein in transgenic mice induces hippocampal and neocortical amyloid-β accumulation and plaque deposition that increases with age. The impact of these effects on neuronal population responses and network activity in sensory cortex is not well understood. We used Voltage Sensitive Dye Imaging, to investigate at high spatial and temporal resolution, the sensory evoked population responses in the barrel cortex of aged transgenic (Tg) mice and of age-matched non-transgenic littermate controls (Ctrl) mice. We found that a whisker deflection evoked abnormal sensory responses in the barrel cortex of Tg mice. The response amplitude and the spatial spread of the cortical responses were significantly larger in Tg than in Ctrl mice. At the network level, spontaneous activity was less synchronized over cortical space than in Ctrl mice, however synchronization during evoked responses induced by whisker deflection did not differ between the two groups. Thus, the presence of elevated Aβ and plaques may alter population responses and disrupts neural synchronization in large-scale networks, leading to abnormalities in sensory processing.
Collapse
Affiliation(s)
- Yossi Maatuf
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002 Israel
| | - Edward A Stern
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002 Israel.,MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Hamutal Slovin
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002 Israel
| |
Collapse
|
22
|
Ferreri F, Vecchio F, Vollero L, Guerra A, Petrichella S, Ponzo D, Määtta S, Mervaala E, Könönen M, Ursini F, Pasqualetti P, Iannello G, Rossini PM, Di Lazzaro V. Sensorimotor cortex excitability and connectivity in Alzheimer's disease: A TMS-EEG Co-registration study. Hum Brain Mapp 2016; 37:2083-96. [PMID: 26945686 DOI: 10.1002/hbm.23158] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 02/13/2016] [Accepted: 02/17/2016] [Indexed: 12/27/2022] Open
Abstract
Several studies have shown that, in spite of the fact that motor symptoms manifest late in the course of Alzheimer's disease (AD), neuropathological progression in the motor cortex parallels that in other brain areas generally considered more specific targets of the neurodegenerative process. It has been suggested that motor cortex excitability is enhanced in AD from the early stages, and that this is related to disease's severity and progression. To investigate the neurophysiological hallmarks of motor cortex functionality in early AD we combined transcranial magnetic stimulation (TMS) with electroencephalography (EEG). We demonstrated that in mild AD the sensorimotor system is hyperexcitable, despite the lack of clinically evident motor manifestations. This phenomenon causes a stronger response to stimulation in a specific time window, possibly due to locally acting reinforcing circuits, while network activity and connectivity is reduced. These changes could be interpreted as a compensatory mechanism allowing for the preservation of sensorimotor programming and execution over a long period of time, regardless of the disease's progression. Hum Brain Mapp 37:2083-2096, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Florinda Ferreri
- Department of Neurology, University Campus Biomedico, Rome, Italy.,Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Fabrizio Vecchio
- Brain Connectivity Laboratory, IRCCS S. Raffaele-Pisana, Rome, Italy
| | - Luca Vollero
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, Rome, Italy
| | - Andrea Guerra
- Department of Neurology, University Campus Biomedico, Rome, Italy
| | - Sara Petrichella
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, Rome, Italy
| | - David Ponzo
- Department of Neurology, University Campus Biomedico, Rome, Italy.,Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Sara Määtta
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Esa Mervaala
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Mervi Könönen
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland.,Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Francesca Ursini
- Department of Neurology, University Campus Biomedico, Rome, Italy
| | - Patrizio Pasqualetti
- Brain Connectivity Laboratory, IRCCS S. Raffaele-Pisana, Rome, Italy.,AFaR Division, Service of Medical Statistics and Information Technology, Fatebenefratelli Foundation for Health Research and Education, Rome, Italy
| | - Giulio Iannello
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, Rome, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, IRCCS S. Raffaele-Pisana, Rome, Italy.,Institute of Neurology, Department of Geriatrics, Neurosciences, Orthopaedics, Policlinic a. Gemelli, Catholic University, Rome, Italy
| | | |
Collapse
|
23
|
Beas BS, McQuail JA, Ban Uelos C, Setlow B, Bizon JL. Prefrontal cortical GABAergic signaling and impaired behavioral flexibility in aged F344 rats. Neuroscience 2016; 345:274-286. [PMID: 26873002 DOI: 10.1016/j.neuroscience.2016.02.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 02/07/2023]
Abstract
The prefrontal cortex (PFC) is critical for the ability to flexibly adapt established patterns of behavior in response to a change in environmental contingencies. Impaired behavioral flexibility results in maladaptive strategies such as perseveration on response options that no longer produce a desired outcome. Pharmacological manipulations of prefrontal cortical GABAergic signaling modulate behavioral flexibility in animal models, and prefrontal cortical interneuron dysfunction is implicated in impaired behavioral flexibility that accompanies neuropsychiatric disease. As deficits in behavioral flexibility also emerge during the normal aging process, the goal of this study was to determine the role of GABAergic signaling, specifically via prefrontal cortical GABA(B) receptors, in such age-related deficits. Young and aged rats were trained in a set shifting task performed in operant chambers. First, rats learned to discriminate between two response levers to obtain a food reward on the basis of a cue light illuminated above the correct lever. Upon acquisition of this initial discrimination, the contingencies were shifted such that rats had to ignore the cue light and respond on the levers according to their left/right positions. Both young and aged rats acquired the initial discrimination similarly; however, aged rats were impaired relative to young following the set shift. Among aged rats, GABA(B) receptor expression in the medial prefrontal cortex (mPFC) was strongly correlated with set shifting, such that lower expression was associated with worse performance. Subsequent experiments showed that intra-mPFC administration of the GABA(B) receptor agonist baclofen enhanced set shifting performance in aged rats. These data directly link GABAergic signaling via GABA(B) receptors to impaired behavioral flexibility associated with normal aging.
Collapse
Affiliation(s)
- B S Beas
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - J A McQuail
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - C Ban Uelos
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - B Setlow
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, United States; Department of Psychology, University of Florida, Gainesville, FL, United States.
| | - J L Bizon
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, United States.
| |
Collapse
|
24
|
Beker S, Kellner V, Chechik G, Stern EA. Learning to classify neural activity from a mouse model of Alzheimer's disease amyloidosis versus controls. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2016; 2:39-48. [PMID: 27239535 PMCID: PMC4879657 DOI: 10.1016/j.dadm.2016.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The mechanisms underlying Alzheimer's disease (AD) onset and progression are not yet elucidated. The extent to which alterations in the activity of individual neurons of an AD model are significant, and the phase at which they can be captured, point to the intensity of the pathology and imply the stage at which it can be detected. Using a machine-learning algorithm, we present a successful cell-by-cell classification of intracellularly recorded neurons from the B6C3 APPswe/PS1dE9 AD model, versus wildtypes controls, at both a late stage and at an early stage, when the plaque pathology and behavioral deficits are absent or rare. These results suggest that the deficits present in neuronal networks of both old and young transgenic animals are large enough to be apparent at the level of individual neurons, and that the pathology could be detected in nearly any given sample, even before pathologic signs.
Collapse
Affiliation(s)
- Shlomit Beker
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Vered Kellner
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Gal Chechik
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Edward A. Stern
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- MassGeneral Institute for Neurological Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
25
|
Antiepileptic drugs as a new therapeutic concept for the prevention of cognitive impairment and Alzheimer’s disease. Recent advances. JOURNAL OF EPILEPTOLOGY 2015. [DOI: 10.1515/joepi-2015-0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
SummaryIntroduction.Excessive accumulation of amyloid-beta (Aβ) peptides in the brain results initially in mild cognitive impairment (MCI) and finally in Alzheimer’s disease (AD). Evidences from experimental and clinical studies show that pathological hyperexcitability of hippocampal neurons is a very early functional impairment observed in progressive memory dysfunctions. Therefore, antiepileptic drugs (AEDs) whose mechanism of action is aimed at inhibition of such neuronal hyperexcitability, seems to be an rationale choice for MCI and AD treatment.Aim.To provide data from experimental and clinical studies on: 1. The unfavorable impact of neuronal hyperexcitability, mainly within the hippocampus, on cognitive processes. 2. Efficacy of AEDs against such abnormally elevated neuronal activity for the prevention of progressive cognitive impairment.Methods.A literature review of publications published within the last fifteen years, was conducted using the PubMed database.Review.The authors describe Aβ-induced hyperexcitability of hippocampal nerve cells as the cause of cognitive deficits, the connection of such activity with an increased risk of seizures and epilepsy in patients with MCI/AD, and finally the efficacy of AEDs: valproic acid (VPA), phenytoin (PHT), topiramate (TPM), lamotrigine (LTG), ethosuximide (ESM) and levetiracetam (LEV) in the prevention of cognitive impairment in experimental models and patients with MCI/AD.Conclusions.The majority of the studied AEDs improve cognitive dysfunction in various experimental models of Aβ-induced brain pathology with accompanied neuronal hyperexcitability. The promising results achieved for LEV in animal models of cognitive impairment were also confirmed in patients with MCI/AD. LEV was well-tolerated and it’s beneficial antidementive effect was confirmed by memory tests and fMRI examination. In conclusion, the use of AEDs could be a novel therapeutic concept for preventing cognitive impairment in patients with Aβ-associated brain pathology.
Collapse
|
26
|
Tamagnini F, Novelia J, Kerrigan TL, Brown JT, Tsaneva-Atanasova K, Randall AD. Altered intrinsic excitability of hippocampal CA1 pyramidal neurons in aged PDAPP mice. Front Cell Neurosci 2015; 9:372. [PMID: 26528126 PMCID: PMC4604241 DOI: 10.3389/fncel.2015.00372] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/07/2015] [Indexed: 12/28/2022] Open
Abstract
Amyloidopathy involves the accumulation of insoluble amyloid β (Aβ) species in the brain's parenchyma and is a key histopathological hallmark of Alzheimer's disease (AD). Work on transgenic mice that overexpress Aβ suggests that elevated Aβ levels in the brain are associated with aberrant epileptiform activity and increased intrinsic excitability (IE) of CA1 hippocampal neurons. In this study we examined if similar changes could be observed in hippocampal CA1 pyramidal neurons from aged PDAPP mice (20-23 month old, Indiana mutation: V717F on APP gene) compared to their age-matched wild-type littermate controls. Whole-cell current clamp recordings revealed that sub-threshold intrinsic properties, such as input resistance, resting membrane potential and hyperpolarization activated "sag" were unaffected, but capacitance was significantly decreased in the transgenic animals. No differences between genotypes were observed in the overall number of action potentials (AP) elicited by 500 ms supra-threshold current stimuli. PDAPP neurons, however, exhibited higher instantaneous firing frequencies after accommodation in response to high intensity current injections. The AP waveform was narrower and shorter in amplitude in PDAPP mice: these changes, according to our in silico model of a CA1/3 pyramidal neuron, depended on the respective increase and reduction of K(+) and Na(+) voltage-gated channels maximal conductances. Finally, the after-hyperpolarization, seen after the first AP evoked by a +300 pA current injection and after 50 Hz AP bursts, was more pronounced in PDAPP mice. These data show that Aβ-overexpression in aged mice altered the capacitance, the neuronal firing and the AP waveform of CA1 pyramidal neurons. Some of these findings are consistent with previous work on younger PDAPP; they also show important differences that can be potentially ascribed to the interaction between amyloidopathy and ageing. Such a change of IE properties over time underlies that the increased incidence of seizure observed in AD patients might rely on different mechanistic pathways during progression of the disease.
Collapse
Affiliation(s)
- Francesco Tamagnini
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - Janet Novelia
- Department of Mathematics, College of Engineering, Mathematics and Physical Sciences, University of Exeter Exeter, UK
| | - Talitha L Kerrigan
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - Jon T Brown
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics, College of Engineering, Mathematics and Physical Sciences, University of Exeter Exeter, UK
| | - Andrew D Randall
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| |
Collapse
|
27
|
Regional cerebral blood flow estimated by early PiB uptake is reduced in mild cognitive impairment and associated with age in an amyloid-dependent manner. Neurobiol Aging 2015; 36:1619-1628. [PMID: 25702957 DOI: 10.1016/j.neurobiolaging.2014.12.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 12/22/2014] [Accepted: 12/26/2014] [Indexed: 12/16/2022]
Abstract
Early uptake of [(11)C]-Pittsburgh Compound B (ePiB, 0-6 minutes) estimates cerebral blood flow. We studied ePiB in 13 PiB-negative and 10 PiB-positive subjects with mild cognitive impairment (MCI, n = 23) and 11 PiB-positive and 74 PiB-negative cognitively healthy elderly control subjects (HCS, n = 85) in 6 bilateral volumes of interest: posterior cingulate cortex (PCC), hippocampus (hipp), temporoparietal region, superior parietal gyrus, parahippocampal gyrus (parahipp), and inferior frontal gyrus (IFG) for the associations with cognitive status, age, amyloid deposition, and apolipoprotein E ε4-allele. We observed no difference in ePiB between PiB-positive and -negative subjects and carriers and noncarriers. EPiB decreased with age in PiB-positive subjects in bilateral superior parietal gyrus, bilateral temporoparietal region, right IFG, right PCC, and left parahippocampal gyrus but not in PiB-negative subjects. MCI had lower ePiB than HCS (left PCC, left IFG, and left and right hipp). Lowest ePiB values were found in MCI of 70 years and older, who also displayed high cortical PiB binding. This suggests that lowered regional cerebral blood flow indicated by ePiB is associated with age in the presence but not in the absence of amyloid pathology.
Collapse
|
28
|
Beker S, Goldin M, Menkes-Caspi N, Kellner V, Chechik G, Stern EA. Amyloid-β disrupts ongoing spontaneous activity in sensory cortex. Brain Struct Funct 2014; 221:1173-88. [PMID: 25523106 DOI: 10.1007/s00429-014-0963-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 12/08/2014] [Indexed: 12/29/2022]
Abstract
UNLABELLED The effect of Alzheimer's disease pathology on activity of individual neocortical neurons in the intact neural network remains obscure. Ongoing spontaneous activity, which constitutes most of neocortical activity, is the background template on which further evoked-activity is superimposed. We compared in vivo intracellular recordings and local field potentials (LFP) of ongoing activity in the barrel cortex of APP/PS1 transgenic mice and age-matched littermate CONTROLS, following significant amyloid-β (Aβ) accumulation and aggregation. We found that membrane potential dynamics of neurons in Aβ-burdened cortex significantly differed from those of nontransgenic CONTROLS durations of the depolarized state were considerably shorter, and transitions to that state frequently failed. The spiking properties of APP/PS1 neurons showed alterations from those of CONTROLS both firing patterns and spike shape were changed in the APP/PS1 group. At the population level, LFP recordings indicated reduced coherence within neuronal assemblies of APP/PS1 mice. In addition to the physiological effects, we show that morphology of neurites within the barrel cortex of the APP/PS1 model is altered compared to CONTROLS. These results are consistent with a process where the effect of Aβ on spontaneous activity of individual neurons amplifies into a network effect, reducing network integrity and leading to a wide cortical dysfunction.
Collapse
Affiliation(s)
- Shlomit Beker
- Gonda Brain Research Center, Bar-Ilan University, 52900, Ramat Gan, Israel
| | - Miri Goldin
- Gonda Brain Research Center, Bar-Ilan University, 52900, Ramat Gan, Israel
| | - Noa Menkes-Caspi
- Gonda Brain Research Center, Bar-Ilan University, 52900, Ramat Gan, Israel
| | - Vered Kellner
- Gonda Brain Research Center, Bar-Ilan University, 52900, Ramat Gan, Israel
| | - Gal Chechik
- Gonda Brain Research Center, Bar-Ilan University, 52900, Ramat Gan, Israel
| | - Edward A Stern
- Gonda Brain Research Center, Bar-Ilan University, 52900, Ramat Gan, Israel.
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|