1
|
Robertson KV, Rodriguez AS, Cartailler JP, Shrestha S, Schleh MW, Schroeder KR, Valenti AM, Kramer AT, Harrison FE, Hasty AH. Knockdown of microglial iron import gene, Slc11a2, worsens cognitive function and alters microglial transcriptional landscape in a sex-specific manner in the APP/PS1 model of Alzheimer's disease. J Neuroinflammation 2024; 21:238. [PMID: 39334471 PMCID: PMC11438269 DOI: 10.1186/s12974-024-03238-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Microglial cell iron load and inflammatory activation are significant hallmarks of late-stage Alzheimer's disease (AD). In vitro, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and excess iron can augment cellular inflammation, suggesting a feed-forward loop between iron import mechanisms and inflammatory signaling. However, it is not understood whether microglial iron import mechanisms directly contribute to inflammatory signaling and chronic disease in vivo. These studies determined the effects of microglial-specific knockdown of Slc11a2 on AD-related cognitive decline and microglial transcriptional phenotype. METHODS In vitro experiments and RT-qPCR were used to assess a role for DMT1 in amyloid-β-associated inflammation. To determine the effects of microglial Slc11a2 knockdown on AD-related phenotypes in vivo, triple-transgenic Cx3cr1Cre-ERT2;Slc11a2flfl;APP/PS1+or - mice were generated and administered corn oil or tamoxifen to induce knockdown at 5-6 months of age. Both sexes underwent behavioral analyses to assess cognition and memory (12-15 months of age). Hippocampal CD11b+ microglia were magnetically isolated from female mice (15-17 months) and bulk RNA-sequencing analysis was conducted. RESULTS DMT1 inhibition in vitro robustly decreased Aβ-induced inflammatory gene expression and cellular iron levels in conditions of excess iron. In vivo, Slc11a2KD APP/PS1 female, but not male, mice displayed a significant worsening of memory function in Morris water maze and a fear conditioning assay, along with significant hyperactivity compared to control WT and APP/PS1 mice. Hippocampal microglia from Slc11a2KD APP/PS1 females displayed significant increases in Enpp2, Ttr, and the iron-export gene, Slc40a1, compared to control APP/PS1 cells. Slc11a2KD cells from APP/PS1 females also exhibited decreased expression of markers associated with subsets of disease-associated microglia (DAMs), such as Apoe, Ctsb, Ly9, Csf1, and Hif1α. CONCLUSIONS This work suggests a sex-specific role for microglial iron import gene Slc11a2 in propagating behavioral and cognitive phenotypes in the APP/PS1 model of AD. These data also highlight an association between loss of a DAM-like phenotype in microglia and cognitive deficits in Slc11a2KD APP/PS1 female mice. Overall, this work illuminates an iron-related pathway in microglia that may serve a protective role during disease and offers insight into mechanisms behind disease-related sex differences.
Collapse
Affiliation(s)
- Katrina Volk Robertson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Alec S Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | | | - Shristi Shrestha
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, TN, USA
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Kyle R Schroeder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Arianna M Valenti
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Alec T Kramer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, 7465 Medical Research Building IV, 2213 Garland Avenue, Nashville, TN, 37232, USA.
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA.
- VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
2
|
Abdollahi Nejat M, Stiedl O, Smit AB, van Kesteren RE. Continuous locomotor activity monitoring to assess animal welfare following intracranial surgery in mice. Front Behav Neurosci 2024; 18:1457894. [PMID: 39296476 PMCID: PMC11408287 DOI: 10.3389/fnbeh.2024.1457894] [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: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
Locomotor activity can serve as a readout to identify discomfort and pain. Therefore, monitoring locomotor activity following interventions that induce potential discomfort may serve as a reliable method for evaluating animal health, complementing conventional methods such as body weight measurement. In this study, we used the digital ventilated cage (DVC®) system for the assessment of circadian locomotor activity, in addition to body weight monitoring, following intracranial stereotaxic surgery in an Alzheimer's disease mouse model (C57BL/6J/APPswe/PSEN1dE9). Stereotaxic surgery did not affect the organization of circadian locomotor activity of both 7-8-week-old and 19-21-week-old mice. However, we observed that both young and old mice exhibited a significant decrease in activity during the dark phase. Also, our study shows that changes in locomotor activity exhibit higher sensitivity in detecting alterations indicative of animal health compared to measuring body weight. In contrast to 7-8-week-old mice, where we observed no genotypic differences in locomotor activity, 19-21-week-old APP/PS1 mice showed increased locomotor activity compared to wild-type mice. Furthermore, our analyses revealed that a subset of the 7-8-week-old mice showed increased locomotor activity during the initial peak of the dark phase. One mouse experienced sudden death early in life, possibly due to epileptic seizures. Altogether, our findings affirm continuous activity measurements as used in the DVC® as a highly valuable objective method for post-surgical welfare monitoring. Its discerning capacity not only facilitates circadian locomotor rhythm assessment but also enables the identification of individual aberrant activity patterns, possibly indicative of epileptic seizures.
Collapse
Affiliation(s)
- Mazyar Abdollahi Nejat
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Oliver Stiedl
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
3
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanjun Liu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chuhui Yin
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yan Zeng
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
4
|
Heuer SE, Bloss EB, Howell GR. Strategies to dissect microglia-synaptic interactions during aging and in Alzheimer's disease. Neuropharmacology 2024; 254:109987. [PMID: 38705570 DOI: 10.1016/j.neuropharm.2024.109987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Age is the largest risk factor for developing Alzheimer's disease (AD), a neurodegenerative disorder that causes a progressive and severe dementia. The underlying cause of cognitive deficits seen in AD is thought to be the disconnection of neural circuits that control memory and executive functions. Insight into the mechanisms by which AD diverges from normal aging will require identifying precisely which cellular events are driven by aging and which are impacted by AD-related pathologies. Since microglia, the brain-resident macrophages, are known to have critical roles in the formation and maintenance of neural circuits through synaptic pruning, they are well-positioned to modulate synaptic connectivity in circuits sensitive to aging or AD. In this review, we provide an overview of the current state of the field and on emerging technologies being employed to elucidate microglia-synaptic interactions in aging and AD. We also discuss the importance of leveraging genetic diversity to study how these interactions are shaped across more realistic contexts. We propose that these approaches will be essential to define specific aging- and disease-relevant trajectories for more personalized therapeutics aimed at reducing the effects of age or AD pathologies on the brain. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Sarah E Heuer
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Erik B Bloss
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| |
Collapse
|
5
|
Zhuang D, Yu N, Han S, Zhang X, Ju C. The Kv7 channel opener Retigabine reduces neuropathology and alleviates behavioral deficits in APP/PS1 transgenic mice. Behav Brain Res 2024; 471:115137. [PMID: 38971432 DOI: 10.1016/j.bbr.2024.115137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Hyperexcitability of neuronal networks is central to the pathogenesis of Alzheimer's disease (AD). Pharmacological activation of Kv7 channels is an effective way to reduce neuronal firing. Our results showed that that pharmacologically activating the Kv7 channel with Retigabine (RTG) can alleviate cognitive impairment in mice without affecting spontaneous activity. RTG could also ameliorate damage to the Nissl bodies in cortex and hippocampal CA and DG regions in 9-month-old APP/PS1 mice. Additionally, RTG could reduce the Aβ plaque number in the hippocampus and cortex of both 6-month-old and 9-month-old mice. By recordings of electroencephalogram, we showed that a decrease in the number of abnormal discharges in the brains of the AD model mice when the Kv7 channel was opened. Moreover, Western blot analysis revealed a reduction in the expression of the p-Tau protein in both the hippocampus and cortex upon Kv7 channel opening. These findings suggest that Kv7 channel opener RTG may ameliorate cognitive impairment in AD, most likely by reducing brain excitability.
Collapse
Affiliation(s)
- Dongpei Zhuang
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Nan Yu
- Department of Pharmacy, Qingdao Eighth People's Hospital, China.
| | - Shuo Han
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Xinyao Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Chuanxia Ju
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| |
Collapse
|
6
|
Lau LA, Zhao Z, Gomperts SN, Staley KJ, Lillis KP. Cellular resolution contributions to ictal population signals. Epilepsia 2024; 65:2165-2178. [PMID: 38752861 PMCID: PMC11251866 DOI: 10.1111/epi.17983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE The increased amplitude of ictal activity is a common feature of epileptic seizures, but the determinants of this amplitude have not been identified. Clinically, ictal amplitudes are measured electrographically (using, e.g., electroencephalography, electrocorticography, and depth electrodes), but these methods do not enable the assessment of the activity of individual neurons. Population signal may increase from three potential sources: (1) increased synchrony (i.e., more coactive neurons); (2) altered active state, from bursts of action potentials and/or paroxysmal depolarizing shifts in membrane potential; and (3) altered subthreshold state, which includes all lower levels of activity. Here, we quantify the fraction of ictal signal from each source. METHODS To identify the cellular determinants of the ictal signal, we measured single cell and population electrical activity and neuronal calcium levels via optical imaging of the genetically encoded calcium indicator (GECI) GCaMP. Spontaneous seizure activity was assessed with microendoscopy in an APP/PS1 mouse with focal cortical injury and via widefield imaging in the organotypic hippocampal slice cultures (OHSCs) model of posttraumatic epilepsy. Single cell calcium signals were linked to a range of electrical activities by performing simultaneous GECI-based calcium imaging and whole-cell patch-clamp recordings in spontaneously seizing OHSCs. Neuronal resolution calcium imaging of spontaneous seizures was then used to quantify the cellular contributions to population-level ictal signal. RESULTS The seizure onset signal was primarily driven by increased subthreshold activity, consistent with either barrages of excitatory postsynaptic potentials or sustained membrane depolarization. Unsurprisingly, more neurons entered the active state as seizure activity progressed. However, the increasing fraction of active cells was primarily driven by synchronous reactivation and not from continued recruitment of new populations of neurons into the seizure. SIGNIFICANCE This work provides a critical link between single neuron activity and population measures of seizure activity.
Collapse
Affiliation(s)
- Lauren A. Lau
- Massachusetts General Hospital, Department of Neurology, Boston, Massachusetts 02114, Harvard Medical School, Boston, Massachusetts 02115
| | - Zhuoyang Zhao
- Massachusetts General Hospital, Department of Neurology, Boston, Massachusetts 02114, Harvard Medical School, Boston, Massachusetts 02115
| | - Stephen N. Gomperts
- Massachusetts General Hospital, Department of Neurology, Boston, Massachusetts 02114, Harvard Medical School, Boston, Massachusetts 02115
| | - Kevin J. Staley
- Massachusetts General Hospital, Department of Neurology, Boston, Massachusetts 02114, Harvard Medical School, Boston, Massachusetts 02115
| | - Kyle P. Lillis
- Massachusetts General Hospital, Department of Neurology, Boston, Massachusetts 02114, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
7
|
Robertson KV, Rodriguez AS, Cartailler JP, Shrestha S, Schroeder KR, Valenti AM, Harrison FE, Hasty AH. Knockdown of microglial iron import gene, DMT1, worsens cognitive function and alters microglial transcriptional landscape in a sex-specific manner in the APP/PS1 model of Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-4559940. [PMID: 38978579 PMCID: PMC11230470 DOI: 10.21203/rs.3.rs-4559940/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: 07/10/2024]
Abstract
Background Microglial cell iron load and inflammatory activation are significant hallmarks of late-stage Alzheimer's disease (AD). In vitro, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and excess iron can augment cellular inflammation, suggesting a feed-forward loop between iron import mechanisms and inflammatory signaling. However, it is not understood whether microglial iron import mechanisms directly contribute to inflammatory signaling and chronic disease in vivo. These studies determined the effects of microglial-specific knockdown of Slc11a2 on AD-related cognitive decline and microglial transcriptional phenotype. Methods In vitro experiments and RT-qPCR were used to assess a role for DMT1 in amyloid-β-associated inflammation. To determine the effects of microglial Slc11a2 knockdown on AD-related phenotypes in vivo, triple-transgenic Cx3cr1 Cre - ERT2 ;Slc11a2 flfl;APP/PS1 + or - mice were generated and administered corn oil or tamoxifen to induce knockdown at 5-6 months of age. Both sexes underwent behavioral analyses to assess cognition and memory (12-15 months of age). Hippocampal CD11b + microglia were magnetically isolated from female mice (15-17 months) and bulk RNA-sequencing analysis was conducted. Results DMT1 inhibition in vitro robustly decreased Aβ-induced inflammatory gene expression and cellular iron levels in conditions of excess iron. In vivo, Slc11a2 KD APP/PS1 female, but not male, mice displayed a significant worsening of memory function in Morris water maze and a fear conditioning assay, along with significant hyperactivity compared to control WT and APP/PS1 mice. Hippocampal microglia from Slc11a2 KD APP/PS1 females displayed significant increases in Enpp2, Ttr, and the iron-export gene, Slc40a1, compared to control APP/PS1 cells. Slc11a2 KD cells from APP/PS1 females also exhibited decreased expression of markers associated with disease-associated microglia (DAMs), such as Apoe, Ctsb, Csf1, and Hif1α. Conclusions This work suggests a sex-specific role for microglial iron import gene Slc11a2 in propagating behavioral and cognitive phenotypes in the APP/PS1 model of AD. These data also highlight an association between loss of a DAM-like phenotype in microglia and cognitive deficits in Slc11a2 KD APP/PS1 female mice. Overall, this work illuminates an iron-related pathway in microglia that may serve a protective role during disease and offers insight into mechanisms behind disease-related sex differences.
Collapse
|
8
|
Yook Y, Lee KY, Kim E, Lizarazo S, Yu X, Tsai NP. Hyperfunction of post-synaptic density protein 95 promotes seizure response in early-stage aβ pathology. EMBO Rep 2024; 25:1233-1255. [PMID: 38413732 PMCID: PMC10933348 DOI: 10.1038/s44319-024-00090-0] [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: 05/30/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
Accumulation of amyloid-beta (Aβ) can lead to the formation of aggregates that contribute to neurodegeneration in Alzheimer's disease (AD). Despite globally reduced neural activity during AD onset, recent studies have suggested that Aβ induces hyperexcitability and seizure-like activity during the early stages of the disease that ultimately exacerbate cognitive decline. However, the underlying mechanism is unknown. Here, we reveal an Aβ-induced elevation of postsynaptic density protein 95 (PSD-95) in cultured neurons in vitro and in an in vivo AD model using APP/PS1 mice at 8 weeks of age. Elevation of PSD-95 occurs as a result of reduced ubiquitination caused by Akt-dependent phosphorylation of E3 ubiquitin ligase murine-double-minute 2 (Mdm2). The elevation of PSD-95 is consistent with the facilitation of excitatory synapses and the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors induced by Aβ. Inhibition of PSD-95 corrects these Aβ-induced synaptic defects and reduces seizure activity in APP/PS1 mice. Our results demonstrate a mechanism underlying elevated seizure activity during early-stage Aβ pathology and suggest that PSD-95 could be an early biomarker and novel therapeutic target for AD.
Collapse
Affiliation(s)
- Yeeun Yook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Eunyoung Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Simon Lizarazo
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xinzhu Yu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
9
|
Mao R, Hu M, Liu X, Ye L, Xu B, Sun M, Xu S, Shao W, Tan Y, Xu Y, Bai F, Shu S. Impairments of GABAergic transmission in hippocampus mediate increased susceptibility of epilepsy in the early stage of Alzheimer's disease. Cell Commun Signal 2024; 22:147. [PMID: 38388921 PMCID: PMC10885444 DOI: 10.1186/s12964-024-01528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Patients with Alzheimer's disease (AD) are often co-morbid with unprovoked seizures, making clinical diagnosis and management difficult. Although it has an important role in both AD and epilepsy, abnormal γ-aminobutyric acid (GABA)ergic transmission is recognized only as a compensative change for glutamatergic damage. Neuregulin 1 (NRG1)-ErbB4 signaling can promote GABA release and suppress epileptogenesis, but its effects on cognition in AD are still controversial. METHODS Four-month-old APPswe/PS1dE9 mice (APP mice) were used as animal models in the early stage of AD in this study. Acute/chronic chemical-kindling epilepsy models were established with pentylenetetrazol. Electroencephalogram and Racine scores were performed to assess seizures. Behavioral tests were used to assess cognition and emotion. Electrophysiology, western blot and immunofluorescence were performed to detect the alterations in synapses, GABAergic system components and NRG1-ErbB4 signaling. Furthermore, NRG1 was administrated intracerebroventricularly into APP mice and then its antiepileptic and cognitive effects were evaluated. RESULTS APP mice had increased susceptibility to epilepsy and resulting hippocampal synaptic damage and cognitive impairment. Electrophysiological analysis revealed decreased GABAergic transmission in the hippocampus. This abnormal GABAergic transmission involved a reduction in the number of parvalbumin interneurons (PV+ Ins) and decreased levels of GABA synthesis and transport. We also found impaired NRG1-ErbB4 signaling which mediated by PV+ Ins loss. And NRG1 administration could effectively reduce seizures and improve cognition in four-month-old APP mice. CONCLUSION Our results indicated that abnormal GABAergic transmission mediated hippocampal hyperexcitability, further excitation/inhibition imbalance, and promoted epileptogenesis in the early stage of AD. Appropriate NRG1 administration could down-regulate seizure susceptibility and rescue cognitive function. Our study provided a potential direction for intervening in the co-morbidity of AD and epilepsy.
Collapse
Affiliation(s)
- Rui Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Mengsha Hu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Lei Ye
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Bingsong Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Wenxuan Shao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yi Tan
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, China.
- Nanjing Neurology Medical Center, Nanjing, China.
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, China.
- Nanjing Neurology Medical Center, Nanjing, China.
| | - Shu Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, China.
- Nanjing Neurology Medical Center, Nanjing, China.
| |
Collapse
|
10
|
Yao AY, Halloran PJ, Ge Y, Singh N, Zhou J, Galske J, He W, Yan R, Hu X. Bace1 Deletion in the Adult Reverses Epileptiform Activity and Sleep-wake Disturbances in AD Mice. J Neurosci 2023; 43:6197-6211. [PMID: 37536983 PMCID: PMC10476643 DOI: 10.1523/jneurosci.2124-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023] Open
Abstract
Alzheimer's disease (AD) increases the risk for seizures and sleep disorders. We show here that germline deletion of β-site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) in neurons, but not in astrocytes, increased epileptiform activity. However, Bace1 deletion at adult ages did not alter the normal EEG waveform, indicating less concern for BACE1 inhibition in patients. Moreover, we showed that deletion of Bace1 in the adult was able to reverse epileptiform activity in 5xFAD mice. Intriguingly, treating 5xFAD and APPNL-G-F/NL-G-F (APP KI) mice of either sex with one BACE1 inhibitor Lanabecestat (AZD3293) dramatically increased epileptiform spiking, likely resulting from an off-target effect. We also monitored sleep-wake pathologies in these mice and showed increased wakefulness, decreased non-rapid eye movement sleep, and rapid eye movement sleep in both 5xFAD and APP KI mice; BACE1 inhibition in the adult 5xFAD mice reversed plaque load and sleep disturbances, but this was not seen in APP KI mice. Further studies with and without BACE1 inhibitor treatment showed different levels of plaque-associated microgliosis and activated microglial proteins in 5xFAD mice compared with APP KI mice. Together, BACE1 inhibition should be developed to avoid off-target effect for achieving benefits in reducing epileptic activity and sleep disturbance in Alzheimer's patients.SIGNIFICANCE STATEMENT BACE1 is widely recognized as a therapeutic target for treating Alzheimer's disease patients. However, BACE1 inhibitors failed in clinical trials because of inability to show cognitive improvement in patients. Here we show that BACE1 inhibition actually reduces sleep disturbances and epileptic seizures; both are seen in AD patients. We further showed that one of clinically tested BACE1 inhibitors does have off-target effects, and development of safer BACE1 inhibitors will be beneficial to AD patients. Results from this study will provide useful guidance for additional drug development.
Collapse
Affiliation(s)
- Annie Y Yao
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Patrick J Halloran
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Yingying Ge
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Neeraj Singh
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - John Zhou
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - James Galske
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Wanxia He
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Xiangyou Hu
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| |
Collapse
|
11
|
Del Pozo A, Knox KM, Lehmann L, Davidson S, Rho S, Jayadev S, Barker-Haliski M. Chronic evoked seizures in young pre-symptomatic APP/PS1 mice induce serotonin changes and accelerate onset on Alzheimer's disease-related neurpathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522897. [PMID: 36711965 PMCID: PMC9881977 DOI: 10.1101/2023.01.05.522897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE People with early-onset Alzheimer's disease (AD) are at elevated seizure risk. Further, chronic seizures in pre-symptomatic stages may disrupt serotonin pathway-related protein expression, precipitating the onset of AD-related pathology and burden of neuropsychiatric comorbidities. METHODS 2-3-month-old APP/PS1, PSEN2-N141I, and transgenic control mice were sham or corneal kindled for 2 weeks to model chronic seizures. Seizure-induced changes in glia, serotonin pathway proteins, and amyloid beta; levels in hippocampus and prefrontal cortex were quantified. RESULTS APP/PS1 mice experienced worsened mortality versus kindled Tg- controls. APP/PS1 females were also more susceptible to chronic kindled seizures. These changes correlated with a marked downregulation of hippocampal tryptophan hydroxylase 2 and monoamine oxidase A protein expression compared to controls; these changes were not detected in PSEN2-N141I mice. Kindled APP/PS1 mice exhibited amyloid beta; overexpression and glial overactivity without plaque deposition. PSEN2 protein expression was AD model-dependent. SIGNIFICANCE Seizures evoked in pre-symptomatic APP/PS1 mice promotes premature mortality in the absence of pathological amyloid deposition. Disruptions in serotonin pathway metabolism are associated with increased glial reactivity and PSEN2 downregulation without amyloid beta; deposition. This study provides the first direct evidence that seizures occurring prior to amyloid beta, plaque accumulation worsen disease burden in an AD genotype-specific manner.
Collapse
|
12
|
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
|
13
|
Soula M, Maslarova A, Harvey RE, Valero M, Brandner S, Hamer H, Fernández-Ruiz A, Buzsáki G. Interictal epileptiform discharges affect memory in an Alzheimer's Disease mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528683. [PMID: 36824810 PMCID: PMC9949089 DOI: 10.1101/2023.02.15.528683] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Interictal epileptiform discharges (IEDs) are transient abnormal electrophysiological events commonly observed in epilepsy patients but are also present in other neurological disease, such as Alzheimer's Disease (AD). Understanding the role IEDs have on the hippocampal circuit is important for our understanding of the cognitive deficits seen in epilepsy and AD. We characterize and compare the IEDs of human epilepsy patients from microwire hippocampal recording with those of AD transgenic mice with implanted multi-layer hippocampal silicon probes. Both the local field potential features and firing patterns of pyramidal cells and interneurons were similar in mouse and human. We found that as IEDs emerged from the CA3-1 circuits, they recruited pyramidal cells and silenced interneurons, followed by post-IED suppression. IEDs suppressed the incidence and altered the properties of physiological sharp-wave ripples (SPW-Rs), altered their physiological properties, and interfered with the replay of place field sequences in a maze. In addition, IEDs in AD mice inversely correlated with daily memory performance. Together, our work implicates that IEDs may present a common and epilepsy-independent phenomenon in neurodegenerative diseases that perturbs hippocampal-cortical communication and interferes with memory. Significant Statement Prevalence of neurodegenerative diseases and the number of people with dementia is increasing steadily. Therefore, novel treatment strategies for learning and memory disorders are urgently necessary. IEDs, apart from being a surrogate for epileptic brain regions, have also been linked to cognitive decline. Here we report that IEDs in human epilepsy patients and AD mouse models have similar local field potential characteristics and associated firing patterns of pyramidal cells and interneurons. Mice with more IEDs displayed fewer hippocampal SPW-Rs, poorer replay of spatial trajectories, and decreased memory performance. IED suppression is an unexplored target to treat cognitive dysfunction in neurodegenerative diseases.
Collapse
|
14
|
Modifiable risk factors of dementia linked to excitation-inhibition imbalance. Ageing Res Rev 2023; 83:101804. [PMID: 36410620 DOI: 10.1016/j.arr.2022.101804] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/04/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Recent evidence identifies 12 potentially modifiable risk factors for dementia to which 40% of dementia cases are attributed. While the recognition of these risk factors has paved the way for the development of new prevention measures, the link between these risk factors and the underlying pathophysiology of dementia is yet not well understood. A growing number of recent clinical and preclinical studies support a role of Excitation-Inhibition (E-I) imbalance in the pathophysiology of dementia. In this review, we aim to propose a conceptual model on the links between the modifiable risk factors and the E-I imbalance in dementia. This model, which aims to address the current gap in the literature, is based on 12 mediating common mechanisms: the hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neuroinflammation, oxidative stress, mitochondrial dysfunction, cerebral hypo-perfusion, blood-brain barrier (BBB) dysfunction, beta-amyloid deposition, elevated homocysteine level, impaired neurogenesis, tau tangles, GABAergic dysfunction, and glutamatergic dysfunction. We believe this model serves as a framework for future studies in this field and facilitates future research on dementia prevention, discovery of new biomarkers, and developing new interventions.
Collapse
|
15
|
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: 7] [Impact Index Per Article: 3.5] [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
|
16
|
Vande Vyver M, Barker‐Haliski M, Aourz N, Nagels G, Bjerke M, Engelborghs S, De Bundel D, Smolders I. Higher susceptibility to 6 Hz corneal kindling and lower responsiveness to antiseizure drugs in mouse models of Alzheimer's disease. Epilepsia 2022; 63:2703-2715. [PMID: 35775150 PMCID: PMC9804582 DOI: 10.1111/epi.17355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Epileptic spikes and seizures seem present early in the disease process of Alzheimer's disease (AD). However, it is unclear how soluble and insoluble amyloid beta (Aβ) and tau proteins affect seizure development in vivo. We aim to contribute to this field by assessing the vulnerability to 6 Hz corneal kindling of young female mice from two well-characterized transgenic AD models and by testing their responsiveness to selected antiseizure drugs (ASDs). METHODS We used 7-week-old triple transgenic (3xTg) mice that have both amyloid and tau mutations, and amyloid precursor protein Swedish/presenillin 1 dE9 (APP/PS1) mice, bearing only amyloid-related mutations. We assessed the absence of plaques via immunohistochemistry and analyzed the concentrations of both soluble and insoluble forms of Aβ1-42 and total tau (t-tau) in brain hippocampal and prefrontal cortical tissue. Seven-week-old mice of the different genotypes were subjected to the 6 Hz corneal kindling model. After kindling acquisition, we tested the anticonvulsant effects of three marketed ASDs (levetiracetam, brivaracetam, and lamotrigine) in fully kindled mice. RESULTS No Aβ plaques were present in either genotype. Soluble Aβ1-42 levels were increased in both AD genotypes, whereas insoluble Aβ1-42 concentrations were only elevated in APP/PS1 mice compared with their respective controls. Soluble and insoluble forms of t-tau were increased in 3xTg mice only. 3xTg and APP/PS1 mice displayed more severe seizures induced by 6 Hz corneal kindling from the first stimulation onward and were more rapidly kindled compared with control mice. In fully kindled AD mice, ASDs had less-pronounced anticonvulsive effects compared with controls. SIGNIFICANCE Mutations increasing Aβ only or both Aβ and tau in the brain enhance susceptibility for seizures and kindling in mice. The effect of ASDs on seizures measured by the Racine scale is less pronounced in both investigated AD models and suggests that seizures of young AD mice are more difficult to treat.
Collapse
Affiliation(s)
- Maxime Vande Vyver
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium,Department of NeurologyUniversitair Ziekenhuis BrusselJetteBelgium,Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium,Neuroprotection and Neuromodulation (NEUR), Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium
| | | | - Najat Aourz
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium
| | - Guy Nagels
- Department of NeurologyUniversitair Ziekenhuis BrusselJetteBelgium,Department of AI Supported Modelling in Clinical Sciences (AIMS)Vrije Universiteit BrusselBrusselsBelgium
| | - Maria Bjerke
- Neuroprotection and Neuromodulation (NEUR), Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium,Department of NeurochemistryUniversitair Ziekenhuis BrusselBrusselsBelgium
| | - Sebastiaan Engelborghs
- Department of NeurologyUniversitair Ziekenhuis BrusselJetteBelgium,Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium,Neuroprotection and Neuromodulation (NEUR), Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for NeurosciencesVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
17
|
Martinsson I, Quintino L, Garcia MG, Konings SC, Torres-Garcia L, Svanbergsson A, Stange O, England R, Deierborg T, Li JY, Lundberg C, Gouras GK. Aβ/Amyloid Precursor Protein-Induced Hyperexcitability and Dysregulation of Homeostatic Synaptic Plasticity in Neuron Models of Alzheimer’s Disease. Front Aging Neurosci 2022; 14:946297. [PMID: 35928998 PMCID: PMC9344931 DOI: 10.3389/fnagi.2022.946297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is increasingly seen as a disease of synapses and diverse evidence has implicated the amyloid-β peptide (Aβ) in synapse damage. The molecular and cellular mechanism(s) by which Aβ and/or its precursor protein, the amyloid precursor protein (APP) can affect synapses remains unclear. Interestingly, early hyperexcitability has been described in human AD and mouse models of AD, which precedes later hypoactivity. Here we show that neurons in culture with either elevated levels of Aβ or with human APP mutated to prevent Aβ generation can both induce hyperactivity as detected by elevated calcium transient frequency and amplitude. Since homeostatic synaptic plasticity (HSP) mechanisms normally maintain a setpoint of activity, we examined whether HSP was altered in AD transgenic neurons. Using methods known to induce HSP, we demonstrate that APP protein levels are regulated by chronic modulation of activity and that AD transgenic neurons have an impaired adaptation of calcium transients to global changes in activity. Further, AD transgenic compared to WT neurons failed to adjust the length of their axon initial segments (AIS), an adaptation known to alter excitability. Thus, we show that both APP and Aβ influence neuronal activity and that mechanisms of HSP are disrupted in primary neuron models of AD.
Collapse
Affiliation(s)
- Isak Martinsson
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Experimental Neuroinflammation Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- *Correspondence: Isak Martinsson,
| | - Luis Quintino
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Megg G. Garcia
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Experimental Neuroinflammation Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sabine C. Konings
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Laura Torres-Garcia
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Neural Plasticity and Repair, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Alexander Svanbergsson
- Neural Plasticity and Repair, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oliver Stange
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rebecca England
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jia-Yi Li
- Neural Plasticity and Repair, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cecilia Lundberg
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Gunnar K. Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Gunnar K. Gouras,
| |
Collapse
|
18
|
B. Szabo A, Cretin B, Gérard F, Curot J, J. Barbeau E, Pariente J, Dahan L, Valton L. Sleep: The Tip of the Iceberg in the Bidirectional Link Between Alzheimer's Disease and Epilepsy. Front Neurol 2022; 13:836292. [PMID: 35481265 PMCID: PMC9035794 DOI: 10.3389/fneur.2022.836292] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The observation that a pathophysiological link might exist between Alzheimer's disease (AD) and epilepsy dates back to the identification of the first cases of the pathology itself and is now strongly supported by an ever-increasing mountain of literature. An overwhelming majority of data suggests not only a higher prevalence of epilepsy in Alzheimer's disease compared to healthy aging, but also that AD patients with a comorbid epileptic syndrome, even subclinical, have a steeper cognitive decline. Moreover, clinical and preclinical investigations have revealed a marked sleep-related increase in the frequency of epileptic activities. This characteristic might provide clues to the pathophysiological pathways underlying this comorbidity. Furthermore, the preferential sleep-related occurrence of epileptic events opens up the possibility that they might hasten cognitive decline by interfering with the delicately orchestrated synchrony of oscillatory activities implicated in sleep-related memory consolidation. Therefore, we scrutinized the literature for mechanisms that might promote sleep-related epileptic activity in AD and, possibly dementia onset in epilepsy, and we also aimed to determine to what degree and through which processes such events might alter the progression of AD. Finally, we discuss the implications for patient care and try to identify a common basis for methodological considerations for future research and clinical practice.
Collapse
Affiliation(s)
- Anna B. Szabo
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- *Correspondence: Anna B. Szabo
| | - Benjamin Cretin
- Clinical Neuropsychology Unit, Neurology Department, CM2R (Memory Resource and Research Centre), University Hospital of Strasbourg, Strasbourg, France
- CNRS, ICube Laboratory, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), Team IMIS, University of Strasbourg, Strasbourg, France
- CMRR d'Alsace, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Pôle Tête et Cou, Strasbourg, France
| | - Fleur Gérard
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jonathan Curot
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Emmanuel J. Barbeau
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
| | - Jérémie Pariente
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Toulouse NeuroImaging Center (ToNIC), INSERM-University of Toulouse Paul Sabatier, Toulouse, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Luc Valton
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Luc Valton
| |
Collapse
|
19
|
Harutyunyan A, Jones NC, Kwan P, Anderson A. Network Preservation Analysis Reveals Dysregulated Synaptic Modules and Regulatory Hubs Shared Between Alzheimer’s Disease and Temporal Lobe Epilepsy. Front Genet 2022; 13:821343. [PMID: 35309145 PMCID: PMC8926077 DOI: 10.3389/fgene.2022.821343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/20/2022] [Indexed: 01/08/2023] Open
Abstract
Background: There is increased prevalence of epilepsy in patients with Alzheimer’s disease (AD). Although shared pathological and clinical features have been identified, the underlying pathophysiology and cause-effect relationships are poorly understood. We aimed to identify commonly dysregulated groups of genes between these two disorders. Methods: Using publicly available transcriptomic data from hippocampal tissue of patients with temporal lobe epilepsy (TLE), late onset AD and non-AD controls, we constructed gene coexpression networks representing all three states. We then employed network preservation statistics to compare the density and connectivity-based preservation of functional gene modules between TLE, AD and controls and used the difference in significance scores as a surrogate quantifier of module preservation. Results: The majority (>90%) of functional gene modules were highly preserved between all coexpression networks, however several modules identified in the TLE network showed various degrees of preservation in the AD network compared to that of control. Of note, two synaptic signalling-associated modules and two metabolic modules showed substantial gain of preservation, while myelination and immune system-associated modules showed significant loss of preservation. The genes SCN3B and EPHA4 were identified as central regulatory hubs of the highly preserved synaptic signalling-associated module. GABRB3 and SCN2A were identified as central regulatory hubs of a smaller neurogenesis-associated module, which was enriched for multiple epileptic activity and seizure-related human phenotype ontologies. Conclusion: We conclude that these hubs and their downstream signalling pathways are common modulators of synaptic activity in the setting of AD and TLE, and may play a critical role in epileptogenesis in AD.
Collapse
Affiliation(s)
- Anna Harutyunyan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Nigel C. Jones
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Patrick Kwan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Alison Anderson
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- *Correspondence: Alison Anderson,
| |
Collapse
|
20
|
Cope ZA, Murai T, Sukoff Rizzo SJ. Emerging Electroencephalographic Biomarkers to Improve Preclinical to Clinical Translation in Alzheimer's Disease. Front Aging Neurosci 2022; 14:805063. [PMID: 35250541 PMCID: PMC8891809 DOI: 10.3389/fnagi.2022.805063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
Abstract
Continually emerging data indicate that sub-clinical, non-convulsive epileptiform activity is not only prevalent in Alzheimer's disease (AD) but is detectable early in the course of the disease and predicts cognitive decline in both humans and animal models. Epileptiform activity and other electroencephalographic (EEG) measures may hold powerful, untapped potential to improve the translational validity of AD-related biomarkers in model animals ranging from mice, to rats, and non-human primates. In this review, we will focus on studies of epileptiform activity, EEG slowing, and theta-gamma coupling in preclinical models, with particular focus on its role in cognitive decline and relevance to AD. Here, each biomarker is described in the context of the contemporary literature and recent findings in AD relevant animal models are discussed.
Collapse
Affiliation(s)
| | | | - Stacey J. Sukoff Rizzo
- Aging Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
21
|
Yu T, Liu X, Wu J, Wang Q. Electrophysiological Biomarkers of Epileptogenicity in Alzheimer's Disease. Front Hum Neurosci 2021; 15:747077. [PMID: 34916917 PMCID: PMC8669481 DOI: 10.3389/fnhum.2021.747077] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Cortical network hyperexcitability is an inextricable feature of Alzheimer’s disease (AD) that also might accelerate its progression. Seizures are reported in 10–22% of patients with AD, and subclinical epileptiform abnormalities have been identified in 21–42% of patients with AD without seizures. Accurate identification of hyperexcitability and appropriate intervention to slow the compromise of cognitive functions of AD might open up a new approach to treatment. Based on the results of several studies, epileptiform discharges, especially those with specific features (including high frequency, robust morphology, right temporal location, and occurrence during awake or rapid eye movement states), frequent small sharp spikes (SSSs), temporal intermittent rhythmic delta activities (TIRDAs), and paroxysmal slow wave events (PSWEs) recorded in long-term scalp electroencephalogram (EEG) provide sufficient sensitivity and specificity in detecting cortical network hyperexcitability and epileptogenicity of AD. In addition, magnetoencephalogram (MEG), foramen ovale (FO) electrodes, and computational approaches help to find subclinical seizures that are invisible on scalp EEGs. We performed a comprehensive analysis of the aforementioned electrophysiological biomarkers of AD-related seizures.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiao Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jianping Wu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Foliaki ST, Schwarz B, Groveman BR, Walters RO, Ferreira NC, Orrù CD, Smith A, Wood A, Schmit OM, Freitag P, Yuan J, Zou W, Bosio CM, Carroll JA, Haigh CL. Neuronal excitatory-to-inhibitory balance is altered in cerebral organoid models of genetic neurological diseases. Mol Brain 2021; 14:156. [PMID: 34635127 PMCID: PMC8507222 DOI: 10.1186/s13041-021-00864-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/29/2021] [Indexed: 12/31/2022] Open
Abstract
The neuro-physiological properties of individuals with genetic pre-disposition to neurological disorders are largely unknown. Here we aimed to explore these properties using cerebral organoids (COs) derived from fibroblasts of individuals with confirmed genetic mutations including PRNPE200K, trisomy 21 (T21), and LRRK2G2019S, which are associated with Creutzfeldt Jakob disease, Down Syndrome, and Parkinson's disease. We utilized no known disease/healthy COs (HC) as normal function controls. At 3-4 and 6-10 months post-differentiation, COs with mutations showed no evidence of disease-related pathology. Electrophysiology assessment showed that all COs exhibited mature neuronal firing at 6-10 months old. At this age, we observed significant changes in the electrophysiology of the COs with disease-associated mutations (dCOs) as compared with the HC, including reduced neuronal network communication, slowing neuronal oscillations, and increased coupling of delta and theta phases to the amplitudes of gamma oscillations. Such changes were linked with the detection of hypersynchronous events like spike-and-wave discharges. These dysfunctions were associated with altered production and release of neurotransmitters, compromised activity of excitatory ionotropic receptors including receptors of kainate, AMPA, and NMDA, and changed levels and function of excitatory glutamatergic synapses and inhibitory GABAergic synapses. Neuronal properties that modulate GABAergic inhibition including the activity of Na-K-Cl cotransport 1 (NKCC1) in Cl- homeostasis and the levels of synaptic and extra-synaptic localization of GABA receptors (GABARs) were altered in the T21 COs only. The neurosteroid allopregnanolone, a positive modulator of GABARs, was downregulated in all the dCOs. Treatment with this neurosteroid significantly improved the neuronal communication in the dCOs, possibly through improving the GABAergic inhibition. Overall, without the manifestation of any disease-related pathology, the genetic mutations PRNPE200K, T21, and LRRK2G2019S significantly altered the neuronal network communication in dCOs by disrupting the excitatory-to-inhibitory balance.
Collapse
Affiliation(s)
- Simote T Foliaki
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Benjamin Schwarz
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Bradley R Groveman
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Ryan O Walters
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Natalia C Ferreira
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Christina D Orrù
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Anna Smith
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Aleksandar Wood
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Olivia M Schmit
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Phoebe Freitag
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Jue Yuan
- Departments of Pathology and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Wenquan Zou
- Departments of Pathology and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Catharine M Bosio
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - James A Carroll
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA.
| |
Collapse
|
23
|
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: 2.7] [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
|
24
|
Romoli M, Sen A, Parnetti L, Calabresi P, Costa C. Amyloid-β: a potential link between epilepsy and cognitive decline. Nat Rev Neurol 2021; 17:469-485. [PMID: 34117482 DOI: 10.1038/s41582-021-00505-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 02/05/2023]
Abstract
People with epilepsy - in particular, late-onset epilepsy of unknown aetiology - have an elevated risk of dementia, and seizures have been detected in the early stages of Alzheimer disease (AD), supporting the concept of an epileptic AD prodrome. However, the relationship between epilepsy and cognitive decline remains controversial, with substantial uncertainties about whether epilepsy drives cognitive decline or vice versa, and whether shared pathways underlie both conditions. Here, we review evidence that amyloid-β (Aβ) forms part of a shared pathway between epilepsy and cognitive decline, particularly in the context of AD. People with epilepsy show an increased burden of Aβ pathology in the brain, and Aβ-mediated epileptogenic alterations have been demonstrated in experimental studies, with evidence suggesting that Aβ pathology might already be pro-epileptogenic at the soluble stage, long before plaque deposition. We discuss the hypothesis that Aβ mediates - or is at least a major determinant of - a continuum spanning epilepsy and cognitive decline. Serial cognitive testing and assessment of Aβ levels might be worthwhile to stratify the risk of developing dementia in people with late-onset epilepsy. If seizures are a clinical harbinger of dementia, people with late-onset epilepsy could be an ideal group in which to implement preventive or therapeutic strategies to slow cognitive decline.
Collapse
Affiliation(s)
- Michele Romoli
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy.,Oxford Epilepsy Research Group, NIHR Biomedical Research Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK.,Neurology and Stroke Unit, "Maurizio Bufalini" Hospital, Cesena, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Neurologia e Rete Stroke Metropolitana, Ospedale Maggiore, Bologna, Italy
| | - Arjune Sen
- Oxford Epilepsy Research Group, NIHR Biomedical Research Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Lucilla Parnetti
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Calabresi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli, IRCCS, UOC Neurologia, Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Rome, Italy
| | - Cinzia Costa
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy.
| |
Collapse
|
25
|
Altered corticostriatal synchronization associated with compulsive-like behavior in APP/PS1 mice. Exp Neurol 2021; 344:113805. [PMID: 34242631 DOI: 10.1016/j.expneurol.2021.113805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/17/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Mild behavioral impairment (MBI), which can include compulsive behavior, is an early sign of Alzheimer's disease (AD), but its underlying neural mechanisms remain unclear. Here, we show that 3-5-month-old APP/PS1 mice display obsessive-compulsive disorder (OCD)-like behavior. The number of parvalbumin-positive (PV) interneurons and level of high gamma (γhigh) oscillation are significantly decreased in the striatum of AD mice. This is accompanied by enhanced β-γhigh coupling and firing rates of putative striatal projection neurons (SPNs), indicating decorrelation between PV interneurons and SPNs. Local field potentials (LFPs) simultaneously recorded in prefrontal cortex (PFC) and striatum (Str) demonstrate a decrease in γhigh-band coherent activity and spike-field coherence in corticostriatal circuits of APP/PS1 mice. Furthermore, levels of GABAB receptor (GABABR), but not GABAA receptor (GABAAR), and glutamatergic receptors, were markedly reduced, in line with presymptomatic AD-related behavioral changes. These findings suggest that MBI occurs as early as 3-5 months in APP/PS1 mice and that altered corticostriatal synchronization may play a role in mediating the behavioral phenotypes observed.
Collapse
|
26
|
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
|
27
|
Jin N, Babiloni C, Drinkenburg WH, Hajós M, Nygaard HB, Tanila H. Recommendations for Preclinical Testing of Treatments Against Alzheimer's Disease-Related Epileptiform Spikes in Transgenic Rodent Models. J Alzheimers Dis 2021; 88:849-865. [PMID: 34092642 DOI: 10.3233/jad-210209] [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] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that about 30%of patients with mild to moderate Alzheimer's disease (AD) without a known diagnosis of epilepsy may display epileptiform spikes during electroencephalographic (EEG) recordings. These abnormal discharges occur predominantly during sleep and may be associated with accelerated disease progression. Subclinical spikes may represent a relevant target for clinical drug interventions, and there is a clear unmet need for preclinical testing of novel disease modifying agents in suitable animal models. Transgenic rodent models of AD pathology exhibit various forms of epileptiform EEG activity related to the abnormal levels of amyloid species in the brain. Among them, large-amplitude cortical and hippocampal EEG spikes in mouse and rat AD models may be reminiscent of the subclinical epileptiform EEG spikes recorded in some AD patients. This article reports the recommendations of a multidisciplinary panel of experts on optimal EEG markers and experimental designs to measure and report epileptiform activities and their response to symptomatic and disease-modifying drugs in transgenic AD model rodents. These recommendations may harmonize future preclinical EEG studies in the drug discovery research and may increase the comparability of experimental outcomes and their translational clinical value.
Collapse
Affiliation(s)
- Nanxiang Jin
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy.,Hospital San Raffaele Cassino, Cassino (FR), Italy
| | - Wilhelmus H Drinkenburg
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.,Groningen Institute for Evolutionary Life Sciences, University of Groningen, The Netherlands
| | - Mihály Hajós
- Cognito Therapeutics, Cambridge, MA, USA.,Yale University School of Medicine, New Haven, CT, USA
| | - Haakon B Nygaard
- Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
28
|
Neuronal Network Excitability in Alzheimer's Disease: The Puzzle of Similar versus Divergent Roles of Amyloid β and Tau. eNeuro 2021; 8:ENEURO.0418-20.2020. [PMID: 33741601 PMCID: PMC8174042 DOI: 10.1523/eneuro.0418-20.2020] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most frequent neurodegenerative disorder that commonly causes dementia in the elderly. Recent evidence indicates that network abnormalities, including hypersynchrony, altered oscillatory rhythmic activity, interneuron dysfunction, and synaptic depression, may be key mediators of cognitive decline in AD. In this review, we discuss characteristics of neuronal network excitability in AD, and the role of Aβ and tau in the induction of network hyperexcitability. Many patients harboring genetic mutations that lead to increased Aβ production suffer from seizures and epilepsy before the development of plaques. Similarly, pathologic accumulation of hyperphosphorylated tau has been associated with hyperexcitability in the hippocampus. We present common and divergent roles of tau and Aβ on neuronal hyperexcitability in AD, and hypotheses that could serve as a template for future experiments.
Collapse
|
29
|
Kelliny S, Lin L, Deng I, Xiong J, Zhou F, Al-Hawwas M, Bobrovskaya L, Zhou XF. A New Approach to Model Sporadic Alzheimer's Disease by Intracerebroventricular Streptozotocin Injection in APP/PS1 Mice. Mol Neurobiol 2021; 58:3692-3711. [PMID: 33797693 DOI: 10.1007/s12035-021-02338-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/22/2021] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia among elderly people. Majority of AD cases are sporadic (SAD) with unknown cause. Transgenic animal models closely reflect the familial (genetic) aspect of the disease but not the sporadic type. However, most new drug candidates which are tested positive in transgenic animal models failed in clinical studies so far. Herein, we aim to develop an AD animal model that combines most of the neuropathological features seen in sporadic AD in humans with amyloid plaques observed in transgenic mice. Four-month-old wild-type and APP/PS1 AD mice were given a single intracerebroventricular (ICV) injection of 3 mg/kg streptozotocin (STZ), a diabetogenic agent. Three weeks later, their cognitive behavior was assessed, and their brain tissues were collected for biochemical and histological analysis. STZ produced cognitive deficits in both non-transgenic mice and AD mice. Biochemical analysis showed a severe decline in synaptic proteins, increase in tau phosphorylation, oxidative stress, disturbed brain insulin signaling with extensive neuroinflammation, and cell death. Significant increase was also observed in the level of the soluble beta amyloid precursor protein (APP) fragments and robust accumulation of amyloid plaques in AD mice compared to the control. These results suggest that STZ ICV treatment causes disturbance in multiple metabolic and cell signaling pathways in the brain that facilitated amyloid plaque accumulation and tau phosphorylation. Therefore, this animal model can be used to evaluate new AD therapeutic agents for clinical translation.
Collapse
Affiliation(s)
- Sally Kelliny
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
- Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Liying Lin
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Isaac Deng
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Jing Xiong
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
- Department of Neurology, The Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan Province, China
| | - Fiona Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Mohammed Al-Hawwas
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
| |
Collapse
|
30
|
Liang CS, Bai YM, Hsu JW, Huang KL, Ko NY, Tsai CK, Yeh TC, Chu HT, Tsai SJ, Chen TJ, Chen MH. The Risk of Epilepsy after Long-term Proton Pump Inhibitor Therapy. Seizure 2021; 87:88-93. [PMID: 33735722 DOI: 10.1016/j.seizure.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Prescription-event monitoring studies have reported incident epilepsy or seizures in proton pump inhibitor (PPI) recipients. We examined the risk of epilepsy after prolonged PPI exposure and determine what age group was at higher risk of epilepsy. METHODS We performed a case-control study nested within a sample of Taiwan National Health Insurance beneficiaries (n = 1,000,000). PPI users with subsequent epilepsy were selected as the case cohort. Controls were PPI users without subsequent epilepsy, matched for age, sex, PPI use indication, enrollment time, end point time, follow-up period, overall systemic health, and comorbidities. The total dose of PPI was defined as the cumulative defined daily dose (cDDD). Prolonged PPI use was defined as a cDDD > 365. A logistic regression analysis was performed. Population attributable risk was calculated. RESULTS Epilepsy occurred 4.13 years after the initiation of PPI use. PPI users with the highest risk of incident epilepsy received a cDDD > 365 [odds ratio = 1.63, 95% confidence interval = 1.37-1.95], followed by 121-365 cDDD (1.33, 1.18-1.51) and 31-120 cDDD (1.15, 1.02-1.29), compared to those receiving a cDDD ≤ 30, after adjusting for potential confounders. Prolonged PPI use increased the risk of epilepsy in all age groups, and the risk was highest for those older than 80 years (3.11, 1.67-5.79). The population attributable risk was 12.2% (> 365 cDDD vs ≤ 30 cDDD). DISCUSSION Prolonged PPI therapy was associated with an increased risk of epilepsy, particularly in the elderly population.
Collapse
Affiliation(s)
- Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ju-Wei Hsu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Kai-Lin Huang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Nai-Ying Ko
- Department of Nursing, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ta-Chuan Yeh
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsuan-Te Chu
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzeng-Ji Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
31
|
Alcantara-Gonzalez D, Chartampila E, Criscuolo C, Scharfman HE. Early changes in synaptic and intrinsic properties of dentate gyrus granule cells in a mouse model of Alzheimer's disease neuropathology and atypical effects of the cholinergic antagonist atropine. Neurobiol Dis 2021; 152:105274. [PMID: 33484828 DOI: 10.1016/j.nbd.2021.105274] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
It has been reported that hyperexcitability occurs in a subset of patients with Alzheimer's disease (AD) and hyperexcitability could contribute to the disease. Several studies have suggested that the hippocampal dentate gyrus (DG) may be an important area where hyperexcitability occurs. Therefore, we tested the hypothesis that the principal DG cell type, granule cells (GCs), would exhibit changes at the single-cell level which would be consistent with hyperexcitability and might help explain it. We used the Tg2576 mouse, where it has been shown that hyperexcitability is robust at 2-3 months of age. GCs from 2 to 3-month-old Tg2576 mice were compared to age-matched wild type (WT) mice. Effects of muscarinic cholinergic antagonism were tested because previously we found that Tg2576 mice exhibited hyperexcitability in vivo that was reduced by the muscarinic cholinergic antagonist atropine, counter to the dogma that in AD one needs to boost cholinergic function. The results showed that GCs from Tg2576 mice exhibited increased frequency of spontaneous excitatory postsynaptic potentials/currents (sEPSP/Cs) and reduced frequency of spontaneous inhibitory synaptic events (sIPSCs) relative to WT, increasing the excitation:inhibition (E:I) ratio. There was an inward NMDA receptor-dependent current that we defined here as a novel synaptic current (nsC) in Tg2576 mice because it was very weak in WT mice. Intrinsic properties were distinct in Tg2576 GCs relative to WT. In summary, GCs of the Tg2576 mouse exhibit early electrophysiological alterations that are consistent with increased synaptic excitation, reduced inhibition, and muscarinic cholinergic dysregulation. The data support previous suggestions that the DG contributes to hyperexcitability and there is cholinergic dysfunction early in life in AD mouse models.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Elissavet Chartampila
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Chiara Criscuolo
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Helen E Scharfman
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY 10016, USA; Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
32
|
Jura B, Młoźniak D, Goszczyńska H, Blinowska K, Biendon N, Macrez N, Meyrand P, Bem T. Reconfiguration of the cortical-hippocampal interaction may compensate for Sharp-Wave Ripple deficits in APP/PS1 mice and support spatial memory formation. PLoS One 2020; 15:e0243767. [PMID: 33382724 PMCID: PMC7774978 DOI: 10.1371/journal.pone.0243767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/25/2020] [Indexed: 12/28/2022] Open
Abstract
Hippocampal-cortical dialogue, during which hippocampal ripple oscillations support information transfer, is necessary for long-term consolidation of spatial memories. Whereas a vast amount of work has been carried out to understand the cellular and molecular mechanisms involved in the impairments of memory formation in Alzheimer's disease (AD), far less work has been accomplished to understand these memory deficiencies at the network-level interaction that may underlie memory processing. We recently demonstrated that freely moving 8 to 9-month-old APP/PS1 mice, a model of AD, are able to learn a spatial reference memory task despite a major deficit in Sharp-Wave Ripples (SWRs), the integrity of which is considered to be crucial for spatial memory formation. In order to test whether reconfiguration of hippocampal-cortical dialogue could be responsible for the maintenance of this ability for memory formation, we undertook a study to identify causal relations between hippocampal and cortical circuits in epochs when SWRs are generated in hippocampus. We analyzed the data set obtained from multielectrode intracranial recording of transgenic and wild-type mice undergoing consolidation of spatial memory reported in our previous study. We applied Directed Transfer Function, a connectivity measure based on Granger causality, in order to determine effective coupling between distributed circuits which express oscillatory activity in multiple frequency bands. Our results showed that hippocampal-cortical coupling in epochs containing SWRs was expressed in the two frequency ranges corresponding to ripple (130-180 Hz) and slow gamma (20-60 Hz) band. The general features of connectivity patterns were similar in the 8 to 9-month-old APP/PS1 and wild-type animals except that the coupling in the slow gamma range was stronger and spread to more cortical sites in APP/PS1 mice than in the wild-type group. During the occurrence of SWRs, the strength of effective coupling from the cortex to hippocampus (CA1) in the ripple band undergoes sharp increase, involving cortical areas that were different in the two groups of animals. In the wild-type group, retrosplenial cortex and posterior cingulate cortex interacted with the hippocampus most strongly, whereas in the APP/PS1 group more anterior structures interacted with the hippocampus, that is, anterior cingulate cortex and prefrontal cortex. This reconfiguration of cortical-hippocampal interaction pattern may be an adaptive mechanism responsible for supporting spatial memory consolidation in AD mice model.
Collapse
Affiliation(s)
- Bartosz Jura
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Dariusz Młoźniak
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Hanna Goszczyńska
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Blinowska
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
- Department of Biomedical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Nathalie Biendon
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Nathalie Macrez
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pierre Meyrand
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- INSERM, Neurocentre Magendie, Bordeaux, France
| | - Tiaza Bem
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
- * E-mail:
| |
Collapse
|
33
|
Sharp PS, Ameen-Ali KE, Boorman L, Harris S, Wharton S, Howarth C, Shabir O, Redgrave P, Berwick J. Neurovascular coupling preserved in a chronic mouse model of Alzheimer's disease: Methodology is critical. J Cereb Blood Flow Metab 2020; 40:2289-2303. [PMID: 31760864 PMCID: PMC7585931 DOI: 10.1177/0271678x19890830] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Impaired neurovascular coupling has been suggested as an early pathogenic factor in Alzheimer's disease (AD), which could serve as an early biomarker of cerebral pathology. We have established an anaesthetic regime to allow repeated measurements of neurovascular function over three months in the J20 mouse model of AD (J20-AD) and wild-type (WT) controls. Animals were 9-12 months old at the start of the experiment. Mice were chronically prepared with a cranial window through which 2-Dimensional optical imaging spectroscopy (2D-OIS) was used to generate functional maps of the cerebral blood volume and saturation changes evoked by whisker stimulation and vascular reactivity challenges. Unexpectedly, the hemodynamic responses were largely preserved in the J20-AD group. This result failed to confirm previous investigations using the J20-AD model. However, a final acute electrophysiology and 2D-OIS experiment was performed to measure both neural and hemodynamic responses concurrently. In this experiment, previously reported deficits in neurovascular coupling in the J20-AD model were observed. This suggests that J20-AD mice may be more susceptible to the physiologically stressing conditions of an acute experimental procedure compared to WT animals. These results therefore highlight the importance of experimental procedure when determining the characteristics of animal models of human disease.
Collapse
Affiliation(s)
- Paul S Sharp
- Nanomedicine Lab, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Department of Psychology, University of Sheffield, Sheffield, UK
| | - Kamar E Ameen-Ali
- Department of Psychology, University of Sheffield, Sheffield, UK.,Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Luke Boorman
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Sam Harris
- Department of Psychology, University of Sheffield, Sheffield, UK.,UK Dementia Research Institute, UCL Institute of Neurology, University College London, London, UK
| | - Stephen Wharton
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Clare Howarth
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Osman Shabir
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Peter Redgrave
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Jason Berwick
- Department of Psychology, University of Sheffield, Sheffield, UK
| |
Collapse
|
34
|
Shabir O, Sharp P, Rebollar MA, Boorman L, Howarth C, Wharton SB, Francis SE, Berwick J. Enhanced Cerebral Blood Volume under Normobaric Hyperoxia in the J20-hAPP Mouse Model of Alzheimer's Disease. Sci Rep 2020; 10:7518. [PMID: 32371859 PMCID: PMC7200762 DOI: 10.1038/s41598-020-64334-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/14/2020] [Indexed: 11/14/2022] Open
Abstract
Early impairments to neurovascular coupling have been proposed to be a key pathogenic factor in the onset and progression of Alzheimer's disease (AD). Studies have shown impaired neurovascular function in several mouse models of AD, including the J20-hAPP mouse. In this study, we aimed to investigate early neurovascular changes using wild-type (WT) controls and J20-hAPP mice at 6 months of age, by measuring cerebral haemodynamics and neural activity to physiological sensory stimulations. A thinned cranial window was prepared to allow access to cortical vasculature and imaged using 2D-optical imaging spectroscopy (2D-OIS). After chronic imaging sessions where the skull was intact, a terminal acute imaging session was performed where an electrode was inserted into the brain to record simultaneous neural activity. We found that cerebral haemodynamic changes were significantly enhanced in J20-hAPP mice compared with controls in response to physiological stimulations, potentially due to the significantly higher neural activity (hyperexcitability) seen in the J20-hAPP mice. Thus, neurovascular coupling remained preserved under a chronic imaging preparation. Further, under hyperoxia, the baseline blood volume and saturation of all vascular compartments in the brains of J20-hAPP mice were substantially enhanced compared to WT controls, but this effect disappeared under normoxic conditions. This study highlights novel findings not previously seen in the J20-hAPP mouse model, and may point towards a potential therapeutic strategy.
Collapse
Affiliation(s)
- Osman Shabir
- The Neurovascular & Neuroimaging Group (Department of Psychology), Alfred Denny Building, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Paul Sharp
- The Neurovascular & Neuroimaging Group (Department of Psychology), Alfred Denny Building, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Monica A Rebollar
- Sheffield Institute for Translational Neuroscience (SITraN), 385a Glossop Road, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Luke Boorman
- The Neurovascular & Neuroimaging Group (Department of Psychology), Alfred Denny Building, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Clare Howarth
- The Neurovascular & Neuroimaging Group (Department of Psychology), Alfred Denny Building, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience (SITraN), 385a Glossop Road, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Sheila E Francis
- Department of Infection, Immunity & Cardiovascular Disease (IICD), University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Jason Berwick
- The Neurovascular & Neuroimaging Group (Department of Psychology), Alfred Denny Building, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
35
|
Vyas Y, Montgomery JM, Cheyne JE. Hippocampal Deficits in Amyloid-β-Related Rodent Models of Alzheimer's Disease. Front Neurosci 2020; 14:266. [PMID: 32317913 PMCID: PMC7154147 DOI: 10.3389/fnins.2020.00266] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/09/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is the most common cause of dementia. Symptoms of AD include memory loss, disorientation, mood and behavior changes, confusion, unfounded suspicions, and eventually, difficulty speaking, swallowing, and walking. These symptoms are caused by neuronal degeneration and cell loss that begins in the hippocampus, and later in disease progression spreading to the rest of the brain. While there are some medications that alleviate initial symptoms, there are currently no treatments that stop disease progression. Hippocampal deficits in amyloid-β-related rodent models of AD have revealed synaptic, behavioral and circuit-level defects. These changes in synaptic function, plasticity, neuronal excitability, brain connectivity, and excitation/inhibition imbalance all have profound effects on circuit function, which in turn could exacerbate disease progression. Despite, the wealth of studies on AD pathology we don't yet have a complete understanding of hippocampal deficits in AD. With the increasing development of in vivo recording techniques in awake and freely moving animals, future studies will extend our current knowledge of the mechanisms underpinning how hippocampal function is altered in AD, and aid in progression of treatment strategies that prevent and/or delay AD symptoms.
Collapse
Affiliation(s)
| | - Johanna M. Montgomery
- Department of Physiology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Juliette E. Cheyne
- Department of Physiology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
36
|
Peters ST, Fahrenkopf A, Choquette JM, Vermilyea SC, Lee MK, Vossel K. Ablating Tau Reduces Hyperexcitability and Moderates Electroencephalographic Slowing in Transgenic Mice Expressing A53T Human α-Synuclein. Front Neurol 2020; 11:563. [PMID: 32636798 PMCID: PMC7316964 DOI: 10.3389/fneur.2020.00563] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Abnormal intraneuronal accumulation of the presynaptic protein α-synuclein (α-syn) is implicated in the etiology of dementia with Lewy bodies (DLB) and Parkinson's disease with dementia (PDD). Recent work revealed that mice expressing human α-syn with the alanine-53-threonine (A53T) mutation have a similar phenotype to the human condition, exhibiting long-term potentiation deficits, learning and memory deficits, and inhibitory hippocampal remodeling, all of which were reversed by genetic ablation of microtubule-associated protein tau. Significantly, memory deficits were associated with histological signs of network hyperactivity/seizures. Electrophysiological abnormalities are often seen in parkinsonian dementias. Baseline electroencephalogram (EEG) slowing is used as a supportive diagnostic feature in DLB and PDD, and patients with these diseases may exhibit indicators of broad network dysfunction such as sleep dysregulation, myoclonus, and seizures. Given the translational significance, we examined whether human A53T α-syn expressing mice exhibit endogenous-tau-dependent EEG abnormalities, as measured with epidural electrodes over the frontal and parietal cortices. Using template-based waveform sorting, we determined that A53T mice have significantly high numbers of epileptiform events as early as 3-4 months of age and throughout life, and this effect is markedly attenuated in the absence of tau. Epileptic myoclonus occurred in half of A53T mice and was markedly reduced by tau ablation. In spectral analysis, tau ablation partially reduced EEG slowing in 6-7 month transgenic mice. We found abnormal sleeping patterns in transgenic mice that were more pronounced in older groups, but did not find evidence that this was influenced by tau genotype. Together, these data support the notion that tau facilitates A53T α-syn-induced hyperexcitability that both precedes and coincides with associated synaptic, cognitive, and behavioral effects. Tau also contributes to some aspects of EEG slowing in A53T mice. Importantly, our work supports tau-based approaches as an effective early intervention in α-synucleinopathies to treat aberrant network activity.
Collapse
Affiliation(s)
- Samuel T Peters
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, United States
| | - Allyssa Fahrenkopf
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, United States
| | - Jessica M Choquette
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, United States
| | - Scott C Vermilyea
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, University of Minnesota, Minneapolis, MN, United States
| | - Keith Vossel
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, United States.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
37
|
SIRT3 Haploinsufficiency Aggravates Loss of GABAergic Interneurons and Neuronal Network Hyperexcitability in an Alzheimer's Disease Model. J Neurosci 2019; 40:694-709. [PMID: 31818974 DOI: 10.1523/jneurosci.1446-19.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/26/2019] [Accepted: 11/05/2019] [Indexed: 01/17/2023] Open
Abstract
Impaired mitochondrial function and aberrant neuronal network activity are believed to be early events in the pathogenesis of Alzheimer's disease (AD), but how mitochondrial alterations contribute to aberrant activity in neuronal circuits is unknown. In this study, we examined the function of mitochondrial protein deacetylase sirtuin 3 (SIRT3) in the pathogenesis of AD. Compared with AppPs1 mice, Sirt3-haploinsufficient AppPs1 mice (Sirt3+/-AppPs1) exhibit early epileptiform EEG activity and seizure. Both male and female Sirt3+/-AppPs1 mice were observed to die prematurely before 5 months of age. When comparing male mice among different genotypes, Sirt3 haploinsufficiency renders GABAergic interneurons in the cerebral cortex vulnerable to degeneration and associated neuronal network hyperexcitability. Feeding Sirt3+/-AppPs1 AD mice with a ketone ester-rich diet increases SIRT3 expression and prevents seizure-related death and the degeneration of GABAergic neurons, indicating that the aggravated GABAergic neuron loss and neuronal network hyperexcitability in Sirt3+/-AppPs1 mice are caused by SIRT3 reduction and can be rescued by increase of SIRT3 expression. Consistent with a protective role in AD, SIRT3 levels are reduced in association with cerebral cortical Aβ pathology in AD patients. In summary, SIRT3 preserves GABAergic interneurons and protects cerebral circuits against hyperexcitability, and this neuroprotective mechanism can be bolstered by dietary ketone esters.SIGNIFICANCE STATEMENT GABAergic neurons provide the main inhibitory control of neuronal activity in the brain. By preserving mitochondrial function, SIRT3 protects parvalbumin and calretinin interneurons against Aβ-associated dysfunction and degeneration in AppPs1 Alzheimer's disease mice, thus restraining neuronal network hyperactivity. The neuronal network dysfunction that occurs in Alzheimer's disease can be partially reversed by physiological, dietary, and pharmacological interventions to increase SIRT3 expression and enhance the functionality of GABAergic interneurons.
Collapse
|
38
|
von Rüden EL, Zellinger C, Gedon J, Walker A, Bierling V, Deeg CA, Hauck SM, Potschka H. Regulation of Alzheimer's disease-associated proteins during epileptogenesis. Neuroscience 2019; 424:102-120. [PMID: 31705965 DOI: 10.1016/j.neuroscience.2019.08.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 07/26/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022]
Abstract
Clinical evidence and pathological studies suggest a bidirectional link between temporal lobe epilepsy and Alzheimer's disease (AD). Data analysis from omic studies offers an excellent opportunity to identify the overlap in molecular alterations between the two pathologies. We have subjected proteomic data sets from a rat model of epileptogenesis to a bioinformatics analysis focused on proteins functionally linked with AD. The data sets have been obtained for hippocampus (HC) and parahippocampal cortex samples collected during the course of epileptogenesis. Our study confirmed a relevant dysregulation of proteins linked with Alzheimer pathogenesis. When comparing the two brain areas, a more prominent regulation was evident in parahippocampal cortex samples as compared to the HC. Dysregulated protein groups comprised those affecting mitochondrial function and calcium homeostasis. Differentially expressed mitochondrial proteins included proteins of the mitochondrial complexes I, III, IV, and V as well as of the accessory subunit of complex I. The analysis also revealed a regulation of the microtubule associated protein Tau in parahippocampal cortex tissue during the latency phase. This was further confirmed by immunohistochemistry. Moreover, we demonstrated a complex epileptogenesis-associated dysregulation of proteins involved in amyloid β processing and its regulation. Among others, the amyloid precursor protein and the α-secretase alpha disintegrin metalloproteinase 17 were included. Our analysis revealed a relevant regulation of key proteins known to be associated with AD pathogenesis. The analysis provides a comprehensive overview of shared molecular alterations characterizing epilepsy development and manifestation as well as AD development and progression.
Collapse
Affiliation(s)
- Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Christina Zellinger
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Julia Gedon
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Andreas Walker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Vera Bierling
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Cornelia A Deeg
- Institute of Animal Physiology, Department of Veterinary Sciences, Ludwig-Maximilians-University (LMU), Munich, Germany; Experimental Ophthalmology, Philipps University of Marburg, Marburg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, Neuherberg, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany.
| |
Collapse
|
39
|
Zelano J, Brigo F, Garcia‐Patek S. Increased risk of epilepsy in patients registered in the Swedish Dementia Registry. Eur J Neurol 2019; 27:129-135. [DOI: 10.1111/ene.14043] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/15/2019] [Indexed: 01/03/2023]
Affiliation(s)
- J. Zelano
- Department of Clinical Neuroscience Institute of Neuroscience and Physiology Sahlgrenska Academy Gothenburg University GothenburgSweden
- Department of Neurology Sahlgrenska University Hospital Gothenburg Sweden
| | - F. Brigo
- Department of Neuroscience, Biomedicine and Movement Science University of Verona VeronaItaly
- Division of Neurology ‘Franz Tappeiner’ Hospital Merano Italy
| | - S. Garcia‐Patek
- Department of Neurobiology, Care Sciences and Society Division of Clinical Geriatrics Karolinska Institutet StockholmSweden
- Internal Medicine Section for Neurology Södersjukhuset Stockholm Sweden
| |
Collapse
|
40
|
Vico Varela E, Etter G, Williams S. Excitatory-inhibitory imbalance in Alzheimer's disease and therapeutic significance. Neurobiol Dis 2019; 127:605-615. [DOI: 10.1016/j.nbd.2019.04.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 11/29/2022] Open
|
41
|
Zegarra-Valdivia JA, Santi A, Fernández de Sevilla ME, Nuñez A, Torres Aleman I. Serum Insulin-Like Growth Factor I Deficiency Associates to Alzheimer’s Disease Co-Morbidities. J Alzheimers Dis 2019; 69:979-987. [DOI: 10.3233/jad-190241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Andrea Santi
- Cajal Institute (CSIC), Madrid, Spain
- CIBERNED, Spain
| | | | - Angel Nuñez
- Department of Neurosciences, School of Medicine, UAM, Madrid, Spain
| | | |
Collapse
|
42
|
Zhou X, Tao H, Cai Y, Cui L, Zhao B, Li K. Stage-dependent involvement of ADAM10 and its significance in epileptic seizures. J Cell Mol Med 2019; 23:4494-4504. [PMID: 31087543 PMCID: PMC6584734 DOI: 10.1111/jcmm.14307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/05/2019] [Accepted: 03/11/2019] [Indexed: 12/22/2022] Open
Abstract
The prevalence of epileptic seizures in Alzheimer's disease (AD) has attracted an increasing amount of attention in recent years, and many cohort studies have found several risk factors associated with the genesis of seizures in AD. Among these factors, young age and severe dementia are seemingly contradictory and independent risk factors, indicating that the pathogenesis of epileptic seizures is, to a certain extent, stage‐dependent. A disintegrin and metalloproteinase domain‐containing protein 10 (ADAM10) is a crucial α‐secretase responsible for ectodomain shedding of its substrates; thus, the function of this protein depends on the biological effects of its substrates. Intriguingly, transgenic models have demonstrated ADAM10 to be associated with epilepsy. Based on the biological effects of its substrates, the potential pathogenic roles of ADAM10 in epileptic seizures can be classified into amyloidogenic processes in the ageing stage and cortical dysplasia in the developmental stage. Therefore, ADAM10 is reviewed here as a stage‐dependent modulator in the pathogenesis of epilepsy. Current data regarding ADAM10 in epileptic seizures were collected and reviewed for potential pathogenic roles (ie amyloidogenic processes and cortical dysplasia) and regulatory mechanisms (ie transcriptional and posttranscriptional regulation). These findings are then discussed in terms of the significance of the stage‐dependent functions of ADAM10 in epilepsy. Several potential targets for seizure control, such as candidate transcription factors and microRNAs that regulate ADAM10, as well as potential genetic screening tools for the early recognition of cortical dysplasia, have been suggested but must be studied in more detail.
Collapse
Affiliation(s)
- Xu Zhou
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua Tao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory of Age-related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yujie Cai
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bin Zhao
- Guangdong Key Laboratory of Age-related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Keshen Li
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Stroke Center, Neurology & Neurosurgery Division, Clinical Medicine Research Institute & the First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
43
|
Kubista H, Boehm S, Hotka M. The Paroxysmal Depolarization Shift: Reconsidering Its Role in Epilepsy, Epileptogenesis and Beyond. Int J Mol Sci 2019; 20:ijms20030577. [PMID: 30699993 PMCID: PMC6387313 DOI: 10.3390/ijms20030577] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/24/2019] [Indexed: 12/29/2022] Open
Abstract
Paroxysmal depolarization shifts (PDS) have been described by epileptologists for the first time several decades ago, but controversy still exists to date regarding their role in epilepsy. In addition to the initial view of a lack of such a role, seemingly opposing hypotheses on epileptogenic and anti-ictogenic effects of PDS have emerged. Hence, PDS may provide novel targets for epilepsy therapy. Evidence for the roles of PDS has often been obtained from investigations of the multi-unit correlate of PDS, an electrographic spike termed “interictal” because of its occurrence during seizure-free periods of epilepsy patients. Meanwhile, interictal spikes have been found to be associated with neuronal diseases other than epilepsy, e.g., Alzheimer’s disease, which may indicate a broader implication of PDS in neuropathologies. In this article, we give an introduction to PDS and review evidence that links PDS to pro- as well as anti-epileptic mechanisms, and to other types of neuronal dysfunction. The perturbation of neuronal membrane voltage and of intracellular Ca2+ that comes with PDS offers many conceivable pathomechanisms of neuronal dysfunction. Out of these, the operation of L-type voltage-gated calcium channels, which play a major role in coupling excitation to long-lasting neuronal changes, is addressed in detail.
Collapse
Affiliation(s)
- Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria.
| | - Stefan Boehm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria.
| | - Matej Hotka
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria.
| |
Collapse
|